Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Nat Immunol ; 21(1): 54-64, 2020 01.
Article in English | MEDLINE | ID: mdl-31819256

ABSTRACT

Ptpn6 is a cytoplasmic phosphatase that functions to prevent autoimmune and interleukin-1 (IL-1) receptor-dependent, caspase-1-independent inflammatory disease. Conditional deletion of Ptpn6 in neutrophils (Ptpn6∆PMN) is sufficient to initiate IL-1 receptor-dependent cutaneous inflammatory disease, but the source of IL-1 and the mechanisms behind IL-1 release remain unclear. Here, we investigate the mechanisms controlling IL-1α/ß release from neutrophils by inhibiting caspase-8-dependent apoptosis and Ripk1-Ripk3-Mlkl-regulated necroptosis. Loss of Ripk1 accelerated disease onset, whereas combined deletion of caspase-8 and either Ripk3 or Mlkl strongly protected Ptpn6∆PMN mice. Ptpn6∆PMN neutrophils displayed increased p38 mitogen-activated protein kinase-dependent Ripk1-independent IL-1 and tumor necrosis factor production, and were prone to cell death. Together, these data emphasize dual functions for Ptpn6 in the negative regulation of p38 mitogen-activated protein kinase activation to control tumor necrosis factor and IL-1α/ß expression, and in maintaining Ripk1 function to prevent caspase-8- and Ripk3-Mlkl-dependent cell death and concomitant IL-1α/ß release.


Subject(s)
Apoptosis/immunology , Caspase 8/immunology , Neutrophils/immunology , Protein Kinases/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/immunology , Animals , Caspase 8/genetics , Cells, Cultured , Gene Deletion , Inflammation/immunology , Interleukin-1/immunology , Interleukin-1alpha/metabolism , Interleukin-1beta/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Receptors, Interleukin-1 Type I/immunology , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
2.
Cell Metab ; 23(1): 155-64, 2016 Jan 12.
Article in English | MEDLINE | ID: mdl-26603191

ABSTRACT

Interleukin-18 (IL-18) is activated by Caspase-1 in inflammasome complexes and has anti-obesity effects; however, it is not known which inflammasome regulates this process. We found that mice lacking the NLRP1 inflammasome phenocopy mice lacking IL-18, with spontaneous obesity due to intrinsic lipid accumulation. This is exacerbated when the mice are fed a high-fat diet (HFD) or a high-protein diet, but not when mice are fed a HFD with low energy density (high fiber). Furthermore, mice with an activating mutation in NLRP1, and hence increased IL-18, have decreased adiposity and are resistant to diet-induced metabolic dysfunction. Feeding these mice a HFD further increased plasma IL-18 concentrations and strikingly resulted in loss of adipose tissue mass and fatal cachexia, which could be prevented by genetic deletion of IL-18. Thus, NLRP1 is an innate immune sensor that functions in the context of metabolic stress to produce IL-18, preventing obesity and metabolic syndrome.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , Inflammasomes/metabolism , Interleukin-18/biosynthesis , Metabolic Syndrome/metabolism , Obesity/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Apoptosis Regulatory Proteins/genetics , Body Weight , Diet, High-Fat/adverse effects , Interleukin-18/genetics , Liver/metabolism , Liver/pathology , Male , Metabolic Syndrome/prevention & control , Mice, Knockout , Obesity/etiology , Obesity/prevention & control
3.
J Exp Med ; 212(6): 927-38, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-26008898

ABSTRACT

Gain-of-function mutations that activate the innate immune system can cause systemic autoinflammatory diseases associated with increased IL-1ß production. This cytokine is activated identically to IL-18 by an intracellular protein complex known as the inflammasome; however, IL-18 has not yet been specifically implicated in the pathogenesis of hereditary autoinflammatory disorders. We have now identified an autoinflammatory disease in mice driven by IL-18, but not IL-1ß, resulting from an inactivating mutation of the actin-depolymerizing cofactor Wdr1. This perturbation of actin polymerization leads to systemic autoinflammation that is reduced when IL-18 is deleted but not when IL-1 signaling is removed. Remarkably, inflammasome activation in mature macrophages is unaltered, but IL-18 production from monocytes is greatly exaggerated, and depletion of monocytes in vivo prevents the disease. Small-molecule inhibition of actin polymerization can remove potential danger signals from the system and prevents monocyte IL-18 production. Finally, we show that the inflammasome sensor of actin dynamics in this system requires caspase-1, apoptosis-associated speck-like protein containing a caspase recruitment domain, and the innate immune receptor pyrin. Previously, perturbation of actin polymerization by pathogens was shown to activate the pyrin inflammasome, so our data now extend this guard hypothesis to host-regulated actin-dependent processes and autoinflammatory disease.


Subject(s)
Actins/physiology , Cytoskeletal Proteins/metabolism , Hereditary Autoinflammatory Diseases/metabolism , Inflammation/metabolism , Interleukin-1beta/metabolism , Microfilament Proteins/metabolism , Actins/chemistry , Animals , Bone Marrow Cells/cytology , Caspase 1/metabolism , Caspases/metabolism , Clodronic Acid/chemistry , Crosses, Genetic , Culture Media, Conditioned/chemistry , Enzyme-Linked Immunosorbent Assay , Interleukin-18/metabolism , Lipopolysaccharides/metabolism , Liposomes/chemistry , Liver/embryology , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Fluorescence , Monocytes/cytology , Pyrin , Signal Transduction
4.
J Leukoc Biol ; 97(2): 321-6, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25473101

ABSTRACT

The regulation of neutrophil lifespan is critical for a circumscribed immune response. Neutrophils are sensitive to Fas/CD95 death receptor signaling in vitro, but it is unknown if Fas regulates neutrophil lifespan in vivo. We hypothesized that FasL-expressing CD8(+) T cells, which kill antigen-stimulated T cells during chronic viral infection, can also induce neutrophil death in tissues during infection. With the use of LysM-Cre Fas(fl/fl) mice, which lack Fas expression in macrophages and neutrophils, we show that Fas regulates neutrophil lifespan during lymphocytic choriomeningitis virus (LCMV) infection in the lung, peripheral blood, and spleen. Fas also contributed to the regulation of neutrophil numbers in the colon of Citrobacter rodentium-infected mice. To examine the effects of infection on Fas activation in neutrophils, we primed neutrophils with TLR ligands or IL-18, resulting in ablation of Fas death receptor signaling. These data provide the first in vivo genetic evidence that neutrophil lifespan is controlled by death receptor signaling and provide a mechanism to account for neutrophil resistance to Fas stimulation during infection.


Subject(s)
Cellular Senescence/immunology , Citrobacter rodentium/immunology , Enterobacteriaceae Infections/immunology , Gene Expression Regulation/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Neutrophils/immunology , fas Receptor/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cellular Senescence/genetics , Enterobacteriaceae Infections/genetics , Enterobacteriaceae Infections/pathology , Fas Ligand Protein/genetics , Fas Ligand Protein/immunology , Gene Expression Regulation/genetics , Interleukin-18/genetics , Interleukin-18/immunology , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/pathology , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Knockout , Neutrophils/pathology , Signal Transduction/genetics , Signal Transduction/immunology , fas Receptor/genetics
5.
Cell ; 157(5): 1175-88, 2014 May 22.
Article in English | MEDLINE | ID: mdl-24813849

ABSTRACT

Upon ligand binding, RIPK1 is recruited to tumor necrosis factor receptor superfamily (TNFRSF) and Toll-like receptor (TLR) complexes promoting prosurvival and inflammatory signaling. RIPK1 also directly regulates caspase-8-mediated apoptosis or, if caspase-8 activity is blocked, RIPK3-MLKL-dependent necroptosis. We show that C57BL/6 Ripk1(-/-) mice die at birth of systemic inflammation that was not transferable by the hematopoietic compartment. However, Ripk1(-/-) progenitors failed to engraft lethally irradiated hosts properly. Blocking TNF reversed this defect in emergency hematopoiesis but, surprisingly, Tnfr1 deficiency did not prevent inflammation in Ripk1(-/-) neonates. Deletion of Ripk3 or Mlkl, but not Casp8, prevented extracellular release of the necroptotic DAMP, IL-33, and reduced Myd88-dependent inflammation. Reduced inflammation in the Ripk1(-/-)Ripk3(-/-), Ripk1(-/-)Mlkl(-/-), and Ripk1(-/-)Myd88(-/-) mice prevented neonatal lethality, but only Ripk1(-/-)Ripk3(-/-)Casp8(-/-) mice survived past weaning. These results reveal a key function for RIPK1 in inhibiting necroptosis and, thereby, a role in limiting, not only promoting, inflammation.


Subject(s)
Genes, Lethal , Hematopoiesis , Inflammation/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Animals , Animals, Newborn , Caspase 8/metabolism , Cell Death , Liver/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Protein Kinases/genetics , Protein Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Tumor Necrosis Factors/metabolism
7.
Immunity ; 37(6): 1009-23, 2012 Dec 14.
Article in English | MEDLINE | ID: mdl-23219391

ABSTRACT

Cytopenias are key prognostic indicators of life-threatening infection, contributing to immunosuppression and mortality. Here we define a role for Caspase-1-dependent death, known as pyroptosis, in infection-induced cytopenias by studying inflammasome activation in hematopoietic progenitor cells. The NLRP1a inflammasome is expressed in hematopoietic progenitor cells and its activation triggers their pyroptotic death. Active NLRP1a induced a lethal systemic inflammatory disease that was driven by Caspase-1 and IL-1ß but was independent of apoptosis-associated speck-like protein containing a CARD (ASC) and ameliorated by IL-18. Surprisingly, in the absence of IL-1ß-driven inflammation, active NLRP1a triggered pyroptosis of hematopoietic progenitor cells resulting in leukopenia at steady state. During periods of hematopoietic stress induced by chemotherapy or lymphocytic choriomeningitis virus (LCMV) infection, active NLRP1a caused prolonged cytopenia, bone marrow hypoplasia, and immunosuppression. Conversely, NLRP1-deficient mice showed enhanced recovery from chemotherapy and LCMV infection, demonstrating that NLRP1 acts as a cellular sentinel to alert Caspase-1 to hematopoietic and infectious stress.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , Apoptosis , Hematopoietic Stem Cells/metabolism , Inflammasomes/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Apoptosis Regulatory Proteins/genetics , CARD Signaling Adaptor Proteins , Caspase 1/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Dermatitis/immunology , Dermatitis/metabolism , Fluorouracil/pharmacology , Hematopoiesis/drug effects , Hematopoiesis/immunology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/virology , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Interferon-gamma/metabolism , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Mice , Mice, Knockout , Mutation , Pancytopenia/immunology , Pancytopenia/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
8.
Immunol Cell Biol ; 90(1): 124-9, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21519345

ABSTRACT

The lack of expression of the suppressor of cytokine signalling-3 (SOCS3) or inactivation of the negative regulatory capacity of SOCS3 has been well documented in rheumatoid arthritis, viral hepatitis and cancer. The specific qualitative and quantitative consequences of SOCS3 deficiency on interleukin-6 (IL-6)-mediated pro- and anti-inflammatory responses remain controversial in vitro and unknown in vivo. Mice with a conditional deletion of SOCS3 in hematopoietic cells develop lethal inflammatory disease during adult life and develop gross histopathological changes during experimental arthritis, typified by elevated IL-6 levels. To clarify the nature of the IL-6 responses in vivo, we generated mice deficient in SOCS3 (SOCS3(-/Δvav)) or both SOCS3 and IL-6 (IL-6(-/-)/SOCS3(-/Δvav)), and examined responses in models of acute and chronic inflammation. Acute responses to IL-1ß were lethal to SOCS3(-/Δvav) mice but not IL-6(-/-)/SOCS3(-/Δvav) mice, indicating that IL-6 was required for the lethal inflammation induced by IL-1ß. Administration of IL-1ß to SOCS3(-/Δvav) mice induced systemic apoptosis of lymphocytes in the thymus, spleen and lymph nodes that was dependent on the presence of IL-6. IL-6 deficiency prolonged survival of SOCS3(-/Δvav) mice and ameliorated spontaneous inflammatory disease developing during adult life. Infection of SOCS3(-/Δvav) mice with LCMV induced a lethal inflammatory response that was dependent on IL-6, despite SOCS3(-/Δvav) mice controlling viral replication. We conclude that SOCS3 is required for survival during inflammatory responses and is a critical regulator of IL-6 in vivo.


Subject(s)
Inflammation/immunology , Interleukin-6/immunology , Signal Transduction/immunology , Suppressor of Cytokine Signaling Proteins/immunology , Acute Disease , Animals , Apoptosis/drug effects , Apoptosis/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Chronic Disease , Cytokines/immunology , Cytokines/metabolism , Female , Flow Cytometry , Inflammation/genetics , Inflammation/metabolism , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Interleukin-1beta/pharmacology , Interleukin-6/deficiency , Interleukin-6/genetics , Lymphocytes/drug effects , Lymphocytes/immunology , Lymphocytes/metabolism , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/deficiency , Suppressor of Cytokine Signaling Proteins/genetics , Survival Analysis
9.
Proc Natl Acad Sci U S A ; 108(32): 13135-40, 2011 Aug 09.
Article in English | MEDLINE | ID: mdl-21768356

ABSTRACT

During immune responses, neutrophils must integrate survival and death signals from multiple sources to regulate their lifespan. Signals that activate either the Bcl-2- or death receptor-regulated apoptosis pathways can provide powerful stimuli for neutrophils to undergo cell death, but whether they act cooperatively in parallel or directly cross-talk in neutrophils is not known. Previous studies suggested that Bcl-2 family proteins are not required for Fas-induced cell death in neutrophils, but did not examine whether they could modulate its rapid onset. By monitoring the rate of change in neutrophil viability associated with activation of the Fas-triggered death receptor pathway using real-time cell imaging, we show that the Bcl-2-related proteins Bid, Bax, and Bak accelerate neutrophil apoptosis but are not essential for cell death. Increased Bcl-2 or Mcl-1 expression prevents efficient induction of apoptosis by Fas stimulation indicating that the Bcl-2-regulated apoptosis pathway can directly interfere with Fas-triggered apoptosis. Fas has been shown to initiate NFκB activation and gene transcription in cell lines, however gene transcription is not altered in Fas-activated Bid(-/-) neutrophils, indicating that apoptosis occurs independently of gene transcription in neutrophils. The specification of kinetics of neutrophil apoptosis by Bid impacts on the magnitude of neutrophil IL-1ß production, implicating a functional role for the Bcl-2-regulated pathway in controlling neutrophil responses to FasL. These data demonstrate that the intrinsic apoptosis pathway directly controls the kinetics of Fas-triggered apoptosis in neutrophils.


Subject(s)
Apoptosis , BH3 Interacting Domain Death Agonist Protein/metabolism , Neutrophils/cytology , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism , fas Receptor/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Biphenyl Compounds/pharmacology , Cell Survival/drug effects , Fas Ligand Protein/pharmacology , Gene Expression Regulation/drug effects , Humans , Imaging, Three-Dimensional , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myeloid Cell Leukemia Sequence 1 Protein , Neutrophils/drug effects , Neutrophils/metabolism , Nitrophenols/pharmacology , Piperazines/pharmacology , Signal Transduction/drug effects , Sulfonamides/pharmacology
10.
J Immunol ; 186(2): 1131-9, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21160041

ABSTRACT

The regulation of neutrophil recruitment, activation, and disposal is pivotal for circumscribed inflammation. SHP1(Y208N/Y208N) mutant mice develop severe cutaneous inflammatory disease that is IL-1R dependent. Genetic reduction in neutrophil numbers and neutrophilic responses to infection is sufficient to prevent the spontaneous initiation of this disease. Neutrophils from SHP1(Y208N/Y208N) mice display increased pro-IL-1ß production due to altered responses to MyD88-dependent and MyD88-independent signals. The IL-1R-dependent inflammatory disease in SHP1(Y208N/Y208N) mice develops independently of caspase 1 and proteinase 3 and neutrophil elastase. In response to Fas ligand, a caspase 1-independent inducer of IL-1ß production, neutrophils from SHP1(Y208N/Y208N) mice produce elevated levels of IL-1ß but display reduced caspase 3 and caspase 7 activation. In neutrophils deficient in SHP1, IL-1ß induces high levels of pro-IL-1ß suggesting the presence of a paracrine IL-1ß loop. These data indicate that the neutrophil- and IL-1-dependent disease in SHP1(Y208N/Y208N) mice is a consequence of loss of negative regulation of TLR and IL-1R signaling.


Subject(s)
Inflammation Mediators/physiology , Interleukin-1beta/biosynthesis , Neutrophils/immunology , Neutrophils/pathology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/physiology , Skin Diseases/pathology , Skin Diseases/prevention & control , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Autoimmune Diseases/prevention & control , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Humans , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Neutrophils/metabolism , Paracrine Communication/genetics , Paracrine Communication/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/deficiency , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Severity of Illness Index , Signal Transduction/genetics , Signal Transduction/immunology , Skin Diseases/immunology , Toll-Like Receptors/antagonists & inhibitors , Toll-Like Receptors/physiology
11.
EMBO Rep ; 11(8): 640-6, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20596075

ABSTRACT

Reports describing the effect of interferon-gamma (IFNgamma) on interleukin-1beta (IL-1beta) production are conflicting. We resolve this controversy by showing that IFNgamma potentiates IL-1beta release from human cells, but transiently inhibits the production of IL-1beta from mouse cells. Release from this inhibition is dependent on suppressor of cytokine signalling 1. IL-1beta and Th17 cells are pathogenic in mouse models for autoimmune disease, which use Mycobacterium tuberculosis (MTB), in which IFNgamma and IFNbeta are anti-inflammatory. We observed that these cytokines suppress IL-1beta production in response to MTB, resulting in a reduced number of IL-17-producing cells. In human cells, IFNgamma increased IL-1beta production, and this might explain why IFNgamma is detrimental for multiple sclerosis. In mice, IFNgamma decreased IL-1beta and subsequently IL-17, indicating that the adaptive immune response can provide a systemic, but transient, signal to limit inflammation.


Subject(s)
Interferon-gamma/pharmacology , Interleukin-17/immunology , Interleukin-1beta/immunology , Macrophages/drug effects , Suppressor of Cytokine Signaling Proteins/immunology , Animals , Cell Line , Dendritic Cells/cytology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Humans , Interferon-gamma/genetics , Interferon-gamma/immunology , Lipopolysaccharides/immunology , Lipopolysaccharides/pharmacology , Macrophages/cytology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mycobacterium/immunology , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins/genetics , T-Lymphocytes/cytology , T-Lymphocytes/immunology
12.
Proc Natl Acad Sci U S A ; 105(39): 15028-33, 2008 Sep 30.
Article in English | MEDLINE | ID: mdl-18806225

ABSTRACT

A recessive phenotype called spin (spontaneous inflammation) was induced by N-ethyl-N-nitrosourea (ENU) mutagenesis in C57BL/6J mice. Homozygotes display chronic inflammatory lesions affecting the feet, salivary glands and lungs, and antichromatin antibodies. They are immunocompetent and show enhanced resistance to infection by Listeria monocytogenes. TLR-induced TNF and IL-1 production are normal in macrophages derived from spin mice. The autoinflammatory phenotype of spin mice is fully suppressed by compound homozygosity for Myd88(poc), Irak4(otiose), and Il1r1-null mutations, but not Ticam1(Lps2), Stat1(m1Btlr), or Tnf-null mutations. Both autoimmune and autoinflammatory phenotypes are suppressed when spin homozygotes are derived into a germ-free environment. The spin phenotype was ascribed to a viable hypomorphic allele of Ptpn6, which encodes the tyrosine phosphatase SHP1, mutated in mice with the classical motheaten alleles me and me-v. Inflammation and autoimmunity caused by SHP1 deficiency are thus conditional. The SHP1-deficient phenotype is driven by microbes, which activate TLR signaling pathways to elicit IL-1 production. IL-1 signaling via MyD88 elicits inflammatory disease.


Subject(s)
Autoimmune Diseases/genetics , Inflammation/genetics , Interleukin-1/immunology , Listeriosis/immunology , Myeloid Differentiation Factor 88/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Alleles , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/microbiology , Autoimmunity/genetics , Ethylnitrosourea/pharmacology , Homozygote , Inflammation/immunology , Inflammation/microbiology , Interleukin-1/genetics , Interleukin-1 Receptor-Associated Kinases/genetics , Interleukin-1 Receptor-Associated Kinases/immunology , Mice , Mice, Mutant Strains , Mutagenesis , Mutation , Myeloid Differentiation Factor 88/genetics , Receptors, Interleukin-1/immunology , Toll-Like Receptors/immunology
13.
Br J Pharmacol ; 144(6): 743-54, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15778698

ABSTRACT

1. Our aim was to determine whether lipoamino acid conjugation of peptides that are high-affinity activators of ryanodine receptor (RyR) channels would (a) render the peptides membrane permeable, (b) alter their structure or (a) reduce their activity. The peptides correspond to the A region of the II-III loop of the skeletal dihydropyridine receptor. 2. The lipoamino acid conjugation increased the apparent permeability of the peptide across the Caco-2 cell monolayer by up to approximately 20-fold. 3. Nuclear magnetic resonance showed that the alpha-helical structure of critical basic residues, required for optimal activation of RyRs, was retained after conjugation. 4. The conjugated peptides were more effective in enhancing resting Ca2+ release, Ca2+-induced Ca2+ release and caffeine-induced Ca2+ release from isolated sarcoplasmic reticulum (SR) than their unconjugated counterparts, and significantly enhanced caffeine-induced Ca2+ release from mechanically skinned extensor digitorum longus (EDL) fibres. 5. The effect of both conjugated and unconjugated peptides on Ca2+ release from skeletal SR was 30-fold greater than their effect on either cardiac Ca2+ release or on the Ca2+ Mg2+ ATPase. 6. A small and very low affinity effect of the peptide in slowing Ca2+ uptake by the Ca2+, Mg2+ ATPase was exacerbated by lipoamino acid conjugation in both isolated SR and in skinned EDL fibres. 7. The results show that lipoamino acid conjugation of A region peptides increases their membrane permeability without impairing their structure or efficacy in activating skeletal and cardiac RyRs.


Subject(s)
Cell Membrane Permeability/drug effects , Peptide Fragments/pharmacology , Ryanodine Receptor Calcium Release Channel/drug effects , Alanine/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Binding Sites , Caco-2 Cells , Caffeine/pharmacology , Calcium/metabolism , Cell Culture Techniques , Cell Membrane Permeability/physiology , Dose-Response Relationship, Drug , Electrophysiology , Humans , Lipid Bilayers , Molecular Sequence Data , Molecular Structure , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Myocardium/metabolism , Nuclear Magnetic Resonance, Biomolecular , Peptide Fragments/chemistry , Peptide Fragments/genetics , Protein Structure, Secondary , Rabbits , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/metabolism , Stereoisomerism , Structure-Activity Relationship
14.
J Biol Chem ; 279(12): 11853-62, 2004 Mar 19.
Article in English | MEDLINE | ID: mdl-14699105

ABSTRACT

Imperatoxin A is a high affinity activator of ryanodine receptors. The toxin contains a positively charged surface structure similar to that of the A fragment of skeletal dihydropyridine receptors (peptide A), suggesting that the toxin and peptide could bind to a common site on the ryanodine receptor. However, the question of a common binding site has not been resolved, and the concentration dependence of the actions of the toxin has not been fully explored. We characterize two novel high affinity actions of the toxin on the transient gating of cardiac and skeletal channels, in addition to the well documented lower affinity induction of prolonged substates. Transient activity was (a) enhanced with 0.2-10 nm toxin and (b) depressed by >50 nm toxin. The toxin at >/=1 nm enhanced Ca(2+) release from SR in a manner consistent with two independent activation processes. The effects of the toxin on transient activity, as well as the toxin-induced substate, were independent of cytoplasmic Ca(2+) or Mg(2+) concentrations or the presence of adenine nucleotide and were seen in diisothiocyanostilbene-2',2'-disulfonic acid-modified channels. Peptide A activated skeletal and cardiac channels with 100 nm cytoplasmic Ca(2+) and competed with Imperatoxin A in the high affinity enhancement of transient channel activity and Ca(2+) release from SR. In contrast to transient activity, prolonged substate openings induced by the toxin were not altered in the presence of peptide A. The results suggest that Imperatoxin A has three independent actions on ryanodine receptor channels and competes with peptide A for at least one action.


Subject(s)
Ryanodine Receptor Calcium Release Channel/metabolism , Scorpion Venoms/metabolism , Animals , Binding Sites , Lipid Bilayers , Rabbits , Ryanodine Receptor Calcium Release Channel/chemistry , Sarcoplasmic Reticulum/metabolism
15.
Biochem J ; 379(Pt 1): 161-72, 2004 Apr 01.
Article in English | MEDLINE | ID: mdl-14678014

ABSTRACT

We show that peptide fragments of the dihydropyridine receptor II-III loop alter cardiac RyR (ryanodine receptor) channel activity in a cytoplasmic Ca2+-dependent manner. The peptides were AC (Thr-793-Ala-812 of the cardiac dihydropyridine receptor), AS (Thr-671-Leu-690 of the skeletal dihydropyridine receptor), and a modified AS peptide [AS(D-R18)], with an extended helical structure. The peptides added to the cytoplasmic side of channels in lipid bilayers at > or = 10 nM activated channels when the cytoplasmic [Ca2+] was 100 nM, but either inhibited or did not affect channel activity when the cytoplasmic [Ca2+] was 10 or 100 microM. Both activation and inhibition were independent of bilayer potential. Activation by AS, but not by AC or AS(D-R18), was reduced at peptide concentrations >1 mM in a voltage-dependent manner (at +40 mV). In control experiments, channels were not activated by the scrambled AS sequence (ASS) or skeletal II-III loop peptide (NB). Resting Ca2+ release from cardiac sarcoplasmic reticulum was not altered by peptide AC, but Ca2+-induced Ca2+ release was depressed. Resting and Ca2+-induced Ca2+ release were enhanced by both the native and modified AS peptides. NMR revealed (i) that the structure of peptide AS(D-R18) is not influenced by [Ca2+] and (ii) that peptide AC adopts a helical structure, particularly in the region containing positively charged residues. This is the first report of specific functional interactions between dihydropyridine receptor A region peptides and cardiac RyR ion channels in lipid bilayers.


Subject(s)
Calcium Channels, L-Type/physiology , Calcium/metabolism , Peptide Fragments/pharmacology , Ryanodine Receptor Calcium Release Channel/drug effects , Sarcoplasmic Reticulum/drug effects , Amino Acid Sequence , Animals , Calcium Channels, L-Type/chemistry , Dose-Response Relationship, Drug , Ion Transport , Lipid Bilayers , Macromolecular Substances , Microsomes/drug effects , Microsomes/metabolism , Molecular Sequence Data , Muscle Contraction , Muscle Proteins/chemistry , Muscle Proteins/physiology , Muscle, Skeletal/chemistry , Myocardium/chemistry , Protein Structure, Secondary , Rabbits , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...