Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
J Clin Invest ; 133(15)2023 08 01.
Article in English | MEDLINE | ID: mdl-37289545

ABSTRACT

Exaggerated Type 2 immune responses play critical roles in the pathogenesis of a variety of diseases including asthma, allergy, and pulmonary fibrosis. Recent studies have highlighted the importance of innate type 2 immune responses and innate lymphoid 2 cells (ILC2s) in these disorders. However, the mechanisms that control the development of pulmonary innate type 2 responses (IT2IR) and the recruitment and/or activation of ILC2 cells are poorly understood. In mouse models of pulmonary IT2IR, we demonstrated that phospholipid scramblase-1 (PLSCR1), a type II transmembrane protein that mediates bidirectional and nonspecific translocation of phospholipids between the inner and outer leaflets of the plasma membrane, was a critical regulator of IT2IR in the lung. We further suggested that (a) PLSCR1 bound to and physically interacted with chemoattractant receptor-homologous molecule(CRTH2), which is a G-protein-coupled receptor that is expressed on TH2 cells and on multiple immune cells and is commonly used to identify ILC2 cells, and (b) the effects of PLSCR1 on ILC2 activation and IT2IR were mediated via CRTH2-dependent mechanisms. Overall, our studies demonstrated that PLSCR1 played an essential role in the pathogenesis of ILC2 responses, providing critical insights into biology and disease pathogenesis and identifying targets that can be manipulated in attempts to control IT2IR in chronic diseases such as asthma.


Subject(s)
Asthma , Immunity, Innate , Animals , Mice , Phospholipid Transfer Proteins/genetics , Phospholipid Transfer Proteins/metabolism , Lymphocytes , Inflammation/pathology , Lung/pathology , Cytokines
3.
medRxiv ; 2023 Feb 23.
Article in English | MEDLINE | ID: mdl-36865305

ABSTRACT

Alzheimer's disease, the most common age-related neurodegenerative disease, is closely associated with both amyloid-ß plaque and neuroinflammation. Two thirds of Alzheimer's disease patients are females and they have a higher disease risk. Moreover, women with Alzheimer's disease have more extensive brain histological changes than men along with more severe cognitive symptoms and neurodegeneration. To identify how sex difference induces structural brain changes, we performed unbiased massively parallel single nucleus RNA sequencing on Alzheimer's disease and control brains focusing on the middle temporal gyrus, a brain region strongly affected by the disease but not previously studied with these methods. We identified a subpopulation of selectively vulnerable layer 2/3 excitatory neurons that that were RORB-negative and CDH9-expressing. This vulnerability differs from that reported for other brain regions, but there was no detectable difference between male and female patterns in middle temporal gyrus samples. Disease-associated, but sex-independent, reactive astrocyte signatures were also present. In clear contrast, the microglia signatures of diseased brains differed between males and females. Combining single cell transcriptomic data with results from genome-wide association studies (GWAS), we identified MERTK genetic variation as a risk factor for Alzheimer's disease selectively in females. Taken together, our single cell dataset revealed a unique cellular-level view of sex-specific transcriptional changes in Alzheimer's disease, illuminating GWAS identification of sex-specific Alzheimer's risk genes. These data serve as a rich resource for interrogation of the molecular and cellular basis of Alzheimer's disease.

4.
J Clin Invest ; 131(21)2021 11 01.
Article in English | MEDLINE | ID: mdl-34720089

ABSTRACT

Evasion of the immune response is a hallmark of cancer, and programmed cell death 1 (PD-1) and PD-1 ligand 1 (PD-L1) are major mediators of this immunosuppression. Chitinase 3-like 1 (CHI3L1) is induced in many cancers, where it portends a poor prognosis and contributes to tumor metastasis and spread. However, the mechanism(s) that CHI3L1 uses in metastasis have not been defined. Here we demonstrate that CHI3L1 regulates the expression of PD-L1, PD-L2, PD-1, LAG3, and TIM3 and plays a critical role in melanoma progression and lymphatic spread. CHI3L1 also contributed to IFN-γ-stimulated macrophage PD-L1 expression, and RIG-like helicase innate immunity suppressed CHI3L1, PD-L1, and melanoma progression. Individual antibodies against CHI3L1 or PD-1 had discrete antitumor effects and additive antitumor responses in metastasis models and T cell-tumor cell cocultures when administered simultaneously. Synergistic cytotoxic tumor cell death was seen in T cell-tumor cell cocultures, and significantly enhanced antitumor responses were seen in in vivo tumor models treated with bispecific antibodies that simultaneously target CHI3L1 and PD-1. CHI3L1 contributes to tumor progression by stimulating the PD-1/PD-L1 axis and other checkpoint molecules. The simultaneous targeting of CHI3L1 and the PD-1/PD-L1 axis with individual and, more powerfully, with bispecific antibodies represents a promising therapy for pulmonary metastasis and progression.


Subject(s)
Antibodies, Bispecific , Antibodies, Neoplasm , B7-H1 Antigen , Chitinase-3-Like Protein 1 , Lung Neoplasms , Neoplasm Proteins , Animals , Antibodies, Bispecific/immunology , Antibodies, Bispecific/pharmacology , Antibodies, Neoplasm/immunology , Antibodies, Neoplasm/pharmacology , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , Chitinase-3-Like Protein 1/antagonists & inhibitors , Chitinase-3-Like Protein 1/immunology , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Melanoma, Experimental/drug therapy , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Knockout , Neoplasm Metastasis , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/immunology
5.
JCI Insight ; 6(21)2021 11 08.
Article in English | MEDLINE | ID: mdl-34747367

ABSTRACT

COVID-19 is caused by SARS-CoV-2 (SC2) and is more prevalent and severe in elderly and patients with comorbid diseases (CM). Because chitinase 3-like-1 (CHI3L1) is induced during aging and CM, the relationships between CHI3L1 and SC2 were investigated. Here, we demonstrate that CHI3L1 is a potent stimulator of the SC2 receptor angiotensin converting enzyme 2 (ACE2) and viral spike protein priming proteases (SPP), that ACE2 and SPP are induced during aging, and that anti-CHI3L1, kasugamycin, and inhibitors of phosphorylation abrogate these ACE2- and SPP-inductive events. Human studies also demonstrate that the levels of circulating CHI3L1 are increased in the elderly and patients with CM, where they correlate with COVID-19 severity. These studies demonstrate that CHI3L1 is a potent stimulator of ACE2 and SPP, that this induction is a major mechanism contributing to the effects of aging during SC2 infection, and that CHI3L1 co-opts the CHI3L1 axis to augment SC2 infection. CHI3L1 plays a critical role in the pathogenesis of and is an attractive therapeutic target in COVID-19.


Subject(s)
Aging , COVID-19/metabolism , Chitinase-3-Like Protein 1/metabolism , Aging/drug effects , Aminoglycosides/pharmacology , Aminoglycosides/therapeutic use , Angiotensin-Converting Enzyme 2/metabolism , Cell Line, Tumor , Chitinase-3-Like Protein 1/antagonists & inhibitors , HEK293 Cells , Humans , SARS-CoV-2/physiology , COVID-19 Drug Treatment
6.
bioRxiv ; 2021 Feb 16.
Article in English | MEDLINE | ID: mdl-33442679

ABSTRACT

COVID-19 is caused by the SARS-CoV-2 (SC2) virus and is more prevalent and severe in the elderly and patients with comorbid diseases (CM). Because chitinase 3-like-1 (CHI3L1) is induced during aging and CM, the relationships between CHI3L1 and SC2 were investigated. Here we demonstrate that CHI3L1 is a potent stimulator of the SC2 receptor ACE2 and viral spike protein priming proteases (SPP), that ACE2 and SPP are induced during aging and that anti-CHI3L1, kasugamycin and inhibitors of phosphorylation, abrogate these ACE2- and SPP- inductive events. Human studies also demonstrated that the levels of circulating CHI3L1 are increased in the elderly and patients with CM where they correlate with COVID-19 severity. These studies demonstrate that CHI3L1 is a potent stimulator of ACE2 and SPP; that this induction is a major mechanism contributing to the effects of aging during SC2 infection and that CHI3L1 coopts the CHI3L1 axis to augment SC2 infection. CHI3L1 plays a critical role in the pathogenesis of and is an attractive therapeutic target in COVID-19.

7.
Am J Respir Cell Mol Biol ; 63(3): 386-395, 2020 09.
Article in English | MEDLINE | ID: mdl-32402213

ABSTRACT

Chitinase 3-like-1 (Chi3l1) and IL-13 are both ligands of IL-13 receptor α2 (IL-13Rα2). The binding of the former activates mitogen-activated protein kinase, AKT, and Wnt/ß-catenin signaling, and plays important roles in innate and adaptive immunity, cellular apoptosis, oxidative injury, allergic inflammation, tumor metastasis and wound healing, fibrosis, and repair in the lung. In contrast, the latter binding is largely a decoy event that diminishes the effects of IL-13. Here, we demonstrate that IL-13Rα2 N-glycosylation is a critical determinant of which ligand binds. Structure-function evaluations demonstrated that Chi3l1-IL-13Rα2 binding was increased when sites of N-glycosylation are mutated, and studies with tunicamycin and Peptide:N-glycosidase F (PNGase F) demonstrated that Chi3l1-IL-13Rα2 binding and signaling were increased when N-glycosylation was diminished. In contrast, structure-function experiments demonstrated that IL-13 binding to IL-13Rα2 was dependent on each of the four sites of N-glycosylation in IL-13Rα2, and experiments with tunicamycin and PNGase F demonstrated that IL-13-IL-13Rα2 binding was decreased when IL-13Rα2 N-glycosylation was diminished. Studies with primary lung epithelial cells also demonstrated that Chi3l1 inhibited, whereas IL-13 stimulated, N-glycosylation as evidenced by the ability of Chi3l1 to inhibit and IL-13 to stimulate the subunits of the oligosaccharide complex A and B (STT3A and STT3B). These studies demonstrate that N-glycosylation is a critical determinant of Chi3l1 and IL-13 binding to IL-13Rα2, and highlight the ability of Chi3l1 and IL-13 to alter key elements of the N-glycosylation apparatus in a manner that would augment their respective binding.


Subject(s)
Epithelial Cells/metabolism , Interleukin-13 Receptor alpha2 Subunit/metabolism , Interleukin-13/metabolism , Receptors, Interleukin-13/metabolism , Animals , Glycosylation , Hexosyltransferases/metabolism , Lung/metabolism , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/metabolism , Wnt Signaling Pathway/physiology
8.
Life Sci Alliance ; 2(3)2019 06.
Article in English | MEDLINE | ID: mdl-31085559

ABSTRACT

TGF-ß1 is a critical mediator of tissue fibrosis in health and disease whose effects are augmented by chitinase 1 (CHIT1). However, the mechanisms that CHIT1 uses to regulate TGF-ß1-mediated fibrotic responses have not been defined. Here, we demonstrate that CHIT1 enhances TGF-ß1-stimulated fibrotic cellular and tissue responses and TGF-ß1 signaling. Importantly, we also demonstrate that these effects are mediated by the ability of CHIT1 to inhibit TGF-ß1 induction of its feedback inhibitor, SMAD7. CHIT1 also interacted with TGF-ß receptor associated protein 1 (TGFBRAP1) and forkhead box O3 (FOXO3) with TGFBRAP1 playing a critical role in CHIT1 enhancement of TGF-ß1 signaling and effector responses and FOXO3 playing a critical role in TGF-ß1 induction of SMAD7. These pathways were disease relevant because the levels of CHIT1 were increased and inversely correlated with SMAD7 in tissues from patients with idiopathic pulmonary fibrosis or scleroderma-associated interstitial lung disease. These studies demonstrate that CHIT1 regulates TGF-ß1/SMAD7 axis via TGFBRAP1 and FOXO3 and highlight the importance of these pathways in the pathogenesis of pulmonary fibrosis.


Subject(s)
Forkhead Box Protein O3/metabolism , Hexosaminidases/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Pulmonary Fibrosis/etiology , Pulmonary Fibrosis/metabolism , Smad7 Protein/metabolism , Transforming Growth Factor beta/metabolism , Fibroblasts/metabolism , Gene Expression Regulation , Genes, Reporter , Hexosaminidases/metabolism , Humans , Immunohistochemistry , Promoter Regions, Genetic , Pulmonary Fibrosis/pathology , RNA, Small Interfering/genetics , Signal Transduction
9.
J Immunol ; 200(6): 2140-2153, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29427412

ABSTRACT

Hermansky-Pudlak syndrome (HPS) comprises a group of inherited disorders caused by mutations that alter the function of lysosome-related organelles. Pulmonary fibrosis is the major cause of morbidity and mortality in HPS-1 and HPS-4 patients. However, the mechanisms that underlie the exaggerated injury and fibroproliferative repair responses in HPS have not been adequately defined. In particular, although Galectin-3 (Gal-3) is dysregulated in HPS, its roles in the pathogenesis of HPS have not been adequately defined. In addition, although chitinase 3-like 1 (CHI3L1) and its receptors play major roles in the injury and repair responses in HPS, the ability of Gal-3 to interact with or alter the function of these moieties has not been evaluated. In this article, we demonstrate that Gal-3 accumulates in exaggerated quantities in bronchoalveolar lavage fluids, and traffics abnormally and accumulates intracellularly in lung fibroblasts and macrophages from bleomycin-treated pale ear, HPS-1-deficient mice. We also demonstrate that Gal-3 drives epithelial apoptosis when in the extracellular space, and stimulates cell proliferation and myofibroblast differentiation when accumulated in fibroblasts and M2-like differentiation when accumulated in macrophages. Biophysical and signaling evaluations also demonstrated that Gal-3 physically interacts with IL-13Rα2 and CHI3L1, and competes with TMEM219 for IL-13Rα2 binding. By doing so, Gal-3 diminishes the antiapoptotic effects of and the antiapoptotic signaling induced by CHI3L1 in epithelial cells while augmenting macrophage Wnt/ß-catenin signaling. Thus, Gal-3 contributes to the exaggerated injury and fibroproliferative repair responses in HPS by altering the antiapoptotic and fibroproliferative effects of CHI3L1 and its receptor complex in a tissue compartment-specific manner.


Subject(s)
Chitinase-3-Like Protein 1/metabolism , Galectin 3/metabolism , Hermanski-Pudlak Syndrome/metabolism , Lung/metabolism , Animals , Apoptosis/drug effects , Bleomycin/pharmacology , Bronchoalveolar Lavage Fluid/chemistry , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Proliferation/physiology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Lung/drug effects , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Pulmonary Fibrosis/metabolism , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism
11.
Nat Commun ; 7: 12752, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27629921

ABSTRACT

Recent studies demonstrated that chitinase 3-like-1 (Chi3l1) binds to and signals via IL-13Rα2. However, the mechanism that IL-13Rα2 uses to mediate the effects of Chi3l1 has not been defined. Here, we demonstrate that the membrane protein, TMEM219, is a binding partner of IL-13Rα2 using yeast two-hybrid, co-immunoprecipitation, co-localization and bimolecular fluorescence complementation assays. Furthermore, fluorescence anisotropy nanodisc assays revealed a direct physical interaction between TMEM219 and IL-13Rα2-Chi3l1 complexes. Null mutations or siRNA silencing of TMEM219 or IL-13Rα2 similarly decreased Chi3l1-stimulated epithelial cell HB-EGF production and macrophage MAPK/Erk and PKB/Akt activation. Null mutations of TMEM219 or IL-13Rα2 also phenocopied one another as regards the ability of Chi3l1 to inhibit oxidant-induced apoptosis and lung injury, promote melanoma metastasis and stimulate TGF-ß1. TMEM219 also contributed to the decoy function of IL-13Rα2. These studies demonstrate that TMEM219 plays a critical role in Chi3l1-induced IL-13Rα2 mediated signalling and tissue responses.


Subject(s)
Chitinase-3-Like Protein 1/metabolism , Interleukin-13 Receptor alpha2 Subunit/metabolism , Membrane Proteins/metabolism , Animals , Apoptosis , Cell Line, Tumor , Heparin-binding EGF-like Growth Factor/metabolism , Humans , Lung Injury/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , MAP Kinase Signaling System , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice, Inbred C57BL , Neoplasm Metastasis , Proto-Oncogene Proteins c-akt/metabolism , Transforming Growth Factor beta1/metabolism , Two-Hybrid System Techniques , Wnt Signaling Pathway
12.
J Clin Invest ; 125(8): 3178-92, 2015 Aug 03.
Article in English | MEDLINE | ID: mdl-26121745

ABSTRACT

Hermansky-Pudlak syndrome (HPS) comprises a group of inherited disorders caused by mutations that alter the function of lysosome-related organelles. Pulmonary fibrosis is the major cause of morbidity and mortality in patients with subtypes HPS-1 and HPS-4, which both result from defects in biogenesis of lysosome-related organelle complex 3 (BLOC-3). The prototypic chitinase-like protein chitinase 3-like-1 (CHI3L1) plays a protective role in the lung by ameliorating cell death and stimulating fibroproliferative repair. Here, we demonstrated that circulating CHI3L1 levels are higher in HPS patients with pulmonary fibrosis compared with those who remain fibrosis free, and that these levels associate with disease severity. Using murine HPS models, we also determined that these animals have a defect in the ability of CHI3L1 to inhibit epithelial apoptosis but exhibit exaggerated CHI3L1-driven fibroproliferation, which together promote HPS fibrosis. These divergent responses resulted from differences in the trafficking and effector functions of two CHI3L1 receptors. Specifically, the enhanced sensitivity to apoptosis was due to abnormal localization of IL-13Rα2 as a consequence of dysfunctional BLOC-3-dependent membrane trafficking. In contrast, the fibrosis was due to interactions between CHI3L1 and the receptor CRTH2, which trafficked normally in BLOC-3 mutant HPS. These data demonstrate that CHI3L1-dependent pathways exacerbate pulmonary fibrosis and suggest CHI3L1 as a potential biomarker for pulmonary fibrosis progression and severity in HPS.


Subject(s)
Adipokines/blood , Apoptosis , Glycoproteins/blood , Hermanski-Pudlak Syndrome/blood , Lectins/blood , Pulmonary Fibrosis/blood , Respiratory Mucosa/metabolism , Adipokines/genetics , Adult , Animals , Biomarkers/blood , Chitinase-3-Like Protein 1 , Disease Models, Animal , Female , Glycoproteins/genetics , Hermanski-Pudlak Syndrome/complications , Hermanski-Pudlak Syndrome/genetics , Hermanski-Pudlak Syndrome/pathology , Humans , Interleukin-13 Receptor alpha2 Subunit/genetics , Interleukin-13 Receptor alpha2 Subunit/metabolism , Lectins/genetics , Male , Mice , Mice, Knockout , Pulmonary Fibrosis/etiology , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/pathology , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Receptors, Prostaglandin/genetics , Receptors, Prostaglandin/metabolism , Respiratory Mucosa/pathology
13.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 45(5): 789-92, 2014 Sep.
Article in Chinese | MEDLINE | ID: mdl-25341341

ABSTRACT

OBJECTIVE: To investigate the effects of fenofibrate, a lipid-lowering drug, on the growth and migration of human ovarian cancer cells SKOV3 in vitro. METHODS: A human ovarian cancer cell line (SKOV3) as the research object, was incubated with serum-free media for 24 h. These cells were then treated by appropriate concentrations of fenofibrate for different time, including control and experimental groups. Cell proliferation was evaluated by MTT assay. Apoptosis was detected by Hoechst/PI and Annexin-V/PI fluorescent assay. The migration of cells was measured by the scratch-wound healing assay. RESULTS: The MTT assay results demonstrated that the fenofibrate (10, 25, 50, 75, 100 micromol/L) could inhibit the proliferation of SKOV3 cells after 24, 48 and 72 h treatment (P < 0.05). The inhibition rate for 24, 48, 72 h-treatment was 55.72% +/- 0.28%, 57.63% +/- 0.47%, 72.41% +/- 0.62% respectively (P < 0.05). The effects increased with the concentrations. Hoechst/PI and Annexin-V/PI fluorescent assay showed that after stimulus for 24 h, fenofibrate induced apoptosis of SKOV3 cells in a concentration-dependent manner was observed. A significant inhibited cells migration distance (P < 0.05) evaluated with scratch-wound healing assay was observed after treatment with fenofibrate (10, 25, 50, 75, 100 micromol/L) for 24 h. CONCLUSION: Lipid-lowering drug fenofibrate can inhibit the growth and migration of human ovarian cancer cell SKOV3 in vitro, to some extent induce apoptosis. But the detailed mechanism need to be further studied.


Subject(s)
Cell Proliferation/drug effects , Fenofibrate/pharmacology , Ovarian Neoplasms/pathology , Apoptosis/drug effects , Cell Line, Tumor/drug effects , Cell Movement/drug effects , Culture Media, Serum-Free , Female , Humans
14.
Cell Rep ; 4(4): 830-41, 2013 Aug 29.
Article in English | MEDLINE | ID: mdl-23972995

ABSTRACT

Members of the 18 glycosyl hydrolase (GH 18) gene family have been conserved over species and time and are dysregulated in inflammatory, infectious, remodeling, and neoplastic disorders. This is particularly striking for the prototypic chitinase-like protein chitinase 3-like 1 (Chi3l1), which plays a critical role in antipathogen responses where it augments bacterial killing while stimulating disease tolerance by controlling cell death, inflammation, and remodeling. However, receptors that mediate the effects of GH 18 moieties have not been defined. Here, we demonstrate that Chi3l1 binds to interleukin-13 receptor α2 (IL-13Rα2) and that Chi3l1, IL-13Rα2, and IL-13 are in a multimeric complex. We also demonstrate that Chi3l1 activates macrophage mitogen-activated protein kinase, protein kinase B/AKT, and Wnt/ß-catenin signaling and regulates oxidant injury, apoptosis, pyroptosis, inflammasome activation, antibacterial responses, melanoma metastasis, and TGF-ß1 production via IL-13Rα2-dependent mechanisms. Thus, IL-13Rα2 is a GH 18 receptor that plays a critical role in Chi3l1 effector responses.


Subject(s)
Glycoproteins/metabolism , Interleukin-13 Receptor alpha2 Subunit/metabolism , Animals , Apoptosis , Chitinase-3-Like Protein 1 , Glycoproteins/genetics , Humans , Inflammasomes/metabolism , Interleukin-13/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , MAP Kinase Signaling System , Macrophages/metabolism , Melanoma/metabolism , Melanoma/secondary , Mice , Mice, Inbred C57BL , Oxidative Stress , Protein Binding , Transforming Growth Factor beta/metabolism , Wnt Signaling Pathway
15.
J Am Soc Nephrol ; 24(2): 309-19, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23291472

ABSTRACT

Kidney hypoperfusion during episodes of systemic hypotension or after surgical procurement for transplantation can lead to tubular cell death via necrosis and apoptosis, which trigger a series of responses that promote repair. The factors that contribute to the repair phase after kidney injury are not well understood. Using a urine proteomic screen in mice, we identified the macrophage-secreted chitinase-like protein Brp-39, the murine protein product of the chitinase 3-like 1 gene, as a critical component of this reparative response that serves to limit tubular cell apoptotic death via activation of Akt, improving animal survival after kidney ischemia/reperfusion. Examination of graded times of renal ischemia revealed a direct correlation between the degree of kidney injury and both Chi3l1/Brp-39 expression in the kidney and its levels in the urine. In samples collected from patients undergoing deceased-donor kidney transplantation, we found higher levels of the orthologous human protein, YKL-40, in urine and blood from allografts subjected to sufficient peri-transplant ischemia to cause delayed graft function than from allografts with slow or immediate graft function. Urinary levels of YKL-40 obtained within hours of transplant predicted the need for subsequent dialysis in these patients. In summary, these data suggest that Brp-39/YKL-40 is a sensor of the degree of injury, a critical mediator of the reparative response, and a possible biomarker to identify patients at greatest risk of sustained renal failure after transplantation.


Subject(s)
Adipokines/metabolism , Delayed Graft Function/metabolism , Glycoproteins/metabolism , Kidney Transplantation , Lectins/metabolism , Reperfusion Injury/metabolism , Adipokines/genetics , Animals , Apoptosis/physiology , Biomarkers/blood , Biomarkers/urine , Cells, Cultured , Chitinase-3-Like Protein 1 , Delayed Graft Function/mortality , Delayed Graft Function/physiopathology , Disease Models, Animal , Epithelial Cells/cytology , Glycoproteins/genetics , Humans , Kidney/cytology , Lectins/genetics , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Phosphatidylinositol 3-Kinases/metabolism , Predictive Value of Tests , Proto-Oncogene Proteins c-akt/metabolism , Reperfusion Injury/mortality , Reperfusion Injury/physiopathology , Signal Transduction/physiology , Transplantation, Homologous
16.
Cell Host Microbe ; 12(1): 34-46, 2012 Jul 19.
Article in English | MEDLINE | ID: mdl-22817986

ABSTRACT

Host antibacterial responses include mechanisms that kill bacteria, but also those that protect or tolerize the host to potentially damaging antibacterial effects. We determined that Chitinase 3-like-1 (Chi3l1), a conserved prototypic chitinase-like protein, is induced by Streptococcus pneumoniae and plays central roles in promoting bacterial clearance and mediating host tolerance. S. pneumoniae-infected Chi3l1 null mice exhibit exaggerated lung injury, inflammation and hemorrhage, more frequent bacterial dissemination, decreased bacterial clearance, and enhanced mortality compared to controls. Chi3l1 augments macrophage bacterial killing by inhibiting caspase-1-dependent macrophage pyroptosis and augments host tolerance by controlling inflammasome activation, ATP accumulation, expression of ATP receptor P2X7R, and production of thymic stromal lymphopoietin and type 1, type 2, and type 17 cytokines. These data demonstrate that Chi3l1 is induced during infection, where it promotes bacterial clearance while simultaneously augmenting host tolerance, and that these roles likely contributed to the retention of Chi3l1 over species and evolutionary time.


Subject(s)
Glycoproteins/metabolism , Host-Pathogen Interactions , Pneumococcal Infections/metabolism , Pneumococcal Infections/microbiology , Streptococcus pneumoniae/pathogenicity , Adenosine Triphosphate/metabolism , Animals , Carrier Proteins/metabolism , Caspase 1/metabolism , Cell Death , Chitinase-3-Like Protein 1 , Cytokines/metabolism , Glycoproteins/genetics , Inflammasomes/metabolism , Lung/microbiology , Lung/pathology , Macrophages/cytology , Macrophages/metabolism , Macrophages/microbiology , Mice , Mice, Mutant Strains , NLR Family, Pyrin Domain-Containing 3 Protein , Phagocytosis , Pneumococcal Infections/immunology , Pneumococcal Infections/mortality , Pneumococcal Infections/pathology , Pneumonia, Pneumococcal/metabolism , Pneumonia, Pneumococcal/physiopathology , Receptors, Purinergic P2X7/metabolism
17.
Proc Am Thorac Soc ; 8(6): 512-5, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22052929

ABSTRACT

Vascular endothelial growth factor (VEGF) is a potent stimulator of vascular angiogenesis, permeability, and remodeling that also plays important roles in wound healing and tissue cytoprotection. To begin to define the roles of VEGF in diseases like asthma and COPD, we characterized the effects of lung-targeted transgenic VEGF(165) and defined the innate immune pathways that regulate VEGF tissue responses. The former studies demonstrated that VEGF plays an important role in Th2 inflammation because, in addition to stimulating angiogenesis and edema, VEGF induced eosinophilic inflammation, mucus metaplasia, subepithelial fibrosis, myocyte hyperplasia, dendritic cell activation, and airways hyperresponsiveness via IL-13-dependent and -independent mechanisms. VEGF was also produced at sites of aeroallergen-induced Th2 inflammation, and VEGF receptor blockade ameliorated adaptive Th2 inflammation and Th2 cytokine elaboration. The latter studies demonstrated that activation of the RIG-like helicase (RLH) innate immune pathway using viral pathogen-associated molecular patterns such as Poly(I:C) or viruses ameliorated VEGF-induced tissue responses. In accord with these findings, Poly(I:C)-induced RLH activation also abrogated aeroallergen-induced Th2 inflammation. When viewed in combination, these studies suggest that VEGF excess can contribute to the pathogenesis of Th2 inflammatory disorders such as asthma and that abrogation of VEGF signaling via RLH activation can contribute to the pathogenesis of viral disorders such as virus-induced COPD exacerbations. They also suggest that RLH activation may be a useful therapeutic strategy in asthma and related disorders.


Subject(s)
Asthma/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Disease Models, Animal , Humans , Inflammation/metabolism , Mice
18.
Annu Rev Physiol ; 73: 479-501, 2011.
Article in English | MEDLINE | ID: mdl-21054166

ABSTRACT

The 18 glycosyl hydrolase family of chitinases is an ancient gene family that is widely expressed from prokaryotes to eukaryotes. In mammals, despite the absence of endogenous chitin, a number of chitinases and chitinase-like proteins (C/CLPs) have been identified. However, their roles have only recently begun to be elucidated. Acidic mammalian chitinase (AMCase) inhibits chitin-induced innate inflammation; augments chitin-free, allergen-induced Th2 inflammation; and mediates effector functions of IL-13. The CLPs BRP-39/YKL-40 (also termed chitinase 3-like 1) inhibit oxidant-induced lung injury, augments adaptive Th2 immunity, regulates apoptosis, stimulates alternative macrophage activation, and contributes to fibrosis and wound healing. In accord with these findings, levels of YKL-40 in the lung and serum are increased in asthma and other inflammatory and remodeling disorders and often correlate with disease severity. Our understanding of the roles of C/CLPs in inflammation, tissue remodeling, and tissue injury in health and disease is reviewed below.


Subject(s)
Airway Remodeling/physiology , Chitin/metabolism , Chitinases/metabolism , Inflammation/enzymology , Adipokines , Animals , Apoptosis/immunology , Atherosclerosis/enzymology , Atherosclerosis/immunology , Chitin/immunology , Chitinase-3-Like Protein 1 , Chitinases/immunology , Diabetes Mellitus/enzymology , Diabetes Mellitus/immunology , Giant Cell Arteritis/enzymology , Giant Cell Arteritis/immunology , Glycoproteins/blood , Glycoproteins/physiology , Humans , Lectins/blood , Lectins/physiology , Lung Diseases/enzymology , Lung Diseases/immunology , Mice , Neoplasms/enzymology , Neoplasms/immunology , Oxidative Stress/drug effects , Oxidative Stress/physiology
19.
J Immunol ; 182(8): 5098-106, 2009 Apr 15.
Article in English | MEDLINE | ID: mdl-19342690

ABSTRACT

Acidic mammalian chitinase (AMCase) is produced during and plays an important role in the pathogenesis of Th2-mediated diseases and antiparasite responses. However, the effector responses of AMCase in these settings have not been adequately defined and the relationship(s) between its chitinolytic and other biologic properties have not been investigated. In these studies, we demonstrate that AMCase protects airway epithelial cells from Fas ligand- and growth factor withdrawal-induced apoptosis. This cytoprotection was associated with Akt phosphorylation and abrogated when the PI3K/Akt pathway was inhibited. Comparable cytoprotection was also seen in experiments comparing wild-type AMCase and mutant AMCase that lacked chitinolytic activity. Importantly, the apoptosis-inhibiting effect of enzymatically active and inactive AMCase was abrogated by treatment with allosamidin. These studies demonstrate that secreted AMCase feeds back in an autocrine and/or paracrine manner to protect pulmonary epithelial cells from growth factor withdrawal- and Fas ligand-induced apoptosis. They also demonstrate that the cytoprotection is mediated via a PI3K/Akt-dependent and allosamidin-sensitive pathway that is independent of the chitinolytic activity of this chitinase.


Subject(s)
Apoptosis , Chitinases/metabolism , Epithelial Cells/cytology , Epithelial Cells/enzymology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Chitin/pharmacology , Chitinases/genetics , Enzyme Activation , Epithelial Cells/drug effects , Gene Expression Regulation, Enzymologic , Humans , Interleukin-13/genetics , Interleukin-13/metabolism , Lung/cytology , Lung/drug effects , Lung/enzymology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Binding , Proto-Oncogene Proteins c-akt/metabolism
20.
J Biol Chem ; 283(48): 33472-82, 2008 Nov 28.
Article in English | MEDLINE | ID: mdl-18824549

ABSTRACT

Acidic mammalian chitinase (AMCase) is expressed in an exaggerated fashion in epithelial cells at sites of pulmonary T helper cell type 2 inflammation and plays important roles in the pathogenesis of anti-parasite and asthma-like responses. However, the mechanisms that control epithelial cell AMCase secretion and its effector responses have not been adequately defined. To address these issues, we used in vivo and in vitro experimental systems to define the pathways of epithelial AMCase secretion and its epithelial regulatory effects. Here we demonstrate that, in murine T helper cell type 2 modeling systems, AMCase colocalizes with the epidermal growth factor receptor (EGFR) and ADAM17 (a membrane disintegrin and metallopeptidase 17) in lung epithelial cells. In vitro cotransfection experiments in A549 cells demonstrated that AMCase and EGFR physically interact with each other. Cotransfection of AMCase and EGFR also increased, whereas EGFR inhibition decreased AMCase secretion. Interestingly, AMCase secretion was not significantly altered by treatment with EGF but was significantly decreased when the upstream EGFR transactivator ADAM17 was inhibited. AMCase secretion was also decreased when the EGFR-downstream Ras was blocked. Transfected and recombinant AMCase induced epithelial cell production of CCL2, CCL17, and CXCL8. These studies demonstrate that lung epithelial cells secrete AMCase via an EGFR-dependent pathway that is activated by ADAM17 and mediates its effects via Ras. They also demonstrate that the AMCase that is secreted feeds back in an autocrine and/or paracrine fashion to stimulate pulmonary epithelial cell chemokine production.


Subject(s)
ADAM Proteins/metabolism , Chemokines/metabolism , Chitinases/metabolism , ErbB Receptors/metabolism , Lung/metabolism , Respiratory Mucosa/metabolism , ADAM Proteins/genetics , ADAM17 Protein , Animals , Cell Line , Chemokines/genetics , Chitinases/genetics , Epidermal Growth Factor/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , ErbB Receptors/genetics , Humans , Inflammation/genetics , Inflammation/metabolism , Lung/cytology , Mice , Mice, Transgenic , Respiratory Mucosa/cytology , Th2 Cells/cytology , Th2 Cells/metabolism , Transfection/methods , ras Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...