Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 138
Filter
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
Cancer Lett ; 370(2): 222-31, 2016 Jan 28.
Article in English | MEDLINE | ID: mdl-26519755

ABSTRACT

Human telomerase reverse transcriptase (hTERT) contributes to tumor progression as well as maintaining telomere length, however, the mechanism by which hTERT promotes invasiveness is not yet completely understood. This study aims to unravel the precise mechanism through which hTERT promotes cancer invasion. We established an hTERT-overexpressed immortalized cell line (IHOK/hTERT). In orthotopic xenograft models, IHOK/hTERT harbors higher tumorigenicity than IHOK/Control. IHOK/hTERT showed much higher migration and invasion activities compared to IHOK/Control. IHOK/hTERT co-cultured with fibroblasts displayed increased invasion compared to IHOK/hTERT without fibroblasts. We screened for genes that play an important role in intermodulation between cancer cells and fibroblasts using a microarray and identified fibroblast activation protein (FAP). hTERT knockdown showed decreased expression of FAP and early growth response (EGR)-1, one of the transcriptional regulators of FAP in IHOK/hTERT and oral cancer cell line YD10B. Furthermore, EGR-1 knockdown in IHOK/hTERT and YD10B showed reduced invasion and reduced cathepsin D expression compared to Control-siRNA cells. Taken together, this study provides evidence that hTERT overexpression is responsible for the upregulation of the cysteine protease cathepsin D by regulating EGR-1 to activate invasiveness in cancer progression.


Subject(s)
Cathepsin D/physiology , Early Growth Response Protein 1/physiology , Mouth Neoplasms/pathology , Telomerase/physiology , Animals , Cell Line, Tumor , Cell Movement , Humans , Male , Mice , Mice, Inbred BALB C , Neoplasm Invasiveness
10.
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
11.
PLoS One ; 10(6): e0130144, 2015.
Article in English | MEDLINE | ID: mdl-26091289

ABSTRACT

In this work, we have investigated the involvement of DmCatD, a cathepsin D-like peptidase, and acid phosphatase in the process of follicular atresia of Dipetalogaster maxima, a hematophagous insect vector of Chagas' disease. For the studies, fat bodies, ovaries and hemolymph were sampled from anautogenous females at representative days of the reproductive cycle: pre-vitellogenesis, vitellogenesis as well as early and late atresia. Real time PCR (qPCR) and western blot assays showed that DmCatD was expressed in fat bodies and ovaries at all reproductive stages, being the expression of its active form significantly higher at the atretic stages. In hemolymph samples, only the immunoreactive band compatible with pro-DmCatD was observed by western blot. Acid phosphatase activity in ovarian tissues significantly increased during follicular atresia in comparison to pre-vitellogenesis and vitellogenesis. A further enzyme characterization with inhibitors showed that the high levels of acid phosphatase activity in atretic ovaries corresponded mainly to a tyrosine phosphatase. Immunofluorescence assays demonstrated that DmCatD and tyrosine phosphatase were associated with yolk bodies in vitellogenic follicles, while in atretic stages they displayed a different cellular distribution. DmCatD and tyrosine phosphatase partially co-localized with vitellin. Moreover, their interaction was supported by FRET analysis. In vitro assays using homogenates of atretic ovaries as the enzyme source and enzyme inhibitors demonstrated that DmCatD, together with a tyrosine phosphatase, were necessary to promote the degradation of vitellin. Taken together, the results strongly suggested that both acid hydrolases play a central role in early vitellin proteolysis during the process of follicular atresia.


Subject(s)
Acid Phosphatase/physiology , Cathepsin D/physiology , Follicular Atresia , Hemiptera/enzymology , Insect Proteins/physiology , Insect Vectors/enzymology , Acid Phosphatase/chemistry , Animals , Cathepsin D/chemistry , Chagas Disease/parasitology , Fat Body/enzymology , Female , Gene Expression , Hemiptera/parasitology , Hemolymph/enzymology , Humans , Hydrogen-Ion Concentration , Insect Proteins/chemistry , Insect Vectors/parasitology , MCF-7 Cells , Male , Organ Specificity , Ovary/enzymology , Proteolysis , Trypanosoma cruzi/physiology , Vitellins/chemistry , Vitellins/metabolism
12.
Cell Cycle ; 14(7): 964-72, 2015.
Article in English | MEDLINE | ID: mdl-25830414

ABSTRACT

Necrotic cell death triggers a range of biological responses including a strong adaptive immune response, yet we know little about the cellular pathways that control necrotic cell death. Inhibitor studies suggest that proteases, and in particular cathepsins, drive necrotic cell death. The cathepsin B-selective inhibitor CA-074-Me blocks all forms of programmed necrosis by an unknown mechanism. We found that cathepsin B deficiency does not prevent induction of pyroptosis and lysosome-mediated necrosis suggesting that CA-074-Me blocks necrotic cell death by targeting cathepsins other than cathepsin B. A single cathepsin, cathepsin C, drives necrotic cell death mediated by the lysosome-destabilizing agent Leu-Leu-OMe (LLOMe). Here we present evidence that cathepsin C-deficiency and CA-074-Me block LLOMe killing in a distinct and cell type-specific fashion. Cathepsin C-deficiency and CA-074-Me block LLOMe killing of all myeloid cells, except for neutrophils. Cathepsin C-deficiency, but not CA-074-Me, blocks LLOMe killing of neutrophils suggesting that CA-074-Me does not target cathepsin C directly, consistent with inhibitor studies using recombinant cathepsin C. Unlike other cathepsins, cathepsin C lacks endoproteolytic activity, and requires activation by other lysosomal proteases, such as cathepsin D. Consistent with this theory, we found that lysosomotropic agents and cathepsin D downregulation by siRNA block LLOMe-mediated necrosis. Our findings indicate that a proteolytic cascade, involving cathepsins C and D, controls LLOMe-mediated necrosis. In contrast, cathepsins C and D were not required for pyroptotic cell death suggesting that distinct cathepsins control pyroptosis and lysosome-mediated necrosis.


Subject(s)
Cathepsin C/physiology , Cathepsin D/physiology , Lysosomes/enzymology , Animals , Apoptosis , Cathepsin B/antagonists & inhibitors , Cathepsin B/physiology , Dipeptides/pharmacology , Lysosomes/drug effects , Mice, Inbred BALB C , Mice, Inbred C57BL , Necrosis
13.
Nefrologia ; 35(1): 72-9, 2015.
Article in English, Spanish | MEDLINE | ID: mdl-25611836

ABSTRACT

We aimed to investigate the role of cathepsin D, an inflammatory and atherosclerotic mediator, in endothelial dysfunction in chronic kidney disease. The study included 65 patients with stage 2–4 chronic kidney disease (35 females, 30 males; mean age, 55.8±15.6 years). Serum creatinine and cathepsin D levels and glomerular filtration rates (GFRs) were determined, and brachial flow-mediated dilation (FMD) percentage was measured by two-dimensional gray scale and color flow Doppler and vascular imaging. FMD ≤6% was considered to indicate endothelial dysfunction. Mean GFR, median creatinine levels, and median cathepsin D levels were 40.2±11.2mL/min/1.73m2, 1.7mg/dL, and 819.75ng/mL, respectively. Endothelial dysfunction was present in 30 of the 65 patients (46.2%). There was a significant difference between groups with and without endothelial dysfunction in terms of cathepsin D (p=0.001) and creatinine (p=0.03) levels, and negative and significant correlations were found between brachial artery FMD% and cathepsin D (r=−0.359, p=0.003) and creatinine (r=−0.304, p=0.014) levels. Cathepsin D, which is known to be associated with atherosclerosis, may play a role in the process of endothelial dysfunction. Further studies are essential to determine the exact function of cathepsin D in endothelial dysfunction in chronic kidney disease and to determine its value as a tool for early diagnosis and target for treatment of cardiovascular diseases in patients with chronic kidney disease.


Subject(s)
Cathepsin D/blood , Creatinine/blood , Endothelium, Vascular/physiopathology , Renal Insufficiency, Chronic/blood , Adult , Aged , Atherosclerosis/blood , Atherosclerosis/physiopathology , Brachial Artery , Cathepsin D/physiology , Female , Glomerular Filtration Rate , Humans , Male , Middle Aged , Prospective Studies , Renal Insufficiency, Chronic/physiopathology , Vasodilation
14.
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
15.
Cancer Lett ; 323(2): 208-14, 2012 Oct 28.
Article in English | MEDLINE | ID: mdl-22542809

ABSTRACT

Cathepsin D (CatD), a lysosomal aspartic protease, plays an essential role in tumor progression and apoptosis. However, the function of CatD in cell death is not yet fully understood. In this study, we identified CatD as one of up-regulated proteins in human malignant glioblastoma M059J cells that lack the catalytic subunit of DNA-PK compared with its isogenic M059K cells with normal DNA-PK activity. M059J cells were relatively more resistant to genotoxic stress than M059K cells. Overexpression of wild-type CatD but not catalytically inactive mutant CatD (D295N) inhibited H(2)O(2)-induced cell death in HeLa cells. Furthermore, knockdown of CatD expression abolished anti-apoptotic effect by CatD in the presence of H(2)O(2). Interestingly, high expression of CatD in HeLa cells significantly activated autophagy: increase of acidic autophagic vacuoles, LC3-II formation, and GFP-LC3 puncta. These results suggest that CatD can function as an anti-apoptotic mediator by inducing autophagy under cellular stress. In conclusion, inhibition of autophagy could be a novel strategy for the adjuvant chemotherapy of CatD-expressing cancers.


Subject(s)
Autophagy , Brain Neoplasms/pathology , Cathepsin D/physiology , Cell Death/physiology , Glioblastoma/pathology , Oxidative Stress/physiology , Base Sequence , Cell Line, Tumor , DNA Primers , Humans , Microscopy, Electron , Real-Time Polymerase Chain Reaction
16.
J Morphol ; 273(7): 791-800, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22505219

ABSTRACT

Programmed cell death during anuran tail resorption is primarily brought about by apoptosis. Cathepsin D, a lysosomal aspartyl protease, is involved in the death of tail tissues. Thus, anuran tail resorption presents an ideal model to study cathepsin-mediated cell death during vertebrate development. Present study describes the trend of specific activity of cathepsin D in the tail of different developmental stages and immunohistochemical localization of cathepsin D in the tail tissues of the common Asian toad, Duttaphrynus melanostictus. Cathepsin D was involved in programmed cell death in epidermis, muscle, spinal cord, and blood cells in the resorbing tail. Interestingly, it was also involved in the pre-resorbing tail before visible tail resorption which indicates initiation of cell death even before actually the tail resorbs. Melanocytes were found to be one of the causative agents in degrading tail tissues and were associated with the degradation of muscle, epidermis and spinal cord of the resorbing tail.


Subject(s)
Apoptosis/physiology , Bufonidae/growth & development , Cathepsin D/physiology , Melanocytes/physiology , Tail/growth & development , Animals , Anura/metabolism , Cathepsins , Epidermis/growth & development , Larva/growth & development , Lysosomes/metabolism , Muscles/physiology , Spinal Cord/growth & development
17.
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
18.
Gulf J Oncolog ; (11): 38-44, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22227544

ABSTRACT

OBJECTIVE: Cathepsin D is a lysosomal acid protease secreted in increased levels in several malignancies. However, its role in salivary gland tumors has not been studied extensively. The present study aims to assess the expression of Cathepsin D in malignant salivary gland tumors and to compare its expression in these tumors. STUDY DESIGN: A total of 30 cases of malignant salivary gland carcinomas which included 16 cases of adenoid cystic carcinoma (ACC), 9 cases of mucoepidermoid carcinoma (MEC), and 5 cases of polymorphous low grade adenocarcinoma (PLGA) were evaluated immunohistochemically using anti-Cathepsin D antibody. RESULT: All the cases showed positivity (100%) for Cathepsin D with intense expression noted in ACC and MEC as compared to PLGA. Comparison of these tumors revealed statistical significant difference in expression between ACC and PLGA. CONCLUSION: Intense expression of Cathepsin D in high grade carcinomas may be a marker for invasive potential and aggressive behavior.


Subject(s)
Cathepsin D/analysis , Salivary Gland Neoplasms/enzymology , Adenocarcinoma/enzymology , Adenocarcinoma/pathology , Carcinoma, Adenoid Cystic/enzymology , Carcinoma, Adenoid Cystic/pathology , Carcinoma, Mucoepidermoid/enzymology , Carcinoma, Mucoepidermoid/pathology , Cathepsin D/physiology , Humans , Immunohistochemistry , Salivary Gland Neoplasms/pathology
19.
Cancer Res ; 71(22): 7091-102, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21948970

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) remains one of the most lethal cancers largely due to disseminated disease at the time of presentation. Here, we investigated the role and mechanism of action of the metastasis-associated protein anterior gradient 2 (AGR2) in the pathogenesis of pancreatic cancer. AGR2 was induced in all sporadic and familial pancreatic intraepithelial precursor lesions (PanIN), PDACs, circulating tumor cells, and metastases studied. Confocal microscopy and flow cytometric analyses indicated that AGR2 localized to the endoplasmic reticulum (ER) and the external surface of tumor cells. Furthermore, induction of AGR2 in tumor cells regulated the expression of several ER chaperones (PDI, CALU, RCN1), proteins of the ubiquitin-proteasome degradation pathway (HIP2, PSMB2, PSMA3, PSMC3, and PSMB4), and lysosomal proteases [cathepsin B (CTSB) and cathepsin D (CTSD)], in addition to promoting the secretion of the precursor form pro-CTSD. Importantly, the invasiveness of pancreatic cancer cells was proportional to the level of AGR2 expression. Functional downstream targets of the proinvasive activity of AGR2 included CTSB and CTSD in vitro, and AGR2, CTSB, and CTSD were essential for the dissemination of pancreatic cancer cells in vivo. Taken together, the results suggest that AGR2 promotes dissemination of pancreatic cancer and that its cell surface targeting may permit new strategies for early detection as well as therapeutic management.


Subject(s)
Adenocarcinoma/pathology , Antigens, Surface/physiology , Carcinoma, Pancreatic Ductal/pathology , Cathepsin B/physiology , Cathepsin D/physiology , Pancreatic Neoplasms/pathology , Proteins/physiology , Animals , Cathepsin B/genetics , Cathepsin D/genetics , Cell Line, Tumor , Endoplasmic Reticulum/metabolism , Humans , Mucoproteins , Neoplasm Invasiveness , Oncogene Proteins , Proteome , Zebrafish
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...