Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 187
Filter
Add more filters

Publication year range
1.
Cell ; 175(5): 1365-1379.e25, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30445040

ABSTRACT

The exchange of metabolites between the mitochondrial matrix and the cytosol depends on ß-barrel channels in the outer membrane and α-helical carrier proteins in the inner membrane. The essential translocase of the inner membrane (TIM) chaperones escort these proteins through the intermembrane space, but the structural and mechanistic details remain elusive. We have used an integrated structural biology approach to reveal the functional principle of TIM chaperones. Multiple clamp-like binding sites hold the mitochondrial membrane proteins in a translocation-competent elongated form, thus mimicking characteristics of co-translational membrane insertion. The bound preprotein undergoes conformational dynamics within the chaperone binding clefts, pointing to a multitude of dynamic local binding events. Mutations in these binding sites cause cell death or growth defects associated with impairment of carrier and ß-barrel protein biogenesis. Our work reveals how a single mitochondrial "transfer-chaperone" system is able to guide α-helical and ß-barrel membrane proteins in a "nascent chain-like" conformation through a ribosome-free compartment.


Subject(s)
Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Molecular Chaperones/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Amino Acid Sequence , Binding Sites , Intracellular Membranes/metabolism , Mitochondrial Membrane Transport Proteins/chemistry , Mitochondrial Membrane Transport Proteins/genetics , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Protein Binding , Protein Domains , Protein Precursors/chemistry , Protein Precursors/metabolism , Protein Structure, Secondary , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Sequence Alignment
2.
J Biol Chem ; 299(8): 104954, 2023 08.
Article in English | MEDLINE | ID: mdl-37354975

ABSTRACT

Human papillomaviruses (HPVs) are DNA tumor viruses that infect mucosal and cutaneous epithelial cells of more than 20 vertebrates. High-risk HPV causes about 5% of human cancers worldwide, and the viral proteins E6 and E7 promote carcinogenesis by interacting with tumor suppressors and interfering with many cellular pathways. As a consequence, they immortalize cells more efficiently in concert than individually. So far, the networks of E6 and E7 with their respective cellular targets have been studied extensively but independently. However, we hypothesized that E6 and E7 might also interact directly with each other in a novel interaction affecting HPV-related carcinogenesis. Here, we report a direct interaction between E6 and E7 proteins from carcinogenic HPV types 16 and 31. We demonstrated this interaction via cellular assays using two orthogonal methods: coimmunoprecipitation and flow cytometry-based FRET assays. Analytical ultracentrifugation of the recombinant proteins revealed that the stoichiometry of the E6/E7 complex involves two E7 molecules and two E6 molecules. In addition, fluorescence polarization showed that (I) E6 binds to E7 with a similar affinity for HPV16 and HPV31 (in the same micromolar range) and (II) that the binding interface involves the unstructured N-terminal region of E7. The direct interaction of these highly conserved papillomaviral oncoproteins may provide a new perspective for studying HPV-associated carcinogenesis and the overall viral life cycle.


Subject(s)
Human papillomavirus 16 , Oncogene Proteins, Viral , Papillomavirus E7 Proteins , Papillomavirus Infections , Animals , Humans , Carcinogenesis , Human papillomavirus 16/genetics , Human papillomavirus 16/metabolism , Human Papillomavirus Viruses , Neoplasms , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/metabolism , Papillomavirus E7 Proteins/genetics , Papillomavirus E7 Proteins/metabolism
3.
Am J Respir Crit Care Med ; 207(1): 38-49, 2023 01 01.
Article in English | MEDLINE | ID: mdl-35926164

ABSTRACT

Rationale: Coronavirus disease 2019 (COVID-19) can lead to acute respiratory distress syndrome with fatal outcomes. Evidence suggests that dysregulated immune responses, including autoimmunity, are key pathogenic factors. Objectives: To assess whether IgA autoantibodies target lung-specific proteins and contribute to disease severity. Methods: We collected 147 blood, 9 lung tissue, and 36 BAL fluid samples from three tertiary hospitals in Switzerland and one in Germany. Severe COVID-19 was defined by the need to administer oxygen. We investigated the presence of IgA autoantibodies and their effects on pulmonary surfactant in COVID-19 using the following methods: immunofluorescence on tissue samples, immunoprecipitations followed by mass spectrometry on BAL fluid samples, enzyme-linked immunosorbent assays on blood samples, and surface tension measurements with medical surfactant. Measurements and Main Results: IgA autoantibodies targeting pulmonary surfactant proteins B and C were elevated in patients with severe COVID-19 but not in patients with influenza or bacterial pneumonia. Notably, pulmonary surfactant failed to reduce surface tension after incubation with either plasma or purified IgA from patients with severe COVID-19. Conclusions: Our data suggest that patients with severe COVID-19 harbor IgA autoantibodies against pulmonary surfactant proteins B and C and that these autoantibodies block the function of lung surfactant, potentially contributing to alveolar collapse and poor oxygenation.


Subject(s)
COVID-19 , Pulmonary Surfactants , Humans , Pulmonary Surfactants/metabolism , Bronchoalveolar Lavage Fluid/chemistry , Surface-Active Agents , Autoantibodies , Immunoglobulin A
4.
Biochem J ; 479(3): 259-272, 2022 02 11.
Article in English | MEDLINE | ID: mdl-35015082

ABSTRACT

Murine cytomegalovirus protein M45 contains a RIP homotypic interaction motif (RHIM) that is sufficient to confer protection of infected cells against necroptotic cell death. Mechanistically, the N-terminal region of M45 drives rapid self-assembly into homo-oligomeric amyloid fibrils, and interacts with the endogenous RHIM domains of receptor-interacting serine/threonine protein kinases (RIPK) 1, RIPK3, Z-DNA-binding protein 1, and Toll/interleukin-1 receptor domain-containing adaptor-inducing interferon-ß. Remarkably, all four aforementioned mammalian proteins harbouring such a RHIM domain are key components of inflammatory signalling and regulated cell death (RCD) processes. Immunogenic cell death by regulated necrosis causes extensive tissue damage in a wide range of diseases, including ischaemia reperfusion injury, myocardial infarction, sepsis, stroke, and solid organ transplantation. To harness the cell death suppression properties of M45 protein in a therapeutically usable manner, we developed a synthetic peptide encompassing only the RHIM domain of M45. To trigger delivery of RHIM into target cells, we fused the transactivator protein transduction domain of human immunodeficiency virus 1 to the N-terminus of the peptide. The fused peptide could efficiently penetrate eukaryotic cells, but unexpectedly it eradicated or destroyed all tested cancer cell lines and primary cells irrespective of species without further stimulus through a necrosis-like cell death. Typical inhibitors of different forms of RCD cannot impede this process, which appears to involve a direct disruption of biomembranes. Nevertheless, our finding has potential clinical relevance; reliable induction of a necrotic form of cell death distinct from all known forms of RCD may offer a novel therapeutic approach to combat resistant tumour cells.


Subject(s)
Gene Products, tat/chemistry , Gene Products, tat/metabolism , Protein Domains , Recombinant Fusion Proteins/metabolism , Ribonucleotide Reductases/chemistry , Ribonucleotide Reductases/metabolism , Signal Transduction/genetics , Viral Proteins/chemistry , Viral Proteins/metabolism , Amino Acid Sequence , Amyloid/metabolism , Animals , Gene Products, tat/genetics , HIV-1/chemistry , HT29 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NIH 3T3 Cells , Necroptosis/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Ribonucleotide Reductases/genetics , U937 Cells , Viral Proteins/genetics
5.
Pflugers Arch ; 474(6): 613-624, 2022 06.
Article in English | MEDLINE | ID: mdl-35312839

ABSTRACT

Experimental nephrotic syndrome leads to activation of the epithelial sodium channel (ENaC) by proteolysis and promotes renal sodium retention. The membrane-anchored serine protease prostasin (CAP1/PRSS8) is expressed in the distal nephron and participates in proteolytic ENaC regulation by serving as a scaffold for other serine proteases. However, it is unknown whether prostasin is also involved in ENaC-mediated sodium retention of experimental nephrotic syndrome. In this study, we used genetically modified knock-in mice with Prss8 mutations abolishing its proteolytic activity (Prss8-S238A) or prostasin activation (Prss8-R44Q) to investigate the development of sodium retention in doxorubicin-induced nephrotic syndrome. Healthy Prss8-S238A and Prss8-R44Q mice had normal ENaC activity as reflected by the natriuretic response to the ENaC blocker triamterene. After doxorubicin injection, all genotypes developed similar proteinuria. In all genotypes, urinary prostasin excretion increased while renal expression was not altered. In nephrotic mice of all genotypes, triamterene response was similarly increased, consistent with ENaC activation. As a consequence, urinary sodium excretion dropped in all genotypes and mice similarly gained body weight by + 25 ± 3% in Prss8-wt, + 20 ± 2% in Prss8-S238A and + 28 ± 3% in Prss8-R44Q mice (p = 0.16). In Western blots, expression of fully cleaved α- and γ-ENaC was similarly increased in nephrotic mice of all genotypes. In conclusion, proteolytic ENaC activation and sodium retention in experimental nephrotic syndrome are independent of the activation of prostasin and its enzymatic activity and are consistent with the action of aberrantly filtered serine proteases or proteasuria.


Subject(s)
Nephrotic Syndrome , Serine Endopeptidases , Sodium , Animals , Doxorubicin/pharmacology , Epithelial Sodium Channels/genetics , Epithelial Sodium Channels/metabolism , Mice , Nephrotic Syndrome/genetics , Nephrotic Syndrome/metabolism , Serine Endopeptidases/metabolism , Serine Proteases/metabolism , Sodium/metabolism , Triamterene
6.
Nature ; 535(7613): 511-6, 2016 07 28.
Article in English | MEDLINE | ID: mdl-27466123

ABSTRACT

The vast majority of systemic bacterial infections are caused by facultative, often antibiotic-resistant, pathogens colonizing human body surfaces. Nasal carriage of Staphylococcus aureus predisposes to invasive infection, but the mechanisms that permit or interfere with pathogen colonization are largely unknown. Whereas soil microbes are known to compete by production of antibiotics, such processes have rarely been reported for human microbiota. We show that nasal Staphylococcus lugdunensis strains produce lugdunin, a novel thiazolidine-containing cyclic peptide antibiotic that prohibits colonization by S. aureus, and a rare example of a non-ribosomally synthesized bioactive compound from human-associated bacteria. Lugdunin is bactericidal against major pathogens, effective in animal models, and not prone to causing development of resistance in S. aureus. Notably, human nasal colonization by S. lugdunensis was associated with a significantly reduced S. aureus carriage rate, suggesting that lugdunin or lugdunin-producing commensal bacteria could be valuable for preventing staphylococcal infections. Moreover, human microbiota should be considered as a source for new antibiotics.


Subject(s)
Anti-Bacterial Agents/metabolism , Peptides, Cyclic/metabolism , Staphylococcal Infections/microbiology , Staphylococcal Infections/prevention & control , Staphylococcus aureus/growth & development , Staphylococcus lugdunensis/metabolism , Symbiosis , Thiazolidines/metabolism , Animals , Anti-Bacterial Agents/biosynthesis , Carrier State/microbiology , Disease Models, Animal , Drug Resistance, Microbial , Female , Humans , Male , Mice , Mice, Inbred C57BL , Microbial Sensitivity Tests , Microbiota/physiology , Nose/microbiology , Sigmodontinae , Staphylococcal Infections/drug therapy , Staphylococcus aureus/pathogenicity
7.
Molecules ; 27(18)2022 Sep 08.
Article in English | MEDLINE | ID: mdl-36144551

ABSTRACT

The prevalence of novel SARS-CoV-2 variants is also accompanied by an increased turnover rate and additional cleavage sites at the positions necessary for priming the Spike (S) protein. Of these priming sites, the proteolytically sensitive polybasic sequence of the activation loop at the S1/S2 interface and the S2' location within the S2 subunit of the S protein are cleaved by furin and TMPRSS2, which are important for the infection of the target cell. Neutrophils, migrating to the site of infection, secrete serine proteases to fight against pathogens. The serine proteases encompass neutrophil elastase (NE), proteinase 3 (PR3), and cathepsin G (CatG), which can hydrolyze the peptide bond adjacent to the S1/S2 interface. SARS-CoV-2 might take the opportunity to hijack proteases from an immune response to support viral entry to the cell. The region near S704L within the S2 subunit, a novel amino acid substitution of SARS-CoV-2 Omicron sublineage BA.2.12.1, is located close to the S1/S2 interface. We found that NE, PR3, and CatG digested the peptide within this region; however, the S704L amino acid substitution altered cleavage sites for PR3. In conclusion, such an amino acid substitution modifies S2 antigen processing and might further impact the major histocompatibility complex (MHC) binding and T cell activation.


Subject(s)
COVID-19 , SARS-CoV-2 , Cathepsin G , Furin/genetics , Humans , Leukocyte Elastase , Myeloblastin , Peptide Hydrolases/metabolism , Peptides , Spike Glycoprotein, Coronavirus/metabolism
8.
J Cell Sci ; 131(1)2018 01 04.
Article in English | MEDLINE | ID: mdl-29222110

ABSTRACT

Casein kinase 1 (CK1) plays central roles in various signal transduction pathways and performs many cellular activities. For many years CK1 was thought to act independently of modulatory subunits and in a constitutive manner. Recently, DEAD box RNA helicases, in particular DEAD box RNA helicase 3 X-linked (DDX3X), were found to stimulate CK1 activity in vitro In order to observe CK1 activity in living cells and to study its interaction with DDX3X, we developed a CK1-specific FRET biosensor. This tool revealed that DDX3X is indeed required for full CK1 activity in living cells. Two counteracting mechanisms control the activity of these enzymes. Phosphorylation by CK1 impairs the ATPase activity of DDX3X and RNA destabilizes the DDX3X-CK1 complex. We identified possible sites of interaction between DDX3X and CK1. While mutations identified in the DDX3X genes of human medulloblastoma patients can enhance CK1 activity in living cells, the mechanism of CK1 activation by DDX3X points to a possible therapeutic approach in CK1-related diseases such as those caused by tumors driven by aberrant Wnt/ß-catenin and Sonic hedgehog (SHH) activation. Indeed, CK1 peptides can reduce CK1 activity.


Subject(s)
Biosensing Techniques , Casein Kinase I/metabolism , DEAD-box RNA Helicases/metabolism , Medulloblastoma/genetics , RNA Helicases/metabolism , Wnt Signaling Pathway , Cerebellar Neoplasms/genetics , DEAD-box RNA Helicases/genetics , Fluorescence Resonance Energy Transfer , Humans , Mutation , Phosphorylation , RNA Helicases/genetics
9.
Biochem Biophys Res Commun ; 527(1): 238-241, 2020 06 18.
Article in English | MEDLINE | ID: mdl-32446374

ABSTRACT

Aspartic proteases are important biomarkers of human disease and interesting targets for modulation of immune response via MHC class II antigen processing inhibition. The lack of inhibitors with sufficient selectivity hampers precise analysis of the role of cathepsin E and napsin A in samples containing the ubiquitous and highly abundant homolog cathepsin D. Grassystatins from marine cyanobacteria show promising selectivity for cathepsin E but contain several ester bonds that make their synthesis cumbersome and thus limit availability of the inhibitors. Herewith, we present grassystatin-derived cathepsin E inhibitors with greatly facilitated synthesis but retained selectivity profile. We demonstrate their affinity and selectivity with both enzyme kinetic assays and streptavidin-based pull-down from cells and mouse organs. Our findings suggest that grassystatin-like inhibitors are useful tools for targeted inhibition of cathepsin E and thus provide a novel approach for cancer and immunology research.


Subject(s)
Cathepsin D/antagonists & inhibitors , Cathepsin E/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Peptides/pharmacology , Cathepsin D/metabolism , Cathepsin E/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , HEK293 Cells , Humans , Molecular Structure , Peptides/chemical synthesis , Peptides/chemistry , Recombinant Proteins/metabolism , Stereoisomerism , Structure-Activity Relationship
10.
Proc Natl Acad Sci U S A ; 114(17): E3507-E3515, 2017 04 25.
Article in English | MEDLINE | ID: mdl-28396446

ABSTRACT

Acid-sensing ion channels (ASICs) are proton-gated Na+ channels that are expressed throughout the nervous system. ASICs have been implicated in several neuronal disorders, like ischemic stroke, neuronal inflammation, and pathological pain. Several toxins from venomous animals have been identified that target ASICs with high specificity and potency. These toxins are extremely useful in providing protein pharmacophores and to characterize function and structure of ASICs. Marine cone snails contain a high diversity of toxins in their venom such as conotoxins, which are short polypeptides stabilized by disulfide bonds, and conopeptides, which have no or only one disulfide bond. Whereas conotoxins selectively target specific neuronal proteins, mainly ion channels, the targets of conopeptides are less well known. Here, we perform an in vitro screen of venoms from 18 cone snail species to identify toxins targeting ASICs. We identified a small conopeptide of only four amino acids from the venom of Conus textile that strongly potentiated currents of ASIC3, which has a specific role in the pain pathway. This peptide, RPRFamide, belongs to the subgroup of cono-RFamides. Electrophysiological characterization of isolated dorsal root ganglion (DRG) neurons revealed that RPRFamide increases their excitability. Moreover, injection of the peptide into the gastrocnemius muscle strongly enhanced acid-induced muscle pain in mice that was abolished by genetic inactivation of ASIC3. In summary, we identified a conopeptide that targets the nociceptor-specific ion channel ASIC3.


Subject(s)
Acid Sensing Ion Channels/metabolism , Conotoxins/chemistry , Conotoxins/toxicity , Conus Snail/chemistry , Ganglia, Spinal/metabolism , Muscle, Skeletal/metabolism , Myalgia/metabolism , Neurons/metabolism , Animals , Ganglia, Spinal/pathology , Mice , Muscle, Skeletal/physiology , Myalgia/chemically induced , Myalgia/pathology , Neurons/pathology , Xenopus laevis
11.
Biochem Biophys Res Commun ; 515(1): 145-148, 2019 07 12.
Article in English | MEDLINE | ID: mdl-31130231

ABSTRACT

Napsin A is an intracellular aspartic protease and biomarker of various malignancies like lung adenocarcinoma and ovarian clear cell carcinoma, but its detection is usually limited to immunohistochemical techniques gaining excellent information on its distribution but missing information about posttranslational modifications (e.g. maturation state) of the protein. We present a protocol for specific enrichment of napsin A from clinical or biological specimens, that facilitates detailed analysis of the protein. By using the exceptionally broad pH range under which napsin A binds to its inhibitor pepstatin A we achieve highly selective binding of napsin A while other aspartic proteases have negligible affinity. Using this method we demonstrate that lung napsin A in many mammals is a heterogeneous enzyme with a characteristic ladder-like appearance in SDS-PAGE that might be caused by proteolytically processed N- and/or C-termini, in contrast to the more homogeneous form found in kidneys and primary lung adenocarcinoma.


Subject(s)
Aspartic Acid Endopeptidases/metabolism , Lung/metabolism , Pepstatins/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Aspartic Acid Endopeptidases/analysis , Aspartic Acid Endopeptidases/genetics , Blotting, Western , Cattle , Guinea Pigs , HEK293 Cells , Humans , Hydrogen-Ion Concentration , Lung/pathology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Pepstatins/genetics , Protein Binding , Rabbits , Rats , Sheep , Species Specificity , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
12.
Int J Mol Sci ; 20(20)2019 Oct 10.
Article in English | MEDLINE | ID: mdl-31658587

ABSTRACT

The peptide TFF3 is a member of a family of secretory lectins, and is typically synthesized by mucous epithelia together with mucins. It is mainly released from intestinal goblet cells as a high-molecular mass heterodimer with IgG Fc binding protein (FCGBP). Herein, we investigated human saliva by fast protein liquid chromatography (FPLC) and proteomics and identified high- and low-molecular-mass forms of TFF3. Whereas the high-molecular-mass forms represent a heterodimer with FCGBP, the low-molecular-mass forms represent homodimeric TFF3 forms. Proteomic analysis also revealed a C-terminally truncated form of TFF3. We hypothesize that salivary TFF3-FCGBP might play a role in the innate immune defense of the oral cavity and that TFF3 might also bind to microbial glycans. The known interaction of TFF3 with the agglutinin DMBT-1, a typical constituent of human saliva, further supports this protective role.


Subject(s)
Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/metabolism , Dimerization , Saliva/metabolism , Trefoil Factor-3/chemistry , Trefoil Factor-3/metabolism , Calcium-Binding Proteins/metabolism , Cell Adhesion Molecules/immunology , DNA-Binding Proteins/metabolism , Humans , Immunity, Innate , Polysaccharides , Protein Interaction Domains and Motifs , Proteomics , Saliva/immunology , Trefoil Factor-3/immunology , Tumor Suppressor Proteins/metabolism
13.
Angew Chem Int Ed Engl ; 58(27): 9234-9238, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31059155

ABSTRACT

Lugdunin, a novel thiazolidine cyclopeptide, exhibits micromolar activity against methicillin-resistant Staphylococcus aureus (MRSA). For structure-activity relationship (SAR) studies, synthetic analogues obtained from alanine and stereo scanning as well as peptides with modified thiazolidine rings were tested for antimicrobial activity. The thiazolidine ring and the alternating d- and l-amino acid backbone are essential. Notably, the non-natural enantiomer displays equal activity, thus indicating the absence of a chiral target. The antibacterial activity strongly correlates with dissipation of the membrane potential in S. aureus. Lugdunin equalizes pH gradients in artificial membrane vesicles, thereby maintaining membrane integrity, which demonstrates that proton translocation is the mode of action (MoA). The incorporation of extra tryptophan or propargyl moieties further expands the diversity of this class of thiazolidine cyclopeptides.


Subject(s)
Anti-Infective Agents/chemical synthesis , Peptides, Cyclic/chemistry , Thiazolidines/chemistry , Alanine/chemistry , Amino Acid Sequence , Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Microbial Sensitivity Tests , Peptides, Cyclic/chemical synthesis , Peptides, Cyclic/pharmacology , Protons , Stereoisomerism , Structure-Activity Relationship , Thiazolidines/chemical synthesis , Thiazolidines/pharmacology
14.
Kidney Int ; 93(1): 159-172, 2018 01.
Article in English | MEDLINE | ID: mdl-29042083

ABSTRACT

Volume retention in nephrotic syndrome has been linked to activation of the epithelial sodium channel (ENaC) by proteolysis of its γ-subunit following urinary excretion of serine proteases such as plasmin. Here we tested whether pharmacological inhibition of urinary serine protease activity might protect from ENaC activation and volume retention in nephrotic syndrome. Urine from both nephrotic mice (induced by doxorubicin injection) and nephrotic patients exhibited high aprotinin-sensitive serine protease activity. Treatment of nephrotic mice with the serine protease inhibitor aprotinin by means of subcutaneous sustained-release pellets normalized urinary serine protease activity and prevented sodium retention, as did treatment with the ENaC inhibitor amiloride. In the kidney cortex from nephrotic mice, immunofluorescence revealed increased apical γ-ENaC staining, normalized by aprotinin treatment. In Xenopus laevis oocytes heterologously expressing murine ENaC, aprotinin had no direct inhibitory effect on channel activity but prevented proteolytic channel activation. Thus, our study shows that volume retention in experimental nephrotic syndrome is related to proteolytic ENaC activation by proteasuria and can be prevented by treatment with aprotinin. Hence, inhibition of urinary serine protease activity might become a therapeutic approach to treat patients with nephrotic-range proteinuria.


Subject(s)
Aprotinin/pharmacology , Edema/drug therapy , Epithelial Sodium Channels/drug effects , Kidney/drug effects , Nephrotic Syndrome/drug therapy , Nephrotic Syndrome/enzymology , Serine Proteases/urine , Serine Proteinase Inhibitors/pharmacology , Water-Electrolyte Balance/drug effects , Animals , Case-Control Studies , Disease Models, Animal , Doxorubicin , Edema/enzymology , Edema/etiology , Edema/physiopathology , Epithelial Sodium Channels/genetics , Epithelial Sodium Channels/metabolism , Humans , Ion Channel Gating/drug effects , Kidney/metabolism , Kidney/pathology , Mice, 129 Strain , Nephrotic Syndrome/chemically induced , Nephrotic Syndrome/physiopathology , Proteolysis , Signal Transduction/drug effects , Xenopus laevis
15.
EMBO Rep ; 17(7): 965-81, 2016 07.
Article in English | MEDLINE | ID: mdl-27226123

ABSTRACT

Mitochondria are separated from the remainder of the eukaryotic cell by the mitochondrial outer membrane (MOM). The MOM plays an important role in different transport processes like lipid trafficking and protein import. In yeast, the ER-mitochondria encounter structure (ERMES) has a central, but poorly defined role in both activities. To understand the functions of the ERMES, we searched for suppressors of the deficiency of one of its components, Mdm10, and identified a novel mitochondrial protein that we named Mdm10 complementing protein 3 (Mcp3). Mcp3 partially rescues a variety of ERMES-related phenotypes. We further demonstrate that Mcp3 is an integral protein of the MOM that follows a unique import pathway. It is recognized initially by the import receptor Tom70 and then crosses the MOM via the translocase of the outer membrane. Mcp3 is next relayed to the TIM23 translocase at the inner membrane, gets processed by the inner membrane peptidase (IMP) and finally integrates into the MOM. Hence, Mcp3 follows a novel biogenesis route where a MOM protein is processed by a peptidase of the inner membrane.


Subject(s)
Fungal Proteins/metabolism , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/metabolism , Peptide Hydrolases/metabolism , Signal Transduction , Carrier Proteins/metabolism , Fungal Proteins/genetics , Gene Deletion , Gene Dosage , Gene Expression , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/ultrastructure , Mitochondrial Precursor Protein Import Complex Proteins , Mitochondrial Proteins/genetics , Multiprotein Complexes , Protein Transport , Proteolysis
16.
J Immunol ; 196(3): 1284-92, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26729806

ABSTRACT

The challenging human pathogen Staphylococcus aureus has highly efficient immune evasion strategies for causing a wide range of diseases, from skin and soft tissue to life-threatening infections. Phenol-soluble modulin (PSM) peptides are major pathogenicity factors of community-associated methicillin-resistant S. aureus strains. In previous work, we demonstrated that PSMs in combination with TLR2 ligand from S. aureus induce tolerogenic dendritic cells (DCs) characterized by the production of high amounts of IL-10, but no proinflammatory cytokines. This in turn promotes the activation of regulatory T cells while impairing Th1 response; however, the signaling pathways modulated by PSMs remain elusive. In this study, we analyzed the effects of PSMs on signaling pathway modulation downstream of TLR2. TLR2 stimulation in combination with PSMα3 led to increased and prolonged phosphorylation of NF-κB, ERK, p38, and CREB in mouse bone marrow-derived DCs compared with single TLR2 activation. Furthermore, inhibition of p38 and downstream MSK1 prevented IL-10 production, which in turn reduced the capacity of DCs to activate regulatory T cells. Interestingly, the modulation of the signaling pathways by PSMs was independent of the known receptor for PSMs, as shown by experiments with DCs lacking the formyl peptide receptor 2. Instead, PSMs penetrate the cell membrane most likely by transient pore formation. Moreover, colocalization of PSMs and p38 was observed near the plasma membrane in the cytosol, indicating a direct interaction. Thus, PSMs from S. aureus directly modulate the signaling pathway p38-CREB in DCs, thereby impairing cytokine production and in consequence T cell priming to increase the tolerance toward the pathogen.


Subject(s)
Bacterial Toxins/immunology , Cyclic AMP Response Element-Binding Protein/immunology , Dendritic Cells/immunology , MAP Kinase Signaling System/immunology , Staphylococcal Infections/immunology , T-Lymphocytes/immunology , Animals , Cytokines/biosynthesis , Disease Models, Animal , Female , Flow Cytometry , Immune Evasion/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Peptides/immunology , Staphylococcus aureus/immunology
17.
Plant Cell ; 26(5): 1838-1847, 2014 May.
Article in English | MEDLINE | ID: mdl-24808051

ABSTRACT

Peptide signals have emerged as an important class of regulators in cell-to-cell communication in plants. Several families of small, secreted proteins with a conserved C-terminal Pro-rich motif have been identified as functional peptide signals in Arabidopsis thaliana. These proteins are presumed to be trimmed proteolytically and undergo posttranslational modifications, such as hydroxylation of Pro residues and glycosylation, to form mature, bioactive signals. Identification and matching of such ligands with their respective receptors remains a major challenge since the genes encoding them often show redundancy and low expression restricted to a few cells or particular developmental stages. To overcome these difficulties, we propose the use of ectopic expression of receptor genes in suitable plant cells like Nicotiana benthamiana for testing ligand candidates in receptor output assays and in binding studies. As an example, we used the IDA peptide HAE/HSL2 receptor signaling system known to regulate floral organ abscission. We demonstrate that the oxidative burst response can be employed as readout for receptor activation by synthetic peptides and that a new, highly sensitive, nonradioactive labeling approach can be used to reveal a direct correlation between peptide activity and receptor affinity. We suggest that these approaches will be of broad value for the field of ligand-receptor studies in plants.

18.
Bioorg Med Chem ; 25(15): 4163-4174, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28642030

ABSTRACT

EAE is induced to susceptible mice using linear peptides of myelin proteins of the central nervous system. Specific peptide motifs within the peptide-binding groove of the MHC peptide-complex determines the affinity of the peptide in each animal and the consequent T-cell receptor recognition and activation of the cell. Altered peptide ligand (APL) vaccination is a novel approach based on an effort to induce T-cell tolerance or alter cytokine profile from pro-inflammatory to anti-inflammatory. In the present study we synthesized the MOG35-55 peptide and altered its 3-dimensional conformation to make it a cyclic one (c-MOG35-55). EAE was induced in C57BL/6 mice and pathology was studied on acute and chronic phase of the disease. Our data indicates that c-MOG35-55 peptide alone induces a mild transient acute phase without chronic axonopathy. Administration of the c-MOG35-55 peptide at a 1:1 ratio during disease induction significantly ameliorates clinical disease and underlying pathology, such as demyelination and axonopathy in the acute and chronic phases. Binding and structural studies revealed milder interactions between the c-MOG35-55 and mouse or human MHC class II alleles (H2-IAb and HLA-DR2). Collectively, we provide data supporting for the first time the concept that the cyclic modification of an established encephalitogenic peptide ameliorates the clinical outcomes and underlying pathological processes of EAE. Such a cyclic modification of linear peptides could provide a novel treatment approach for future, patient-selective, immunomodulative treatments of multiple sclerosis.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Peptides, Cyclic/therapeutic use , Animals , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Histocompatibility Antigens Class II/immunology , Immune Tolerance , Mice , Mice, Inbred C57BL , Receptors, Antigen, T-Cell/immunology
20.
J Autoimmun ; 75: 82-95, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27484364

ABSTRACT

Cathepsin S (CTSS) is a lysosomal protease whose activity regulation is important for MHC-II signaling and subsequent activation of CD4+ T cell mediated immune responses. Dysregulation of its enzymatic activity or enhanced secretion into extracellular environments is associated with the induction or progression of several autoimmune diseases. Here we demonstrate that commensal intestinal bacteria influence secretion rates and intracellular activity of host CTSS and that symbiotic bacteria, i.e. Bacteroides vulgatus mpk, may actively regulate this process and help to maintain physiological levels of CTSS activities in order to prevent from induction of pathological inflammation. The symbiont-controlled regulation of CTSS activity is mediated by anticipating reactive oxygen species induction in dendritic cells which, in turn, maintains cystatin C (CysC) monomer binding to CTSS. CysC monomers are potent endogenous CTSS inhibitors. This Bacteroides vulgatus caused and CysC dependent CTSS activity regulation is involved in the generation of tolerant intestinal dendritic cells contributing to prevention of T-cell mediated induction of colonic inflammation. Taken together, we demonstrate that symbionts of the intestinal microbiota regulate host CTSS activity and secretion and might therefore be an attractive approach to deal with CTSS associated autoimmune diseases.


Subject(s)
Bacteria/immunology , Cathepsins/immunology , Gastrointestinal Microbiome/immunology , Symbiosis/immunology , Animals , Bacteroides/immunology , Bacteroides/physiology , Bacteroides Infections/immunology , Bacteroides Infections/microbiology , Benzopyrans/pharmacology , Blotting, Western , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Bone Marrow Cells/microbiology , Carbamates/pharmacology , Cathepsins/antagonists & inhibitors , Cathepsins/genetics , Cells, Cultured , Colitis/immunology , Colitis/metabolism , Cytokines/immunology , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/microbiology , Gastrointestinal Microbiome/physiology , Gene Expression/immunology , Host-Pathogen Interactions/immunology , Immune Tolerance/immunology , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL