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










Publication year range
1.
J Virol ; 95(15): e0032721, 2021 07 12.
Article in English | MEDLINE | ID: mdl-33963054

ABSTRACT

The human protein-coding gene ILRUN (inflammation and lipid regulator with UBA-like and NBR1-like domains; previously C6orf106) was identified as a proviral factor for Hendra virus infection and was recently characterized to function as an inhibitor of type I interferon expression. Here, we have utilized transcriptome sequencing (RNA-seq) to define cellular pathways regulated by ILRUN in the context of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection of Caco-2 cells. We find that inhibition of ILRUN expression by RNA interference alters transcription profiles of numerous cellular pathways, including upregulation of the SARS-CoV-2 entry receptor ACE2 and several other members of the renin-angiotensin aldosterone system. In addition, transcripts of the SARS-CoV-2 coreceptors TMPRSS2 and CTSL were also upregulated. Inhibition of ILRUN also resulted in increased SARS-CoV-2 replication, while overexpression of ILRUN had the opposite effect, identifying ILRUN as a novel antiviral factor for SARS-CoV-2 replication. This represents, to our knowledge, the first report of ILRUN as a regulator of the renin-angiotensin-aldosterone system (RAAS). IMPORTANCE There is no doubt that the current rapid global spread of COVID-19 has had significant and far-reaching impacts on our health and economy and will continue to do so. Research in emerging infectious diseases, such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is growing rapidly, with new breakthroughs in the understanding of host-virus interactions to assist with the development of innovative and exciting therapeutic strategies. Here, we present the first evidence that modulation of the human protein-coding gene ILRUN functions as an antiviral factor for SARS-CoV-2 infection, likely through its newly identified role in regulating the expression of SARS-CoV-2 entry receptors ACE2, TMPRSS2, and CTSL. These data improve our understanding of biological pathways that regulate host factors critical to SARS-CoV-2 infection, contributing to the development of antiviral strategies to deal with the current SARS-CoV-2 pandemic.


Subject(s)
Angiotensin-Converting Enzyme 2/biosynthesis , COVID-19/metabolism , Down-Regulation , Gene Expression Regulation, Enzymologic , Neoplasm Proteins/metabolism , SARS-CoV-2/metabolism , Angiotensin-Converting Enzyme 2/genetics , Animals , COVID-19/genetics , Caco-2 Cells , Cathepsin L/biosynthesis , Cathepsin L/genetics , Chlorocebus aethiops , Humans , Neoplasm Proteins/genetics , Renin-Angiotensin System , SARS-CoV-2/genetics , Serine Endopeptidases/biosynthesis , Serine Endopeptidases/genetics , Vero Cells
2.
J Neurochem ; 157(3): 764-780, 2021 05.
Article in English | MEDLINE | ID: mdl-33368303

ABSTRACT

Neuronal ceroid lipofuscinoses (NCLs) are a group of inherited childhood neurodegenerative disorders. In addition to the accumulation of auto-fluorescent storage material in lysosomes, NCLs are largely characterised by region-specific neuroinflammation that can predict neuron loss. These phenotypes suggest alterations in the extracellular environment-making the secretome an area of significant interest. This study investigated the secretome in the CLN6 (ceroid-lipofuscinosis neuronal protein 6) variant of NCL. To investigate the CLN6 secretome, we co-cultured neurons and glia isolated from Cln6nclf or Cln6± mice, and utilised mass spectrometry to compare protein constituents of conditioned media. The significant changes noted in cathepsin enzymes, were investigated further via western blotting and enzyme activity assays. Viral-mediated gene therapy was used to try and rescue the wild-type phenotype and restore the secretome-both in vitro in co-cultures and in vivo in mouse plasma. In Cln6nclf cells, proteomics revealed a marked increase in catabolic and cytoskeletal-associated proteins-revealing new similarities between the pathogenic signatures of NCLs with other neurodegenerative disorders. These changes were, in part, corrected by gene therapy intervention, suggesting these proteins as candidate in vitro biomarkers. Importantly, these in vitro changes show promise for in vivo translation, with Cathepsin L (CTSL) activity reduced in both co-cultures and Cln6nclf plasma samples post gene-therapy. This work suggests the secretome plays a role in CLN6 pathogenesis and highlights its potential use as an in vitro model. Proteomic changes present a list of candidate biomarkers for monitoring disease and assessing potential therapeutics in future studies.


Subject(s)
Membrane Proteins/genetics , Neuronal Ceroid-Lipofuscinoses/genetics , Animals , Biomarkers , Cathepsin L/biosynthesis , Coculture Techniques , Computational Biology , Disease Models, Animal , Female , Gene Expression Regulation , Genetic Therapy , Male , Mice , Mice, Knockout , Neuroglia/metabolism , Neuronal Ceroid-Lipofuscinoses/diagnosis , Neuronal Ceroid-Lipofuscinoses/drug therapy , Neurons/metabolism , Primary Cell Culture , Proteomics
3.
Sci Rep ; 9(1): 491, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30679571

ABSTRACT

Pulmonary fibrosis is a progressive disease characterized by a widespread accumulation of myofibroblasts and extracellular matrix components. Growing evidences support that cysteine cathepsins, embracing cathepsin B (CatB) that affects TGF-ß1-driven Smad pathway, along with their extracellular inhibitor cystatin C, participate in myofibrogenesis. Here we established that curcumin, a potent antifibrotic drug used in traditional Asian medicine, impaired the expression of both α-smooth muscle actin and mature TGF-ß1 and inhibited the differentiation of human lung fibroblasts (CCD-19Lu cells). Curcumin induced a compelling upregulation of CatB and CatL. Conversely cystatin C was downregulated, which allowed the recovery of the peptidase activity of secreted cathepsins and the restoration of the proteolytic balance. Consistently, the amount of both insoluble and soluble type I collagen decreased, reaching levels similar to those observed for undifferentiated fibroblasts. The signaling pathways activated by curcumin were further examined. Curcumin triggered the expression of nuclear peroxisome proliferator-activated receptor γ (PPARγ). Contrariwise PPARγ inhibition, either by an antagonist (2-chloro-5-nitro-N-4-pyridinyl-benzamide) or by RNA silencing, restored TGF-ß1-driven differentiation of curcumin-treated CCD-19Lu cells. PPARγ response element (PPRE)-like sequences were identified in the promoter regions of both CatB and CatL. Finally, we established that the transcriptional induction of CatB and CatL depends on the binding of PPARγ to PPRE sequences as a PPARγ/Retinoid X Receptor-α heterodimer.


Subject(s)
Cathepsin B/biosynthesis , Cathepsin L/biosynthesis , Cell Differentiation/drug effects , Curcumin/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Lung/metabolism , PPAR gamma/metabolism , Transforming Growth Factor beta1/pharmacology , Up-Regulation/drug effects , Cell Line , Humans
4.
Cancer Res ; 78(10): 2524-2535, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29510992

ABSTRACT

CHIP/STUB1 ubiquitin ligase is a negative co-chaperone for HSP90/HSC70, and its expression is reduced or lost in several cancers, including breast cancer. Using an extensive and well-annotated breast cancer tissue collection, we identified the loss of nuclear but not cytoplasmic CHIP to predict more aggressive tumorigenesis and shorter patient survival, with loss of CHIP in two thirds of ErbB2+ and triple-negative breast cancers (TNBC) and in one third of ER+ breast cancers. Reduced CHIP expression was seen in breast cancer patient-derived xenograft tumors and in ErbB2+ and TNBC cell lines. Ectopic CHIP expression in ErbB2+ lines suppressed in vitro oncogenic traits and in vivo xenograft tumor growth. An unbiased screen for CHIP-regulated nuclear transcription factors identified many candidates whose DNA-binding activity was up- or downregulated by CHIP. We characterized myeloid zinc finger 1 (MZF1) as a CHIP target, given its recently identified role as a positive regulator of cathepsin B/L (CTSB/L)-mediated tumor cell invasion downstream of ErbB2. We show that CHIP negatively regulates CTSB/L expression in ErbB2+ and other breast cancer cell lines. CTSB inhibition abrogates invasion and matrix degradation in vitro and halts ErbB2+ breast cancer cell line xenograft growth. We conclude that loss of CHIP remodels the cellular transcriptome to unleash critical pro-oncogenic pathways, such as the matrix-degrading enzymes of the cathepsin family, whose components can provide new therapeutic opportunities in breast and other cancers with loss of CHIP expression.Significance: These findings reveal a novel targetable pathway of breast oncogenesis unleashed by the loss of tumor suppressor ubiquitin ligase CHIP/STUB1. Cancer Res; 78(10); 2524-35. ©2018 AACR.


Subject(s)
Cathepsin B/metabolism , Cathepsin L/metabolism , Cell Transformation, Neoplastic/genetics , Triple Negative Breast Neoplasms/pathology , Ubiquitin-Protein Ligases/metabolism , Cathepsin B/biosynthesis , Cathepsin L/biosynthesis , Cell Line, Tumor , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic/genetics , HEK293 Cells , Humans , Kruppel-Like Transcription Factors/metabolism , MCF-7 Cells , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Signal Transduction , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/mortality , Ubiquitin-Protein Ligases/genetics , Xenograft Model Antitumor Assays
5.
Nat Prod Commun ; 12(1): 107-109, 2017 Jan.
Article in English | MEDLINE | ID: mdl-30549840

ABSTRACT

Overexpression of a putative type III polyketide synthase (PKSIII) from the marine myxobacterium Enhygromyxa salina SWB007 in Streptoinyces coelicolor MI 146 led to the accumulation of a novel monoketopiperazine consisting of phenylalanine and isoleucine. This compound was named phileucin and shows high structural similarity to phevalin (aureusimine B). The protease inhibiting activity was tested against human cathepsin L, human leukocyte elastase; bovine trypsin and bovine chymotrypsin. In contrast to phevalin, no protease inhibition was observed.


Subject(s)
Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Pyrazines/chemistry , Streptomyces coelicolor/chemistry , Animals , Cathepsin L/antagonists & inhibitors , Cathepsin L/biosynthesis , Cattle , Chymotrypsin/antagonists & inhibitors , Humans , Magnetic Resonance Spectroscopy , Myxococcales/enzymology , Polyketide Synthases/metabolism , Proteinase Inhibitory Proteins, Secretory/chemistry , Proteinase Inhibitory Proteins, Secretory/pharmacology , Spectrometry, Mass, Electrospray Ionization , Spectrophotometry, Infrared , Spectrophotometry, Ultraviolet , Trypsin Inhibitors/chemistry , Trypsin Inhibitors/pharmacology
6.
Oncotarget ; 7(29): 45995-46001, 2016 Jul 19.
Article in English | MEDLINE | ID: mdl-27351223

ABSTRACT

Paclitaxel is recommended as a first-line chemotherapeutic agent against, ovarian cancer, however, the development of chemoresistance is a major obstacle in patients with aggressive ovarian cancer and results in recurrence after conventional therapy. The key molecule or mechanism associated with paclitaxel resistance in ovarian cancer still remains unclear. Cathepsin L (CTSL) is overexpressed in various cancers, however, the association between CTSL expression and paclitaxel resistance remains unclear. In the present study, we investigated the role of CTSL in paclitaxel-resistant SKOV3/TAX cells by CTSL silencing. Expression of CTSL was examined by immunohistochemistry and qRT-PCR in 58 clinical samples, and in SKOV3 cells and SKOV3/TAX cells. Effects of CTSL knockdown on ovarian cancer cell proliferation, apoptosis, migration, and invasion were also studied. The IHC and real-time PCR results showed that the difference of CTSL expression between ovarian cancer and the adjacent non-tumourous ovarian tissues was statistically significant. Western blot analysis showed that the CTSL was overexpressed in SKOV3/TAX cells and weakly detectable in paclitaxel-sensitive SKOV3 cells. Knocking-down of CTSL in ovarian cancer cells could decrease cell proliferation, migration, and invasion, and potentiate apoptosis induced by paclitaxel, suggesting CTSL may contribute to Paclitaxel resistance in ovarian cancer.


Subject(s)
Cathepsin L/biosynthesis , Drug Resistance, Neoplasm/genetics , Neoplasm Invasiveness/genetics , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/pathology , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Female , Humans
7.
Clin Transl Oncol ; 18(7): 722-7, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26474873

ABSTRACT

Lung cancer, the most common malignancy, is still the leading cause of cancer-related death worldwide. Non-small-cell lung cancer (NSCLC) accounts for 80 % of all lung cancers. Recent studies showed Cathepsin L (CTSL) is overexpressed in various cancerous tissues; however, the association between CTSL expression and EGFR-TKI resistance remains unknown. In this study, we investigated the expression of CTSL in lung cancer specimens and matched normal tissues by quantitative real-time PCR and IHC. The functional role of CTSL in resistant PC-9/GR cell line was investigated by proliferation and apoptosis analysis compared with control PC-9 cells. Our results found that the level of CTSL expression was higher in NSCLC tissues compared with matched normal adjacent tissue samples, and CTSL was more highly expressed in PC-9/GR cells compared to PC-9 cells. Knocking-down of CTSL in PC-9/GR cells could decrease cell proliferation and potentiate apoptosis induced by gefitinib, suggesting CTSL may contribute to gefitinib resistance in NSCLC. CTSL might be explored as a candidate of therapeutic target for modulating EGFR-TKI sensitivity in NSCLC.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/pathology , Cathepsin L/biosynthesis , Drug Resistance, Neoplasm/physiology , Lung Neoplasms/pathology , Quinazolines/therapeutic use , Apoptosis/drug effects , Biomarkers, Tumor/analysis , Blotting, Western , Cathepsin L/analysis , Cell Proliferation/drug effects , Gefitinib , Humans , Immunohistochemistry , Real-Time Polymerase Chain Reaction
8.
J Mol Cell Cardiol ; 86: 32-41, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26163874

ABSTRACT

AIMS: Macrophage inflammation response is important in the pathogenesis of atherosclerosis. We investigated the role and mechanism of cellular repressor of E1A-stimulated genes (CREG) in regulating TNF-α induced inflammation response in macrophages and explore whether CREG might be a therapeutic target for atherosclerosis. METHOD AND RESULTS: Immunostaining and western blotting showed that expression of CREG was reduced in human atherosclerotic coronary artery. In vivo experiments demonstrated that supplementation of recombinant CREG protein to ApoE(-/-) mice fed with high fat diet alleviated aortic atherosclerosis development and inflammation. In vitro, macrophage from ApoE(-/-) mice fed with high fat diet had lower level of CREG compared to control mice fed with normal diet. Immunohistochemical staining and western blotting further confirmed that CREG inhibited inflammatory response of macrophages induced by TNF-α. Supplementation of exogenous recombinant CREG protein or CREG gene silencing showed that CREG promoted autophagy in TNF-α treated macrophages. The use of autophagy inhibitors, 3-methyladenine and bafilomycin A, identified that CREG attenuated TNF-α induced inflammation by activate autophagy. In addition, supplementation of exogenous CREG protein stimulated expression and maturity of cathepsin B and cathepsin L and induced lysosome formation, whereas CREG deficiency reduced lysosomal formation. CONCLUSION: CREG inhibits inflammation and promotes autophagy mediated by lysosome formation; it might be a potential therapeutic target in atherosclerosis.


Subject(s)
Atherosclerosis/genetics , Inflammation/genetics , Repressor Proteins/biosynthesis , Tumor Necrosis Factor-alpha/administration & dosage , Animals , Apolipoproteins E/genetics , Atherosclerosis/pathology , Cathepsin B/biosynthesis , Cathepsin L/biosynthesis , Humans , Inflammation/chemically induced , Inflammation/pathology , Lysosomes/metabolism , Macrophages/metabolism , Macrophages/pathology , Mice , Repressor Proteins/genetics , Tumor Necrosis Factor-alpha/metabolism
9.
Carcinogenesis ; 36(9): 1019-27, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26069256

ABSTRACT

To develop new and effective chemopreventive agents against bone metastasis, we assessed the effects of muscadine grape skin extract (MSKE), whose main bioactive component is anthocyanin, on bone turnover, using prostate and breast cancer cell models overexpressing Snail transcription factor. MSKE has been shown previously to promote apoptosis in prostate cancer cells without affecting normal prostate epithelial cells. Snail is overexpressed in prostate and breast cancer, and is associated with increased invasion, migration and bone turnover/osteoclastogenesis. Cathepsin L (CatL) is a cysteine cathepsin protease that is overexpressed in cancer and involved in bone turnover. Snail overexpression in prostate (LNCaP, ARCaP-E) and breast (MCF-7) cancer cells led to increased CatL expression/activity and phosphorylated STAT-3 (pSTAT-3), compared to Neo vector controls, while the reverse was observed in C4-2 (the aggressive subline of LNCaP) cells with Snail knockdown. Moreover, CatL expression was higher in prostate and breast tumor tissue compared to normal tissue. MSKE decreased Snail and pSTAT3 expression, and abrogated Snail-mediated CatL activity, migration and invasion. Additionally, Snail overexpression promoted osteoclastogenesis, which was significantly inhibited by the MSKE as effectively as Z-FY-CHO, a CatL-specific inhibitor, or osteoprotegerin, a receptor activator of nuclear factor kappa B ligand (RANKL) antagonist. Overall, these novel findings suggest that Snail regulation of CatL may occur via STAT-3 signaling and can be antagonized by MSKE, leading to decreased cell invasion, migration and bone turnover. Therefore, inhibition using a natural product such as MSKE could potentially be a promising bioactive compound for bone metastatic cancer.


Subject(s)
Anticarcinogenic Agents/pharmacology , Bone Neoplasms/prevention & control , Breast Neoplasms/pathology , Cathepsin L/antagonists & inhibitors , Plant Extracts/pharmacology , Prostatic Neoplasms/pathology , Transcription Factors/antagonists & inhibitors , Vitis/chemistry , Animals , Anticarcinogenic Agents/therapeutic use , Apoptosis/drug effects , Bone Neoplasms/secondary , Cathepsin L/biosynthesis , Cathepsin L/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Chemoprevention/methods , Female , Humans , MCF-7 Cells , Male , Mice , Mice, Nude , Neoplasm Invasiveness , Osteoclasts/cytology , Osteogenesis/drug effects , Osteoprotegerin/pharmacology , Plant Extracts/therapeutic use , RANK Ligand/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Snail Family Transcription Factors , Transcription Factors/biosynthesis
10.
J Biol Chem ; 290(25): 15758-15769, 2015 Jun 19.
Article in English | MEDLINE | ID: mdl-25957406

ABSTRACT

The cysteine protease cathepsin L (CTSL) is often thought to act as a tumor promoter by enhancing tumor progression and metastasis. This goes along with increased CTSL activity in various tumor entities; however, the mechanisms leading to high CTSL levels are incompletely understood. With the help of the polyoma middle T oncogene driven breast cancer mouse model expressing a human CTSL genomic transgene, we show that CTSL indeed promotes breast cancer metastasis to the lung. During tumor formation and progression high expression levels of CTSL are maintained by enduring translation of CTSL mRNA. Interestingly, human breast cancer specimens expressed the same pattern of 5' untranslated region (UTR) splice variants as the transgenic mice and the human cancer cell line MDA-MB 321. By polyribosome profiling of tumor tissues and human breast cancer cells, we observe an intrinsic resistance of CTSL to stress-induced shutdown of translation. This ability can be attributed to all 5' UTR variants of CTSL and is not dependent on a previously described internal ribosomal entry site motif. In conclusion, we provide in vivo functional evidence for overexpressed CTSL as a promoter of lung metastasis, whereas high CTSL levels are maintained during tumor progression due to stress-resistant mRNA translation.


Subject(s)
5' Untranslated Regions , Breast Neoplasms/metabolism , Cathepsin L/biosynthesis , Lung Neoplasms/metabolism , Mammary Neoplasms, Experimental/metabolism , Protein Biosynthesis , RNA, Neoplasm/metabolism , Stress, Physiological , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cathepsin L/genetics , Cell Line, Tumor , Female , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Transgenic , Neoplasm Metastasis , RNA, Neoplasm/genetics
11.
Free Radic Biol Med ; 65: 1155-1163, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23999505

ABSTRACT

Advanced glycation end product (AGE)-modified proteins are formed by the nonenzymatic glycation of free amino groups of proteins and, along with lipofuscin (a highly oxidized aggregate of covalently cross-linked proteins, sugars, and lipids), have been found to accumulate during aging and in several age-related diseases. As the in vivo effects of diet-derived AGEs or lipofuscin remain elusive, we sought to study the impact of oral administration of glucose-, fructose-, or ribose-modified albumin or of artificial lipofuscin in a genetically tractable model organism. We report herein that continuous feeding of young Drosophila flies with culture medium enriched in AGEs or in lipofuscin resulted in reduced locomotor performance and in accelerated rates of AGE-modified proteins and carbonylated proteins accumulation in the somatic tissues and hemolymph of flies, as well as in a significant reduction of flies health span and life span. These phenotypic effects were accompanied by reduced proteasome peptidase activities in both the hemolymph and the somatic tissues of flies and higher levels of oxidative stress; furthermore, oral administration of AGEs or lipofuscin in flies triggered an upregulation of the lysosomal cathepsin B, L activities. Finally, RNAi-mediated cathepsin D knockdown reduced flies longevity and significantly augmented the deleterious effects of AGEs and lipofuscin, indicating that lysosomal cathepsins reduce the toxicity of diet-derived AGEs or lipofuscin. Our in vivo studies demonstrate that chronic ingestion of AGEs or lipofuscin disrupts proteostasis and accelerates the functional decline that occurs with normal aging.


Subject(s)
Aging/drug effects , Drosophila melanogaster/metabolism , Glycation End Products, Advanced/pharmacology , Lipofuscin/pharmacology , Protein Folding/drug effects , Albumins/chemistry , Animals , Animals, Genetically Modified , Cathepsin B/biosynthesis , Cathepsin B/metabolism , Cathepsin D/genetics , Cathepsin L/biosynthesis , Cathepsin L/metabolism , Diet , Fructose/chemistry , Glucose/chemistry , Glycation End Products, Advanced/administration & dosage , Glycation End Products, Advanced/chemistry , Glycosylation , Lipofuscin/administration & dosage , Lipofuscin/chemistry , Longevity/drug effects , Oxidation-Reduction , Oxidative Stress/drug effects , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors , RNA Interference , RNA, Small Interfering , Ribose/chemistry , Up-Regulation
12.
Cancer Invest ; 31(7): 461-71, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23915070

ABSTRACT

In the present study, we assessed the expression of extracellular matrix (ECM) degrading proteases-cathepsin L and matrix metalloprotease-2 (MMP-2) in pancreatic cancer tissue and correlated their levels with clinicopathological parameters and survival. Both the proteases were expressed in the majority of the tumor tissues examined. Staining intensity of cathepsin L was significantly higher in the tumor stroma compared to tumor epithelium while MMP-2 staining showed no such difference. Both proteases showed correlation with some of the clinicopathological parameters but only cathepsin L expression in tumor epithelium predicted a poor prognosis for the disease.


Subject(s)
Adenocarcinoma/enzymology , Biomarkers, Tumor/analysis , Cathepsin L/analysis , Matrix Metalloproteinase 2/analysis , Pancreatic Neoplasms/enzymology , Adenocarcinoma/mortality , Adult , Aged , Cathepsin L/biosynthesis , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Matrix Metalloproteinase 2/biosynthesis , Middle Aged , Pancreatic Neoplasms/mortality , Prognosis , Proportional Hazards Models
13.
J Immunol ; 191(3): 1200-9, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23794633

ABSTRACT

Thymic epithelial cells (TECs) help orchestrate thymopoiesis, and TEC differentiation relies on bidirectional interactions with thymocytes. Although the molecular mediators that stimulate medullary thymic epithelial cell (mTEC) maturation are partially elucidated, the signals that regulate cortical thymic epithelial cell (cTEC) homeostasis remain elusive. Using IL-7 reporter mice, we show that TECs coexpressing high levels of IL-7 (Il7(YFP+) TECs) reside within a subset of CD205(+)Ly51(+)CD40(low) cTECs that coexpresses Dll4, Ccl25, Ccrl1, Ctsl, Psmb11, and Prss16 and segregates from CD80(+)CD40(high) mTECs expressing Tnfrsf11a, Ctss, and Aire. As the frequency of Il7(YFP+) TECs gradually declines as mTEC development unfolds, we explored the relationship between Il7(YFP+) TECs and mTECs. In thymic organotypic cultures, the thymocyte-induced reduction in Il7(YFP+) TECs dissociates from the receptor activator of NF-κB-mediated differentiation of CD80(+) mTECs. Still, Il7(YFP+) TECs can generate some CD80(+) mTECs in a stepwise differentiation process via YFP(-)Ly51(low)CD80(low) intermediates. Il7(YFP+) TECs are sustained in Rag2(-/-) mice, even following in vivo anti-CD3ε treatment that mimics the process of pre-TCR ß-selection of thymocytes to the double positive (DP) stage. Using Marilyn-Rag2(-/-) TCR transgenic, we find that positive selection into the CD4 lineage moderately reduces the frequency of Il7(YFP+) TECs, whereas negative selection provokes a striking loss of Il7(YFP+) TECs. These results imply that the strength of MHC/peptide-TCR interactions between TECs and thymocytes during selection constitutes a novel rheostat that controls the maintenance of IL-7-expressing cTECs.


Subject(s)
Epithelial Cells/immunology , Interleukin-7/biosynthesis , Thymus Gland/immunology , Adaptor Proteins, Signal Transducing , Animals , Antigens, CD/metabolism , B7-1 Antigen/metabolism , CD3 Complex/immunology , CD40 Antigens/metabolism , Calcium-Binding Proteins , Cathepsin L/biosynthesis , Cathepsins/biosynthesis , Cell Differentiation/immunology , Cell Movement , Cells, Cultured , Chemokines, CC/biosynthesis , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Epithelial Cells/metabolism , Homeostasis , Interleukin-7/metabolism , Intracellular Signaling Peptides and Proteins/biosynthesis , Lectins, C-Type/metabolism , Lymphocyte Activation , Membrane Proteins/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Minor Histocompatibility Antigens , Organ Culture Techniques , Receptor Activator of Nuclear Factor-kappa B/biosynthesis , Receptor Activator of Nuclear Factor-kappa B/metabolism , Receptors, CCR/biosynthesis , Receptors, Cell Surface/metabolism , Serine Endopeptidases/biosynthesis , Thymocytes/metabolism , Thymus Gland/metabolism , Transcription Factors/biosynthesis , AIRE Protein
14.
Clin Lab ; 57(7-8): 535-41, 2011.
Article in English | MEDLINE | ID: mdl-21888018

ABSTRACT

BACKGROUND: Cyclosporin A (CsA) is an immunosuppressant with side effects including gingival hyperplasia. Sarcoidosis is a systemic disease characterized by granulomas. Here, we report on a rare case of sarcoidosis with gingival hyperplasia to clarify whether clinical observation corresponds to in vitro results. METHODS: Gingival fibroblasts (HGFs) were isolated from healthy gingiva and cultured with CsA. Total RNA was collected and expression of mRNAs examined using semi-quantitative RT-PCR analysis. Cathepsin B, D, and L expression in overgrown gingiva of the patient was examined by immunohistochemistry. RESULTS: Cathepsin D, L, and vascular endothelial growth factor (VEGF)165 mRNA were markedly suppressed in CsA-treated HGFs, whereas cathepsin B, matrix metalloproteinase-1 (MMP-1) and tissue inhibitor of metalloproteinase-1 (TIMP-1) mRNA were not reduced. Next, the decrease of cathepsin B and L expression in enlarged gingiva was observed, whereas an increase of cathepsin D expression was observed. Clinically, the enlarged gingival lesions were fully resolved by performing oral infection control. CONCLUSIONS: Cathepsins regulation might be an important factor in the development of CsA-mediated gingival hyperplasia.


Subject(s)
Cathepsin B/genetics , Cathepsin D/genetics , Cathepsin L/genetics , Cyclosporine/adverse effects , Gene Expression Regulation/drug effects , Gingival Hyperplasia/metabolism , Immunosuppressive Agents/adverse effects , Sarcoidosis/drug therapy , Vascular Endothelial Growth Factor A/genetics , Adrenal Cortex Hormones/administration & dosage , Adrenal Cortex Hormones/therapeutic use , Bacteroidaceae Infections/complications , Cathepsin B/biosynthesis , Cathepsin D/biosynthesis , Cathepsin L/biosynthesis , Cyclosporine/administration & dosage , Cyclosporine/pharmacology , Cyclosporine/therapeutic use , Dental Scaling , Drug Therapy, Combination , Enzyme Induction/drug effects , Female , Gingival Hyperplasia/chemically induced , Gingival Hyperplasia/etiology , Gingival Hyperplasia/prevention & control , Gingivitis/complications , Gingivitis/microbiology , Gingivitis/therapy , Humans , Immunocompromised Host , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Matrix Metalloproteinase 1/biosynthesis , Matrix Metalloproteinase 1/genetics , Middle Aged , Oral Hygiene , Porphyromonas gingivalis/isolation & purification , Sarcoidosis/complications , Tissue Inhibitor of Metalloproteinase-1/biosynthesis , Tissue Inhibitor of Metalloproteinase-1/genetics , Treponema denticola/isolation & purification , Vascular Endothelial Growth Factor A/biosynthesis
15.
Blood ; 118(15): 4199-208, 2011 Oct 13.
Article in English | MEDLINE | ID: mdl-21846901

ABSTRACT

Alternatively activated macrophages, generated in a T-helper 2 environment, have demonstrated roles in wound repair and tissue remodeling in addition to being charged with immune tasks. Because the hydrolytic chemistries of the phagosomal lumen are central to many of these functions, we investigated their modification after alternative activation with IL-4 and IL-13. Most significantly, we found striking up-regulation of the proteolytic levels within the phagosome of IL-4-activated macrophages. Two synergistic mechanisms were determined to underlie this up-regulation. First, IL-4-activated macrophages displayed increased expression of cathepsin S and L, providing greater proteolytic machinery to the phagosome despite unchanged rates of lysosomal contribution. Secondly, decreased phagosomal NADPH oxidase (NOX2) activity, at least partially resulting from decreased expression of the NOX2 subunit gp91(phox), resulted in a more reductive lumenal microenvironment, which in turn, enhanced activities of local cysteine cathepsins. Decreased NOX2 activity additionally increased the phagosome's ability to reduce disulfides, further enhancing the efficiency of the macrophage to degrade proteins containing disulfide bonds. Together, these changes initiated by IL-4 act synergistically to rapidly and dramatically enhance the macrophage's ability to degrade phagocytosed protein, which, we reason, better equips this cell for its roles in wound repair and tissue remodeling.


Subject(s)
Interleukin-4/immunology , Macrophage Activation/immunology , Macrophages/immunology , Phagosomes/immunology , Proteolysis , Th2 Cells/immunology , Animals , Cathepsin L/biosynthesis , Cathepsin L/genetics , Cathepsin L/immunology , Cathepsins/biosynthesis , Cathepsins/genetics , Cathepsins/immunology , Gene Expression Regulation, Enzymologic/genetics , Gene Expression Regulation, Enzymologic/immunology , Interleukin-13/genetics , Interleukin-13/immunology , Interleukin-13/metabolism , Interleukin-4/genetics , Interleukin-4/metabolism , Macrophage Activation/genetics , Macrophages/enzymology , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Knockout , NADPH Oxidase 2 , NADPH Oxidases/biosynthesis , NADPH Oxidases/genetics , NADPH Oxidases/immunology , Phagosomes/enzymology , Phagosomes/genetics , Th2 Cells/metabolism , Wound Healing/genetics , Wound Healing/immunology
16.
EMBO J ; 30(16): 3383-96, 2011 Jul 12.
Article in English | MEDLINE | ID: mdl-21750527

ABSTRACT

Genomic instability due to telomere dysfunction and defective repair of DNA double-strand breaks (DSBs) is an underlying cause of ageing-related diseases. 53BP1 is a key factor in DNA DSBs repair and its deficiency is associated with genomic instability and cancer progression. Here, we uncover a novel pathway regulating the stability of 53BP1. We demonstrate an unprecedented role for the cysteine protease Cathepsin L (CTSL) in the degradation of 53BP1. Overexpression of CTSL in wild-type fibroblasts leads to decreased 53BP1 protein levels and changes in its cellular distribution, resulting in defective repair of DNA DSBs. Importantly, we show that the defects in DNA repair associated with 53BP1 deficiency upon loss of A-type lamins are due to upregulation of CTSL. Furthermore, we demonstrate that treatment with vitamin D stabilizes 53BP1 and promotes DNA DSBs repair via inhibition of CTSL, providing an as yet unsuspected link between vitamin D action and DNA repair. Given that CTSL upregulation is a hallmark of cancer and progeria, regulation of this pathway could be of great therapeutic significance for these diseases.


Subject(s)
Cathepsin L/physiology , Chromosomal Proteins, Non-Histone/metabolism , DNA Repair/physiology , DNA-Binding Proteins/metabolism , Lamin Type A/physiology , Vitamin D/physiology , Animals , Calcitriol/pharmacology , Cathepsin L/antagonists & inhibitors , Cathepsin L/biosynthesis , Cathepsin L/genetics , Cell Line , Enzyme Precursors/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Lamin Type A/deficiency , Lamin Type A/genetics , Leupeptins/pharmacology , Mice , Proteasome Endopeptidase Complex/metabolism , Protein Stability , Recombinant Fusion Proteins/physiology , Species Specificity , Transfection , Tumor Suppressor p53-Binding Protein 1
17.
Fish Shellfish Immunol ; 29(6): 1010-8, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20732429

ABSTRACT

Cathepsins, a superfamily of hydrolytic enzymes produced and enclosed within lysosomes, function in immune response in vertebrates; however, their function within the innate immune system of invertebrates remains largely unknown. Therefore, we investigated the immune functionality of cathepsin L (catL) in Chinese mitten crab (Eriocheir sinensis), a commercially important and disease vulnerable aquaculture species. The full-length catL cDNA (1274 bp) was cloned via PCR based upon an initial expressed sequence tag (EST) isolated from a hepatopancreatic cDNA library. The catL cDNA contained a 978 bp open reading frame (ORF) that encoded a putative 325 amino acid (aa) protein. Comparisons with other reported invertebrate and vertebrate sequences revealed conserved gene structure and enzyme active sites common among papain-like cysteine proteases, and high percent identity among other invertebrate cathepsins. CatL mRNA expression in E. sinensis was (a) tissue-specific, with the highest expression observed in hepatotpancreas, gill, stomach, and hemocytes, and (b) responsive in hemocytes to a Vibrio anguillarum challenge, the catL expression level and enzyme activity both with peak exposure observed 8 h post-injection. Collectively, data demonstrate the successful isolation of catL from the Chinese mitten crab, and its involvement in the innate immune system of an invertebrate.


Subject(s)
Brachyura/enzymology , Brachyura/genetics , Cathepsin L/genetics , Amino Acid Sequence , Animals , Base Sequence , Brachyura/immunology , Brachyura/microbiology , Cathepsin L/biosynthesis , Cathepsin L/immunology , Cloning, Molecular , Cricetinae , Cricetulus , Immunity, Innate/genetics , Immunity, Innate/immunology , Molecular Sequence Data , Phylogeny , RNA/chemistry , RNA/genetics , Random Amplified Polymorphic DNA Technique , Reverse Transcriptase Polymerase Chain Reaction , Sequence Alignment , Sequence Analysis, DNA , Vibrio/immunology , Vibrio Infections/immunology , Vibrio Infections/microbiology
18.
Exp Parasitol ; 125(4): 371-9, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20214898

ABSTRACT

The digestive tract of Fasciola gigantica is composed of the oral sucker, buccal tube, pharynx, esophagus, and caecum. The tegumental-type epithelium lines the first four parts of the digestive tract while the caecal-type epithelium lines the remaining parts from the caecal bifurcation. The caecal-epithelial cells are classified into 3 types according to their staining properties and ultrastructural characteristics, as related to the amount of food contents in the caecal lumen. All caecal-type epithelial cells synthesize and secrete cathepsin L, a major group of enzymes in the digestive tract, as detected by in situ hybridization and immunolocalization. Moreover, the secreted cathepsin L is also adsorbed on the outer surface of the tegument and the glycocalyx coating of the surface of the tegument, whereas the tegumental cells and tegumental syncytium covering the parasite's body and lining the proximal part of the digestive tract exhibit no in situ hybridization signal and immunostaining for cathepsin L.


Subject(s)
Cathepsin L/genetics , Fasciola/anatomy & histology , Animals , Cathepsin L/biosynthesis , Cattle , Fasciola/enzymology , Fasciola/genetics , Fluorescent Antibody Technique , Gastrointestinal Tract/anatomy & histology , Gastrointestinal Tract/ultrastructure , Gene Expression Regulation, Enzymologic , Immunohistochemistry , In Situ Hybridization , Microscopy, Electron, Transmission , RNA, Messenger/metabolism
19.
Atherosclerosis ; 208(1): 83-9, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19671471

ABSTRACT

OBJECTIVE: The cysteine protease, legumain, is thought to have a role in the processing and activation of proteases such as cathepsin-L, which have been implicated in plaque rupture. This study aimed to determine: if legumain activity is up-regulated in unstable areas of plaque; the effect of legumain over-expression on the activity of cathepsin-L and the effect of mutation of the legumain RGD sequence on its cellular location. METHODS AND RESULTS: Legumain was measured in human carotid plaque extracts (n=17) using a novel ELISA and modified activity assay. Unstable regions of plaque contained more than twice the amount of legumain protein (P<0.001) and activity (P<0.03) compared with stable regions of the same plaque. Over-expression of legumain in THP-1 macrophages using an adenoviral construct resulted in the processing of cathepsin-L from its 30kDa to its 25kDa form compared with controls. CONCLUSION: Unstable regions of plaque contain increased levels of active legumain. Over-expression of legumain in macrophages alters intracellular processing of cathepsin-L to its mature 25kDa form. This may be a means by which legumain could contribute to plaque instability.


Subject(s)
Carotid Artery Diseases/metabolism , Cathepsin L/biosynthesis , Cysteine Endopeptidases/biosynthesis , Humans , In Vitro Techniques
20.
Am J Physiol Regul Integr Comp Physiol ; 298(2): R341-50, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20007517

ABSTRACT

The effects of insulin-like growth factor-I (IGF-I), insulin, and leucine on protein turnover and pathways that regulate proteolytic gene expression and protein polyubiquitination were investigated in primary cultures of 4-day-old rainbow trout myocytes. Supplementing media with 100 nM IGF-I increased protein synthesis by 13% (P < 0.05) and decreased protein degradation by 14% (P < 0.05). Treatment with 1 microM insulin increased protein synthesis by 13% (P < 0.05) and decreased protein degradation by 17% (P < 0.05). Supplementing media containing 0.6 mM leucine with an additional 2.5 mM leucine did not increase protein synthesis rates but reduced rates of protein degradation by 8% (P < 0.05). IGF-I (1 nM-100 nM) and insulin (1 nM-1 microM) independently reduced the abundance of ubiquitin ligase mRNA in a dose-dependent manner, with maximal reductions of approximately 70% for muscle atrophy F-box (Fbx) 32, 40% for Fbx25, and 25% for muscle RING finger-1 (MuRF1, P < 0.05). IGF-I and insulin stimulated phosphorylation of FOXO1 and FOXO4 (P < 0.05), which was inhibited by the phosphatidylinositol 3-kinase (PI 3-kinase) inhibitor wortmannin, and decreased the abundance of polyubiquitinated proteins by 10-20% (P < 0.05). Supplementing media with leucine reduced Fbx32 expression by 25% (P < 0.05) but did not affect Fbx25 nor MuRF1 transcript abundance. Serum deprivation decreased rates of protein synthesis by 60% (P < 0.05), increased protein degradation by 40% (P < 0.05), and increased expression of all ubiquitin ligases. These data suggest that, similar to mammals, the inhibitory effects of IGF-I and insulin on proteolysis occur via P I3-kinase/protein kinase B signaling and are partially responsible for the ability of these compounds to promote protein accretion.


Subject(s)
Hypoglycemic Agents/pharmacology , Insulin-Like Growth Factor I/pharmacology , Insulin/pharmacology , Leucine/pharmacology , Monocytes/metabolism , Oncorhynchus mykiss/metabolism , Proteins/metabolism , Ubiquitin-Protein Ligases/biosynthesis , Anabolic Agents/pharmacology , Animals , Blotting, Western , Cathepsin D/biosynthesis , Cathepsin D/genetics , Cathepsin L/biosynthesis , Cathepsin L/genetics , Cell Separation , Culture Media , Culture Media, Serum-Free , Dose-Response Relationship, Drug , F-Box Proteins/metabolism , Monocytes/drug effects , Monocytes/enzymology , Phosphatidylinositol 3-Kinases/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...