Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 138
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Biochim Biophys Acta Mol Cell Res ; 1865(1): 25-33, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29024694

ABSTRACT

Epithelial ovarian cancer (EOC) frequently metastasises to the omentum, a process that requires pro-angiogenic activation of human omental microvascular endothelial cells (HOMECs) by tumour-secreted factors. We have previously shown that ovarian cancer cells secrete a range of factors that induce pro-angiogenic responses e.g. migration, in HOMECs including the lysosomal protease cathepsin D (CathD). However, the cellular mechanism by which CathD induces these cellular responses is not understood. The aim of this study was to further examine the pro-angiogenic effects of CathD in HOMECs i.e. proliferation and migration, to investigate whether these effects are dependent on CathD catalytic activity and to delineate the intracellular signalling kinases activated by CathD. We report, for the first time, that CathD significantly increases HOMEC proliferation and migration via a non-proteolytic mechanism resulting in activation of ERK1/2 and AKT. These data suggest that EOC cancer secreted CathD acts as an extracellular ligand and may play an important pro-angiogenic, and thus pro-metastatic, role by activating the omental microvasculature during EOC metastasis to the omentum.


Subject(s)
Cathepsin D/physiology , Cell Movement/genetics , Cell Proliferation/genetics , Endothelial Cells/physiology , Omentum/cytology , Carcinoma, Ovarian Epithelial , Cathepsin D/genetics , Cells, Cultured , Endothelial Cells/cytology , Female , Humans , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase 3/metabolism , Neoplasm Metastasis , Neoplasms, Glandular and Epithelial/metabolism , Neoplasms, Glandular and Epithelial/pathology , Omentum/blood supply , Omentum/pathology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Peritoneal Neoplasms/genetics , Peritoneal Neoplasms/metabolism , Peritoneal Neoplasms/secondary , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology
2.
J Am Soc Nephrol ; 27(9): 2685-700, 2016 09.
Article in English | MEDLINE | ID: mdl-26823550

ABSTRACT

Studies have revealed many analogies between podocytes and neurons, and these analogies may be key to elucidating the pathogenesis of podocyte injury. Cathepsin D (CD) is a representative aspartic proteinase in lysosomes. Central nervous system neurons in CD-deficient mice exhibit a form of lysosomal storage disease with a phenotype resembling neuronal ceroid lipofuscinoses. In the kidney, the role of CD in podocytes has not been fully explored. Herein, we generated podocyte-specific CD-knockout mice that developed proteinuria at 5 months of age and ESRD by 20-22 months of age. Immunohistochemical analysis of these mice showed apoptotic podocyte death followed by proteinuria and glomerulosclerosis with aging. Using electron microscopy, we identified, in podocytes, granular osmiophilic deposits (GRODs), autophagosome/autolysosome-like bodies, and fingerprint profiles, typical hallmarks of CD-deficient neurons. CD deficiency in podocytes also led to the cessation of autolysosomal degradation and accumulation of proteins indicative of autophagy impairment and the mitochondrial ATP synthase subunit c accumulation in the GRODs, again similar to changes reported in CD-deficient neurons. Furthermore, both podocin and nephrin, two essential components of the slit diaphragm, translocated to Rab7- and lysosome-associated membrane glycoprotein 1-positive amphisomes/autolysosomes that accumulated in podocyte cell bodies in podocyte-specific CD-knockout mice. We hypothesize that defective lysosomal activity resulting in foot process effacement caused this accumulation of podocin and nephrin. Overall, our results suggest that loss of CD in podocytes causes autophagy impairment, triggering the accumulation of toxic subunit c-positive lipofuscins as well as slit diaphragm proteins followed by apoptotic cell death.


Subject(s)
Cathepsin D/physiology , Podocytes , Proteinuria/etiology , Renal Insufficiency, Chronic/etiology , Animals , Mice , Mice, Knockout , Podocytes/pathology
3.
Tumour Biol ; 37(8): 10685-96, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26867770

ABSTRACT

Cathepsin D (CD), a ubiquitously expressed lysosomal aspartic protease, is upregulated in human breast carcinoma and many other tumor types. CD has been repeatedly reported to act as key mediator of apoptosis induced by various chemotherapeutics. However, there is still controversy over the role of enzymatic/proteolytic versus protein-protein interaction activities of CD in apoptotic signaling. The elucidation of molecular mechanism responsible for the effect of CD in the chemotherapy-induced cell death is crucial for development of an appropriate strategy to target this protease in cancer treatment. Therefore, the objective of this study was to investigate the molecular mechanism behind the CD-mediated regulation of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced cell death. For this purpose, MDA-MB-231 breast carcinoma cells with an increased level of wt CD (CD) or mutant enzymatically inactive CD (ΔCD) were subjected to TRAIL and the frequency of apoptosis was determined. Our results show that CD facilitates the TRAIL-induced apoptosis of MDA-MB-231 breast cancer cells in enzymatic activity-dependent manner. Moreover, the importance of endosomal/lysosomal acidification in this process was documented. Analysis of the potential substrates specifically cleaved by CD during the TRAIL-induced apoptosis confirmed caspase-8 and Bid proteins as the CD targets. Moreover, in search for protein regulators of apoptosis that can be cleaved by CD at physiologically relevant pH, we identified the Bcl-2 protein as a suitable candidate. The modulatory role of CD in cell response to TRAIL was also confirmed in another breast cancer cell line SKBR3. These experiments identified the CD enzymatic activity as a new factor affecting sensitivity of breast cancer cells to TRAIL.


Subject(s)
Adenocarcinoma/pathology , Breast Neoplasms/pathology , Cathepsin D/physiology , Neoplasm Proteins/physiology , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Adenocarcinoma/enzymology , Apoptosis/drug effects , BH3 Interacting Domain Death Agonist Protein/metabolism , Breast Neoplasms/enzymology , Caspase 8/metabolism , Cathepsin D/antagonists & inhibitors , Cathepsin D/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm , Endosomes/metabolism , Enzyme Activation , Female , Humans , Hydrogen-Ion Concentration , Lysosomes/metabolism , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA Interference , RNA, Small Interfering/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Transfection
4.
J Sci Food Agric ; 96(9): 3233-9, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26493936

ABSTRACT

BACKGROUND: Texture deterioration often negatively affects sensory attributes and commercial values of ice-stored fish fillets. The mechanism of softening of fish fillets during chilling storage is not fully resolved. Grass carp is a predominant freshwater fish species in China. The objective of the present study was to investigate the differential role of endogenous cathepsin and microorganisms in texture softening of ice-stored grass carp fillets. RESULTS: The fillets were immersed in either NaN3 solution to reduce microbial activity or in iodoacetic acid solution to exclude cathepsin activity before ice storage. Treatment with NaN3 reduced microbial load of fillets below 2 log CFU g(-1) muscle during the entire storage period, and had no significant influence on the cathepsin activity and proteolysis. But the shear force of fillets treated with NaN3 decreased by 66% after 21 days of storage. Meanwhile, treatment with iodoacetic acid inactivated cathepsin B and B + L but did not significantly affect the microbial growth of fillets. Compared to NaN3 treatment, iodoacetic acid effectively alleviated softening and inhibited the increase in TCA-soluble peptides during storage. CONCLUSION: This study demonstrated that proteolysis induced by endogenous cathepsins, rather than microorganisms, plays an important role in texture softening of ice-stored grass carp fillets. © 2015 Society of Chemical Industry.


Subject(s)
Carps/microbiology , Cathepsins/metabolism , Cathepsins/physiology , Food Preservation/methods , Seafood , Animals , Bacteria/drug effects , Cathepsin B/metabolism , Cathepsin D/metabolism , Cathepsin D/physiology , Cathepsin L/metabolism , Cathepsin L/pharmacology , China , Cold Temperature , Fish Proteins/metabolism , Food Storage , Ice , Iodoacetic Acid/pharmacology , Proteolysis , Sodium Azide/pharmacology
5.
Gastroenterology ; 142(5): 1160-71, 2012 May.
Article in English | MEDLINE | ID: mdl-22333951

ABSTRACT

BACKGROUND & AIMS: The Helicobacter pylori toxin vacuolating cytotoxin (VacA) promotes gastric colonization, and its presence (VacA(+)) is associated with more-severe disease. The exact mechanisms by which VacA contributes to infection are unclear. We previously found that limited exposure to VacA induces autophagy of gastric cells, which eliminates the toxin; we investigated whether autophagy serves as a defense mechanism against H pylori infection. METHODS: We investigated the effect of VacA on autophagy in human gastric epithelial cells and primary gastric cells from mice. Expression of p62, a marker of autophagy, was also assessed in gastric tissues from patients infected with toxigenic (VacA(+)) or nontoxigenic strains. We analyzed the effect of VacA on autophagy in peripheral blood monocytes obtained from subjects with different genotypes of ATG16L1, which regulates autophagy. We performed genotyping for ATG16L1 in 2 cohorts of infected and uninfected subjects. RESULTS: Prolonged exposure of human gastric epithelial cells and mouse gastric cells to VacA disrupted induction of autophagy in response to the toxin, because the cells lacked cathepsin D in autophagosomes. Loss of autophagy resulted in the accumulation of p62 and reactive oxygen species. Gastric biopsy samples from patients infected with VacA(+), but not nontoxigenic strains of H pylori, had increased levels of p62. Peripheral blood monocytes isolated from individuals with polymorphisms in ATG16L1 that increase susceptibility to Crohn's disease had reduced induction of autophagy in response to VacA(+) compared to cells from individuals that did not have these polymorphisms. The presence of the ATG16L1 Crohn's disease risk variant increased susceptibility to H pylori infection in 2 separate cohorts. CONCLUSIONS: Autophagy protects against infection with H pylori; the toxin VacA disrupts autophagy to promote infection, which could contribute to inflammation and eventual carcinogenesis.


Subject(s)
Autophagy/physiology , Bacterial Proteins/physiology , Helicobacter Infections/etiology , Helicobacter pylori , Alleles , Animals , Bacterial Proteins/genetics , Cathepsin D/physiology , Crohn Disease/etiology , Crohn Disease/genetics , Genotype , Humans , Immunity, Innate , Mice , Phagosomes/physiology
6.
J Immunol ; 187(3): 1475-85, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21709160

ABSTRACT

The peptide F2L was previously characterized as a high-affinity natural agonist for the human formyl peptide receptor (FPR) 3. F2L is an acetylated 21-aa peptide corresponding with the N terminus of the intracellular heme-binding protein 1 (HEBP1). In the current work, we have investigated which proteases were able to generate the F2L peptide from its precursor HEBP1. Structure-function analysis of F2L identified three amino acids, G(3), N(7), and S(8), as the most important for interaction of the peptide with FPR3. We expressed a C-terminally His-tagged form of human HEBP1 in yeast and purified it to homogeneity. The purified protein was used as substrate to identify proteases generating bioactive peptides for FPR3-expressing cells. A conditioned medium from human monocyte-derived macrophages was able to generate bioactivity from HEBP1, and this activity was inhibited by pepstatin A. Cathepsin D was characterized as the protease responsible for HEBP1 processing, and the bioactive product was identified as F2L. We have therefore determined how F2L, the specific agonist of FPR3, is generated from the intracellular protein HEBP1, although it is unknown in which compartment the processing by cathepsin D occurs in vivo.


Subject(s)
Carrier Proteins/metabolism , Cathepsin D/physiology , Chemotactic Factors/agonists , Hemeproteins/metabolism , Peptides/agonists , Protein Precursors/metabolism , Protein Processing, Post-Translational/immunology , Receptors, Formyl Peptide/metabolism , Amino Acid Sequence , Animals , CHO Cells , Carrier Proteins/biosynthesis , Cathepsin D/deficiency , Cells, Cultured , Chemotactic Factors/biosynthesis , Chemotactic Factors/metabolism , Cricetinae , Cricetulus , Heme-Binding Proteins , Hemeproteins/biosynthesis , Humans , Ligands , Macrophages/enzymology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Neutrophils/enzymology , Neutrophils/immunology , Neutrophils/metabolism , Peptides/metabolism , Protein Binding/immunology , Protein Precursors/biosynthesis , Receptors, Formyl Peptide/biosynthesis
7.
J Immunol ; 187(1): 240-7, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21632707

ABSTRACT

It is well established that dendritic cells (DCs) take up, process, and present lipid Ags in complex with CD1d molecules to invariant NKT cells. The lipid-activated transcription factor, peroxisome proliferator-activated receptor γ (PPARγ), has previously been shown to regulate CD1d expression in human monocyte-derived DCs, providing a link between lipid metabolism and lipid Ag presentation. We report that PPARγ regulates the expression of a lysosomal protease, cathepsin D (CatD), in human monocyte-derived DCs. Inhibition of CatD specifically reduced the expansion of invariant NKT cells and furthermore resulted in decreased maturation of saposins, a group of lipid transfer proteins required for lysosomal lipid Ag processing and loading. These results reveal a novel mechanism of lipid Ag presentation and identify CatD as a key component of this machinery and firmly place PPARγ as the transcriptional regulator linking lipid metabolism and lipid Ag processing.


Subject(s)
Antigen Presentation/immunology , Cathepsin D/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Lipoproteins/metabolism , PPAR gamma/physiology , Antigens, CD1d/immunology , Antigens, CD1d/metabolism , Cathepsin D/biosynthesis , Cathepsin D/physiology , Cells, Cultured , Coculture Techniques , Humans , Lipid Metabolism/immunology , Lipoproteins/immunology , Lysosomes/enzymology , Lysosomes/metabolism , Monocytes/immunology , Monocytes/metabolism , Natural Killer T-Cells/enzymology , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Saposins/metabolism , Saposins/physiology , Signal Transduction/immunology , Up-Regulation/immunology
8.
PLoS Genet ; 6(7): e1001026, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20664788

ABSTRACT

Expansion of the lysosomal system, including cathepsin D upregulation, is an early and prominent finding in Alzheimer's disease brain. Cell culture studies, however, have provided differing perspectives on the lysosomal connection to Alzheimer's disease, including both protective and detrimental influences. We sought to clarify and molecularly define the connection in vivo in a genetically tractable model organism. Cathepsin D is upregulated with age in a Drosophila model of Alzheimer's disease and related tauopathies. Genetic analysis reveals that cathepsin D plays a neuroprotective role because genetic ablation of cathepsin D markedly potentiates tau-induced neurotoxicity. Further, generation of a C-terminally truncated form of tau found in Alzheimer's disease patients is significantly increased in the absence of cathepsin D. We show that truncated tau has markedly increased neurotoxicity, while solubility of truncated tau is decreased. Importantly, the toxicity of truncated tau is not affected by removal of cathepsin D, providing genetic evidence that modulation of neurotoxicity by cathepsin D is mediated through C-terminal cleavage of tau. We demonstrate that removing cathepsin D in adult postmitotic neurons leads to aberrant lysosomal expansion and caspase activation in vivo, suggesting a mechanism for C-terminal truncation of tau. We also demonstrate that both cathepsin D knockout mice and cathepsin D-deficient sheep show abnormal C-terminal truncation of tau and accompanying caspase activation. Thus, caspase cleavage of tau may be a molecular mechanism through which lysosomal dysfunction and neurodegeneration are causally linked in Alzheimer's disease.


Subject(s)
Cathepsin D/physiology , Lysosomes/pathology , Neurotoxicity Syndromes/etiology , tau Proteins/metabolism , Alzheimer Disease , Animals , Caspases/metabolism , Drosophila , Lysosomes/metabolism , Mice , Mice, Knockout , Neurons/pathology , Sheep
9.
Poult Sci ; 91(9): 2288-93, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22912465

ABSTRACT

Egg yolk constitutes the main storage compartment of the avian egg and the first nutritional source that supports embryonic growth. Most egg yolk components are synthesized by the liver of laying hens at sexual maturity and are secreted into the blood to be further transferred into the ovarian oocyte (yolky follicle) by receptor-mediated endocytosis. Egg yolk proteins are secreted as precursors and must undergo proteolytic processing to be bioactive. It is assumed that chicken cathepsin D, an aspartic protease, is a key enzyme in this process. Very recently, a novel aspartic protease, namely "similar to nothepsin," has been identified in the egg yolk. Previous experiments conducted in Antarctic fish have shown that the expression of nothepsin is tissue- and sex-specific. To gain insight into the specificities of expression of both cathepsin D and "similar to nothepsin" in Gallus gallus, we compared their distribution in various tissues, in male and females. Cathepsin D is ubiquitously expressed in all tissues examined, including liver of both male and female adults, and its expression is stable during sexual maturation. In contrast, "similar to nothepsin" expression is unique to the liver of adult females and is sex steroid-dependent as it increases gradually in the liver of hens during sexual maturation. The sexual dimorphic expression of the "similar to nothepsin" gene suggests that the activity of this protein is regulated by the steroid environment of laying hens and is specifically adapted for inclusion in the yolk. Further studies are needed to assess whether "similar to nothepsin" assists cathepsin D in the proteolytic processing of egg yolk proteins during follicular growth.


Subject(s)
Cathepsin D/physiology , Chickens/growth & development , Chickens/metabolism , Egg Yolk/physiology , Amino Acid Sequence , Animals , Female , Genes, Developmental , Liver/metabolism , Male , Molecular Sequence Data , Sex Factors , Sexual Maturation/physiology
10.
Gut ; 60(1): 55-65, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21076125

ABSTRACT

BACKGROUND: The sphingolipid sphingomyelin is a constituent in food derived from animals. Digestive breakdown of sphingomyelin results in ceramide, recently suggested to be involved in activation of cathepsin D as a novel mediator of apoptosis. Damage of the epithelial barrier was detected in patients with inflammatory bowel disease (IBD) due to increased rates of intestinal epithelial cell (IEC) apoptosis. METHODS: Acute colitis was induced in C57-BL/6 mice with 2.0% dextran sulfate sodium (DSS) over 7 days. Spontaneous colitis was developed in B6-IL10tm1Cgn (interleukin 10-negative (IL-10(-/-))) mice. Mice received 4 or 8 mg sphingomyelin/day by oral gavage. IECs were isolated ex vivo. Apoptosis was determined by propidium iodide (PI) and terminal deoxynucleotidyl transferase dUTP nick end labelling (TUNEL) staining. Execution of apoptosis was confirmed by analysis of active cathepsin D, caspase-3 and caspase-9 with western blot and immunohistochemistry (IHC). RESULTS: Following DSS-mediated colitis, fluorescence-activated cell sorting (FACS) analysis indicated increased apoptosis of IECs under dietary sphingomyelin. The mean sub-G(1) portion increased from 8.7±2.5% under a normal diet to 14.0±3.1% under dietary sphingomyelin. Cathepsin activity was significantly increased in isolated IECs after gavage of 4 mg of sphingomyelin per day. Western blot and IHC revealed execution of the apoptotic cascade via activated caspase-3 and caspase-9. Dietary sphingomyelin in the IL-10(-/-) model confirmed aggravation of mucosal inflammation. CONCLUSION: Apoptosis of IEC induced by dietary sphingomyelin is mediated via ceramide and cathepsin D activation. This shortens the physiological life cycle of IECs and impairs crucial functions of the intestinal mucosa: barrier, defence and nutrient absorption. The findings provide evidence that dietary sphingomyelin may increase intestinal inflammation.


Subject(s)
Apoptosis/drug effects , Cathepsin D/physiology , Colitis/pathology , Intestinal Mucosa/pathology , Sphingomyelins/pharmacology , Animals , Apoptosis/physiology , Colitis/chemically induced , Colitis/metabolism , Colonoscopy , Dextran Sulfate , Dietary Fats/pharmacokinetics , Dietary Fats/pharmacology , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial Cells/pathology , Feces/chemistry , Female , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C57BL , Signal Transduction/drug effects , Sphingomyelins/pharmacokinetics , Weight Loss/drug effects
11.
J Immunol ; 183(1): 543-51, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19542465

ABSTRACT

There is an abundance of antimicrobial peptides in cystic fibrosis (CF) lungs. Despite this, individuals with CF are susceptible to microbial colonization and infection. In this study, we investigated the antimicrobial response within the CF lung, focusing on the human cathelicidin LL-37. We demonstrate the presence of the LL-37 precursor, human cathelicidin precursor protein designated 18-kDa cationic antimicrobial protein, in the CF lung along with evidence that it is processed to active LL-37 by proteinase-3. We demonstrate that despite supranormal levels of LL-37, the lung fluid from CF patients exhibits no demonstrable antimicrobial activity. Furthermore Pseudomonas killing by physiological concentrations of exogenous LL-37 is inhibited by CF bronchoalveolar lavage (BAL) fluid due to proteolytic degradation of LL-37 by neutrophil elastase and cathepsin D. The endogenous LL-37 in CF BAL fluid is protected from this proteolysis by interactions with glycosaminoglycans, but while this protects LL-37 from proteolysis it results in inactivation of LL-37 antimicrobial activity. By digesting glycosaminoglycans in CF BAL fluid, endogenous LL-37 is liberated and the antimicrobial properties of CF BAL fluid restored. High sodium concentrations also liberate LL-37 in CF BAL fluid in vitro. This is also seen in vivo in CF sputum where LL-37 is complexed to glycosaminoglycans but is liberated following nebulized hypertonic saline resulting in increased antimicrobial effect. These data suggest glycosaminoglycan-LL-37 complexes to be potential therapeutic targets. Factors that disrupt glycosaminoglycan-LL-37 aggregates promote the antimicrobial effects of LL-37 with the caveat that concomitant administration of antiproteases may be needed to protect the now liberated LL-37 from proteolytic cleavage.


Subject(s)
Antimicrobial Cationic Peptides/antagonists & inhibitors , Antimicrobial Cationic Peptides/metabolism , Cystic Fibrosis/immunology , Cystic Fibrosis/metabolism , Glycosaminoglycans/metabolism , Lung/immunology , Lung/metabolism , Saline Solution, Hypertonic/pharmacology , Adjuvants, Immunologic/physiology , Adolescent , Antibody Specificity/physiology , Antimicrobial Cationic Peptides/immunology , Antimicrobial Cationic Peptides/physiology , Cathepsin D/physiology , Child , Cystic Fibrosis/enzymology , Cystic Fibrosis/microbiology , Glycosaminoglycans/physiology , Humans , Hydrolysis , Leukocyte Elastase/physiology , Lung/enzymology , Lung/microbiology , Macromolecular Substances/antagonists & inhibitors , Macromolecular Substances/immunology , Macromolecular Substances/metabolism , Molecular Weight , Myeloblastin/metabolism , Nebulizers and Vaporizers , Protein Precursors/metabolism , Protein Processing, Post-Translational , Saline Solution, Hypertonic/administration & dosage , Solubility , Sputum/enzymology , Sputum/immunology , Sputum/microbiology , Cathelicidins
12.
J Zhejiang Univ Sci B ; 22(11): 893-905, 2021 Nov 15.
Article in English | MEDLINE | ID: mdl-34783220

ABSTRACT

Cathepsin D (CTSD), the major lysosomal aspartic protease that is widely expressed in different tissues, potentially regulates the biological behaviors of various cells. Follicular granulosa cells are responsive to the increase of ovulation number, hence indirectly influencing litter size. However, the mechanism underlying the effect of CTSD on the behaviors of goat granulosa cells has not been fully elucidated. This study used immunohistochemistry to analyze CTSD localization in goat ovarian tissues. Moreover, western blotting was applied to examine the differential expression of CTSD in the ovarian tissues of monotocous and polytocous goats. Subsequently, the effects of CTSD knockdown on cell proliferation, apoptosis, cell cycle, and the expression of candidate genes of the prolific traits, including bone morphogenetic protein receptor IB (BMPR-IB), follicle-stimulating hormone (FSHR), and inhibin α (INHA), were determined in granulosa cells. Results showed that CTSD was expressed in corpus luteum, follicle, and granulosa cells. Notably, CTSD expression in the monotocous group was significantly higher than that in the polytocous group. In addition, CTSD knockdown could improve granulosa cell proliferation, inhibit cell apoptosis, and significantly elevate the expression of proliferating cell nuclear antigen (PCNA) and B cell lymphoma 2 (Bcl-2), but it lowered the expression of Bcl-2-associated X (Bax) and caspase-3. Furthermore, CTSD knockdown significantly reduced the ratios of cells in the G0/G1 and G2/M phases but substantially increased the ratio of cells in the S phase. The expression levels of cyclin D2 and cyclin E were elevated followed by the obvious decline of cyclin A1 expression. However, the expression levels of BMPR-IB, FSHR, and INHA clearly increased as a result of CTSD knockdown. Hence, our findings demonstrate that CTSD is an important factor affecting the litter size trait in goats by regulating the granulosa cell proliferation, apoptosis, cell cycle, and the expression of candidate genes of the prolific trait.


Subject(s)
Cathepsin D/physiology , Granulosa Cells/physiology , Litter Size , Animals , Apoptosis , Cathepsin D/analysis , Cell Proliferation , Cells, Cultured , Female , Goats , Ovary/chemistry
13.
Front Immunol ; 12: 675535, 2021.
Article in English | MEDLINE | ID: mdl-34335574

ABSTRACT

Background & Aims: The lysosomal enzyme, cathepsin D (CTSD) has been implicated in the pathogenesis of non-alcoholic steatohepatitis (NASH), a disease characterised by hepatic steatosis and inflammation. We have previously demonstrated that specific inhibition of the extracellular CTSD leads to improved metabolic features in Sprague-Dawley rats with steatosis. However, the individual roles of extracellular and intracellular CTSD in NASH are not yet known. In the current study, we evaluated the underlying mechanisms of extracellular and intracellular CTSD fractions in NASH-related metabolic inflammation using specific small-molecule inhibitors. Methods: Low-density lipoprotein receptor knock out (Ldlr-/-) mice were fed a high-fat, high cholesterol (HFC) diet for ten weeks to induce NASH. Further, to investigate the effects of CTSD inhibition, mice were injected either with an intracellular (GA-12) or extracellular (CTD-002) CTSD inhibitor or vehicle control at doses of 50 mg/kg body weight subcutaneously once in two days for ten weeks. Results: Ldlr-/- mice treated with extracellular CTSD inhibitor showed reduced hepatic lipid accumulation and an associated increase in faecal bile acid levels as compared to intracellular CTSD inhibitor-treated mice. Furthermore, in contrast to intracellular CTSD inhibition, extracellular CTSD inhibition switched the systemic immune status of the mice to an anti-inflammatory profile. In line, label-free mass spectrometry-based proteomics revealed that extra- and intracellular CTSD fractions modulate proteins belonging to distinct metabolic pathways. Conclusion: We have provided clinically translatable evidence that extracellular CTSD inhibition shows some beneficial metabolic and systemic inflammatory effects which are distinct from intracellular CTSD inhibition. Considering that intracellular CTSD inhibition is involved in essential physiological processes, specific inhibitors capable of blocking extracellular CTSD activity, can be promising and safe NASH drugs.


Subject(s)
Cathepsin D/physiology , Inflammation/etiology , Lipid Metabolism , Liver/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Animals , Bile Acids and Salts/analysis , Cathepsin D/antagonists & inhibitors , Female , Inflammation/prevention & control , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/drug therapy , Proteomics , Receptors, LDL/physiology
14.
J Exp Med ; 186(4): 549-60, 1997 Aug 18.
Article in English | MEDLINE | ID: mdl-9254653

ABSTRACT

Antigen-presenting cells (APC) degrade endocytosed antigens into peptides that are bound and presented to T cells by major histocompatibility complex (MHC) class II molecules. Class II molecules are delivered to endocytic compartments by the class II accessory molecule invariant chain (Ii), which itself must be eliminated to allow peptide binding. The cellular location of Ii degradation, as well as the enzymology of this event, are important in determining the sets of antigenic peptides that will bind to class II molecules. Here, we show that the cysteine protease cathepsin S acts in a concerted fashion with other cysteine and noncysteine proteases to degrade mouse Ii in a stepwise fashion. Inactivation of cysteine proteases results in incomplete degradation of Ii, but the extent to which peptide loading is blocked by such treatment varies widely among MHC class II allelic products. These observations suggest that, first, class II molecules associated with larger Ii remnants can be converted efficiently to class II-peptide complexes and, second, that most class II-associated peptides can still be generated in cells treated with inhibitors of cysteine proteases. Surprisingly, maturation of MHC class II in mice deficient in cathepsin D is unaffected, showing that this major aspartyl protease is not involved in degradation of Ii or in generation of the bulk of antigenic peptides.


Subject(s)
Antigens, Differentiation, B-Lymphocyte/metabolism , Cathepsin D/physiology , Cathepsins/physiology , Histocompatibility Antigens Class II/metabolism , Histocompatibility Antigens Class II/physiology , Amino Acid Sequence , Animals , Cricetinae , Cysteine Proteinase Inhibitors/pharmacology , Leupeptins/pharmacology , Mice , Molecular Sequence Data , Polymorphism, Genetic , Rabbits
15.
Hepatology ; 49(4): 1297-307, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19116891

ABSTRACT

UNLABELLED: Cathepsins have been best characterized in tumorigenesis and cell death and implicated in liver fibrosis; however, whether cathepsins directly regulate hepatic stellate cell (HSC) activation and proliferation, hence modulating their fibrogenic potential, is largely unknown. Here, we show that expression of cathepsin B (CtsB) and cathepsin D (CtsD) is negligible in quiescent HSCs but parallels the increase of alpha-smooth muscle actin and transforming growth factor-beta during in vitro mouse HSC activation. Both cathepsins are necessary for HSC transdifferentiation into myofibroblasts, because their silencing or inhibition decreased HSC proliferation and the expression of phenotypic markers of HSC activation, with similar results observed with the human HSC cell line LX2. CtsB inhibition blunted AKT phosphorylation in activated HSCs in response to platelet-derived growth factor. Moreover, during in vivo liver fibrogenesis caused by CCl(4) administration, CtsB expression increased in HSCs but not in hepatocytes, and its inactivation mitigated CCl(4)-induced inflammation, HSC activation, and collagen deposition. CONCLUSION: These findings support a critical role for cathepsins in HSC activation, suggesting that the antagonism of cathepsins in HSCs may be of relevance for the treatment of liver fibrosis.


Subject(s)
Cathepsin B/physiology , Cathepsin D/physiology , Cell Transdifferentiation , Hepatic Stellate Cells/physiology , Liver Cirrhosis/physiopathology , Actins/metabolism , Animals , Carbon Tetrachloride , Cell Line , Cell Proliferation , Down-Regulation , Gene Expression , Gene Silencing , Humans , Liver Cirrhosis/chemically induced , Liver Cirrhosis/metabolism , Mice , Mice, Inbred C57BL , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Transforming Growth Factor beta/metabolism , Up-Regulation
16.
J Cell Biol ; 168(3): 489-99, 2005 Jan 31.
Article in English | MEDLINE | ID: mdl-15668295

ABSTRACT

The aspartyl-protease cathepsin D (cath-D) is overexpressed and hypersecreted by epithelial breast cancer cells and stimulates their proliferation. As tumor epithelial-fibroblast cell interactions are important events in cancer progression, we investigated whether cath-D overexpression affects also fibroblast behavior. We demonstrate a requirement of cath-D for fibroblast invasive growth using a three-dimensional (3D) coculture assay with cancer cells secreting or not pro-cath-D. Ectopic expression of cath-D in cath-D-deficient fibroblasts stimulates 3D outgrowth that is associated with a significant increase in fibroblast proliferation, survival, motility, and invasive capacity, accompanied by activation of the ras-MAPK pathway. Interestingly, all these stimulatory effects on fibroblasts are independent of cath-D proteolytic activity. Finally, we show that pro-cath-D secreted by cancer cells is captured by fibroblasts and partially mimics effects of transfected cath-D. We conclude that cath-D is crucial for fibroblast invasive outgrowth and could act as a key paracrine communicator between cancer and stromal cells, independently of its catalytic activity.


Subject(s)
Cathepsin D/physiology , Cell Movement/physiology , Fibroblasts/cytology , Animals , Apoptosis/physiology , Butadienes/pharmacology , Cathepsin D/genetics , Cathepsin D/metabolism , Cell Enlargement/drug effects , Cell Growth Processes/drug effects , Cell Growth Processes/physiology , Cell Line , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/physiology , Coculture Techniques , Culture Media, Conditioned/pharmacology , Endocytosis/drug effects , Enzyme Inhibitors/pharmacology , Enzyme Precursors/metabolism , Enzyme Precursors/physiology , Fibroblasts/drug effects , Fibroblasts/ultrastructure , Humans , Mannosephosphates/pharmacology , Mice , Microscopy, Electron , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neoplasm Invasiveness , Neoplasms, Glandular and Epithelial/enzymology , Neoplasms, Glandular and Epithelial/metabolism , Neoplasms, Glandular and Epithelial/pathology , Nitriles/pharmacology , Paracrine Communication/physiology , Phosphorylation/drug effects , RNA, Small Interfering/genetics , Transfection , Wound Healing
17.
Neurosci Bull ; 36(10): 1147-1157, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32170568

ABSTRACT

The main lysosomal protease cathepsin D (cathD) is essential for maintaining tissue homeostasis via its degradative function, and its loss leads to ceroid accumulation in the mammalian nervous system, which results in progressive neurodegeneration. Increasing evidence implies non-proteolytic roles of cathD in regulating various biological processes such as apoptosis, cell proliferation, and migration. Along these lines, we here showed that cathD is required for modulating dendritic architecture in the nervous system independent of its traditional degradative function. Upon cathD depletion, class I and class III arborization (da) neurons in Drosophila larvae exhibited aberrant dendritic morphology, including over-branching, aberrant turning, and elongation defects. Re-introduction of wild-type cathD or its proteolytically-inactive mutant dramatically abolished these morphological defects. Moreover, cathD knockdown also led to dendritic defects in the adult mushroom bodies, suggesting that cathD-mediated processes are required in both the peripheral and central nervous systems. Taken together, our results demonstrate a critical role of cathD in shaping dendritic architecture independent of its proteolytic function.


Subject(s)
Cathepsin D/physiology , Dendrites/physiology , Drosophila Proteins , Lysosomes/enzymology , Animals , Central Nervous System , Drosophila , Drosophila Proteins/physiology
18.
Biochim Biophys Acta ; 1783(12): 2294-300, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18775751

ABSTRACT

The present investigation was undertaken to measure the relative abilities of pro-death versus pro-survival proteases in degrading each other and to determine how this might influence cellular susceptibility to death. For this, we first carried out in vitro experiments in which recombinant pro-death proteases (caspase-3 or cathepsin D) were incubated with the pro-survival protease (cathepsin L) in their respective optimal conditions and determined the effects of these reactions on enzyme integrity and activity. The results indicated that cathepsin L was able to degrade cathepsin D, which in turn cleaves caspase-3, however the later enzyme was unable to degrade any of the cathepsins. The consequences of this proteolytic sequence on cellular ability to undergo apoptosis or other types of cell death were studied in cells subjected to treatment with a specific inhibitor of cathepsin L or the corresponding siRNA. Both treatments resulted in suppression of cellular proliferation and the induction of a cell death with no detectable caspase-3 activation or DNA fragmentation, however, it was associated with increased accumulation of cathepsin D, cellular vaculolization, expression of the mannose-6-phosphate receptor, and the autophagy marker LC3-II, all of which are believed to be associated with autophagy. Genetic manipulations leading either to the gain or loss of cathepsin D expression implicated this enzyme as a key player in the switch from apoptosis to autophagy. Overall, these findings suggest that a hierarchy between pro-survival and pro-death proteases may have important consequences on cell fate.


Subject(s)
Apoptosis/physiology , Autophagy , Caspase 3/physiology , Cathepsin D/physiology , Cathepsins/physiology , Cysteine Endopeptidases/physiology , Blotting, Western , Cathepsin L , Cell Survival , Humans , Microscopy, Fluorescence , RNA, Small Interfering/pharmacology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Tumor Cells, Cultured/enzymology , Tumor Cells, Cultured/pathology
19.
Neuromolecular Med ; 21(3): 287-294, 2019 09.
Article in English | MEDLINE | ID: mdl-31134487

ABSTRACT

Parkinson's disease (PD) is a progressive neurodegenerative disease with complex etiology. Both genetic and environmental factors play significant role. Apart from candidate genes, some modifier genes have been reported to be associated with the altered risk of PD. Previous studies have identified Apolipoprotein E (APOE), Cathepsin D (CTSD), and Brain-Derived Neurotrophic Factor (BDNF) as key players of neurodegenerative pathways with their variants associated with different neurodegenerative diseases. Hence, this study aims to identify the potential role of these modifier genes in the pathogenesis of PD among Eastern Indian PD patients. A case-control study was performed using 302 clinically diagnosed PD patients and 304 ethnically matched controls. Promoter SNPs of APOE (rs449647, rs405509) and BDNF (rs56164415), and coding SNPs of APOE (rs429358, rs7412 resulting in ε2, ε3, and ε4 alleles), CTSD (rs17571), and BDNF (rs6265) were analyzed by PCR-RFLP and bidirectional sequencing. The effect of rs56164415 on BDNF expression was characterized by Luciferase assay. APOEε4 allele was significantly overrepresented (p value = 0.0003) among PD patients, whereas ε3 allele was predominant in the control population. The promoter haplotype (A-rs449647, G-rs405509) of APOE was preponderant among female PD patients posing risk. No association was found for CTSD polymorphism. The 'T/T' genotype of BDNF rs56164415 was overrepresented (p-value = 0.02) among early onset PD patients. Expression of BDNF for the 'T/T' variant was significantly lower (p-value = 0.012) than the 'C/C' variant, suggesting a possible role in PD pathogenesis. This study suggests that APOE and BDNF may serve as modifier loci among eastern Indian PD patients.


Subject(s)
Apolipoproteins E/physiology , Brain-Derived Neurotrophic Factor/physiology , Cathepsin D/physiology , Parkinson Disease/genetics , Adolescent , Adult , Age of Onset , Aged , Alleles , Case-Control Studies , Child , Female , Gene Frequency , Genotype , Humans , India/epidemiology , Male , Middle Aged , Parkinson Disease/epidemiology , Parkinson Disease/metabolism , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , Protein Isoforms/genetics , Protein Isoforms/physiology , Young Adult
20.
Brain Pathol ; 29(1): 63-74, 2019 01.
Article in English | MEDLINE | ID: mdl-30051532

ABSTRACT

Alzheimer's disease (AD) is characterized by accumulation of ß-amyloid plaques (AP) and neurofibrillary tangles (NFT) in the cortex, together with synaptic loss and amyloid angiopathy. Perturbations in the brain lysosomal system, including the cathepsin family of proteases, have been implicated in AD where they may be involved in proteolytic clearance of misfolded and abnormally aggregated peptides. However, the status of cathepsin D (catD) is unclear in Lewy body dementia, the second most common form of neurodegenerative dementia after AD, and characterized by Lewy bodies (LB) containing aggregated α-synuclein. Furthermore, earlier reports of catD changes in AD have not been entirely consistent. We measured CatD immunoreactivities in the temporal (Brodmann area BA21) and parietal (BA40) cortices of well characterized AD brains as well as two clinical subtypes of Lewy body dementia, namely Parkinson disease dementia (PDD) and dementia with Lewy bodies (DLB), known to show varying degrees of concomitant AD pathology. Increased catD immunoreactivities in AD were found for both neocortical regions measured, where they also correlated with neuropathological NFT scores and phosphorylated pSer396 tau burden, and appeared to co-localize at least partly to NFT-containing neurons. In contrast, catD was increased only in BA40 in DLB and not at all in PDD, did not correlate with LB scores, and did not appreciably co-localize with α-synuclein inclusions. Our study suggests that catD upregulation may be an adaptive response to AD-related processes leading to neurofibrillary degeneration, but may not be directly associated with formation of α-synuclein inclusions in Lewy body dementia.


Subject(s)
Alzheimer Disease/metabolism , Cathepsin D/physiology , Neocortex/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Biomarkers , Cathepsin D/genetics , Cathepsin D/metabolism , Female , Humans , Lewy Bodies/pathology , Lewy Body Disease/pathology , Male , Neocortex/physiology , Neurodegenerative Diseases/physiopathology , Neurofibrillary Tangles/metabolism , Neurons/pathology , Parietal Lobe/pathology , Plaque, Amyloid/pathology , Temporal Lobe/pathology , alpha-Synuclein/metabolism , tau Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL