1
|
Shen Q, Pan X, Li Y, Li J, Zhang C, Jiang X, Liu F, Pang B. Lysosomes, curcumin, and anti-tumor effects: how are they linked? Front Pharmacol 2023; 14:1220983. [PMID: 37484013 PMCID: PMC10359997 DOI: 10.3389/fphar.2023.1220983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 06/27/2023] [Indexed: 07/25/2023] Open
Abstract
Curcumin is a natural active ingredient from traditional Chinese medicine (TCM) that has multi-target characteristics to exert extensive pharmacological activities and thus has been applied in the treatment of various diseases such as cancer, cardiovascular diseases, nervous system, and autoimmune disorders. As an important class of membranous organelles in the intracellular membrane system, lysosomes are involved in biological processes such as programmed cell death, cell metabolism, and immune regulation, thus affecting tumor initiation and progression. It has been shown that curcumin can modulate lysosomal function through the aforementioned pathways, thereby affecting tumor proliferation, invasion, metastasis, drug resistance, and immune function. This review briefly elaborated the regulatory mechanisms of lysosome biogenesis and summarized curcumin-related studies with its anti-tumor effect, providing a reference for the clinical application of curcumin and anti-tumor research targeting lysosomes.
Collapse
Affiliation(s)
- Qian Shen
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xue Pan
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yi Li
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Junchen Li
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Chuanlong Zhang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaochen Jiang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fudong Liu
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bo Pang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
2
|
Deshpande NP, Riordan SM, Gorman CJ, Nielsen S, Russell TL, Correa-Ospina C, Fernando BSM, Waters SA, Castaño-Rodríguez N, Man SM, Tedla N, Wilkins MR, Kaakoush NO. Multi-omics of the esophageal microenvironment identifies signatures associated with progression of Barrett's esophagus. Genome Med 2021; 13:133. [PMID: 34412659 PMCID: PMC8375061 DOI: 10.1186/s13073-021-00951-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 08/11/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The enrichment of Gram-negative bacteria of oral origin in the esophageal microbiome has been associated with the development of metaplasia. However, to date, no study has comprehensively assessed the relationships between the esophageal microbiome and the host. METHODS Here, we examine the esophageal microenvironment in gastro-esophageal reflux disease and metaplasia using multi-omics strategies targeting the microbiome and host transcriptome, followed by targeted culture, comparative genomics, and host-microbial interaction studies of bacterial signatures of interest. RESULTS Profiling of the host transcriptome from esophageal mucosal biopsies revealed profound changes during metaplasia. Importantly, five biomarkers showed consistent longitudinal changes with disease progression from reflux disease to metaplasia. We showed for the first time that the esophageal microbiome is distinct from the salivary microbiome and the enrichment of Campylobacter species as a consistent signature in disease across two independent cohorts. Shape fitting and matrix correlation identified associations between the microbiome and host transcriptome profiles, with a novel co-exclusion relationship found between Campylobacter and napsin B aspartic peptidase. Targeted culture of Campylobacter species from the same cohort revealed a subset of isolates to have a higher capacity to survive within primary human macrophages. Comparative genomic analyses showed these isolates could be differentiated by specific genomic features, one of which was validated to be associated with intracellular fitness. Screening for these Campylobacter strain-specific signatures in shotgun metagenomics data from another cohort showed an increase in prevalence with disease progression. Comparative transcriptomic analyses of primary esophageal epithelial cells exposed to the Campylobacter isolates revealed expression changes within those infected with strains with high intracellular fitness that could explain the increased likelihood of disease progression. CONCLUSIONS We provide a comprehensive assessment of the esophageal microenvironment, identifying bacterial strain-specific signatures with high relevance to progression of metaplasia.
Collapse
Affiliation(s)
- Nandan P Deshpande
- School of Biotechnology and Biomolecular Sciences, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Stephen M Riordan
- Gastrointestinal and Liver Unit, The Prince of Wales Hospital, Randwick, NSW, 2031, Australia
| | - Claire J Gorman
- School of Medical Sciences, Faculty of Medicine, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Shaun Nielsen
- School of Medical Sciences, Faculty of Medicine, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Tonia L Russell
- Ramaciotti Centre for Genomics, UNSW Sydney, Sydney, NSW, 2052, Australia
| | | | - Bentotage S M Fernando
- School of Medical Sciences, Faculty of Medicine, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Shafagh A Waters
- School of Women's and Children's Health, UNSW Sydney, Sydney, NSW, 2052, Australia
| | | | - Si Ming Man
- The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, 2601, Australia
| | - Nicodemus Tedla
- School of Medical Sciences, Faculty of Medicine, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Marc R Wilkins
- School of Biotechnology and Biomolecular Sciences, UNSW Sydney, Sydney, NSW, 2052, Australia
- Ramaciotti Centre for Genomics, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Nadeem O Kaakoush
- School of Medical Sciences, Faculty of Medicine, UNSW Sydney, Sydney, NSW, 2052, Australia.
| |
Collapse
|
3
|
Zhang Z, Yue P, Lu T, Wang Y, Wei Y, Wei X. Role of lysosomes in physiological activities, diseases, and therapy. J Hematol Oncol 2021; 14:79. [PMID: 33990205 PMCID: PMC8120021 DOI: 10.1186/s13045-021-01087-1] [Citation(s) in RCA: 159] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 05/03/2021] [Indexed: 02/07/2023] Open
Abstract
Long known as digestive organelles, lysosomes have now emerged as multifaceted centers responsible for degradation, nutrient sensing, and immunity. Growing evidence also implicates role of lysosome-related mechanisms in pathologic process. In this review, we discuss physiological function of lysosomes and, more importantly, how the homeostasis of lysosomes is disrupted in several diseases, including atherosclerosis, neurodegenerative diseases, autoimmune disorders, pancreatitis, lysosomal storage disorders, and malignant tumors. In atherosclerosis and Gaucher disease, dysfunction of lysosomes changes cytokine secretion from macrophages, partially through inflammasome activation. In neurodegenerative diseases, defect autophagy facilitates accumulation of toxic protein and dysfunctional organelles leading to neuron death. Lysosomal dysfunction has been demonstrated in pathology of pancreatitis. Abnormal autophagy activation or inhibition has been revealed in autoimmune disorders. In tumor microenvironment, malignant phenotypes, including tumorigenesis, growth regulation, invasion, drug resistance, and radiotherapy resistance, of tumor cells and behaviors of tumor-associated macrophages, fibroblasts, dendritic cells, and T cells are also mediated by lysosomes. Based on these findings, a series of therapeutic methods targeting lysosomal proteins and processes have been developed from bench to bedside. In a word, present researches corroborate lysosomes to be pivotal organelles for understanding pathology of atherosclerosis, neurodegenerative diseases, autoimmune disorders, pancreatitis, and lysosomal storage disorders, and malignant tumors and developing novel therapeutic strategies.
Collapse
Affiliation(s)
- Ziqi Zhang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan People’s Republic of China
| | - Pengfei Yue
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan People’s Republic of China
| | - Tianqi Lu
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan People’s Republic of China
| | - Yang Wang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan People’s Republic of China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan People’s Republic of China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan People’s Republic of China
| |
Collapse
|
4
|
Luo Y, Chen L, Zhou Q, Xiong Y, Wang G, Liu X, Xiao Y, Ju L, Wang X. Identification of a prognostic gene signature based on an immunogenomic landscape analysis of bladder cancer. J Cell Mol Med 2020; 24:13370-13382. [PMID: 33048468 PMCID: PMC7701570 DOI: 10.1111/jcmm.15960] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 06/27/2020] [Accepted: 08/05/2020] [Indexed: 12/22/2022] Open
Abstract
Cancer immune plays a critical role in cancer progression. Tumour immunology and immunotherapy are one of the exciting areas in bladder cancer research. In this study, we aimed to develop an immune‐related gene signature to improve the prognostic prediction of bladder cancer. Firstly, we identified 392 differentially expressed immune‐related genes (IRGs) based on TCGA and ImmPort databases. Functional enrichment analysis revealed that these genes were enriched in inflammatory and immune‐related pathways, including in ‘regulation of signaling receptor activity’, ‘cytokine‐cytokine receptor interaction’ and ‘GPCR ligand binding’. Then, we separated all samples in TCGA data set into the training cohort and the testing cohort in a ratio of 3:1 randomly. Data set GSE13507 was set as the validation cohort. We constructed a prognostic six‐IRG signature with LASSO Cox regression in the training cohort, including AHNAK, OAS1, APOBEC3H, SCG2, CTSE and KIR2DS4. Six IRGs reflected the microenvironment of bladder cancer, especially immune cell infiltration. The prognostic value of six‐IRG signature was further validated in the testing cohort and the validation cohort. The results of multivariable Cox regression and subgroup analysis revealed that six‐IRG signature was a clinically independent prognostic factor for bladder cancer patients. Further, we constructed a nomogram based on six‐IRG signature and other clinicopathological risk factors, and it performed well in predict patients' survival. Finally, we found six‐IRG signature showed significant difference in different molecular subtypes of bladder cancer. In conclusions, our research provided a novel immune‐related gene signature to estimate prognosis for patients' survival with bladder cancer.
Collapse
Affiliation(s)
- Yongwen Luo
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China.,Human Genetics Resource Preservation Center of Hubei Province, Wuhan, China
| | - Liang Chen
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Qiang Zhou
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yaoyi Xiong
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Gang Wang
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China.,Human Genetics Resource Preservation Center of Hubei Province, Wuhan, China.,Laboratory of Precision Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China.,Cancer Precision Diagnosis and Treatment and Translational Medicine, Hubei Engineering Research Center, Wuhan, China
| | - Xuefeng Liu
- Department of Pathology, Lombardi Comprehensive Cancer Center, Georgetown University Medical School, Washington, DC, USA
| | - Yu Xiao
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China.,Human Genetics Resource Preservation Center of Hubei Province, Wuhan, China.,Laboratory of Precision Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Lingao Ju
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China.,Human Genetics Resource Preservation Center of Hubei Province, Wuhan, China.,Laboratory of Precision Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xinghuan Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Human Genetics Resource Preservation Center of Hubei Province, Wuhan, China.,Cancer Precision Diagnosis and Treatment and Translational Medicine, Hubei Engineering Research Center, Wuhan, China
| |
Collapse
|
5
|
Correia ACP, Calpe S, Mostafavi N, Hoefnagel SJM, Sancho-Serra MDC, de Koning PS, Krishnadath KK. Detection of circulating BMP5 as a risk factor for Barrett's esophagus. Sci Rep 2020; 10:15579. [PMID: 32968094 PMCID: PMC7511298 DOI: 10.1038/s41598-020-70760-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 06/08/2020] [Indexed: 12/11/2022] Open
Abstract
Barrett's esophagus (BE) predisposes for the malignant condition of esophageal adenocarcinoma (EAC). Since BE patients have few or no symptoms, most of these patients are not identified and not included in surveillance programs. These BE patients are at risk of developing advanced-stage EAC. At present, non-invasive tests to identify BE patients from the general population are lacking. We and others showed that Bone Morphogenetic Protein 4 (BMP4), and other BMPs are upregulated in BE. We aimed to determine if circulating BMPs can be identified and used as blood biomarkers to identify BE patients at high risk in the general population. In this study, we could detect the different BMPs in the blood of 112 BE patients and 134 age- and sex-matched controls. Concentration levels of BMP2, BMP4, and BMP5 were elevated in BE patients, with BMP2 and BMP5 significantly increased. BMP5 remained significant after multivariate analysis and was associated with an increased risk for BE with an OR of 1.49 (p value 0.01). Per log (pg/mL) of BMP5, the odds of having BE increased by 50%. Future optimization and validation studies might be needed to prove its utility as a non-invasive method for the detection of BE in high-risk populations and screening programs.
Collapse
Affiliation(s)
- Ana C P Correia
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Center, Location AMC, Amsterdam, The Netherlands.,Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
| | - Silvia Calpe
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Center, Location AMC, Amsterdam, The Netherlands.,Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
| | - Nahid Mostafavi
- Department of Gastroenterology and Hepatology, Subdivision Statistics, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
| | - Sanne Johanna Maria Hoefnagel
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Center, Location AMC, Amsterdam, The Netherlands.,Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
| | - Maria Del Carmen Sancho-Serra
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Center, Location AMC, Amsterdam, The Netherlands.,Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands.,Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK
| | - Patricia S de Koning
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
| | - Kausilia K Krishnadath
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Center, Location AMC, Amsterdam, The Netherlands. .,Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands.
| |
Collapse
|
6
|
Li L, Wu J, Liu D, Du G, Liu Y, Shang Y, Liu X. Transcriptional Profiles of Murine Bone Marrow-Derived Dendritic Cells in Response to Peste des Petits Ruminants Virus. Vet Sci 2019; 6:vetsci6040095. [PMID: 31795377 PMCID: PMC6958494 DOI: 10.3390/vetsci6040095] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 11/20/2019] [Accepted: 11/26/2019] [Indexed: 11/16/2022] Open
Abstract
Background: Peste des petits ruminants virus (PPRV) is the causative agent of PPR, which can cause an acute, highly contagious and fatal disease of sheep and goats, resulting in significant economic losses for commercial animal husbandry due to its high mortality and morbidity. As professional antigen-presenting cells, dendritic cells (DCs) play a unique role in innate immunity. This study aimed to gain a deeper understanding of the transcriptional response of bone marrow-derived dendritic cells (BMDCs) stimulated with PPRV. Results: Transcriptional profiling was performed using RNA sequencing. Herein, we reported that compared to untreatedBMDCs, 4492 differentially expressed genes (DEGs) were identified following PPRV stimulation, out of these DEGs 2311 were upregulated and 2181 were downregulated, respectively. A total of three gene ontology (GO) term clusters of biological process, cell component and molecular function were significantly enriched in 963 GO terms in the PPRV-stimulated BMDCs. These GO clusters were related to inflammatory response, cell division and vacuole, anchoring junction, positive regulation of cellular component and nucleoside binding. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways of DEGs were enriched in a chemokine signaling pathway, protein processing in endoplasmic reticulum, cell cycle and mTOR signaling pathway. Additionally, identified DEGs of BMDCs were further validated by qRT-PCR and the results were in accordance with the change of the genes. This study suggested the effects of PPRV stimulation on the maturation and function of BMDCs. Conclusion: We found that the dramatic BMDCs transcriptome changes triggered were predominantly related to an inflammatory response and chemokine signaling pathway.
Collapse
|
7
|
Pontious C, Kaul S, Hong M, Hart PA, Krishna SG, Lara LF, Conwell DL, Cruz-Monserrate Z. Cathepsin E expression and activity: Role in the detection and treatment of pancreatic cancer. Pancreatology 2019; 19:951-956. [PMID: 31582345 PMCID: PMC6829043 DOI: 10.1016/j.pan.2019.09.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 09/19/2019] [Indexed: 12/11/2022]
Abstract
Cathepsin E (CTSE) is an intracellular, hydrolytic aspartic protease found to be expressed in cells of the immune and gastrointestinal systems, lymphoid tissues, erythrocytes, and cancer cells. The precise functions are not fully understood; however, various studies have investigated its numerous cell-type specific roles. CTSE expression has been shown to be a potential early biomarker for pancreatic ductal adenocarcinoma (PDAC). PDAC patients have low survival rates mostly due to the lack of early detection methods. CTSE-specific activity probes have been developed and tested to assist in tumor imaging and functional studies investigating the role of CTSE expression in PDAC tumors. Furthermore, a CTSE protease-specific, photodynamic therapy pro-drug was developed to explore its potential use to treat tumors that express CTSE. Since CTSE is expressed in pancreatic diseases that are risk factors for PDAC, such as pancreatic cysts and chronic pancreatitis, learning about its function in these disease types could assist in early PDAC detection and in understanding the biology of PDAC progression. Overall, CTSE expression and activity shows potential to detect PDAC and other pancreatic diseases. Further research is needed to fully understand its functions and potential translational applicability.
Collapse
Affiliation(s)
- Corbin Pontious
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH, USA; The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Sabrina Kaul
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH, USA; The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Marcus Hong
- The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Kenyon College, Gambier, OH, USA
| | - Phil A Hart
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Somashekar G Krishna
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH, USA; The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Luis F Lara
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Darwin L Conwell
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH, USA; The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Zobeida Cruz-Monserrate
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH, USA; The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
8
|
Proteomic analysis of murine bone marrow derived dendritic cells in response to peste des petits ruminants virus. Res Vet Sci 2019; 125:195-204. [PMID: 31260839 DOI: 10.1016/j.rvsc.2019.06.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 06/11/2019] [Accepted: 06/18/2019] [Indexed: 02/03/2023]
Abstract
Peste des petits ruminants virus (PPRV) poses a great threat to livestock husbandry, especially goat farming due to its high mortality and morbidity. Dendritic cells (DCs), as the principal stimulators of naive Th cells were widely used in antigen processing and presenting. In the previous study, we tested the effects of PPRV on murine bone marrow derived dendritic cells (BMDCs) including surface markers and cytokines. While the aim of this study is to detect the proteomic profile of BMDCs stimulated with PPRV towards key proteins involved in. Following PPRV stimulation, 110 differentially expressed proteins (DEPs) were identified through iTRAQ labelling with LC-MS/MS approach, of which 94 DEPs were up-regulated and 16 DEPs were down-regulated, respectively. Among them 15 out of 110 DGPs were related to innate immune system, three were involved in cell apoptosis, RPS15a and Smox were related to translation of viral mRNA. Additionally, western blot analysis showed identical results to iTRAQ analysis. There will be profound significance for understanding antigen-presenting of BMDCs after stimulation with PPRV.
Collapse
|
9
|
Abstract
Cathepsins (CTS) are mainly lysosomal acid hydrolases extensively involved in the prognosis of different diseases, and having a distinct role in tumor progression by regulating cell proliferation, autophagy, angiogenesis, invasion, and metastasis. As all these processes conjunctively lead to cancer progression, their site-specific regulation might be beneficial for cancer treatment. CTS regulate activation of the proteolytic cascade and protein turnover, while extracellular CTS is involved in promoting extracellular matrix degradation and angiogenesis, thereby stimulating invasion and metastasis. Despite cancer regulation, the involvement of CTS in cellular adaptation toward chemotherapy and radiotherapy augments their therapeutic potential. However, lysosomal permeabilization mediated cytosolic translocation of CTS induces programmed cell death. This complex behavior of CTS generates the need to discuss the different aspects of CTS associated with cancer regulation. In this review, we mainly focused on the significance of each cathepsin in cancer signaling and their targeting which would provide noteworthy information in the context of cancer biology and therapeutics.
Collapse
Affiliation(s)
- Tejinder Pal Khaket
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk 38453, Republic of Korea
| | - Taeg Kyu Kwon
- Department of Immunology, School of Medicine, Keimyung University, Dalseo-Gu, Daegu 704-701, Republic of Korea.
| | - Sun Chul Kang
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk 38453, Republic of Korea.
| |
Collapse
|
10
|
Creemers A, Ebbing EA, Pelgrim TC, Lagarde SM, van Etten-Jamaludin FS, van Berge Henegouwen MI, Hulshof MCCM, Krishnadath KK, Meijer SL, Bijlsma MF, van Oijen MGH, van Laarhoven HWM. A systematic review and meta-analysis of prognostic biomarkers in resectable esophageal adenocarcinomas. Sci Rep 2018; 8:13281. [PMID: 30185893 PMCID: PMC6125467 DOI: 10.1038/s41598-018-31548-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 08/20/2018] [Indexed: 02/07/2023] Open
Abstract
Targeted therapy is lagging behind in esophageal adenocarcinoma (EAC). To guide the development of new treatment strategies, we provide an overview of the prognostic biomarkers in resectable EAC treated with curative intent. The Medline, Cochrane and EMBASE databases were systematically searched, focusing on overall survival (OS). The quality of the studies was assessed using a scoring system ranging from 0-7 points based on modified REMARK criteria. To evaluate all identified prognostic biomarkers, the hallmarks of cancer were adapted to fit all biomarkers based on their biological function in EAC, resulting in the features angiogenesis, cell adhesion and extra-cellular matrix remodeling, cell cycle, immune, invasion and metastasis, proliferation, and self-renewal. Pooled hazard ratios (HR) and 95% confidence intervals (CI) were derived by random effects meta-analyses performed on each hallmarks of cancer feature. Of the 3298 unique articles identified, 84 were included, with a mean quality of 5.9 points (range 3.5-7). The hallmarks of cancer feature 'immune' was most significantly associated with worse OS (HR 1.88, (95%CI 1.20-2.93)). Of the 82 unique prognostic biomarkers identified, meta-analyses showed prominent biomarkers, including COX-2, PAK-1, p14ARF, PD-L1, MET, LC3B, IGFBP7 and LGR5, associated to each hallmark of cancer.
Collapse
Affiliation(s)
- Aafke Creemers
- Laboratory of Experimental Oncology and Radiobiology, Amsterdam UMC, Univ of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands.
- Department of Medical Oncology, Amsterdam UMC, Univ of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands.
| | - Eva A Ebbing
- Laboratory of Experimental Oncology and Radiobiology, Amsterdam UMC, Univ of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Medical Oncology, Amsterdam UMC, Univ of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Thomas C Pelgrim
- Department of Medical Oncology, Amsterdam UMC, Univ of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Sjoerd M Lagarde
- Department of Surgery, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Faridi S van Etten-Jamaludin
- Department of Medical Library Science, Amsterdam UMC, Univ of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | | | - Maarten C C M Hulshof
- Department of Radiotherapy, Amsterdam UMC, Univ of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Kausilia K Krishnadath
- Laboratory of Experimental Oncology and Radiobiology, Amsterdam UMC, Univ of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Gastroenterology, Amsterdam UMC, Univ of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Sybren L Meijer
- Department of Pathology, Amsterdam UMC, Univ of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Maarten F Bijlsma
- Laboratory of Experimental Oncology and Radiobiology, Amsterdam UMC, Univ of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Martijn G H van Oijen
- Department of Medical Oncology, Amsterdam UMC, Univ of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Hanneke W M van Laarhoven
- Laboratory of Experimental Oncology and Radiobiology, Amsterdam UMC, Univ of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Medical Oncology, Amsterdam UMC, Univ of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
11
|
Abstract
Cathepsins are lysosomal peptidases belonging to the papain family, and based on their catalytic sites, these enzymes can be divided into serine, cysteine and aspartic proteases. The studies conducted to date have identified, 15 types of cathepsins that are widely distributed in intracellular and extracellular spaces. These proteases participate in various pathological activities, including the occurrence and development of human cancers. Several recent studies suggest that cathepsins, particularly cathepsins B, D, E and L, contribute to digestive tumorigenesis. Cathepsins were found to promote the development of most digestive cancers except liver cancer, in which they might have the opposite effects. Due to their important roles in digestive tumors, cathepsins might be therapeutic targets for the treatment of digestive cancers.
Collapse
|
12
|
Mengwasser J, Babes L, Cordes S, Mertlitz S, Riesner K, Shi Y, McGearey A, Kalupa M, Reinheckel T, Penack O. Cathepsin E Deficiency Ameliorates Graft-versus-Host Disease and Modifies Dendritic Cell Motility. Front Immunol 2017; 8:203. [PMID: 28298913 PMCID: PMC5331043 DOI: 10.3389/fimmu.2017.00203] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 02/14/2017] [Indexed: 01/15/2023] Open
Abstract
Microbial products influence immunity after allogeneic hematopoietic stem cell transplantation (allo-SCT). In this context, the role of cathepsin E (Ctse), an aspartate protease known to cleave bacterial peptides for antigen presentation in dendritic cells (DCs), has not been studied. During experimental acute graft-versus-host disease (GVHD), we found infiltration by Ctse-positive immune cells leading to higher Ctse RNA- and protein levels in target organs. In Ctse-deficient allo-SCT recipients, we found ameliorated GVHD, improved survival, and lower numbers of tissue-infiltrating DCs. Donor T cell proliferation was not different in Ctse-deficient vs. wild-type allo-SCT recipients in MHC-matched and MHC-mismatched models. Furthermore, Ctse-deficient DCs had an intact ability to induce allogeneic T cell proliferation, suggesting that its role in antigen presentation may not be the main mechanism how Ctse impacts GVHD. We found that Ctse deficiency significantly decreases DC motility in vivo, reduces adhesion to extracellular matrix (ECM), and diminishes invasion through ECM. We conclude that Ctse has a previously unrecognized role in regulating DC motility that possibly contributes to reduced DC counts and ameliorated inflammation in GVHD target organs of Ctse-deficient allo-SCT recipients. However, our data do not provide definite proof that the observed effect of Ctse−/− deficiency is exclusively mediated by DCs. A contribution of Ctse−/−-mediated functions in other recipient cell types, e.g., macrophages, cannot be excluded.
Collapse
Affiliation(s)
- Jörg Mengwasser
- Medical Department, Division of Hematology, Oncology and Tumor Immunology, Charité University Medicine , Berlin , Germany
| | - Liane Babes
- Faculty of Medicine, BIOSS Centre for Biological Signalling Studies, Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University Freiburg , Freiburg , Germany
| | - Steffen Cordes
- Medical Department, Division of Hematology, Oncology and Tumor Immunology, Charité University Medicine , Berlin , Germany
| | - Sarah Mertlitz
- Medical Department, Division of Hematology, Oncology and Tumor Immunology, Charité University Medicine , Berlin , Germany
| | - Katarina Riesner
- Medical Department, Division of Hematology, Oncology and Tumor Immunology, Charité University Medicine , Berlin , Germany
| | - Yu Shi
- Medical Department, Division of Hematology, Oncology and Tumor Immunology, Charité University Medicine , Berlin , Germany
| | - Aleixandria McGearey
- Medical Department, Division of Hematology, Oncology and Tumor Immunology, Charité University Medicine , Berlin , Germany
| | - Martina Kalupa
- Medical Department, Division of Hematology, Oncology and Tumor Immunology, Charité University Medicine , Berlin , Germany
| | - Thomas Reinheckel
- Faculty of Medicine, BIOSS Centre for Biological Signalling Studies, Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University Freiburg , Freiburg , Germany
| | - Olaf Penack
- Medical Department, Division of Hematology, Oncology and Tumor Immunology, Charité University Medicine , Berlin , Germany
| |
Collapse
|