Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Invest Dermatol ; 139(2): 412-421, 2019 02.
Article in English | MEDLINE | ID: mdl-30287285

ABSTRACT

PAR2 has been proposed to contribute to lesion formation and intense itch in atopic dermatitis. Here, we tested the ability of a cell-penetrating pepducin, PZ-235, to mitigate the potentially deleterious effects of PAR2 in models of atopic dermatitis. PZ-235 significantly inhibited PAR2-mediated expression of inflammatory factors NF-κB, TSLP, TNF-α, and differentiation marker K10 by 94%-98% (P < 0.001) in human keratinocytes and suppressed IL-4 and IL-13 by 68%-83% (P < 0.05) in mast cells. In delayed pepducin treatment models of oxazolone- and DNFB-induced dermatitis, PZ-235 significantly attenuated skin thickening by 43%-100% (P < 0.01) and leukocyte crusting by 57% (P < 0.05), and it inhibited ex vivo chemotaxis of leukocytes toward PAR2 agonists. Daily PZ-235 treatment of filaggrin-deficient mice exposed to dust mite allergens for 8 weeks significantly suppressed total leukocyte and T-cell infiltration by 50%-68%; epidermal thickness by 60%-77%; and skin thickening, scaling, excoriation, and total lesion severity score by 46%-56%. PZ-235 significantly reduced itching caused by wasp venom peptide degranulation of mast cells in mice by 51% (P < 0.05), which was comparable to the protective effects conferred by PAR2 deficiency. Taken together, these results suggest that a PAR2 pepducin may confer broad therapeutic benefits as a disease-modifying treatment for atopic dermatitis and itch.


Subject(s)
Cell-Penetrating Peptides/pharmacology , Dermatitis, Atopic/drug therapy , Pruritus/drug therapy , Receptor, PAR-2/antagonists & inhibitors , Animals , Cell-Penetrating Peptides/therapeutic use , Dermatitis, Atopic/etiology , Dermatitis, Atopic/pathology , Disease Models, Animal , Drug Evaluation, Preclinical , Filaggrin Proteins , Humans , Keratinocytes , Male , Mice , Pruritus/etiology , Pruritus/pathology , Receptor, PAR-2/immunology , Receptor, PAR-2/metabolism
2.
Blood ; 128(1): 93-103, 2016 07 07.
Article in English | MEDLINE | ID: mdl-27073223

ABSTRACT

Dematin is a relatively low abundance actin binding and bundling protein associated with the spectrin-actin junctions of mature erythrocytes. Primary structure of dematin includes a loosely folded core domain and a compact headpiece domain that was originally identified in villin. Dematin's actin binding properties are regulated by phosphorylation of its headpiece domain by cyclic adenosine monophosphate-dependent protein kinase. Here, we used a novel gene disruption strategy to generate the whole body dematin gene knockout mouse model (FLKO). FLKO mice, while born at a normal Mendelian ratio, developed severe anemia and exhibited profound aberrations of erythrocyte morphology and membrane stability. Having no apparent effect on primitive erythropoiesis, FLKO mice show significant enhancement of erythroblast enucleation during definitive erythropoiesis. Using membrane protein analysis, domain mapping, electron microscopy, and dynamic deformability measurements, we investigated the mechanism of membrane instability in FLKO erythrocytes. Although many membrane and cytoskeletal proteins remained at their normal levels, the major peripheral membrane proteins spectrin, adducin, and actin were greatly reduced in FLKO erythrocytes. Our results demonstrate that dematin plays a critical role in maintaining the fundamental properties of the membrane cytoskeleton complex.


Subject(s)
Anemia, Hemolytic , Cytoskeletal Proteins/genetics , Cytoskeleton , Erythrocyte Membrane , Gene Deletion , Anemia, Hemolytic/genetics , Anemia, Hemolytic/metabolism , Anemia, Hemolytic/pathology , Animals , Calmodulin-Binding Proteins/genetics , Calmodulin-Binding Proteins/metabolism , Cytoskeletal Proteins/metabolism , Cytoskeleton/genetics , Cytoskeleton/metabolism , Cytoskeleton/pathology , Erythrocyte Membrane/genetics , Erythrocyte Membrane/metabolism , Erythrocyte Membrane/pathology , Female , Male , Mice , Mice, Knockout , Spectrin/genetics , Spectrin/metabolism
3.
J Immunol ; 192(11): 5130-9, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24760147

ABSTRACT

Mast cells play a central role in allergy through secretion of both preformed and newly synthesized mediators. Mast cell mediator secretion is controlled by a complex network of signaling events. Despite intensive studies, signaling pathways in the regulation of mast cell mediator secretion remain incompletely defined. In this study, we examined the role of calpain in IgE-dependent mast cell activation. IgE-mediated activation of mouse bone marrow-derived mast cells enhanced calpain activity. Inhibition of calpain activity by a number of calpain inhibitors reduced IgE-mediated mast cell degranulation both in vitro and in vivo. Calpain inhibitors blocked IgE-mediated TNF and IL-6 production in vitro and reduced late-phase allergic response in vivo. Importantly, mouse calpain-1 null bone marrow-derived mast cells showed reduced IgE-mediated mast cell degranulation in vitro and in vivo, diminished cytokine and chemokine production in vitro, and impaired late-phase allergic response in vivo. Further studies revealed that calpain-1 deficiency led to specific attenuation of IκB-NF-κB pathway and IKK-SNAP23 pathway, whereas calcium flux, MAPK, Akt, and NFAT pathway proceed normally in IgE-activated calpain-1 null mast cells. Thus, calpain-1 is identified as a novel regulator in IgE-mediated mast cell activation and could serve as a potential therapeutic target for the management of allergic inflammation.


Subject(s)
Bone Marrow Cells/immunology , Calpain/immunology , Cell Degranulation/immunology , Hypersensitivity/immunology , Immunoglobulin E/immunology , Mast Cells/immunology , Animals , Bone Marrow Cells/pathology , Calpain/genetics , Cell Degranulation/genetics , Hypersensitivity/genetics , Hypersensitivity/pathology , I-kappa B Kinase/genetics , I-kappa B Kinase/immunology , Immunoglobulin E/genetics , Interleukin-6/genetics , Interleukin-6/immunology , Mast Cells/pathology , Mice , Mice, Mutant Strains , NFATC Transcription Factors/genetics , NFATC Transcription Factors/immunology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , Qb-SNARE Proteins/genetics , Qb-SNARE Proteins/immunology , Qc-SNARE Proteins/genetics , Qc-SNARE Proteins/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
4.
J Biol Chem ; 288(19): 13325-36, 2013 May 10.
Article in English | MEDLINE | ID: mdl-23519467

ABSTRACT

BACKGROUND: Inactivation of integrin αIIbß3 reverses platelet aggregate formation upon coagulation. RESULTS AND CONCLUSION: Platelets from patient (Scott) and mouse (Capn1(-/-) and Ppif(-/-)) blood reveal a dual mechanism of αIIbß3 inactivation: by calpain-2 cleavage of integrin-associated proteins and by cyclophilin D/TMEM16F-dependent phospholipid scrambling. SIGNIFICANCE: These data provide novel insight into the switch mechanisms from aggregating to procoagulant platelets. Aggregation of platelets via activated integrin αIIbß3 is a prerequisite for thrombus formation. Phosphatidylserine-exposing platelets with a key role in the coagulation process disconnect from a thrombus by integrin inactivation via an unknown mechanism. Here we show that αIIbß3 inactivation in procoagulant platelets relies on a sustained high intracellular Ca(2+), stimulating intracellular cleavage of the ß3 chain, talin, and Src kinase. Inhibition of calpain activity abolished protein cleavage, but only partly suppressed αIIbß3 inactivation. Integrin αIIbß3 inactivation was unchanged in platelets from Capn1(-/-) mice, suggesting a role of the calpain-2 isoform. Scott syndrome platelets, lacking the transmembrane protein TMEM16F and having low phosphatidylserine exposure, displayed reduced αIIbß3 inactivation with the remaining activity fully dependent on calpain. In platelets from Ppif(-/-) mice, lacking mitochondrial permeability transition pore (mPTP) formation, agonist-induced phosphatidylserine exposure and αIIbß3 inactivation were reduced. Treatment of human platelets with cyclosporin A gave a similar phenotype. Together, these data point to a dual mechanism of αIIbß3 inactivation via calpain(-2) cleavage of integrin-associated proteins and via TMEM16F-dependent phospholipid scrambling with an assistant role of mPTP formation.


Subject(s)
Blood Platelets/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Animals , Anoctamins , Blood Platelets/drug effects , Blood Platelets/physiology , CD36 Antigens/metabolism , Calcium Signaling , Calpain/antagonists & inhibitors , Calpain/metabolism , Cell Membrane/metabolism , Crotalid Venoms/pharmacology , Dipeptides/pharmacology , Humans , Lectins, C-Type , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Phosphatidylserines/metabolism , Phospholipid Transfer Proteins/metabolism , Platelet Aggregation , Protein Structure, Quaternary , Proteolysis , Talin/metabolism , Thrombin/pharmacology , Thrombin/physiology , src-Family Kinases/metabolism
5.
FASEB J ; 27(2): 750-9, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23085996

ABSTRACT

Sickle cell disease (SCD) is a globally distributed hereditary red blood cell (RBC) disorder. One of the hallmarks of SCD is the presence of circulating dense RBCs, which are important in SCD-related clinical manifestations. In human dense sickle cells, we found reduced calpastatin activity and protein expression compared to either healthy RBCs or unfractionated sickle cells, suggesting an imbalance between activator and inhibitor of calpain-1 in favor of activator in dense sickle cells. Calpain-1 is a nonlysosomal cysteine proteinase that modulates multiple cell functions through the selective cleavage of proteins. To investigate the relevance of this observation in vivo, we evaluated the effects of the orally active inhibitor of calpain-1, BDA-410 (30 mg/kg/d), on RBCs from SAD mice, a mouse model for SCD. In SAD mice, BDA-410 improved RBC morphology, reduced RBC density (D(20); from 1106 ± 0.001 to 1100 ± 0.001 g/ml; P<0.05) and increased RBC-K(+) content (from 364 ± 10 to 429 ± 12.3 mmol/kg Hb; P<0.05), markedly reduced the activity of the Ca(2+)-activated K(+)channel (Gardos channel), and decreased membrane association of peroxiredoxin-2. The inhibitory effect of calphostin C, a specific inhibitor of protein kinase C (PKC), on the Gardos channel was eliminated after BDA-410 treatment, which suggests that calpain-1 inhibition affects the PKC-dependent fraction of the Gardos channel. BDA-410 prevented hypoxia-induced RBC dehydration and K(+) loss in SAD mice. These data suggest a potential role of BDA-410 as a novel therapeutic agent for treatment of SCD.


Subject(s)
Anemia, Sickle Cell/blood , Anemia, Sickle Cell/drug therapy , Calpain/antagonists & inhibitors , Erythrocytes, Abnormal/drug effects , Erythrocytes, Abnormal/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/blood , Anemia, Sickle Cell/genetics , Animals , Calcium-Binding Proteins/blood , Calpain/blood , Cysteine Proteinase Inhibitors/pharmacology , Dehydration/blood , Dehydration/prevention & control , Disease Models, Animal , Humans , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Mice , Mice, Mutant Strains , Mice, Transgenic , Sulfonamides/pharmacology
6.
J Biol Chem ; 287(49): 41218-31, 2012 Nov 30.
Article in English | MEDLINE | ID: mdl-23060452

ABSTRACT

Dematin is a broadly expressed membrane cytoskeletal protein that has been well characterized in erythrocytes and to a lesser extent in non-erythroid cells. However, dematin's function in platelets is not known. Here, we show that dematin is abundantly expressed in both human and mouse platelets. Platelets harvested from the dematin headpiece knock-out (HPKO) mouse model exhibit a striking defect in the mobilization of calcium in response to multiple agonists of platelet activation. The reduced calcium mobilization in HPKO platelets is associated with concomitant inhibition of platelet aggregation and granule secretion. Integrin α(IIb)ß(3) activation in response to agonists is attenuated in the HPKO platelets. The mutant platelets show nearly normal spreading on fibrinogen and an unaltered basal cAMP level; however, the clot retraction was compromised in the mutant mice. Immunofluorescence analysis indicated that dematin is present both at the dense tubular system and plasma membrane fractions of platelets. Proteomic analysis of dematin-associated proteins in human platelets identified inositol 1,4,5-trisphosphate 3-kinase isoform B (IP3KB) as a binding partner, which was confirmed by immunoprecipitation analysis. IP3KB, a dense tubular system protein, is a major regulator of calcium homeostasis. Loss of the dematin headpiece resulted in a decrease of IP3KB at the membrane and increased levels of IP3KB in the cytosol. Collectively, these findings unveil dematin as a novel regulator of internal calcium mobilization in platelets affecting multiple signaling and cytoskeletal functions. Implications of a conserved role of dematin in the regulation of calcium homeostasis in other cell types will be discussed.


Subject(s)
Blood Platelets/metabolism , Calcium/chemistry , Microfilament Proteins/chemistry , Animals , Blood Platelets/cytology , Calcium Signaling , Cardiovascular Diseases/metabolism , Cytoskeletal Proteins/metabolism , Cytosol/chemistry , Cytosol/metabolism , Erythrocyte Membrane/metabolism , Hemostasis , Humans , Mice , Models, Biological , Mutation , Platelet Activation , Protein Structure, Tertiary , Signal Transduction
7.
J Biol Chem ; 287(16): 13182-93, 2012 Apr 13.
Article in English | MEDLINE | ID: mdl-22367208

ABSTRACT

Calpains are calcium-regulated cysteine proteases that have been implicated in the regulation of cell death pathways. Here, we used our calpain-1 null mouse model to evaluate the function of calpain-1 in neural degeneration following a rodent model of traumatic brain injury. In vivo, calpain-1 null mice show significantly less neural degeneration and apoptosis and a smaller contusion 3 days post-injury than wild type littermates. Protection from traumatic brain injury corroborated with the resistance of calpain-1 neurons to apoptosis induced by oxidative stress. Biochemical analysis revealed that caspase-3 activation, extracellular calcium entry, mitochondrial membrane permeability, and release of apoptosis-inducing factor from mitochondria are partially blocked in the calpain-1 null neurons. These findings suggest that the calpain-1 knock-out mice may serve as a useful model system for neuronal protection and apoptosis in traumatic brain injury and other neurodegenerative disorders in which oxidative stress plays a role.


Subject(s)
Apoptosis/physiology , Brain Injuries/metabolism , Brain Injuries/pathology , Calpain/genetics , Calpain/metabolism , Oxidative Stress/physiology , Animals , Apoptosis Inducing Factor/metabolism , Calcium/metabolism , Caspase 3/metabolism , Disease Models, Animal , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Membranes/metabolism , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Neurons/metabolism , Neurons/pathology , RNA, Small Interfering/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...