Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 92
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mol Pharmacol ; 99(4): 277-285, 2021 04.
Article in English | MEDLINE | ID: mdl-33436521

ABSTRACT

Acetaminophen (APAP) is a commonly used pain and fever reliever but is also the most frequent cause of drug-induced liver injury. The mechanism pertaining acetaminophen toxicity has been well documented, whereas mechanisms of hepatotoxicity are not well established. Serine (or cysteine) peptidase inhibitor, clade A, member 3N (SerpinA3N), a serine protease inhibitor, is synthesized in the liver but the role of SerpinA3N in relation to APAP-induced liver injury is not known. Wild-type and hepatocyte-specific SerpinA3N knockout (HKO) mice were injected intraperitoneally with a single dose of PBS or APAP (400 mg/kg) for 12 hours, and markers of liver injury, cell death, and inflammation were assessed. SerpinA3N expression was highly induced in mice with APAP overdose. SerpinA3N HKO mice had diminished liver injury and necrosis as shown by lower alanine aminotransferase and interleukin-6 levels, accompanied by suppressed inflammatory cytokines and reduced neutrophil infiltration. The reduced oxidative stress was associated with enhanced antioxidant enzyme capabilities. Taken together, hepatocyte SerpinA3N deficiency reduced APAP-induced liver injury by ameliorating inflammation and modulating the 5' AMP-activated protein kinase-unc-51-like autophagy activating kinase 1 signaling pathway. Our study provides novel insights into a potential role for SerpinA3N in APAP-induced liver injury. SIGNIFICANCE STATEMENT: Our studies indicate that serine (or cysteine) peptidase inhibitor, clade A, member 3N (SerpinA3N) may have a pathophysiological role in modulating acetaminophen (APAP)-induced liver injury. More specifically, mice with hepatic deletion of SerpinA3N suppressed inflammation and liver injury to reduce APAP-induced hepatotoxicity. Controlling the inflammatory response offers possible approaches for novel therapeutics; therefore, understanding the pathophysiological role of SerpinA3N in inducing liver injury may add to the development of more efficacious treatments.


Subject(s)
Acetaminophen/toxicity , Acute-Phase Proteins/deficiency , Analgesics, Non-Narcotic/toxicity , Chemical and Drug Induced Liver Injury/metabolism , Serpins/deficiency , Acute-Phase Proteins/genetics , Animals , Chemical and Drug Induced Liver Injury/genetics , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Serpins/genetics
2.
Exp Eye Res ; 198: 108121, 2020 09.
Article in English | MEDLINE | ID: mdl-32721425

ABSTRACT

The SERPINF1 gene encodes pigment epithelium-derived factor (PEDF), a member of the serpin superfamily with neurotrophic and antiangiogenic properties in the retina. We hypothesized that absence of PEDF would lead to increased stress-associated retinal degeneration in Serpinf1 null mice. Accordingly, using a Serpinf1 null mouse model, we investigated the impact of PEDF absence on retinal morphology, and susceptibility to induced and inherited retinal degeneration. We studied the pattern of Serpinf1 expression in the mouse retina layers. PEDF protein was detected by western blotting. Transmission electron microscopy was performed on mouse retina. Serpinf1 null mice and wild type littermates were injected with NaIO3 (30 mg/kg body weight) intraperitonially. At post-injection day 1, 3, 4, 6 and 8 mice were euthanized, and eyes were enucleated. Serpinf1 null and rd10 double mutant mice were generated and their eyes enucleated at different time points from post-natal day 15 to post-natal day 28. Enucleated eyes were processed for hematoxylin and eosin staining and histopathological evaluations. We found that Serpinf1 was expressed in the retinal pigment epithelium, in the inner nuclear layer and in the ganglion cell layer, but undetectable in the outer nuclear layer of wild type mice. Plasma PEDF protein levels were undetectable in Serpinf1 null animals. RPE atrophy and retinal thinning were observed in NaIO3-treated wild type mice that progressed with time post-injection. NaIO3-treated Serpinf1 null mice showed comparatively better retinal morphology than wild type mice at day 4 post-injection. However, the absence of PEDF in Serpinf1 null x rd10 mice increased the susceptibility to retinal degeneration relative to that of rd10 mice. We concluded that histopathological evaluation of retinas lacking PEDF showed that removal of the Serpinf1 gene may activate PEDF-independent compensatory mechanisms to protect the retina against oxidative stress, while it increases the susceptibility to degenerate the retina in inherited retinal degeneration models.


Subject(s)
Nerve Growth Factors/deficiency , Retinal Degeneration/metabolism , Serpins/deficiency , Animals , Blotting, Western , Disease Models, Animal , Disease Progression , Eye Proteins/genetics , Eye Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/pathology , Retinal Degeneration/pathology , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Serpins/genetics , Serpins/metabolism
3.
Am J Physiol Cell Physiol ; 314(4): C456-C472, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29351407

ABSTRACT

Age-related macular degeneration (AMD) is the leading cause of vision loss among the elderly population, and is associated with severe macular degeneration and choroidal neovascularization (CNV). Although the pathogenesis of AMD is associated with choroidal dysfunction and CNV, the detailed underlying mechanisms remain unresolved. Altered production of pigment epithelium-derived factor (PEDF), a neuroprotective and antiangiogenic factor, contributes to CNV. Furthermore, exogenous PEDF mitigates angiogenesis in preclinical CNV models. How PEDF expression affects choroidal endothelial cell (ChEC) function is unknown. Here we isolated ChECs from PEDF+/+ and PEDF-deficient (PEDF-/-) mice and determined the impact of PEDF expression on the proangiogenic and pro-inflammatory properties of ChECs. We showed that PEDF expression significantly affects the proliferation, migration, adhesion, and oxidative and inflammatory state of ChECs. The PEDF-/- ChECs were, however, more sensitive to H2O2 challenge and exhibited increased rate of apoptosis and oxidative stress. We also observed a significant increase in production of cytokines with a primary role in inflammation and angiogenesis including vascular endothelial growth factor (VEGF) and osteopontin, and a reprograming of chemokines and cytokines expression profiles in PEDF-/- ChECs. Collectively, our results indicate that PEDF expression has a significant impact on oxidative and inflammatory properties of ChECs, whose alteration could contribute to pathogenesis of chronic inflammatory diseases including exudative AMD.


Subject(s)
Choroid/blood supply , Choroidal Neovascularization/metabolism , Cytokines/metabolism , Endothelial Cells/metabolism , Eye Proteins/metabolism , Inflammation Mediators/metabolism , Macular Degeneration/metabolism , Nerve Growth Factors/metabolism , Oxidative Stress , Serpins/metabolism , Animals , Apoptosis , Cell Adhesion , Cell Movement , Cell Proliferation , Cells, Cultured , Choroidal Neovascularization/genetics , Choroidal Neovascularization/pathology , Endothelial Cells/pathology , Eye Proteins/genetics , Macular Degeneration/genetics , Macular Degeneration/pathology , Mice, Inbred C57BL , Mice, Knockout , Nerve Growth Factors/deficiency , Nerve Growth Factors/genetics , Osteopontin/metabolism , Serpins/deficiency , Serpins/genetics , Vascular Endothelial Growth Factor A/metabolism
4.
J Cell Mol Med ; 22(9): 4387-4398, 2018 09.
Article in English | MEDLINE | ID: mdl-29992759

ABSTRACT

Kallistatin, a plasma protein, protects against vascular and organ injury. This study is aimed to investigate the role and mechanism of kallistatin in endothelial senescence. Kallistatin inhibited H2 O2 -induced senescence in human endothelial cells, as indicated by reduced senescence-associated-ß-galactosidase activity, p16INK4a and plasminogen activator inhibitor-1 expression, and elevated telomerase activity. Kallistatin blocked H2 O2 -induced superoxide formation, NADPH oxidase levels and VCAM-1, ICAM-1, IL-6 and miR-34a synthesis. Kallistatin reversed H2 O2 -mediated inhibition of endothelial nitric oxide synthase (eNOS), SIRT1, catalase and superoxide dismutase (SOD)-2 expression, and kallistatin alone stimulated the synthesis of these antioxidant enzymes. Moreover, kallistatin's anti-senescence and anti-oxidant effects were attributed to SIRT1-mediated eNOS pathway. Kallistatin, via interaction with tyrosine kinase, up-regulated Let-7g, whereas Let-7g inhibitor abolished kallistatin's effects on miR-34a and SIRT1/eNOS synthesis, leading to inhibition of senescence, oxidative stress and inflammation. Furthermore, lung endothelial cells isolated from endothelium-specific kallistatin knockout mice displayed marked reduction in mouse kallistatin levels. Kallistatin deficiency in mouse endothelial cells exacerbated senescence, oxidative stress and inflammation compared to wild-type mouse endothelial cells, and H2 O2 treatment further magnified these effects. Kallistatin deficiency caused marked reduction in Let-7g, SIRT1, eNOS, catalase and SOD-1 mRNA levels, and elevated miR-34a synthesis in mouse endothelial cells. These findings indicate that endogenous kallistatin through novel mechanisms protects against endothelial senescence by modulating Let-7g-mediated miR-34a-SIRT1-eNOS pathway.


Subject(s)
Endothelial Cells/metabolism , MicroRNAs/genetics , Nitric Oxide Synthase Type III/genetics , Serpins/genetics , Sirtuin 1/genetics , Animals , Catalase/genetics , Catalase/metabolism , Cellular Senescence/drug effects , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Endothelial Cells/cytology , Endothelial Cells/drug effects , Gene Expression Regulation , HEK293 Cells , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hydrogen Peroxide/pharmacology , Lung/cytology , Lung/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/metabolism , Nitric Oxide Synthase Type III/metabolism , Primary Cell Culture , Serpin E2/genetics , Serpin E2/metabolism , Serpins/deficiency , Serpins/pharmacology , Signal Transduction , Sirtuin 1/metabolism , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism , Telomerase/genetics , Telomerase/metabolism , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
5.
Learn Mem ; 24(12): 650-659, 2017 12.
Article in English | MEDLINE | ID: mdl-29142062

ABSTRACT

The serine protease inhibitor neuroserpin regulates the activity of tissue-type plasminogen activator (tPA) in the nervous system. Neuroserpin expression is particularly prominent at late stages of neuronal development in most regions of the central nervous system (CNS), whereas it is restricted to regions related to learning and memory in the adult brain. The physiological expression pattern of neuroserpin, its high degree of colocalization with tPA within the CNS, together with its dysregulation in neuropsychiatric disorders, suggest a role in formation and refinement of synapses. In fact, studies in cell culture and mice point to a role for neuroserpin in dendritic branching, spine morphology, and modulation of behavior. In this study, we investigated the physiological role of neuroserpin in the regulation of synaptic density, synaptic plasticity, and behavior in neuroserpin-deficient mice. In the absence of neuroserpin, mice show a significant decrease in spine-synapse density in the CA1 region of the hippocampus, while expression of the key postsynaptic scaffold protein PSD-95 is increased in this region. Neuroserpin-deficient mice show decreased synaptic potentiation, as indicated by reduced long-term potentiation (LTP), whereas presynaptic paired-pulse facilitation (PPF) is unaffected. Consistent with altered synaptic plasticity, neuroserpin-deficient mice exhibit cognitive and sociability deficits in behavioral assays. However, although synaptic dysfunction is implicated in neuropsychiatric disorders, we do not detect alterations in expression of neuroserpin in fusiform gyrus of autism patients or in dorsolateral prefrontal cortex of schizophrenia patients. Our results identify neuroserpin as a modulator of synaptic plasticity, and point to a role for neuroserpin in learning and memory.


Subject(s)
Gene Expression Regulation/genetics , Neuronal Plasticity/genetics , Neuropeptides/deficiency , Serine Proteinase Inhibitors/metabolism , Serpins/deficiency , Social Behavior , Synapses/genetics , Adolescent , Adult , Animals , Autistic Disorder/genetics , Autistic Disorder/pathology , Autistic Disorder/psychology , Child , Exploratory Behavior/physiology , Hippocampus/physiology , Hippocampus/ultrastructure , Humans , Long-Term Potentiation/genetics , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Neuropeptides/genetics , Serpins/genetics , Synapses/physiology , Synapses/ultrastructure , Synaptosomal-Associated Protein 25/metabolism , Young Adult , Neuroserpin
6.
Am J Physiol Cell Physiol ; 313(4): C405-C420, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28747334

ABSTRACT

Pigment epithelium-derived factor (PEDF) is an endogenous inhibitor of angiogenesis. Although various ocular cell types including retinal endothelial cells (EC) produce PEDF, we know very little about cell autonomous effects of PEDF in these cell types. Here we determined how PEDF expression affects retinal EC proangiogenic properties. Retinal EC were prepared from wild-type (PEDF+/+) and PEDF-deficient (PEDF-/-) mice. The identity of EC was confirmed by staining for specific markers including vascular endothelial cadherin, CD31, and B4-lectin. Retinal EC also expressed VEGF receptor 1 and endoglin, as well as ICAM-1, ICAM-2, and VCAM-1. PEDF-/- retinal EC were more proliferative, less apoptotic when challenged with H2O2, less migratory, and less adherent compared with PEDF+/+ EC. These changes could be associated, at least in part, with increased levels of tenascin-C, fibronectin, thrombospondin-1 and collagen IV, and lower amounts of osteopontin. PEDF-/- EC also exhibited alterations in expression of a number of integrins including α2, αv, ß1, ß8, and αvß3, and cell-cell adhesion molecules including CD31, zonula occluden-1, and occludin. These observations correlated with attenuation of capillary morphogenesis and increased levels of oxidative stress in PEDF-/- EC. PEDF-/- EC also produced lower levels of VEGF compared with PEDF+/+ cells. Thus, PEDF deficiency has a significant impact on retinal EC adhesion and migration, perhaps through altered production of extracellular matrix and junctional proteins in response to increased oxidative stress affecting their proangiogenic activity.


Subject(s)
Cell Adhesion , Endothelial Cells/metabolism , Eye Proteins/metabolism , Neovascularization, Physiologic , Nerve Growth Factors/metabolism , Retinal Vessels/metabolism , Serpins/metabolism , Animals , Apoptosis , Cell Adhesion Molecules/metabolism , Cell Movement , Cell Proliferation , Cells, Cultured , Endoglin/metabolism , Endothelial Cells/pathology , Extracellular Matrix/metabolism , Eye Proteins/genetics , Female , Genotype , Humans , Integrins/metabolism , Male , Mice, Knockout , Nerve Growth Factors/deficiency , Nerve Growth Factors/genetics , Oxidative Stress , Phenotype , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Retinal Vessels/pathology , Serpins/deficiency , Serpins/genetics , Signal Transduction , Tight Junction Proteins/metabolism , Time Factors , Transfection , Vascular Endothelial Growth Factor Receptor-1/metabolism
7.
Kidney Int ; 91(3): 642-657, 2017 03.
Article in English | MEDLINE | ID: mdl-27914705

ABSTRACT

Pigment epithelium-derived factor (PEDF) expression is downregulated in the kidneys of diabetic rats, and delivery of PEDF suppressed renal fibrotic factors in these animals. PEDF has multiple functions including anti-angiogenic, anti-inflammatory and antifibrotic activities. Since the mechanism underlying its antifibrotic effect remains unclear, we studied this in several murine models of renal disease. Renal PEDF levels were significantly reduced in genetic models of type 1 and type 2 diabetes (Akita and db/db, respectively), negatively correlating with Wnt signaling activity in the kidneys. In unilateral ureteral obstruction, an acute renal injury model, there were significant decreases of renal PEDF levels. The kidneys of PEDF knockout mice with ureteral obstruction displayed exacerbated expression of fibrotic and inflammatory factors, oxidative stress, tubulointerstitial fibrosis, and tubule epithelial cell apoptosis, compared to the kidneys of wild-type mice with obstruction. PEDF knockout enhanced Wnt signaling activation induced by obstruction, while PEDF inhibited the Wnt pathway-mediated fibrosis in primary renal proximal tubule epithelial cells. Additionally, oxidative stress was aggravated in renal proximal tubule epithelial cells isolated from knockout mice and suppressed by PEDF treatment of renal proximal tubule epithelial cells. PEDF also reduced oxidation-induced apoptosis in renal proximal tubule epithelial cells. Thus, the renoprotective effects of PEDF are mediated, at least partially, by inhibition of the Wnt pathway. Hence, restoration of renal PEDF levels may have therapeutic potential for renal fibrosis.


Subject(s)
Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 2/metabolism , Epithelial Cells/metabolism , Eye Proteins/metabolism , Kidney Diseases/prevention & control , Kidney Tubules, Proximal/metabolism , Nerve Growth Factors/metabolism , Serpins/metabolism , Ureteral Obstruction/metabolism , Wnt Signaling Pathway , Animals , Apoptosis , Axin Protein/genetics , Axin Protein/metabolism , Cell Line , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Diabetic Nephropathies/genetics , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Diabetic Nephropathies/prevention & control , Disease Models, Animal , Epithelial Cells/pathology , Eye Proteins/genetics , Fibrosis , Gene Expression Regulation , Genetic Predisposition to Disease , Humans , Inflammation Mediators/metabolism , Kidney Diseases/genetics , Kidney Diseases/metabolism , Kidney Diseases/pathology , Kidney Tubules, Proximal/pathology , Mice, Inbred C57BL , Mice, Knockout , Nerve Growth Factors/deficiency , Nerve Growth Factors/genetics , Oxidative Stress , Phenotype , Serpins/deficiency , Serpins/genetics , Time Factors , Transfection , Ureteral Obstruction/complications , Ureteral Obstruction/genetics , Ureteral Obstruction/pathology
8.
BMC Musculoskelet Disord ; 18(1): 39, 2017 01 25.
Article in English | MEDLINE | ID: mdl-28122611

ABSTRACT

BACKGROUND: Inflammation is a major cause of cartilage destruction and leads to the imbalance of metabolic activities in the arthritic joint. Pigment epithelium-derived factor (PEDF) has been reported to have both pro- and anti-inflammatory activities in various cell types and to be upregulated in the arthritic joint, but its role in joint destruction is unclear. Our aim was to investigate the role of PEDF in cartilage degeneration under inflammatory conditions. METHODS: PEDF was ectopically expressed in primary human articular chondrocytes, and catabolic gene expression and protein secretion in response to the pro-inflammatory cytokine interleukin 1 beta (IL-1ß) were evaluated. Metatarsal bones from PEDF-deficient and wild type mice were cultured in the presence or absence of IL-1ß. Cartilage matrix integrity and matrix metalloproteinases MMP-1, MMP-3, and MMP-13 were evaluated. PEDF-deficient and wild type mice were evaluated in the monosodium iodoacetate (MIA) inflammatory joint destruction animal model to determine the role of PEDF in inflammatory arthritis in vivo. Student's t-tests and Mann-Whitney tests were employed where appropriate, for parametric and non-parametric data, respectively. RESULTS: We showed that PEDF protein levels were higher in human osteoarthritis samples compared to normal samples. We demonstrated that ectopic PEDF expression in primary human articular chondrocytes exacerbated catabolic gene expression in the presence of IL-1ß. In whole bone organ cultures, IL-1ß induced MMP-1, MMP-3 and MMP-13 protein production, and caused significant cartilage matrix loss. Interestingly, Toluidine Blue staining showed that PEDF-deficient bones from 29 week old animals, but not 10 week old animals, had reduced matrix loss in response to IL-1ß compared to their wild type counterparts. In addition, PEDF-deficiency in 29 week old animals preserved matrix integrity and protected against cell loss in the MIA joint destruction model in vivo. CONCLUSION: We conclude that PEDF exacerbates cartilage degeneration in an age-dependent manner under an inflammatory setting. This is the first study identifying a specific role for PEDF in joint inflammation and highlights the multi-faceted activities of PEDF.


Subject(s)
Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Eye Proteins/biosynthesis , Nerve Growth Factors/biosynthesis , Serpins/biosynthesis , Age Factors , Aged , Animals , Cells, Cultured , Female , Humans , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Knockout , Middle Aged , Nerve Growth Factors/deficiency , Serpins/deficiency
9.
Cancer Sci ; 107(5): 619-28, 2016 May.
Article in English | MEDLINE | ID: mdl-26892864

ABSTRACT

An increasingly accepted concept is that the progression of colorectal cancer is accompanied by epithelial-mesenchymal transition (EMT). In our study, in order to characterize the properties of EMT in 16 colorectal cancer cell lines, the cells were first orthotopically implanted into nude mice, and the tumors in vivo, as well as cells cultured in vitro, were immunostained for EMT markers. The immunostaining revealed that seven of the cells had an epithelial phenotype with a high expression of E-cadherin, whereas other cells showed opposite patterns, such as a high expression of vimentin (CX-1, COLO205, CloneA, HCT116, and SW48). Among the cells expressing vimentin, some expressed vimentin in the orthotopic tumors but not in the cultured cells (SW480, SW620, and COLO320). We evaluated these findings in combination with microarray analyses, and selected five genes: CHST11, SERPINI1, AGR2, FBP1, and FOXA1. Next, we downregulated the expression of SERPINI1 with siRNA in the cells, the results of which showed reverse-EMT changes at the protein level and in the cellular morphology. Along with immunohistochemical analyses, we confirmed the effect of the intracellular and secreted SERPINI1 protein of SW620 cells, which supported the importance of SERPINI1 in EMT. The development of therapeutic strategies targeting EMT is ongoing, including methods targeting the transforming growth factor-ß signaling pathway as well as the Wnt pathway. SERPINI1 is an important regulator of EMT. Our findings help to elucidate the signaling pathways of EMT, hopefully clarifying therapeutic pathways as well.


Subject(s)
Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Epithelial-Mesenchymal Transition , Models, Biological , Neoplasm Transplantation , Neuropeptides/metabolism , Serpins/metabolism , Animals , Cadherins/metabolism , Cell Line, Tumor , Colorectal Neoplasms/genetics , Disease Progression , Gene Knockdown Techniques , Humans , Immunohistochemistry , Male , Mice , Neuropeptides/deficiency , Neuropeptides/genetics , Oligonucleotide Array Sequence Analysis , Serpins/deficiency , Serpins/genetics , Sulfotransferases/deficiency , Sulfotransferases/genetics , Sulfotransferases/metabolism , Vimentin/metabolism , Wnt Signaling Pathway , Neuroserpin
10.
Biol Chem ; 397(9): 897-905, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27107834

ABSTRACT

Serpinb1 is an inhibitor of neutrophil granule serine proteases cathepsin G, proteinase-3 and elastase. One of its core physiological functions is to protect neutrophils from granule protease-mediated cell death. Mice lacking Serpinb1a (Sb1a-/-), its mouse ortholog, have reduced bone marrow neutrophil numbers due to cell death mediated by cathepsin G and the mice show increased susceptibility to lung infections. Here, we show that conditional deletion of Serpinb1a using the Lyz2-cre and Cebpa-cre knock-in mice effectively leads to recombination-mediated deletion in neutrophils but protein-null neutrophils were only obtained using the latter recombinase-expressing strain. Absence of Serpinb1a protein in neutrophils caused neutropenia and increased granule permeabilization-induced cell death. We then generated transgenic mice expressing human Serpinb1 in neutrophils under the human MRP8 (S100A8) promoter. Serpinb1a expression levels in founder lines correlated positively with increased neutrophil survival when crossed with Sb1a-/- mice, which had their defective neutrophil phenotype rescued in the higher expressing transgenic line. Using new conditional and transgenic mouse models, our study demonstrates the presence of a relatively low Serpinb1a protein threshold in neutrophils that is required for sustained survival. These models will also be helpful in delineating recently described functions of Serpinb1 in metabolism and cancer.


Subject(s)
Gene Deletion , Myeloid Cells/metabolism , Neutrophils/cytology , Serpins/deficiency , Serpins/genetics , Alleles , Animals , Cell Survival , Gene Knock-In Techniques , Humans , Mice, Transgenic , Recombination, Genetic
11.
Biol Chem ; 397(12): 1287-1292, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27543783

ABSTRACT

Netherton syndrome (NS) is caused by mutations in the SPINK5 gene. Several Spink5-deficient mouse models were generated to understand the mechanisms of NS in vivo. However, Spink5-deficiency in mice is associated with postnatal lethality that hampers further analysis. Here we present a viable mouse model for NS generated by mosaic inactivation of the Spink5 gene. We propose that these mice are a valuable experimental tool to study NS, especially for long-term studies evaluating potential therapeutic compounds. Furthermore, we show that mosaic inactivation of a gene using TALENs or CRISPR/Cas9 systems can be used to study lethal phenotypes in adult mice.


Subject(s)
Disease Models, Animal , Gene Silencing , Mosaicism , Netherton Syndrome/genetics , Serpins/deficiency , Serpins/genetics , Animals , Female , Male , Mice , Mice, Inbred C57BL , Phenotype , Serine Peptidase Inhibitor Kazal-Type 5
12.
Mol Genet Metab ; 117(3): 378-82, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26693895

ABSTRACT

Osteogenesis imperfecta (OI) is a group of genetic disorders characterized by bone fragility and deformity. OI type VI is unique owing to the mineralization defects observed in patient biopsies. Furthermore, it has been reported to respond less well to standard therapy with bisphosphonates [1]. Others and we have previously identified SERPINF1 mutations in patients with OI type VI. SERPINF1 encodes pigment epithelium derived factor (PEDF), a secreted collagen-binding glycoprotein that is absent in the sera of patients with OI type VI. Serpinf1 null mice show increased osteoid and decreased bone mass, and thus recapitulate the OI type VI phenotype. We tested whether restoration of circulating PEDF in the blood could correct the phenotype of OI type VI in the context of protein replacement. To do so, we utilized a helper-dependent adenoviral vector (HDAd) to express human SERPINF1 in the mouse liver and assessed whether PEDF secreted from the liver was able to rescue the bone phenotype observed in Serpinf1(-/-) mice. We confirmed that expression of SERPINF1 in the liver restored the serum level of PEDF. We also demonstrated that PEDF secreted from the liver was biologically active by showing the expected metabolic effects of increased adiposity and impaired glucose tolerance in Serpinf1(-/-) mice. Interestingly, overexpression of PEDF in vitro increased mineralization with a concomitant increase in the expression of bone gamma-carboxyglutamate protein, alkaline phosphatase and collagen, type I, alpha I, but the increased serum PEDF level did not improve the bone phenotype of Serpinf1(-/-) mice. These results suggest that PEDF may function in a context-dependent and paracrine fashion in bone homeostasis.


Subject(s)
Bone and Bones/physiology , Eye Proteins/blood , Eye Proteins/genetics , Liver/metabolism , Nerve Growth Factors/blood , Nerve Growth Factors/genetics , Osteogenesis Imperfecta/physiopathology , Osteogenesis Imperfecta/therapy , Serpins/blood , Serpins/genetics , 1-Carboxyglutamic Acid/genetics , Adenoviridae/genetics , Alkaline Phosphatase/genetics , Animals , Bone Density , Collagen Type I/genetics , Gene Transfer Techniques , Glucose Intolerance , HEK293 Cells , Homeostasis , Humans , Mice , Mice, Knockout , Mutation , Nerve Growth Factors/deficiency , Phenotype , Serpins/deficiency
13.
Am J Physiol Lung Cell Mol Physiol ; 306(7): L620-34, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24318110

ABSTRACT

Pigment epithelium-derived factor (PEDF) is a multifunctional protein with important roles in regulation of inflammation and angiogenesis. It is produced by various cell types, including endothelial cells (EC). However, the cell autonomous impact of PEDF on EC function needs further investigation. Lung EC prepared from PEDF-deficient (PEDF-/-) mice were more migratory and failed to undergo capillary morphogenesis in Matrigel compared with wild type (PEDF+/+) EC. Although no significant differences were observed in the rates of apoptosis in PEDF-/- EC compared with PEDF+/+ cells under basal or stress conditions, PEDF-/- EC proliferated at a slower rate. PEDF-/- EC also expressed increased levels of proinflammatory markers, including vascular endothelial growth factor, inducible nitric oxide synthase, vascular cell adhesion molecule-1, as well as altered cellular junctional organization, and nuclear localization of ß-catenin. The PEDF-/- EC were also more adhesive, expressed decreased levels of thrombospondin-2, tenascin-C, and osteopontin, and increased fibronectin. Furthermore, we showed lungs from PEDF-/- mice exhibited increased expression of macrophage marker F4/80, along with increased thickness of the vascular walls, consistent with a proinflammatory phenotype. Together, our data suggest that the PEDF expression makes significant contribution to modulation of the inflammatory and angiogenic phenotype of the lung endothelium.


Subject(s)
Endothelial Cells/physiology , Eye Proteins/biosynthesis , Neovascularization, Physiologic/drug effects , Nerve Growth Factors/biosynthesis , Serpins/biosynthesis , Animals , Antigens, CD/biosynthesis , Antigens, Differentiation/biosynthesis , Apoptosis , Cadherins/biosynthesis , Cell Adhesion/drug effects , Cell Movement , Cell Proliferation , Lung/blood supply , Lung/cytology , Mice , Nerve Growth Factors/deficiency , Nitric Oxide Synthase Type II/biosynthesis , Platelet Endothelial Cell Adhesion Molecule-1/biosynthesis , Serpins/deficiency , Vascular Endothelial Growth Factor A/biosynthesis , beta Catenin/biosynthesis
14.
Am J Pathol ; 183(1): 49-59, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23669344

ABSTRACT

A homozygous mutation of SERPINB6, a gene encoding an intracellular protease inhibitor, has recently been associated with post-lingual, autosomal-recessive, nonsyndromic hearing loss in humans (DFNB91). Herein, we describe the physiological changes underlying SERPINB6 deficiency by analyzing mutant mice in which the orthologous gene is replaced by enhanced green fluorescent protein. SERPINB6A is present in the neurosensory epithelium, lateral wall, and spiral limbus of the cochlea, with highest levels in the inner and outer hair cells of the organ of Corti, cells lining the inner sulcus, and supporting cells distributed along the epithelial gap junction layer to the outer sulcus. Measurements of hearing thresholds in these mice demonstrated age-related hearing loss in all homozygous-null, but not heterozygous, mice. Hearing impairment was first detected at 3 weeks of age, affecting only high frequencies before spreading to other frequencies as the mice aged. The defect is associated with progressive cellular degeneration within the cochlea. This begins with the hair cells, then involves the primary auditory neurons, and, finally, the fibrocytes in the lateral wall. These findings establish these mutant mice as a suitable model system to elucidate how SERPINB6 deficiency causes deafness in humans.


Subject(s)
Hearing Loss, Sensorineural/metabolism , Serpins/deficiency , Age Factors , Animals , Biomarkers/metabolism , Cochlea/metabolism , Cochlea/pathology , Hearing Loss, Sensorineural/pathology , Mice , Mice, Knockout , Organ of Corti/metabolism , Organ of Corti/pathology , Serpins/metabolism
15.
J Immunol ; 189(3): 1133-43, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22745374

ABSTRACT

The persistence of memory T lymphocytes confers lifelong protection from pathogens. Memory T cells survive and undergo homeostatic proliferation (HSP) in the absence of Ag, although the cell-intrinsic mechanisms by which cytokines drive the HSP of memory T cells are not well understood. In this study we report that lysosome stability limits the long-term maintenance of memory CD8(+) T cell populations. Serine protease inhibitor (Spi) 2A, an anti-apoptotic cytosolic cathepsin inhibitor, is induced by both IL-15 and IL-7. Mice deficient in Spi2A developed fewer memory phenotype CD44(hi)CD8(+) T cells with age, which underwent reduced HSP in the bone marrow. Spi2A was also required for the maintenance of central memory CD8(+) T cell populations after acute infection with lymphocytic choriomeningitis virus. Spi2A-deficient Ag-specific CD8(+) T cell populations declined more than wild-type competitors after viral infection, and they were eroded further after successive infections. Spi2A protected memory cells from lysosomal breakdown by inhibiting cathepsin B. The impaired maintenance of Spi2A-deficient memory CD8(+) T cells was rescued by concomitant cathepsin B deficiency, demonstrating that cathepsin B was a physiological target of Spi2A in memory CD8(+) T cell survival. Our findings support a model in which protection from lysosomal rupture through cytokine-induced expression of Spi2A determines the long-term persistence of memory CD8(+) T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cathepsin B/physiology , Immunologic Memory , Animals , CD8-Positive T-Lymphocytes/enzymology , CD8-Positive T-Lymphocytes/metabolism , Cathepsin B/antagonists & inhibitors , Cathepsin B/genetics , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Proliferation , Cell Survival/genetics , Cell Survival/immunology , Female , Homeostasis/genetics , Homeostasis/immunology , Immunologic Memory/genetics , Lysosomes/enzymology , Lysosomes/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Mice, Transgenic , Serpins/deficiency , Serpins/physiology
16.
Nat Med ; 13(8): 975-80, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17676051

ABSTRACT

Acne rosacea is an inflammatory skin disease that affects 3% of the US population over 30 years of age and is characterized by erythema, papulopustules and telangiectasia. The etiology of this disorder is unknown, although symptoms are exacerbated by factors that trigger innate immune responses, such as the release of cathelicidin antimicrobial peptides. Here we show that individuals with rosacea express abnormally high levels of cathelicidin in their facial skin and that the proteolytically processed forms of cathelicidin peptides found in rosacea are different from those present in normal individuals. These cathelicidin peptides are a result of a post-translational processing abnormality associated with an increase in stratum corneum tryptic enzyme (SCTE) in the epidermis. In mice, injection of the cathelicidin peptides found in rosacea, addition of SCTE, and increasing protease activity by targeted deletion of the serine protease inhibitor gene Spink5 each increases inflammation in mouse skin. The role of cathelicidin in enabling SCTE-mediated inflammation is verified in mice with a targeted deletion of Camp, the gene encoding cathelicidin. These findings confirm the role of cathelicidin in skin inflammatory responses and suggest an explanation for the pathogenesis of rosacea by demonstrating that an exacerbated innate immune response can reproduce elements of this disease.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Rosacea/metabolism , Rosacea/pathology , Serine Endopeptidases/metabolism , Animals , Antimicrobial Cationic Peptides/deficiency , Antimicrobial Cationic Peptides/genetics , Biopsy , Cells, Cultured , Cytokines/biosynthesis , Enzyme Activation , Humans , Inflammation/metabolism , Inflammation/pathology , Keratinocytes/cytology , Keratinocytes/metabolism , Mice , Mice, Knockout , Serine Peptidase Inhibitor Kazal-Type 5 , Serpins/deficiency , Serpins/genetics , Serpins/metabolism , Cathelicidins
17.
J Neurosci ; 32(35): 12152-64, 2012 Aug 29.
Article in English | MEDLINE | ID: mdl-22933798

ABSTRACT

Pigment epithelium-derived factor (PEDF) is a serine protease inhibitor (serpin) protein with well established neuroprotective and anti-angiogenic properties. Recent studies have also shown that PEDF enhances renewal of adult subventricular zone (SVZ) neural precursors. In neurosphere cultures prepared from the SVZ of adult mice, we found that addition of recombinant PEDF to the medium enhanced expressions of oligodendroglial lineage markers (NG2 and PDGFrα) and transcription factors (Olig1, Olig2, and Sox10). Similarly, continuous PEDF administration into the lateral ventricles of adult glial fibrillary acidic protein:green fluorescent protein (GFAP:GFP) transgenic mice increased the proportions of GFAP:GFP+ and GFAP:GFP- SVZ neural precursors coexpressing oligodendroglial lineage markers and transcription factors. Notably, PEDF infusion also resulted in an induction of doublecortin- and Sox10 double-positive cells in the adult SVZ. Immunoreactive PEDF receptor was detectable in multiple cell types in both adult SVZ and corpus callosum. Furthermore, PEDF intracerebral infusion enhanced survival and maturation of newly born oligodendroglial progenitor cells in the normal corpus callosum, and accelerated oligodendroglial regeneration in lysolecithin-induced corpus callosum demyelinative lesions. Western blot analysis showed a robust upregulation of endogenous PEDF in the corpus callosum upon lysolecithin-induced demyelination. Our results document previously unrecognized oligodendrotrophic effects of recombinant PEDF on the adult SVZ and corpus callosum, demonstrate induction of endogenous CNS PEDF production following demyelination, and make PEDF a strong candidate for pharmacological intervention in demyelinative diseases.


Subject(s)
Corpus Callosum/physiology , Eye Proteins/administration & dosage , Lateral Ventricles/physiology , Morphogenesis/physiology , Nerve Growth Factors/administration & dosage , Oligodendroglia/physiology , Serpins/administration & dosage , Animals , Cells, Cultured , Corpus Callosum/cytology , Eye Proteins/genetics , Female , Infusions, Intraventricular , Lateral Ventricles/cytology , Male , Mice , Mice, Knockout , Mice, Transgenic , Morphogenesis/genetics , Nerve Growth Factors/deficiency , Nerve Growth Factors/genetics , Serpins/deficiency , Serpins/genetics
18.
Am J Physiol Lung Cell Mol Physiol ; 305(12): L981-9, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24163143

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is characterized by emphysema and chronic bronchitis and is a leading cause of morbidity and mortality worldwide. Tobacco smoke and deficiency in α1-antitrypsin (AAT) are the most prominent environmental and genetic risk factors, respectively. Yet the pathogenesis of COPD is not completely elucidated. Disease progression appears to include a vicious circle driven by self-perpetuating lung inflammation, endothelial and epithelial cell death, and proteolytic degradation of extracellular matrix proteins. Like AAT, serpinB1 is a potent inhibitor of serine proteases including neutrophil elastase and cathepsin G. Because serpinB1 is expressed in myeloid and lung epithelial cells and is protective during lung infections, we investigated the role of serpinB1 in preventing age-related and cigarette smoke-induced emphysema in mice. Fifteen-month-old mice showed increased lung volume and decreased pulmonary function compared with young adult mice (3 mo old), but no differences were observed between serpinB1-deficient (KO) and wild-type (WT) mice. Chronic exposure to secondhand cigarette smoke resulted in structural emphysematous changes compared with respective control mice, but no difference in lung morphometry was observed between genotypes. Of note, the different pattern of stereological changes induced by age and cigarette smoke suggest distinct mechanisms leading to increased airway volume. Finally, expression of intracellular and extracellular protease inhibitors were differently regulated in lungs of WT and KO mice following smoke exposure; however, activity of proteases was not significantly altered. In conclusion, we showed that, although AAT and serpinB1 are similarly potent inhibitors of neutrophil proteases, serpinB1 deficiency is not associated with more severe emphysema.


Subject(s)
Pulmonary Emphysema/genetics , Pulmonary Emphysema/metabolism , Pulmonary Emphysema/pathology , Serpins/metabolism , Animals , Bronchoalveolar Lavage Fluid/cytology , Disease Models, Animal , Disease Susceptibility/metabolism , Disease Susceptibility/pathology , Mice , Mice, 129 Strain , Mice, Knockout , Mutation/genetics , Pneumonia/chemically induced , Pneumonia/genetics , Pneumonia/pathology , Serpins/deficiency , Smoke/adverse effects
19.
Reproduction ; 146(2): R73-80, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23690629

ABSTRACT

Protein Z (PZ) is a vitamin K-dependent factor characterized by its homology to other vitamin K-dependent factors (factors VII, IX, and X, protein C and protein S), but lacks any enzymatic activity. Instead, PZ acts as a cofactor for the inhibition of factor Xa through the serpin PZ-dependent protease inhibitor (ZPI). PZ deficiency is associated with a procoagulant state, highlighted by excessive FXa secretion and thrombin production, and is linked with several thrombotic disorders, including arterial vascular and venous thromboembolic diseases. A role for the PZ-ZPI complex in the regulation of physiological pregnancy has been demonstrated, highlighted by the progressive elevation in PZ levels in the first trimester of gestation, which then steadily decline toward delivery. An association between altered plasma PZ concentrations and adverse pregnancy outcomes (recurrent miscarriage, stillbirth, preeclampsia, intrauterine growth restriction, and placental abruption) has been reported. The mechanism by which PZ deficiency leads to adverse pregnancy outcomes is not clear, but it is multifactorial. It may be attributed to the anti-PZ IgG and IgM autoantibodies, which apparently act independently of classical antiphospholipid antibodies (lupus anticoagulant, anticardiolipin, and anti-ß2-glycoprotein I antibodies). PZ deficiency has also been reported to be constitutional, and a number of variants in the PROZ (PZ) gene and SERPINA10 (ZPI) gene are linked with specific adverse pregnancy complications. This review summarizes the relationship between adverse pregnancy outcomes and acquired and constitutional PZ-ZPI deficiency, in order to understand whether or not PZ deficiency could be considered as a risk factor for poor pregnancy outcomes.


Subject(s)
Blood Proteins/metabolism , Pregnancy Maintenance , Autoantibodies/analysis , Blood Coagulation , Blood Coagulation Disorders/blood , Blood Coagulation Disorders/genetics , Blood Coagulation Disorders/immunology , Blood Coagulation Disorders/metabolism , Blood Proteins/chemistry , Blood Proteins/deficiency , Blood Proteins/genetics , Female , Genetic Variation , Humans , Molecular Conformation , Pregnancy , Pregnancy Complications, Hematologic/blood , Pregnancy Complications, Hematologic/genetics , Pregnancy Complications, Hematologic/immunology , Pregnancy Complications, Hematologic/metabolism , Serpins/blood , Serpins/deficiency , Serpins/genetics , Serpins/metabolism
20.
J Immunol ; 186(1): 411-22, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21098233

ABSTRACT

Leishmania major is a protozoan parasite that causes skin ulcerations in cutaneous leishmaniasis. In the mammalian host, the parasite resides in professional phagocytes and has evolved to avoid killing by macrophages. We identified L. major genes encoding inhibitors of serine peptidases (ISPs), which are orthologs of bacterial ecotins, and found that ISP2 inhibits trypsin-fold S1A family peptidases. In this study, we show that L. major mutants deficient in ISP2 and ISP3 (Δisp2/3) trigger higher phagocytosis by macrophages through a combined action of the complement type 3 receptor, TLR4, and unregulated activity of neutrophil elastase (NE), leading to parasite killing. Whereas all three components are required to mediate enhanced parasite uptake, only TLR4 and NE are necessary to promote parasite killing postinfection. We found that the production of superoxide by macrophages in the absence of ISP2 is the main mechanism controlling the intracellular infection. Furthermore, we show that NE modulates macrophage infection in vivo, and that the lack of ISP leads to reduced parasite burdens at later stages of the infection. Our findings support the hypothesis that ISPs function to prevent the activation of TLR4 by NE during the Leishmania-macrophage interaction to promote parasite survival and growth.


Subject(s)
Intracellular Fluid/parasitology , Leishmania major/enzymology , Leishmania major/growth & development , Leukocyte Elastase/physiology , Macrophages, Peritoneal/parasitology , Serine Proteinase Inhibitors/physiology , Serpins/physiology , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/metabolism , Animals , Cells, Cultured , Host-Parasite Interactions/immunology , Intracellular Fluid/enzymology , Intracellular Fluid/immunology , Leishmania major/immunology , Leukocyte Elastase/antagonists & inhibitors , Macrophages, Peritoneal/enzymology , Macrophages, Peritoneal/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Oxidative Stress/immunology , Serine Proteinase Inhibitors/deficiency , Serine Proteinase Inhibitors/genetics , Serpins/deficiency , Serpins/genetics , Toll-Like Receptor 4/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL