cathepsin s
Recently Published Documents


TOTAL DOCUMENTS

588
(FIVE YEARS 107)

H-INDEX

58
(FIVE YEARS 7)

Author(s):  
Satyendra Kumar Sonkar ◽  
Prashant Kumar Singh ◽  
Sharad Chandra ◽  
Gyanendra Kumar Sonkar ◽  
Vivek Bhosale ◽  
...  

Abstract Introduction: A high incidence of cardiovascular disease (CVD) events and premature mortality is observed in patients with chronic kidney disease (CKD). Thus, new biomarkers that may help predict the development of CVD in early stages of CKD are being investigated along with other traditional risk factors. Objective: To investigate cathepsin S as an early biomarker for CVD in patients with CKD. Methods: A total of 64 patients with CKD were included and classified into 2 groups: CKD patients with established CVD and CKD patients with non-established CVD. All patients were submitted to routine investigations including complete blood count, random blood sugar, glycated hemoglobin (HbA1c), serum electrolytes, urea, creatinine, total protein, total albumin, calcium total, phosphorous, uric acid, vitamin D, parathormone, lipid profile, liver function test, measurement of serum cathepsin S (Cat S), and 2D Echo of the heart. Results: The level of serum Cat S was increased in CKD patients with CVD (p <0.05) as well as in later stages of CKD (p <0.05). CVD was also more common in patients in early stage CKD. In early stages CKD, Cat S and CVD were positively correlated. Conclusion: These findings suggest that serum Cat S might be useful as an early biomarker for CVD in CKD patients.


2022 ◽  
Vol 12 ◽  
Author(s):  
Xiang-Yang Zhang ◽  
Xinyue Zhuo ◽  
Jie Cheng ◽  
Xiaohong Wang ◽  
Kexin Liang ◽  
...  

Different morphologies have been detected in teleost macrophages. In this study, two macrophage cell lines were sub-cloned from a large yellow croaker head kidney cell line, LYCK. One type of sub-cloned cells was fusiform but the other was round, named LYC-FM and LYC-RM cells respectively, based on their morphologies. Both types showed the characteristics of macrophages, including expression of macrophage-specific marker genes, possession of phagocytic and bactericidal activities, and production of reactive oxygen species (ROS) and nitric oxide (NO). The transcription factor PU.1, crucial for the development of macrophages in mammals, was found to exist in two transcripts, PU.1a and PU.1b, in large yellow croaker, and constitutively expressed in LYC-FM and LYC-RM cells. The expression levels of PU.1a and PU.1b could be upregulated by recombinant large yellow croaker IFN-γ protein (rLcIFN-γ). Further studies showed that both PU.1a and PU.1b increased the expression of cathepsin S (CTSS) by binding to different E26−transformation−specific (Ets) motifs of the CTSS promoter. Additionally, we demonstrated that all three domains of PU.1a and PU.1b were essential for initiating CTSS expression by truncated mutation experiments. Our results therefore provide the first evidence that teleost PU.1 has a role in regulating the expression of CTSS.


2022 ◽  
pp. 109811
Author(s):  
Lei Yuan ◽  
Juanping Zhao ◽  
Songkun Zhao ◽  
Tianyi Dong ◽  
Ruitong Dong ◽  
...  

2021 ◽  
Author(s):  
Lexing Xie ◽  
Shuang Zhang ◽  
Li Huang ◽  
Zhouzhou Peng ◽  
Hui Lu ◽  
...  

Abstract Background: Stroke persists as a major cause of morbidity and mortality worldwide. After a stroke, peripheral immune cells are rapidly activated and then infiltrate the central nervous system to cause inflammation in the brain. However, it is not clear when and how these peripheral immune cells affect the central inflammatory response and whether there are intervention targets that can alleviate ischemia-reperfusion injury. In this study, we collected mouse peripheral blood samples at different time points after stroke for single-cell sequencing to reveal the dynamic changes in peripheral immune cells. Methods: We performed single-cell sequencing on peripheral blood of mice at 1, 3, 7, and 14 days after ischemia-reperfusion to analyze the changes of subpopulations after cerebral ischemia-reperfusion; Real-time PCR, western blot and enzyme‑linked immunoabsorbent assay were used to perform mRNA and protein levels verification; Immunoprecipitation verifies the interaction of proteins and between junctional adhesion molecule (JAM-A) and Cathepsin S (CTSS) protein, in vitro enzyme digestion and silver staining method to detect the protease digestion effect of CTSS.Results: Peripheral monocyte subpopulations increased significantly after ischemia-reperfusion. Pseudo-time trajectory analysis and gene function analysis further suggested that CTSS may play an important role in regulating monocyte activation and leading to proteolysis. Next, we found that the expression of CTSS was significantly increased in monocytes after I/R in mice. Then, we used CTSS inhibitors and knockout mouse experiments to prove that inhibiting its expression can significantly reduce infarct volume and reduce blood–brain barrier (BBB) leakage. In addition, we found that CTSS destroys BBB by binding to JAM family proteins to cause them degradation. Conclusion: Inhibition of Cathepsin S attenuated cerebral ischemia reperfusion injury and Cathepsin S can be used as a novel target for drug intervention after stroke.


2021 ◽  
pp. 108895
Author(s):  
Zhiyuan Yu ◽  
Jinmiao Li ◽  
Gowthaman Govindarajan ◽  
Sarah Hamm-Alvarez ◽  
Jahan Alam ◽  
...  
Keyword(s):  
Dry Eye ◽  

2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Beatrice Romier ◽  
Cédric Dray ◽  
Laetitia Vanalderwiert ◽  
Amandine Wahart ◽  
Thinhinane Hocine ◽  
...  

AbstractNumerous recent studies have shown that in the continuum of cardiovascular diseases, the measurement of arterial stiffness has powerful predictive value in cardiovascular risk and mortality and that this value is independent of other conventional risk factors, such as age, cholesterol levels, diabetes, smoking, or average blood pressure. Vascular stiffening is often the main cause of arterial hypertension (AHT), which is common in the presence of obesity. However, the mechanisms leading to vascular stiffening, as well as preventive factors, remain unclear. The aim of the present study was to investigate the consequences of apelin deficiency on the vascular stiffening and wall remodeling of aorta in mice. This factor freed by visceral adipose tissue, is known for its homeostasic role in lipid and vascular metabolisms, or again in inflammation. We compared the level of metabolic markers, inflammation of white adipose tissue (WAT), and aortic wall remodeling from functional and structural approaches in apelin-deficient and wild-type (WT) mice. Apelin-deficient mice were generated by knockout of the apelin gene (APL-KO). From 8 mice by groups, aortic stiffness was analyzed by pulse wave velocity measurements and by characterizations of collagen and elastic fibers. Mann–Whitney statistical test determined the significant data (p < 5%) between groups. The APL-KO mice developed inflammation, which was associated with significant remodeling of visceral WAT, such as neutrophil elastase and cathepsin S expressions. In vitro, cathepsin S activity was detected in conditioned medium prepared from adipose tissue of the APL-KO mice, and cathepsin S activity induced high fragmentations of elastic fiber of wild-type aorta, suggesting that the WAT secretome could play a major role in vascular stiffening. In vivo, remodeling of the extracellular matrix (ECM), such as collagen accumulation and elastolysis, was observed in the aortic walls of the APL-KO mice, with the latter associated with high cathepsin S activity. In addition, pulse wave velocity (PWV) and AHT were increased in the APL-KO mice. The latter could explain aortic wall remodeling in the APL-KO mice. The absence of apelin expression, particularly in WAT, modified the adipocyte secretome and facilitated remodeling of the ECM of the aortic wall. Thus, elastolysis of elastic fibers and collagen accumulation contributed to vascular stiffening and AHT. Therefore, apelin expression could be a major element to preserve vascular homeostasis.


2021 ◽  
pp. 112245
Author(s):  
YoungJo Yoo ◽  
Eun Choi ◽  
Yejin Kim ◽  
Yunyoung Cha ◽  
Eunhye Um ◽  
...  

2021 ◽  
Author(s):  
Lingwei Kong ◽  
Hairu Ji ◽  
Xintian Gan ◽  
Sheng Cao ◽  
Zhehong Li ◽  
...  

Abstract BackgroundOsteosarcoma (OS) is a malignant bone tumour of mesenchymal origin. These tumours are characterised by rich vascularisation, therefore promoting rapid proliferation and facilitating metastasis. CD44 has been reported to be involved in OS, but its role and molecular mechanisms in the pathogenesis of the disease are not fully determined. MethodsIn this study, we investigated the antitumor effect of CD44 on the development of OS and further explored the molecular mechanisms. The expression of CD44, cathepsin S and MMP-9 was detected by Western blot (WB) and reverse transcription-polymerase chain reaction (RT-qPCR) in different cell lines (MG63, U2OS OS and hFOB 1.19). To elucidate the role of CD44 in OS, MG63 and U2OS cells were treated with small interference RNA (siRNA) to knock down CD44, and the knockdown efficiency was validated with GFP and RT-qPCR. Furthermore, cell proliferation was assayed using Cell Counting Kit‑8 (CCK-8) and colony formation assays, and cell migration and invasion were assayed by transwell and wound-healing assays. ResultsWe found that CD44 expression in the MG63 and U2OS OS cell lines was markedly increased compared to that of the human osteoblast hFOB 1.19 cell line. Knockdown of CD44 inhibited proliferation, migration, and invasion of MG63 and U2OS cells, possibly by regulating the expression of cathepsin S in OS. ConclusionTaken together, our data reinforced the evidence that CD44 knockdown inhibited cell proliferation, migration, and invasion of OS cells accompanied by altered expression of cathepsin S. These findings offer new clues for OS development and progression, suggesting CD44 as a potential therapeutic target for OS.


Cancers ◽  
2021 ◽  
Vol 13 (18) ◽  
pp. 4697
Author(s):  
Nguyen Huu Tu ◽  
Kenji Inoue ◽  
Elyssa Chen ◽  
Bethany M. Anderson ◽  
Caroline M. Sawicki ◽  
...  

Oral squamous cell carcinoma (SCC) pain is more prevalent and severe than pain generated by any other form of cancer. We previously showed that protease-activated receptor-2 (PAR2) contributes to oral SCC pain. Cathepsin S is a lysosomal cysteine protease released during injury and disease that can activate PAR2. We report here a role for cathepsin S in PAR2-dependent cancer pain. We report that cathepsin S was more active in human oral SCC than matched normal tissue, and in an orthotopic xenograft tongue cancer model than normal tongue. The multiplex immunolocalization of cathepsin S in human oral cancers suggests that carcinoma and macrophages generate cathepsin S in the oral cancer microenvironment. After cheek or paw injection, cathepsin S evoked nociception in wild-type mice but not in mice lacking PAR2 in Nav1.8-positive neurons (Par2Nav1.8), nor in mice treated with LY3000328 or an endogenous cathepsin S inhibitor (cystatin C). The human oral SCC cell line (HSC-3) with homozygous deletion of the gene for cathepsin S (CTSS) with CRISPR/Cas9 provoked significantly less mechanical allodynia and thermal hyperalgesia, as did those treated with LY3000328, compared to the control cancer mice. Our results indicate that cathepsin S is activated in oral SCC, and that cathepsin S contributes to cancer pain through PAR2 on neurons.


Sign in / Sign up

Export Citation Format

Share Document