1
|
Ngana GS, Di Bernardo MA, Surette MG, MacNeil LT. Actinomyces viscosus promotes neuroprotection in C. elegans models of Parkinson's disease. Mech Ageing Dev 2025; 225:112061. [PMID: 40258426 DOI: 10.1016/j.mad.2025.112061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 04/04/2025] [Accepted: 04/11/2025] [Indexed: 04/23/2025]
Abstract
Parkinson's Disease is characterized by selective degeneration of dopaminergic neurons, primarily in the substantia nigra pars compacta, as well as accumulation of alpha-synuclein enriched protein aggregates within neurons. The pathogenesis of PD is still not completely understood, and no treatments exist that alter disease progression. Obvious genetic causes are detected in only a small number of PD patients (5-10 %), suggesting that environmental factors play a significant role the development of PD. Correlative studies suggest that the microbiota could be an important environmental modifier of neurodegeneration. We identified a microbiotal isolate, Actinomyces viscosus, that reduced neurodegeneration in C. elegans expressing a pathological mutant form (G2019S) of leucine-rich repeat kinase 2 (LRRK2) in dopaminergic neurons. A. viscosus also suppressed autophagic dysfunction in these animals and reduced alpha-synuclein aggregation in a synucleinopathy model. Global gene expression analysis revealed increased expression of aspartic cathepsins in response to A. viscosus. Consistent with the involvement of these proteins in neuroprotection, we found that reducing aspartic cathepsin function increased neurodegeneration in the LRRK2 transgenic model. Our findings contribute to the current understanding of how the gut microbiota may influence PD, elucidating one potential mechanism of microbiota-mediated neuroprotection.
Collapse
Affiliation(s)
- G Sophie Ngana
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main St W., Hamilton, ON, Canada
| | - Mercedes A Di Bernardo
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main St W., Hamilton, ON, Canada
| | - Michael G Surette
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main St W., Hamilton, ON, Canada; Farncombe Family Digestive Health Research Institute, McMaster University, 1280 Main St W, Hamilton, ON, Canada; Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, 1280 Main St W, Hamilton ON, Canada; Department of Medicine, McMaster University, 1280 Main St W, Hamilton, ON, Canada
| | - Lesley T MacNeil
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main St W., Hamilton, ON, Canada; Farncombe Family Digestive Health Research Institute, McMaster University, 1280 Main St W, Hamilton, ON, Canada; Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, 1280 Main St W, Hamilton ON, Canada.
| |
Collapse
|
2
|
Giri A, Hong IS, Kwon TK, Kang JS, Jeong JH, Kweon S, Yook S. Exploring therapeutic and diagnostic potential of cysteine cathepsin as targets for cancer therapy with nanomedicine. Int J Biol Macromol 2025; 315:144324. [PMID: 40398760 DOI: 10.1016/j.ijbiomac.2025.144324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2025] [Revised: 05/05/2025] [Accepted: 05/15/2025] [Indexed: 05/23/2025]
Abstract
Cysteine cathepsins have been discovered to be substantially expressed in multiple types of cancer. They play a key role in the progression and growth of these cancers, rendering them appealing targets for nanoscale delivery and noninvasive diagnostic imaging. This review explores cathepsins from the papain-like enzyme family (C1) within the cysteine peptidase clan (CA), emphasizing the role of cathepsin-responsive nanoparticles in tumor growth. Furthermore, it also explores how nanotechnology can harness cathepsin activity to enable targeted drug delivery, improve tumor imaging, and reduce systemic toxicity. By examining the molecular mechanisms governing cathepsin function and evaluating different nanocarrier systems, this work aims to enhance our understanding of targeted cancer treatment. Despite significant advances, challenges remain in translating these nanomedicine platforms into clinical use, including improving delivery efficiency, biocompatibility, long-term safety, and addressing issues such as interspecies protease variability and scalable nanomanufacturing. Future advancement, integrating advanced biomaterials, patient-derived organoid models, bispecific immune-protease targeting, CRISPR-based cathepsin editing, and artificial intelligence-driven pharmacokinetic modeling and analysis will be critical to fully realizing the clinical potential of cathepsin targeted nanomedicines. These innovations hold promises for advancing precision oncology by overcoming current limitations and improving patient outcomes.
Collapse
Affiliation(s)
- Anil Giri
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - In-Sun Hong
- Department of Molecular Medicine, School of Medicine, Gachon University, Incheon 406-840, Republic of Korea
| | - Taeg Kyu Kwon
- Department of Immunology, School of Medicine, Keimyung University, Daegu 42601, Republic of Korea; Center for Forensic Pharmaceutical Science, Keimyung University, Daegu, 42601, Republic of Korea
| | - Jong-Sun Kang
- Department of Molecular Cell Biology, School of Medicine, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jee-Heon Jeong
- Department of Precision Medicine, School of Medicine, Sungkyunkwan University, Suwon, Gyeonggi 16419, Republic of Korea
| | - Seho Kweon
- College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea.
| | - Simmyung Yook
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea; School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
3
|
Lufrano D, Gong C, Cecarini V, Cuccioloni M, Bonfili L, Sturaro C, Bettegazzi B, Ruzza C, Perelló M, Angeletti M, Eleuteri AM. An Insight into Neuronal Processing of Ghrelin: Effects of a Bioactive Ghrelin Derivative on Proteolytic Pathways and Mitophagy. Mol Neurobiol 2025:10.1007/s12035-025-04976-5. [PMID: 40285938 DOI: 10.1007/s12035-025-04976-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 04/16/2025] [Indexed: 04/29/2025]
Abstract
Protein homeostasis (proteostasis) is preserved by an orchestrated network of molecular mechanisms that regulate protein synthesis, folding, and degradation, ensuring cellular integrity and function. Proteostasis declines with age and is related to pathologies such as neurodegenerative diseases and cardiac disorders, which are accompanied by the accumulation of toxic protein aggregates. In this context, therapeutic strategies enhancing the two primary degradative systems involved in the cellular clearance of those abnormal proteins, namely ubiquitin-proteasome system and autophagy-lysosomal pathway, represent a promising approach to counteract the collapse of proteostasis in such pathological conditions. In this work, we explored the processing of ghrelin, a pleiotropic peptide hormone linked to energy metabolism and higher brain functions, which is reported to modulate the protein degradative mechanisms. According to our data, ghrelin is processed by serine hydrolases secreted into the conditioned medium of SH-SY5Y neuroblastoma cell line, commonly used in neurotoxicology and neuroscience research. Ghrelin processing leads to the formation of a shorter peptide (ghrelin(1-11)) that stimulates both the cell proteasome system and autophagy-lysosomal pathway, encompassing the selective autophagy of mitochondria. Our findings suggest that ghrelin processing may contribute to the maintenance of neuronal proteostasis.
Collapse
Affiliation(s)
- Daniela Lufrano
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032, Camerino, Italy.
- Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional La Plata (UNLP), CONICET, B1900 AVW, La Plata, Argentina.
| | - Chunmei Gong
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032, Camerino, Italy
- Key Laboratory of Tropical Translational Medicine of the Ministry of Education, Hainan Medical University, 571199, Haikou, China
| | - Valentina Cecarini
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032, Camerino, Italy
| | - Massimiliano Cuccioloni
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032, Camerino, Italy
| | - Laura Bonfili
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032, Camerino, Italy
| | - Chiara Sturaro
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121, Ferrara, Italy
| | | | - Chiara Ruzza
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121, Ferrara, Italy
| | - Mario Perelló
- Grupo de Neurofisiología, Instituto Multidisciplinario de Biología Celular (IMBICE), Universidad Nacional La Plata (UNLP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) y Comisión de Investigaciones Científicas de La Provincia de Buenos Aires (CIC-PBA), B1906 APM, La Plata, Argentina
| | - Mauro Angeletti
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032, Camerino, Italy
| | - Anna Maria Eleuteri
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032, Camerino, Italy.
| |
Collapse
|
4
|
Zhang Z, Wang J, Shi Y, Wang B, Wang D. Cathepsin L promotes oesophageal squamous cell carcinoma development and may be associated with tumour-associated macrophages. Heliyon 2024; 10:e29273. [PMID: 38601581 PMCID: PMC11004422 DOI: 10.1016/j.heliyon.2024.e29273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/02/2024] [Accepted: 04/03/2024] [Indexed: 04/12/2024] Open
Abstract
Background Oesophageal squamous cell carcinoma (ESCC) is a leading cause of cancer-related deaths worldwide because existing treatments are often insufficient. Therefore, new, reliable biomarkers must be identified. CTSL overexpression is closely associated with tumour progression and poor prognosis. However, the role and mechanism of CTSL as an oncogene in ESCC remain unclear. Methods Genome-wide association study (GWAS) data were used for Mendelian randomization analysis to investigate the possible relationships between CTSL and ESCC. The correlation between CTSL expression and prognosis was analysed using GEO, TCGA, and GEPIA data. We compared CTSL expression among the cell types using single-cell sequencing. Correlations between CTSL and the tumour microenvironment, immune cell infiltration, tumour mutational load, immunological checkpoints, and treatment sensitivity in patients with ESCC were investigated. Finally, using mouse models and cellular investigations, we assessed the effects of CTSL on the growth, apoptosis, and metastasis of ESCC tumour cells. Results CTSL was overexpressed in ESCC and correlated with prognosis. We also discovered its close association with cell immunity, especially with tumour-associated macrophages and immune checkpoints in the tumour microenvironment. CTSL may play a key role in ESCC development by affecting M2 macrophage polarisation. CTSL and the M2 macrophage marker genes showed significant positive correlations. Mendelian randomization analysis confirmed a relationship between CTSL and ESCC. Finally, our in vitro and in vivo experiments demonstrated that CTSL promoted the proliferation and migration of ESCC cells, validating our bioinformatic analysis. Conclusion CTSL emerged as a crucial gene in ESCC that influences patient prognosis and immunity, particularly in association with M2 macrophages. Therefore, targeting or modulating CTSL levels may provide new therapeutic strategies for patients with ESCC.
Collapse
Affiliation(s)
- Zhenhu Zhang
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
| | - Jianyu Wang
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
| | - Yamin Shi
- School of Foreign Languages, Shandong University of Finance and Economics, Jinan, 250014, China
| | - Ben Wang
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
| | - Dong Wang
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
| |
Collapse
|
5
|
Eriksson I, Öllinger K. Lysosomes in Cancer-At the Crossroad of Good and Evil. Cells 2024; 13:459. [PMID: 38474423 PMCID: PMC10930463 DOI: 10.3390/cells13050459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/27/2024] [Accepted: 03/01/2024] [Indexed: 03/14/2024] Open
Abstract
Although it has been known for decades that lysosomes are central for degradation and recycling in the cell, their pivotal role as nutrient sensing signaling hubs has recently become of central interest. Since lysosomes are highly dynamic and in constant change regarding content and intracellular position, fusion/fission events allow communication between organelles in the cell, as well as cell-to-cell communication via exocytosis of lysosomal content and release of extracellular vesicles. Lysosomes also mediate different forms of regulated cell death by permeabilization of the lysosomal membrane and release of their content to the cytosol. In cancer cells, lysosomal biogenesis and autophagy are increased to support the increased metabolism and allow growth even under nutrient- and oxygen-poor conditions. Tumor cells also induce exocytosis of lysosomal content to the extracellular space to promote invasion and metastasis. However, due to the enhanced lysosomal function, cancer cells are often more susceptible to lysosomal membrane permeabilization, providing an alternative strategy to induce cell death. This review summarizes the current knowledge of cancer-associated alterations in lysosomal structure and function and illustrates how lysosomal exocytosis and release of extracellular vesicles affect disease progression. We focus on functional differences depending on lysosomal localization and the regulation of intracellular transport, and lastly provide insight how new therapeutic strategies can exploit the power of the lysosome and improve cancer treatment.
Collapse
Affiliation(s)
- Ida Eriksson
- Division of Cell Biology, Department of Biomedical and Clinical Sciences, Linköping University, 58185 Linköping, Sweden;
| | | |
Collapse
|
6
|
Xu J, Xu W, Choi J, Brhane Y, Christiani DC, Kothari J, McKay J, Field JK, Davies MPA, Liu G, Amos CI, Hung RJ, Briollais L. Large-scale whole exome sequencing studies identify two genes,CTSL and APOE, associated with lung cancer. PLoS Genet 2023; 19:e1010902. [PMID: 37738239 PMCID: PMC10516417 DOI: 10.1371/journal.pgen.1010902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 08/07/2023] [Indexed: 09/24/2023] Open
Abstract
Common genetic variants associated with lung cancer have been well studied in the past decade. However, only 12.3% heritability has been explained by these variants. In this study, we investigate the contribution of rare variants (RVs) (minor allele frequency <0.01) to lung cancer through two large whole exome sequencing case-control studies. We first performed gene-based association tests using a novel Bayes Factor statistic in the International Lung Cancer Consortium, the discovery study (European, 1042 cases vs. 881 controls). The top genes identified are further assessed in the UK Biobank (European, 630 cases vs. 172 864 controls), the replication study. After controlling for the false discovery rate, we found two genes, CTSL and APOE, significantly associated with lung cancer in both studies. Single variant tests in UK Biobank identified 4 RVs (3 missense variants) in CTSL and 2 RVs (1 missense variant) in APOE stongly associated with lung cancer (OR between 2.0 and 139.0). The role of these genetic variants in the regulation of CTSL or APOE expression remains unclear. If such a role is established, this could have important therapeutic implications for lung cancer patients.
Collapse
Affiliation(s)
- Jingxiong Xu
- Prosserman Centre for Population Health Research, Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
| | - Wei Xu
- Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Jiyeon Choi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Yonathan Brhane
- Prosserman Centre for Population Health Research, Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
| | - David C. Christiani
- T. H. Chan School of Public Health, Harvard University, Boston, Massachusetts, United States of America
| | - Jui Kothari
- Department of Environmental Health, T. H. Chan School of Public Health, Harvard University, Boston, Massachusetts, United States of America
| | - James McKay
- International Agency for Research on Cancer, Lyon, France
| | - John K. Field
- Department of Molecular and Clinical Cancer Medicine, The University of Liverpool, Liverpool, United Kingdom
| | - Michael P. A. Davies
- Department of Molecular and Clinical Cancer Medicine, The University of Liverpool, Liverpool, United Kingdom
| | - Geoffrey Liu
- Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Christopher I. Amos
- Dan L. Duncan Comprehensive Cancer Center, Department of Medicine, Baylor College of Medicine, Houston, Texas, United States of America
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, Texas, United States of America
| | - Rayjean J. Hung
- Prosserman Centre for Population Health Research, Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Laurent Briollais
- Prosserman Centre for Population Health Research, Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
7
|
Mustafa A, Elkhamisy F, Arghiani N, Pranjol MZI. Potential crosstalk between pericytes and cathepsins in the tumour microenvironment. Biomed Pharmacother 2023; 164:114932. [PMID: 37236029 DOI: 10.1016/j.biopha.2023.114932] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/17/2023] [Accepted: 05/22/2023] [Indexed: 05/28/2023] Open
Abstract
Cancer remains a formidable global health challenge, and as such, investigators are constantly exploring underlying mechanisms that drive its progression. One area of interest is the role of lysosomal enzymes, such as cathepsins, in regulating cancer growth and development in the tumour microenvironment (TME). Pericytes, a key component of vasculature, play a key role in regulating blood vessel formation in the TME, have been shown to be influenced by cathepsins and their activity. Although cathepsins such as cathepsins D and L have been shown to induce angiogenesis, currently no direct link is known between pericytes and cathepsins interaction. This review aims to shed light on the potential interplay between pericytes and cathepsins in the TME, highlighting the possible implications for cancer therapy and future research directions.
Collapse
Affiliation(s)
- A Mustafa
- School of Life Sciences, University of Sussex, Falmer, Brighton BN1 9QG, UK
| | - F Elkhamisy
- School of Life Sciences, University of Sussex, Falmer, Brighton BN1 9QG, UK
| | - N Arghiani
- School of Life Sciences, University of Sussex, Falmer, Brighton BN1 9QG, UK; Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden.
| | - M Z I Pranjol
- School of Life Sciences, University of Sussex, Falmer, Brighton BN1 9QG, UK.
| |
Collapse
|
8
|
Di Sante M, Antonucci S, Pontarollo L, Cappellaro I, Segat F, Deshwal S, Greotti E, Grilo LF, Menabò R, Di Lisa F, Kaludercic N. Monoamine oxidase A-dependent ROS formation modulates human cardiomyocyte differentiation through AKT and WNT activation. Basic Res Cardiol 2023; 118:4. [PMID: 36670288 PMCID: PMC9859871 DOI: 10.1007/s00395-023-00977-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 12/21/2022] [Accepted: 01/07/2023] [Indexed: 01/21/2023]
Abstract
During embryonic development, cardiomyocytes undergo differentiation and maturation, processes that are tightly regulated by tissue-specific signaling cascades. Although redox signaling pathways involved in cardiomyogenesis are established, the exact sources responsible for reactive oxygen species (ROS) formation remain elusive. The present study investigates whether ROS produced by the mitochondrial flavoenzyme monoamine oxidase A (MAO-A) play a role in cardiomyocyte differentiation from human induced pluripotent stem cells (hiPSCs). Wild type (WT) and MAO-A knock out (KO) hiPSCs were generated by CRISPR/Cas9 genome editing and subjected to cardiomyocyte differentiation. Mitochondrial ROS levels were lower in MAO-A KO compared to the WT cells throughout the differentiation process. MAO-A KO hiPSC-derived cardiomyocytes (hiPSC-CMs) displayed sarcomere disarray, reduced α- to β-myosin heavy chain ratio, GATA4 upregulation and lower macroautophagy levels. Functionally, genetic ablation of MAO-A negatively affected intracellular Ca2+ homeostasis in hiPSC-CMs. Mechanistically, MAO-A generated ROS contributed to the activation of AKT signaling that was considerably attenuated in KO cells. In addition, MAO-A ablation caused a reduction in WNT pathway gene expression consistent with its reported stimulation by ROS. As a result of WNT downregulation, expression of MESP1 and NKX2.5 was significantly decreased in MAO-A KO cells. Finally, MAO-A re-expression during differentiation rescued expression levels of cardiac transcription factors, contractile structure, and intracellular Ca2+ homeostasis. Taken together, these results suggest that MAO-A mediated ROS generation is necessary for the activation of AKT and WNT signaling pathways during cardiac lineage commitment and for the differentiation of fully functional human cardiomyocytes.
Collapse
Affiliation(s)
- Moises Di Sante
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi 58/B, 35131, Padua, Italy
| | - Salvatore Antonucci
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi 58/B, 35131, Padua, Italy
| | - Laura Pontarollo
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi 58/B, 35131, Padua, Italy
| | - Ilaria Cappellaro
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi 58/B, 35131, Padua, Italy
| | - Francesca Segat
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi 58/B, 35131, Padua, Italy
| | - Soni Deshwal
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi 58/B, 35131, Padua, Italy
- Max Planck Institute for Biology of Ageing, 50931, Cologne, Germany
| | - Elisa Greotti
- Neuroscience Institute, National Research Council of Italy (CNR), Via Ugo Bassi 58/B, 35131, Padua, Italy
| | - Luis F Grilo
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
| | - Roberta Menabò
- Neuroscience Institute, National Research Council of Italy (CNR), Via Ugo Bassi 58/B, 35131, Padua, Italy
| | - Fabio Di Lisa
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi 58/B, 35131, Padua, Italy.
- Neuroscience Institute, National Research Council of Italy (CNR), Via Ugo Bassi 58/B, 35131, Padua, Italy.
| | - Nina Kaludercic
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi 58/B, 35131, Padua, Italy.
- Neuroscience Institute, National Research Council of Italy (CNR), Via Ugo Bassi 58/B, 35131, Padua, Italy.
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza (IRP), 35127, Padua, Italy.
| |
Collapse
|
9
|
Huertas J, Lee HT. Multi‑faceted roles of cathepsins in ischemia reperfusion injury (Review). Mol Med Rep 2022; 26:368. [PMID: 36300202 PMCID: PMC9644425 DOI: 10.3892/mmr.2022.12885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 10/06/2022] [Indexed: 11/05/2022] Open
Abstract
Cathepsins are one of the most abundant proteases within the lysosomes with diverse physiological effects ranging from immune responses, cell death and intracellular protein degradation. Cathepsins are involved in extracellular and systemic functions such as systemic inflammation and extracellular matrix degradation. Ischemia reperfusion (IR) injury is responsible for numerous diseases including myocardial infarction, acute kidney injury, stroke and acute graft failure after transplant surgery. Inflammation plays a major role in the reperfusion phase of IR injury and previous research has shown that cathepsins are key mediators of the inflammation cascade as well as apoptosis. Taken together, cathepsins modulation could provide potential therapeutic approaches to attenuate IR injury. The present review summarized the current understanding of various cathepsin subtypes, their major physiologic functions, their roles in multi‑organ IR injury and detailed selective cathepsin inhibitors with therapeutic potential.
Collapse
Affiliation(s)
- Jaime Huertas
- Department of Anesthesiology, College of Physicians and Surgeons of Columbia University, New York, NY 10032-3784, USA
| | - H. Thomas Lee
- Department of Anesthesiology, College of Physicians and Surgeons of Columbia University, New York, NY 10032-3784, USA
| |
Collapse
|
10
|
Shi Q, Shen Q, Liu Y, Shi Y, Huang W, Wang X, Li Z, Chai Y, Wang H, Hu X, Li N, Zhang Q, Cao X. Increased glucose metabolism in TAMs fuels O-GlcNAcylation of lysosomal Cathepsin B to promote cancer metastasis and chemoresistance. Cancer Cell 2022; 40:1207-1222.e10. [PMID: 36084651 DOI: 10.1016/j.ccell.2022.08.012] [Citation(s) in RCA: 160] [Impact Index Per Article: 53.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 07/06/2022] [Accepted: 08/15/2022] [Indexed: 12/24/2022]
Abstract
How glucose metabolism remodels pro-tumor functions of tumor-associated macrophages (TAMs) needs further investigation. Here we show that M2-like TAMs bear the highest individual capacity to take up intratumoral glucose. Their increased glucose uptake fuels hexosamine biosynthetic pathway-dependent O-GlcNAcylation to promote cancer metastasis and chemoresistance. Glucose metabolism promotes O-GlcNAcylation of the lysosome-encapsulated protease Cathepsin B at serine 210, mediated by lysosome-localized O-GlcNAc transferase (OGT), elevating mature Cathepsin B in macrophages and its secretion in the tumor microenvironment (TME). Loss of OGT in macrophages reduces O-GlcNAcylation and mature Cathepsin B in the TME and disrupts cancer metastasis and chemoresistance. Human TAMs with high OGT are positively correlated with Cathepsin B expression, and both levels predict chemotherapy response and prognosis of individuals with cancer. Our study reports the biological and potential clinical significance of glucose metabolism in tumor-promoting TAMs and reveals insights into the underlying mechanisms.
Collapse
Affiliation(s)
- Qingzhu Shi
- Department of Immunology, Institute of Basic Medical Research, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100005, China; National Key Laboratory of Medical Immunology, Institute of Immunology, Naval Medical University, Shanghai 200433, China
| | - Qicong Shen
- National Key Laboratory of Medical Immunology, Institute of Immunology, Naval Medical University, Shanghai 200433, China
| | - Yanfang Liu
- Department of Pathology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Yang Shi
- Department of Immunology, Institute of Basic Medical Research, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100005, China; National Key Laboratory of Medical Immunology, Institute of Immunology, Naval Medical University, Shanghai 200433, China
| | - Wenwen Huang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xi Wang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Zhiqing Li
- National Key Laboratory of Medical Immunology, Institute of Immunology, Naval Medical University, Shanghai 200433, China
| | - Yangyang Chai
- Department of Immunology, Institute of Basic Medical Research, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Hao Wang
- Department of Colorectal Surgery, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Xiangjia Hu
- National Key Laboratory of Medical Immunology, Institute of Immunology, Naval Medical University, Shanghai 200433, China
| | - Nan Li
- National Key Laboratory of Medical Immunology, Institute of Immunology, Naval Medical University, Shanghai 200433, China
| | - Qian Zhang
- National Key Laboratory of Medical Immunology, Institute of Immunology, Naval Medical University, Shanghai 200433, China.
| | - Xuetao Cao
- Department of Immunology, Institute of Basic Medical Research, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100005, China; National Key Laboratory of Medical Immunology, Institute of Immunology, Naval Medical University, Shanghai 200433, China; Frontier Research Center for Cell Response, Institute of Immunology, College of Life Sciences, Nankai University, Tianjin 300071, China.
| |
Collapse
|
11
|
The Key Role of Lysosomal Protease Cathepsins in Viral Infections. Int J Mol Sci 2022; 23:ijms23169089. [PMID: 36012353 PMCID: PMC9409221 DOI: 10.3390/ijms23169089] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/10/2022] [Accepted: 08/12/2022] [Indexed: 11/17/2022] Open
Abstract
Cathepsins encompass a family of lysosomal proteases that mediate protein degradation and turnover. Although mainly localized in the endolysosomal compartment, cathepsins are also found in the cytoplasm, nucleus, and extracellular space, where they are involved in cell signaling, extracellular matrix assembly/disassembly, and protein processing and trafficking through the plasma and nuclear membrane and between intracellular organelles. Ubiquitously expressed in the body, cathepsins play regulatory roles in a wide range of physiological processes including coagulation, hormone secretion, immune responses, and others. A dysregulation of cathepsin expression and/or activity has been associated with many human diseases, including cancer, diabetes, obesity, cardiovascular and inflammatory diseases, kidney dysfunctions, and neurodegenerative disorders, as well as infectious diseases. In viral infections, cathepsins may promote (1) activation of the viral attachment glycoproteins and entry of the virus into target cells; (2) antigen processing and presentation, enabling the virus to replicate in infected cells; (3) up-regulation and processing of heparanase that facilitates the release of viral progeny and the spread of infection; and (4) activation of cell death that may either favor viral clearance or assist viral propagation. In this review, we report the most relevant findings on the molecular mechanisms underlying cathepsin involvement in viral infection physiopathology, and we discuss the potential of cathepsin inhibitors for therapeutical applications in viral infectious diseases.
Collapse
|
12
|
Hsu CH, Lee KJ, Chiu YH, Huang KC, Wang GS, Chen LP, Liao KW, Lin CS. The Lysosome in Malignant Melanoma: Biology, Function and Therapeutic Applications. Cells 2022; 11:1492. [PMID: 35563798 PMCID: PMC9103375 DOI: 10.3390/cells11091492] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 04/21/2022] [Accepted: 04/26/2022] [Indexed: 02/04/2023] Open
Abstract
Lysosomes are membrane-bound vesicles that play roles in the degradation and recycling of cellular waste and homeostasis maintenance within cells. False alterations of lysosomal functions can lead to broad detrimental effects and cause various diseases, including cancers. Cancer cells that are rapidly proliferative and invasive are highly dependent on effective lysosomal function. Malignant melanoma is the most lethal form of skin cancer, with high metastasis characteristics, drug resistance, and aggressiveness. It is critical to understand the role of lysosomes in melanoma pathogenesis in order to improve the outcomes of melanoma patients. In this mini-review, we compile our current knowledge of lysosomes' role in tumorigenesis, progression, therapy resistance, and the current treatment strategies related to lysosomes in melanoma.
Collapse
Affiliation(s)
- Chia-Hsin Hsu
- Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei 10617, Taiwan;
- Department of Biomedical Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Keng-Jung Lee
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA;
| | - Yi-Han Chiu
- Department of Microbiology, Soochow University, Taipei 10617, Taiwan;
| | - Kuo-Ching Huang
- Holistic Education Center, Mackay Medical College, New Taipei City 25245, Taiwan;
| | - Guo-Shou Wang
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan; (G.-S.W.); (K.-W.L.)
| | - Lei-Po Chen
- Ph.D. Degree Program of Biomedical Science and Engineering, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan;
| | - Kuang-Wen Liao
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan; (G.-S.W.); (K.-W.L.)
| | - Chen-Si Lin
- Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei 10617, Taiwan;
| |
Collapse
|
13
|
Integrated analysis of single-cell and bulk RNA-sequencing identifies a signature based on B cell marker genes to predict prognosis and immunotherapy response in lung adenocarcinoma. Cancer Immunol Immunother 2022; 71:2341-2354. [PMID: 35152302 DOI: 10.1007/s00262-022-03143-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 12/27/2021] [Indexed: 02/07/2023]
Abstract
As an essential component of the tumor microenvironment, B cells exist in all stages of tumor and exert important roles in anti-tumor immunity and shaping tumor development. We aimed to explore the expression profile of B cell marker genes and construct a prognostic signature based on these genes in Lung adenocarcinoma (LUAD). A total of 1268 LUAD patients from different cohorts were enrolled in this study. We performed an analysis of single-cell RNA-sequencing (scRNA-seq) data from Gene expression omnibus (GEO) database to identify B cell marker genes in LUAD. TCGA database was used to construct signature, and six cohorts from GEO database were used for validation. We also investigated the association between this signature and immunotherapy response. Based on 258 B cell marker genes identified by scRNA-seq analysis, a nine-gene signature was constructed for prognostic prediction in TCGA dataset, which classified patients into high-risk and low-risk groups according to overall survival. The multivariate analysis demonstrated that the signature was an independent prognostic factor. The signature's predictive power was verified in other six independent cohorts and different clinical subgroups. Analysis of immune profiles showed that high-risk groups presented discriminative immune-cell infiltrations and immune-suppressive states. More importantly, risk scores of the signature were closely correlated with PD-L1, tumor mutation burden, neoantigens, and tumor immune dysfunction and exclusion score. Our study proposed a novel prognostic signature based on B cell marker genes for LUAD patients. The signature could effectively indicate LUAD patients' survival and serve as a predictor for immunotherapy.
Collapse
|
14
|
Chou C, Chin MT. Pathogenic Mechanisms of Hypertrophic Cardiomyopathy beyond Sarcomere Dysfunction. Int J Mol Sci 2021; 22:ijms22168933. [PMID: 34445638 PMCID: PMC8396307 DOI: 10.3390/ijms22168933] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/16/2021] [Accepted: 08/18/2021] [Indexed: 01/23/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is the most common inherited cardiovascular disorder, affecting 1 in 500 people in the general population. Although characterized by asymmetric left ventricular hypertrophy, cardiomyocyte disarray, and cardiac fibrosis, HCM is in fact a highly complex disease with heterogenous clinical presentation, onset, and complications. While HCM is generally accepted as a disease of the sarcomere, variable penetrance in families with identical genetic mutations challenges the monogenic origin of HCM and instead implies a multifactorial cause. Furthermore, large-scale genome sequencing studies revealed that many genes previously reported as causative of HCM in fact have little or no evidence of disease association. These findings thus call for a re-evaluation of the sarcomere-centered view of HCM pathogenesis. Here, we summarize our current understanding of sarcomere-independent mechanisms of cardiomyocyte hypertrophy, highlight the role of extracellular signals in cardiac fibrosis, and propose an alternative but integrated model of HCM pathogenesis.
Collapse
Affiliation(s)
- Chun Chou
- Department of Medicine, Tufts University School of Medicine, Boston, MA 02111, USA;
| | - Michael T. Chin
- Department of Medicine, Tufts University School of Medicine, Boston, MA 02111, USA;
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA
- Correspondence: ; Tel.: +1-617-636-8776
| |
Collapse
|
15
|
Phan HAT, Giannakoulias SG, Barrett TM, Liu C, Petersson EJ. Rational design of thioamide peptides as selective inhibitors of cysteine protease cathepsin L. Chem Sci 2021; 12:10825-10835. [PMID: 35355937 PMCID: PMC8901119 DOI: 10.1039/d1sc00785h] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 06/30/2021] [Indexed: 12/24/2022] Open
Abstract
Aberrant levels of cathepsin L (Cts L), a ubiquitously expressed endosomal cysteine protease, have been implicated in many diseases such as cancer and diabetes. Significantly, Cts L has been identified as a potential target for the treatment of COVID-19 due to its recently unveiled critical role in SARS-CoV-2 entry into the host cells. However, there are currently no clinically approved specific inhibitors of Cts L, as it is often challenging to obtain specificity against the many highly homologous cathepsin family cysteine proteases. Peptide-based agents are often promising protease inhibitors as they offer high selectivity and potency, but unfortunately are subject to degradation in vivo. Thioamide substitution, a single-atom O-to-S modification in the peptide backbone, has been shown to improve the proteolytic stability of peptides addressing this issue. Utilizing this approach, we demonstrate herein that good peptidyl substrates can be converted into sub-micromolar inhibitors of Cts L by a single thioamide substitution in the peptide backbone. We have designed and scanned several thioamide stabilized peptide scaffolds, in which one peptide, RS 1A, was stabilized against proteolysis by all five cathepsins (Cts L, Cts V, Cts K, Cts S, and Cts B) while inhibiting Cts L with >25-fold specificity against the other cathepsins. We further showed that this stabilized RS 1A peptide could inhibit Cts L in human liver carcinoma lysates (IC50 = 19 μM). Our study demonstrates that one can rationally design a stabilized, specific peptidyl protease inhibitor by strategic placement of a thioamide and reaffirms the place of this single-atom modification in the toolbox of peptide-based rational drug design.
Collapse
Affiliation(s)
- Hoang Anh T Phan
- Department of Chemistry, University of Pennsylvania Philadelphia Pennsylvania 19104 USA
| | - Sam G Giannakoulias
- Department of Chemistry, University of Pennsylvania Philadelphia Pennsylvania 19104 USA
| | - Taylor M Barrett
- Department of Chemistry, University of Pennsylvania Philadelphia Pennsylvania 19104 USA
| | - Chunxiao Liu
- Department of Chemistry, University of Pennsylvania Philadelphia Pennsylvania 19104 USA
- Key Laboratory for Northern Urban Agriculture of Ministry of Agriculture and Rural Affairs, Beijing University of Agriculture Beijing 102206 P. R. China
| | - E James Petersson
- Department of Chemistry, University of Pennsylvania Philadelphia Pennsylvania 19104 USA
| |
Collapse
|
16
|
Dai J, Zhang Q, Wan C, Liu J, Zhang Q, Yu Y, Wang J. Significances of viable synergistic autophagy-associated cathepsin B and cathepsin D (CTSB/CTSD) as potential biomarkers for sudden cardiac death. BMC Cardiovasc Disord 2021; 21:233. [PMID: 33964876 PMCID: PMC8106142 DOI: 10.1186/s12872-021-02040-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 04/26/2021] [Indexed: 01/06/2023] Open
Abstract
Background The Cathepsins family, including cathepsin B and cathepsin D, potentially affects the entire processes involved in atherosclerosis. Although coronary heart disease (CHD) has been widely studied as the basis of Sudden Cardiac Death (SCD), the relationship between CHD and CTSB/D remains unclear. Methods We screened for differentially expressed proteins (DEPs) associated with autophagy by limma package in R. For the genes corresponding to the DEPs after screening, we used various databases to carry out functional enrichment of related DEGs to explore their possible influence on a specific aspect of the disease. Functional enrichment analysis of DEGs was performed by DAVID, Metascape and GSEA. STRING and Cytoscape were obtained the hub genes, the analysis of interaction networks through the GENMANIA and Networkanalyst. Western Blot was used to validate the protein expression level of target genes. TF and miRNA prediction were performed using Networkanalyst and visualized using Cytoscape. Results The expression levels of members of the cathepsin family were up regulated in CHD tissues compared with the control. GO and KEGG revealed that cathepsin was markedly enriched in endopeptidase activities, immune responses, lysosome pathways, et al. The correlation analysis showed that in patients with CHD, the CTSB/CTSD expression were negatively correlated with ATG4D and BNIP3, but positively with BCL2L1, CAPNS1, and TP53. In the TF-mRNA-miRNA network, has-miR-24-3p and has-miR-128-3p had higher degrees, CTSB/CTSD could be targeted by them. Conclusions Our findings elucidated the expression and regulatory role of cathepsins in coronary heart disease induced SCD and might further explore the potential mechanisms of autophagy in CHD.
Collapse
Affiliation(s)
- Jialin Dai
- School of Forensic Medicine, Guizhou Medical University, 4 Beijing Road, Guiyang, 550001, Guizhou, China
| | - Qiong Zhang
- School of Forensic Medicine, Guizhou Medical University, 4 Beijing Road, Guiyang, 550001, Guizhou, China
| | - Changwu Wan
- School of Forensic Medicine, Guizhou Medical University, 4 Beijing Road, Guiyang, 550001, Guizhou, China
| | - Jiangjin Liu
- School of Forensic Medicine, Guizhou Medical University, 4 Beijing Road, Guiyang, 550001, Guizhou, China
| | - Qiaojun Zhang
- School of Forensic Medicine, Guizhou Medical University, 4 Beijing Road, Guiyang, 550001, Guizhou, China
| | - Yanni Yu
- School of Forensic Medicine, Guizhou Medical University, 4 Beijing Road, Guiyang, 550001, Guizhou, China.
| | - Jie Wang
- School of Forensic Medicine, Guizhou Medical University, 4 Beijing Road, Guiyang, 550001, Guizhou, China.
| |
Collapse
|
17
|
Wang X, Li X, Lin F, Sun H, Lin Y, Wang Z, Wang X. The lnc-CTSLP8 upregulates CTSL1 as a competitive endogenous RNA and promotes ovarian cancer metastasis. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:151. [PMID: 33933142 PMCID: PMC8088648 DOI: 10.1186/s13046-021-01957-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 04/20/2021] [Indexed: 01/05/2023]
Abstract
Background Ovarian cancer is highly lethal and has a poor prognosis due to metastasis. Long non-coding RNAs (lncRNAs) are key regulators of tumor development, but their role in ovarian cancer metastasis remains unclear. Methods The expression of lnc-CTSLP8 in ovarian cancer was analyzed in public databases (TCGA and GEO) and validated via qRT-PCR. Lnc-CTSLP8 overexpression and knockout cell lines were constructed using a lentiviral vector and the CRISP/Cas9 system. Cell proliferation, colony formation, migration, and invasion were analyzed. An ovarian orthotopic tumor mouse model was used for the in vivo study. Changes in autophagosomes, autolysosomes, and mitochondria in ovarian cancer cells were observed via transmission electron microscopy. EMT markers were detected by immunoblotting and immunofluorescence assays. RNA immunoprecipitation, RNA pull-down, and dual luciferase reporter assays were performed to confirm the interaction between lnc-CTSLP8 and miR-199a-5p. Results A novel pseudogene, lnc-CTSLP8, was identified in ovarian cancer, with significantly elevated expression in metastatic tumor tissues compared to primary ovarian tumors. When overexpressed, lnc-CTSLP8 promoted ovarian cancer in vitro and in vivo by acting as a sponge for miR-199a-5p. Autophagy and EMT in ovarian cancer were also enhanced by lnc-CTSLP8. Mechanistically, lnc-CTSLP8 upregulated CTSL1 as a competitive endogenous RNA and exhibited oncogenic effects. Moreover, CTSL1 inhibitor treatment and miR-199a-5p overexpression abrogated the effects of lnc-CTSLP8 overexpression. Conclusions lnc-CTSLP8 acts as a ceRNA in ovarian cancer and represents a potential therapeutic target for metastatic ovarian cancer. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-021-01957-z.
Collapse
Affiliation(s)
- Xinjing Wang
- Department of Gynecology and Obstetrics, XinHua Hospital, Shanghai JiaoTong University School of Medicine, 1665 Kongjiang Rd, Yangpu District, Shanghai, 200092, China
| | - Xiaoduan Li
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Feikai Lin
- Department of Gynecology and Obstetrics, XinHua Hospital, Shanghai JiaoTong University School of Medicine, 1665 Kongjiang Rd, Yangpu District, Shanghai, 200092, China
| | - Huizhen Sun
- Department of Gynecology and Obstetrics, XinHua Hospital, Shanghai JiaoTong University School of Medicine, 1665 Kongjiang Rd, Yangpu District, Shanghai, 200092, China
| | - Yingying Lin
- Department of Neurosurgery, Renji Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Ziliang Wang
- Department of Gynecology and Obstetrics, XinHua Hospital, Shanghai JiaoTong University School of Medicine, 1665 Kongjiang Rd, Yangpu District, Shanghai, 200092, China.
| | - Xipeng Wang
- Department of Gynecology and Obstetrics, XinHua Hospital, Shanghai JiaoTong University School of Medicine, 1665 Kongjiang Rd, Yangpu District, Shanghai, 200092, China.
| |
Collapse
|
18
|
Quinacrine-Induced Autophagy in Ovarian Cancer Triggers Cathepsin-L Mediated Lysosomal/Mitochondrial Membrane Permeabilization and Cell Death. Cancers (Basel) 2021; 13:cancers13092004. [PMID: 33919392 PMCID: PMC8122252 DOI: 10.3390/cancers13092004] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/01/2021] [Accepted: 04/17/2021] [Indexed: 01/08/2023] Open
Abstract
Simple Summary Ovarian cancer (OC) is the most common cause of cancer-related deaths among women worldwide, and its incidence has been increasing and has continued to prove resistant to a variety of therapeutics. This observation is principally disturbing given the amount of money invested in identifying novel therapies for this disease. A comparatively rapid and economical pipeline for identification of novel drugs is drug repurposing. We reported earlier that the antimalarial drug Quinacrine (QC) also has anticancer activity and here we discovered that QC significantly upregulates cathepsin L (CTSL) and promoting autophagic flux in ovarian cancer. QC-induced CTSL activation promotes lysosomal membrane permeability resulting in active CTSL release into the cytosol, which promotes Bid cleavage, mitochondrial membrane permeability, cytochrome-c release and cell death in both in-vitro and in-vivo models. Therefore, QC is a promising candidate for OC treatment. Abstract We previously reported that the antimalarial compound quinacrine (QC) induces autophagy in ovarian cancer cells. In the current study, we uncovered that QC significantly upregulates cathepsin L (CTSL) but not cathepsin B and D levels, implicating the specific role of CTSL in promoting QC-induced autophagic flux and apoptotic cell death in OC cells. Using a Magic Red® cathepsin L activity assay and LysoTracker red, we discerned that QC-induced CTSL activation promotes lysosomal membrane permeability (LMP) resulting in the release of active CTSL into the cytosol to promote apoptotic cell death. We found that QC-induced LMP and CTSL activation promotes Bid cleavage, mitochondrial outer membrane permeabilization (MOMP), and mitochondrial cytochrome-c release. Genetic (shRNA) and pharmacological (Z-FY(tBU)-DMK) inhibition of CTSL markedly reduces QC-induced autophagy, LMP, MOMP, apoptosis, and cell death; whereas induced overexpression of CTSL in ovarian cancer cell lines has an opposite effect. Using recombinant CTSL, we identified p62/SQSTM1 as a novel substrate of CTSL, suggesting that CTSL promotes QC-induced autophagic flux. CTSL activation is specific to QC-induced autophagy since no CTSL activation is seen in ATG5 knockout cells or with the anti-malarial autophagy-inhibiting drug chloroquine. Importantly, we showed that upregulation of CTSL in QC-treated HeyA8MDR xenografts corresponds with attenuation of p62, upregulation of LC3BII, cytochrome-c, tBid, cleaved PARP, and caspase3. Taken together, the data suggest that QC-induced autophagy and CTSL upregulation promote a positive feedback loop leading to excessive autophagic flux, LMP, and MOMP to promote QC-induced cell death in ovarian cancer cells.
Collapse
|
19
|
O'Toole D, Zaeri AAI, Nicklin SA, French AT, Loughrey CM, Martin TP. Signalling pathways linking cysteine cathepsins to adverse cardiac remodelling. Cell Signal 2020; 76:109770. [PMID: 32891693 DOI: 10.1016/j.cellsig.2020.109770] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/27/2020] [Accepted: 08/31/2020] [Indexed: 12/12/2022]
Abstract
Adverse cardiac remodelling clinically manifests as deleterious changes to heart architecture (size, mass and geometry) and function. These changes, which include alterations to ventricular wall thickness, chamber dilation and poor contractility, are important because they progressively drive patients with cardiac disease towards heart failure and are associated with poor prognosis. Cysteine cathepsins contribute to key signalling pathways involved in adverse cardiac remodelling including synthesis and degradation of the cardiac extracellular matrix (ECM), cardiomyocyte hypertrophy, impaired cardiomyocyte contractility and apoptosis. In this review, we highlight the role of cathepsins in these signalling pathways as well as their translational potential as therapeutic targets in cardiac disease.
Collapse
Affiliation(s)
- Dylan O'Toole
- British Heart Foundation Glasgow Cardiovascular Research Centre, Institute of Cardiovascular & Medical Sciences, University of Glasgow, UK
| | - Ali Abdullah I Zaeri
- British Heart Foundation Glasgow Cardiovascular Research Centre, Institute of Cardiovascular & Medical Sciences, University of Glasgow, UK
| | - Stuart A Nicklin
- British Heart Foundation Glasgow Cardiovascular Research Centre, Institute of Cardiovascular & Medical Sciences, University of Glasgow, UK
| | - Anne T French
- Clinical Sciences Department, Ross University School of Veterinary Medicine, Basseterre, St. Kitts, West Indies, Saint Kitts and Nevis
| | - Christopher M Loughrey
- British Heart Foundation Glasgow Cardiovascular Research Centre, Institute of Cardiovascular & Medical Sciences, University of Glasgow, UK.
| | - Tamara P Martin
- British Heart Foundation Glasgow Cardiovascular Research Centre, Institute of Cardiovascular & Medical Sciences, University of Glasgow, UK.
| |
Collapse
|
20
|
Xu T, Nicolson S, Sandow JJ, Dayan S, Jiang X, Manning JA, Webb AI, Kumar S, Denton D. Cp1/cathepsin L is required for autolysosomal clearance in Drosophila. Autophagy 2020; 17:2734-2749. [PMID: 33112206 DOI: 10.1080/15548627.2020.1838105] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Macroautophagy/autophagy is a highly conserved lysosomal degradative pathway important for maintaining cellular homeostasis. Much of our current knowledge of autophagy is focused on the initiation steps in this process. Recently, an understanding of later steps, particularly lysosomal fusion leading to autolysosome formation and the subsequent role of lysosomal enzymes in degradation and recycling, is becoming evident. Autophagy can function in both cell survival and cell death, however, the mechanisms that distinguish adaptive/survival autophagy from autophagy-dependent cell death remain to be established. Here, using proteomic analysis of Drosophila larval midguts during degradation, we identify a group of proteins with peptidase activity, suggesting a role in autophagy-dependent cell death. We show that Cp1/cathepsin L-deficient larval midgut cells accumulate aberrant autophagic vesicles due to a block in autophagic flux, yet later stages of midgut degradation are not compromised. The accumulation of large aberrant autolysosomes in the absence of Cp1 appears to be the consequence of decreased degradative capacity as they contain undigested cytoplasmic material, rather than a defect in autophagosome-lysosome fusion. Finally, we find that other cathepsins may also contribute to proper autolysosomal degradation in Drosophila larval midgut cells. Our findings provide evidence that cathepsins play an essential role in the autolysosome to maintain basal autophagy flux by balancing autophagosome production and turnover.
Collapse
Affiliation(s)
- Tianqi Xu
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia
| | - Shannon Nicolson
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia
| | - Jarrod J Sandow
- Advanced Technology and Biology, The Walter & Eliza Hall Institute of Medical Research, Parkville, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Sonia Dayan
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia
| | - Xin Jiang
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia
| | - Jantina A Manning
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia
| | - Andrew I Webb
- Advanced Technology and Biology, The Walter & Eliza Hall Institute of Medical Research, Parkville, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Sharad Kumar
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia
| | - Donna Denton
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia
| |
Collapse
|
21
|
Yadati T, Houben T, Bitorina A, Shiri-Sverdlov R. The Ins and Outs of Cathepsins: Physiological Function and Role in Disease Management. Cells 2020; 9:cells9071679. [PMID: 32668602 PMCID: PMC7407943 DOI: 10.3390/cells9071679] [Citation(s) in RCA: 264] [Impact Index Per Article: 52.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/10/2020] [Accepted: 07/11/2020] [Indexed: 12/14/2022] Open
Abstract
Cathepsins are the most abundant lysosomal proteases that are mainly found in acidic endo/lysosomal compartments where they play a vital role in intracellular protein degradation, energy metabolism, and immune responses among a host of other functions. The discovery that cathepsins are secreted and remain functionally active outside of the lysosome has caused a paradigm shift. Contemporary research has unraveled many versatile functions of cathepsins in extralysosomal locations including cytosol and extracellular space. Nevertheless, extracellular cathepsins are majorly upregulated in pathological states and are implicated in a wide range of diseases including cancer and cardiovascular diseases. Taking advantage of the differential expression of the cathepsins during pathological conditions, much research is focused on using cathepsins as diagnostic markers and therapeutic targets. A tailored therapeutic approach using selective cathepsin inhibitors is constantly emerging to be safe and efficient. Moreover, recent development of proteomic-based approaches for the identification of novel physiological substrates offers a major opportunity to understand the mechanism of cathepsin action. In this review, we summarize the available evidence regarding the role of cathepsins in health and disease, discuss their potential as biomarkers of disease progression, and shed light on the potential of extracellular cathepsin inhibitors as safe therapeutic tools.
Collapse
|
22
|
Hu B, Zhu X, Lu J. Cathepsin A knockdown decreases the proliferation and invasion of A549 lung adenocarcinoma cells. Mol Med Rep 2020; 21:2553-2559. [PMID: 32323791 PMCID: PMC7185279 DOI: 10.3892/mmr.2020.11068] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 03/13/2020] [Indexed: 02/07/2023] Open
Abstract
Cathepsin A (CTSA) is a lysosomal protease that is abnormally expressed in various types of cancer; however, the function of CTSA in lung adenocarcinoma (LUAD) is unknown. The aim of the present study was to investigate the role of CTSA during LUAD development in vitro. The Cancer Genome Atlas (TCGA) database was used to analyze the expression of CTSA mRNA in LUAD tissues. CTSA was significantly upregulated in LUAD tissues compared with normal lung tissues. To explore the effect of CTSA on LUAD in vitro, LUAD A549 cells were transfected with CTSA small interfering RNA and the hallmarks of tumorigenesis were investigated using cell proliferation, cell cycle, wound healing, invasion and western blot assays. Following CTSA knockdown, proliferation of LUAD cells was decreased and an increased proportion of LUAD cells were arrested at the G0/G1 phase, with altered expression of critical cell cycle and proliferative marker proteins, including p53, p21 and proliferating cell nuclear antigen. Moreover, CTSA knockdown decreased the migration and invasion of A549 cells, as determined by wound healing, invasion, and western blotting assays. The expression levels of key proteins involved in epithelial-mesenchymal transition were analyzed by western blotting. CTSA knockdown enhanced the expression of E-cadherin, but decreased the expression of N-cadherin and β-catenin in A549 cells. To the best of our knowledge, the present study suggested for the first time it has been identified that CTSA may serve as a tumor promoter in LUAD, enhancing the malignant progression of LUAD cells by promoting cell proliferation, migration and invasion. The results suggested that CTSA may serve as a novel therapeutic target for LUAD.
Collapse
Affiliation(s)
- Bo Hu
- Department of Thoracic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Xike Zhu
- Department of Research Center, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Jibin Lu
- Department of Thoracic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| |
Collapse
|
23
|
Toxoplasma Cathepsin Protease B and Aspartyl Protease 1 Are Dispensable for Endolysosomal Protein Digestion. mSphere 2020; 5:5/1/e00869-19. [PMID: 32051238 PMCID: PMC7021471 DOI: 10.1128/msphere.00869-19] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Roughly one-third of the human population is chronically infected with the intracellular single-celled parasite Toxoplasma gondii, but little is known about how this organism persists inside people. Previous research suggested that a parasite proteolytic enzyme, termed cathepsin protease L, is important for Toxoplasma persistence; however, it remained possible that other associated proteolytic enzymes could also be involved in the long-term survival of the parasite during infection. Here, we show that two proteolytic enzymes associated with cathepsin protease L play dispensable roles and are dependent on cathepsin L to reach maturity, which differs from the corresponding enzymes in humans. These findings establish a divergent hierarchy of proteases and help focus attention principally on cathepsin protease L as a potential target for interrupting Toxoplasma chronic infection. The lysosome-like vacuolar compartment (VAC) is a major site of proteolysis in the intracellular parasite Toxoplasma gondii. Previous studies have shown that genetic ablation of a VAC-residing cysteine protease, cathepsin protease L (CPL), resulted in the accumulation of undigested protein in the VAC and loss of parasite viability during the chronic stage of infection. However, since the maturation of another VAC localizing protease, cathepsin protease B (CPB), is dependent on CPL, it remained unknown whether these defects result directly from ablation of CPL or indirectly from a lack of CPB maturation. Likewise, although a previously described cathepsin D-like aspartyl protease 1 (ASP1) could also play a role in proteolysis, its definitive residence and function in the Toxoplasma endolysosomal system were not well defined. Here, we demonstrate that CPB is not necessary for protein turnover in the VAC and that CPB-deficient parasites have normal growth and viability in both the acute and chronic stages of infection. We also show that ASP1 depends on CPL for correct maturation, and it resides in the T. gondii VAC, where, similar to CPB, it plays a dispensable role in protein digestion. Taken together with previous work, our findings suggest that CPL is the dominant protease in a hierarchy of proteolytic enzymes within the VAC. This unusual lack of redundancy for CPL in T. gondii makes it a single exploitable target for disrupting chronic toxoplasmosis. IMPORTANCE Roughly one-third of the human population is chronically infected with the intracellular single-celled parasite Toxoplasma gondii, but little is known about how this organism persists inside people. Previous research suggested that a parasite proteolytic enzyme, termed cathepsin protease L, is important for Toxoplasma persistence; however, it remained possible that other associated proteolytic enzymes could also be involved in the long-term survival of the parasite during infection. Here, we show that two proteolytic enzymes associated with cathepsin protease L play dispensable roles and are dependent on cathepsin L to reach maturity, which differs from the corresponding enzymes in humans. These findings establish a divergent hierarchy of proteases and help focus attention principally on cathepsin protease L as a potential target for interrupting Toxoplasma chronic infection.
Collapse
|
24
|
Sproles AE, Oakley CA, Matthews JL, Peng L, Owen JG, Grossman AR, Weis VM, Davy SK. Proteomics quantifies protein expression changes in a model cnidarian colonised by a thermally tolerant but suboptimal symbiont. THE ISME JOURNAL 2019; 13:2334-2345. [PMID: 31118473 PMCID: PMC6775970 DOI: 10.1038/s41396-019-0437-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 04/23/2019] [Accepted: 05/03/2019] [Indexed: 11/09/2022]
Abstract
The acquisition of thermally tolerant algal symbionts by corals has been proposed as a natural or assisted mechanism of increasing coral reef resilience to anthropogenic climate change, but the cell-level processes determining the performance of new symbiotic associations are poorly understood. We used liquid chromatography-mass spectrometry to investigate the effects of an experimentally induced symbiosis on the host proteome of the model sea anemone Exaiptasia pallida. Aposymbiotic specimens were colonised by either the homologous dinoflagellate symbiont (Breviolum minutum) or a thermally tolerant, ecologically invasive heterologous symbiont (Durusdinium trenchii). Anemones containing D. trenchii exhibited minimal expression of Niemann-Pick C2 proteins, which have predicted biochemical roles in sterol transport and cell recognition, and glutamine synthetases, which are thought to be involved in nitrogen assimilation and recycling between partners. D. trenchii-colonised anemones had higher expression of methionine-synthesising betaine-homocysteine S-methyltransferases and proteins with predicted oxidative stress response functions. Multiple lysosome-associated proteins were less abundant in both symbiotic treatments compared with the aposymbiotic treatment. The differentially abundant proteins are predicted to represent pathways that may be involved in nutrient transport or resource allocation between partners. These results provide targets for specific experiments to elucidate the mechanisms underpinning compensatory physiology in the coral-dinoflagellate symbiosis.
Collapse
Affiliation(s)
- Ashley E Sproles
- School of Biological Sciences, Victoria University of Wellington, Wellington, 6140, New Zealand
| | - Clinton A Oakley
- School of Biological Sciences, Victoria University of Wellington, Wellington, 6140, New Zealand
| | - Jennifer L Matthews
- School of Biological Sciences, Victoria University of Wellington, Wellington, 6140, New Zealand
| | - Lifeng Peng
- School of Biological Sciences, Victoria University of Wellington, Wellington, 6140, New Zealand
| | - Jeremy G Owen
- School of Biological Sciences, Victoria University of Wellington, Wellington, 6140, New Zealand
| | - Arthur R Grossman
- Department of Plant Biology, The Carnegie Institution for Science, Stanford, CA, 94305, USA
| | - Virginia M Weis
- Department of Integrative Biology, Oregon State University, Corvallis, OR, 97331, USA
| | - Simon K Davy
- School of Biological Sciences, Victoria University of Wellington, Wellington, 6140, New Zealand.
| |
Collapse
|
25
|
Weiss-Sadan T, Itzhak G, Kaschani F, Yu Z, Mahameed M, Anaki A, Ben-Nun Y, Merquiol E, Tirosh B, Kessler B, Kaiser M, Blum G. Cathepsin L Regulates Metabolic Networks Controlling Rapid Cell Growth and Proliferation. Mol Cell Proteomics 2019; 18:1330-1344. [PMID: 31010818 PMCID: PMC6601214 DOI: 10.1074/mcp.ra119.001392] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 04/07/2019] [Indexed: 01/07/2023] Open
Abstract
Rapidly proliferating cells reshape their metabolism to satisfy their ever-lasting need for cellular building blocks. This phenomenon is exemplified in certain malignant conditions such as cancer but also during embryonic development when cells rely heavily on glycolytic metabolism to exploit its metabolic intermediates for biosynthetic processes. How cells reshape their metabolism is not fully understood. Here we report that loss of cathepsin L (Cts L) is associated with a fast proliferation rate and enhanced glycolytic metabolism that depend on lactate dehydrogenase A (LDHA) activity. Using mass spectrometry analysis of cells treated with a pan cathepsin inhibitor, we observed an increased abundance of proteins involved in central carbon metabolism. Further inspection of putative Cts L targets revealed an enrichment for glycolytic metabolism that was independently confirmed by metabolomic and biochemical analyses. Moreover, proteomic analysis of Cts L-knockout cells identified LDHA overexpression that was demonstrated to be a key metabolic junction in these cells. Lastly, we show that Cts L inhibition led to increased LDHA protein expression, suggesting a causal relationship between LDHA expression and function. In conclusion, we propose that Cts L regulates this metabolic circuit to keep cell division under control, suggesting the therapeutic potential of targeting this protein and its networks in cancer.
Collapse
Affiliation(s)
- Tommy Weiss-Sadan
- From the ‡Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University, Jerusalem, Israel, 9112001
| | - Gal Itzhak
- From the ‡Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University, Jerusalem, Israel, 9112001
| | - Farnusch Kaschani
- §Department of Chemical Biology, University of Duisburg-Essen, Center for Medical Biotechnology, Faculty of Biology, Essen, Germany
| | - Zhanru Yu
- ¶Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Mohamed Mahameed
- From the ‡Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University, Jerusalem, Israel, 9112001
| | - Adi Anaki
- From the ‡Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University, Jerusalem, Israel, 9112001
| | - Yael Ben-Nun
- From the ‡Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University, Jerusalem, Israel, 9112001
| | - Emmanuelle Merquiol
- From the ‡Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University, Jerusalem, Israel, 9112001
| | - Boaz Tirosh
- From the ‡Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University, Jerusalem, Israel, 9112001
| | - Benedikt Kessler
- ¶Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Markus Kaiser
- §Department of Chemical Biology, University of Duisburg-Essen, Center for Medical Biotechnology, Faculty of Biology, Essen, Germany
| | - Galia Blum
- From the ‡Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University, Jerusalem, Israel, 9112001;.
| |
Collapse
|
26
|
Identification and characterization of the lamprey cathepsin genes. Immunogenetics 2019; 71:421-432. [PMID: 31089760 DOI: 10.1007/s00251-019-01117-w] [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: 02/02/2019] [Accepted: 04/10/2019] [Indexed: 10/26/2022]
Abstract
Cathepsins are key mammalian proteases that play an important role in the immune response. Several studies have revealed the versatile and critical functions of cathepsins. Here, we obtained ten kinds of cathepsin homologs and identified seven homologs with complete coding sequences. Phylogenetic analysis verified their identities and supported the classification of cathepsins into seven families, which is similar to other vertebrates. Tissue-specific expression analysis showed that all lamprey cathepsins (L-cathepsins) are present in the supraneural body (SB), kidney, gill, intestine, brain, heart, and liver, but their relative abundance varied among tissues. Additionally, we focused on the lamprey cathepsin L (L-cathepsin L) and used recombinant L-cathepsin L protein (rL-cathepsin L) to prepare anti rL-cathepsin L polyclonal antibodies, which were used to detect its distribution in lamprey tissues. The L-cathepsin L protein was primarily detected in the SB, kidney, gill, intestine, brain, and liver via western blot and immunohistochemistry assays. Importantly, quantitative real-time PCR (RT-PCR) revealed that the expression level of L-cathepsins mRNA significantly increased after exposure to three different stimuli (poly I:C, Staphylococcus aureus (S.a) and Vibro anguilarum (V.an)). This suggested that L-cathepsins may participate in defense processes. These results revealed that L-cathepsins may play key roles in the immune response to exogenous stimuli. The findings provide important information for future studies aiming to understand the molecular mechanisms underlying the immune response to pathogen invasion in lamprey.
Collapse
|
27
|
Ost GS, Ng'ang'a PN, Lang AE, Aktories K. Photorhabdus luminescens
Tc toxin is inhibited by the protease inhibitor MG132 and activated by protease cleavage resulting in increased binding to target cells. Cell Microbiol 2018; 21:e12978. [DOI: 10.1111/cmi.12978] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 10/19/2018] [Accepted: 11/04/2018] [Indexed: 12/24/2022]
Affiliation(s)
- Gerhard Stefan Ost
- Institute for Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine; University of Freiburg; Freiburg Germany
- Faculty of Biology; University of Freiburg; Freiburg Germany
| | - Peter Njenga Ng'ang'a
- Institute for Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine; University of Freiburg; Freiburg Germany
- Faculty of Biology; University of Freiburg; Freiburg Germany
- Spemann Graduate School of Biology and Medicine (SGBM); University of Freiburg; Freiburg Germany
| | - Alexander E. Lang
- Institute for Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine; University of Freiburg; Freiburg Germany
| | - Klaus Aktories
- Institute for Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine; University of Freiburg; Freiburg Germany
- Centre for Biological Signalling Studies (BIOSS); University of Freiburg; Freiburg Germany
| |
Collapse
|
28
|
Sharif U, Mahmud NM, Kay P, Yang YC, Harding SP, Grierson I, Kamalden TA, Jackson MJ, Paraoan L. Advanced glycation end products-related modulation of cathepsin L and NF-κB signalling effectors in retinal pigment epithelium lead to augmented response to TNFα. J Cell Mol Med 2018; 23:405-416. [PMID: 30338926 PMCID: PMC6307775 DOI: 10.1111/jcmm.13944] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 09/07/2018] [Accepted: 09/10/2018] [Indexed: 01/02/2023] Open
Abstract
The retinal pigment epithelium (RPE) plays a central role in neuroretinal homoeostasis throughout life. Altered proteolysis and inflammatory processes involving RPE contribute to the pathophysiology of age‐related macular degeneration (AMD), but the link between these remains elusive. We report for the first time the effect of advanced glycation end products (AGE)—known to accumulate on the ageing RPE's underlying Bruch's membrane in situ—on both key lysosomal cathepsins and NF‐κB signalling in RPE. Cathepsin L activity and NF‐κB effector levels decreased significantly following 2‐week AGE exposure. Chemical cathepsin L inhibition also decreased total p65 protein levels, indicating that AGE‐related change of NF‐κB effectors in RPE cells may be modulated by cathepsin L. However, upon TNFα stimulation, AGE‐exposed cells had significantly higher ratio of phospho‐p65(Ser536)/total p65 compared to non‐AGEd controls, with an even higher fold increase than in the presence of cathepsin L inhibition alone. Increased proportion of active p65 indicates an AGE‐related activation of NF‐κB signalling in a higher proportion of cells and/or an enhanced response to TNFα. Thus, NF‐κB signalling modulation in the AGEd environment, partially regulated via cathepsin L, is employed by RPE cells as a protective (para‐inflammatory) mechanism but renders them more responsive to pro‐inflammatory stimuli.
Collapse
Affiliation(s)
- Umar Sharif
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | - Nur Musfirah Mahmud
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK.,Eye Research Centre, University of Malaya, Kuala Lumpur, Malaysia
| | - Paul Kay
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | - Yit C Yang
- Ophthalmology, The Royal Wolverhampton NHS Trust, Wolverhampton, UK
| | - Simon P Harding
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | - Ian Grierson
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | | | - Malcolm J Jackson
- Department of Musculoskeletal Science, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | - Luminita Paraoan
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| |
Collapse
|
29
|
Tan J, Qian X, Song B, An X, Cai T, Zuo Z, Ding D, Lu Y, Li H. Integrated bioinformatics analysis reveals that the expression of cathepsin S is associated with lymph node metastasis and poor prognosis in papillary thyroid cancer. Oncol Rep 2018; 40:111-122. [PMID: 29749483 PMCID: PMC6059735 DOI: 10.3892/or.2018.6428] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 04/26/2018] [Indexed: 12/20/2022] Open
Abstract
The prognosis of the majority of patients with papillary thyroid cancer (PTC) is excellent, although there are patients who experience disease recurrence and progression. The aim of the present study was to identify potential prognostic risk markers in PTC. Differentially expressed genes (DEGs), identified from four Genome Expression Omnibus cohorts were subjected to functional enrichment analyses with Gene Ontology terms and the Kyoto Encyclopedia of Genes and Genome pathways. Hub genes, filtered from cytoHubba, were validated using the The Cancer Genome Atlas (TCGA) cohort, and their associations with clinicopathological features and prognosis were analyzed. A total of 277 DEGs were identified following data preprocessing. DEGs were primarily enriched in 'small cell lung cancer', 'ECM-receptor interaction', 'pathways in cancer'and 'tyrosine metabolism'. Hub genes [APOE, cathepsin S (CTSS), insulin receptor substrate 1 (IRS1), KIT, LGALS3, RUNX2 and TGFBR1] were extracted from cytoHubba. Their expression in the TCGA cohort was consistent with that in the GEO cohorts. CTSS (P=0.006) and IRS1 (P=0.005) were associated with disease‑free survival, as determined using the Kaplan-Meier analysis. CTSS was an independent risk factor for poor disease‑free survival (HR, 2.649; 95% CI, 1.095-6.409; P=0.031). Patients with high expression of CTSS exhibited different histological types (increased tall-cell subtype and reduced follicular subtype; P<0.001), more frequent lymph node metastasis (P<0.001) and advanced tumor-node-metastasis stages (P=0.049) compared with the low-expression group. High expression of CTSS was independently associated with lymph node metastasis (OR, 2.015; 95% CI, 1.225-3.315; P=0.006). Therefore, CTSS may serve as a predictive risk marker for the progression and prognosis of PTC.
Collapse
Affiliation(s)
- Juan Tan
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
- Department of Gerontology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| | - Xiaoxiao Qian
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Bin Song
- Department of Endocrinology, Clinical Medical College of Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Xiumin An
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Tingting Cai
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Zhihua Zuo
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Dafa Ding
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Yibing Lu
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Hong Li
- Medical Examination Center, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| |
Collapse
|
30
|
Zhang X, Xu R, Zhang C, Xu Y, Han M, Huang B, Chen A, Qiu C, Thorsen F, Prestegarden L, Bjerkvig R, Wang J, Li X. Trifluoperazine, a novel autophagy inhibitor, increases radiosensitivity in glioblastoma by impairing homologous recombination. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2017; 36:118. [PMID: 28870216 PMCID: PMC5584019 DOI: 10.1186/s13046-017-0588-z] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Accepted: 08/28/2017] [Indexed: 01/16/2023]
Abstract
BACKGROUND Resistance to adjuvant radiotherapy is a major cause of treatment failure in patients with glioblastoma (GBM). Autophagy inhibitors have been shown to enhance the efficacy of radiotherapy for certain solid tumors. However, current inhibitors do not penetrate the blood-brain-barrier (BBB). Here, we assessed the radiosensitivity effects of the antipsychotic drug trifluoperazine (TFP) on GBM in vitro and in vivo. METHODS U251 and U87 GBM cell lines as well as GBM cells from a primary human biopsy (P3), were used in vitro and in vivo to evaluate the efficacy of TFP treatment. Viability and cytotoxicity was evaluated by CCK-8 and clonogenic formation assays. Molecular studies using immunohistochemistry, western blots, immunofluorescence and qPCR were used to gain mechanistic insight into the biological activity of TFP. Preclinical therapeutic efficacy was evaluated in orthotopic xenograft mouse models. RESULTS IC50 values of U251, U87 and P3 cells treated with TFP were 16, 15 and 15.5 μM, respectively. TFP increased the expression of LC3B-II and p62, indicating a potential disruption of autophagy flux. These results were further substantiated by a decreased Lysotracker Red uptake, indicating impaired acidification of the lysosomes. We show that TFP and radiation had an additive effect when combined. This effect was in part due to impaired TFP-induced homologous recombination. Mechanistically we show that down-regulation of cathepsin L might explain the radiosensitivity effect of TFP. Finally, combining TFP and radiation resulted in a significant antitumor effect in orthotopic GBM xenograft models. CONCLUSIONS This study provides a strong rationale for further clinical studies exploring the combination therapy of TFP and radiation to treat GBM patients.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Jinan, 250012, People's Republic of China
| | - Ran Xu
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Jinan, 250012, People's Republic of China
| | - Chao Zhang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Jinan, 250012, People's Republic of China
| | - Yangyang Xu
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Jinan, 250012, People's Republic of China
| | - Mingzhi Han
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Jinan, 250012, People's Republic of China
| | - Bin Huang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Jinan, 250012, People's Republic of China
| | - Anjing Chen
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Jinan, 250012, People's Republic of China
| | - Chen Qiu
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, 250012, People's Republic of China
| | - Frits Thorsen
- Kristian Gerhard Jebsen Brain Tumour Research Centre, Department of Biomedicine, University of Bergen, 5009, Bergen, Norway.,The Molecular Imaging Center, Department of Biomedicine, University of Bergen, 5009, Bergen, Norway
| | - Lars Prestegarden
- Kristian Gerhard Jebsen Brain Tumour Research Centre, Department of Biomedicine, University of Bergen, 5009, Bergen, Norway.,Department of Dermatology, Haukeland University Hospital, 5009, Bergen, Norway
| | - Rolf Bjerkvig
- Kristian Gerhard Jebsen Brain Tumour Research Centre, Department of Biomedicine, University of Bergen, 5009, Bergen, Norway.,Department of Oncology, Luxembourg Institute of Health, L-1526, Strassen, Luxembourg
| | - Jian Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Jinan, 250012, People's Republic of China. .,Kristian Gerhard Jebsen Brain Tumour Research Centre, Department of Biomedicine, University of Bergen, 5009, Bergen, Norway.
| | - Xingang Li
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Jinan, 250012, People's Republic of China.
| |
Collapse
|
31
|
Nagakannan P, Eftekharpour E. Differential redox sensitivity of cathepsin B and L holds the key to autophagy-apoptosis interplay after Thioredoxin reductase inhibition in nutritionally stressed SH-SY5Y cells. Free Radic Biol Med 2017; 108:819-831. [PMID: 28478025 DOI: 10.1016/j.freeradbiomed.2017.05.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 04/25/2017] [Accepted: 05/02/2017] [Indexed: 01/10/2023]
Abstract
Reactive oxygen species (ROS) are essential for induction of protective autophagy, however unexpected rise in cellular ROS levels overpowers the cellular defense and therefore promotes the programmed apoptotic cell death. We recently reported that inhibition of thioredoxin reductase (TrxR) in starving SH-SY5Y cells interrupted autophagy flux by induction of lysosomal deficiency and promoted apoptosis. (Free Radic Biol Med. 2016: 101:53-70). Here, we aimed to elucidate the underlying mechanisms during autophagy-apoptosis interplay, and focused on regulation of cathepsin B (CTSB) and L (CTSL), the pro-apoptotic and pro-autophagy cathepsins respectively. Inhibition of TrxR by Auranofin, caused lysosomal membrane permeabilization (LMP) that was associated with a significant upregulation of CTSB activity, despite no significant changes in CTSB protein level. Conversely, a significant rise in CTSL protein levels was observed without any apparent change in CTSL activity. Using thiol-trapping techniques to examine the differential sensitivity of cathepsins to oxidative stress, we discovered that Auranofin-mediated oxidative stress interferes with CTSL processing and thereby interrupts its pro-autophagy function. No evidence of CTSB susceptibility to oxidative stress was observed. Our data suggest that cellular fate in these conditions is mediated by two concurrent systems: while oxidative stress prevents the protective autophagy by inhibition of CTSL processing, concomitantly, apoptosis is induced by increasing lysosomal membrane permeability and leakage of CTSB into cytoplasm. Inhibition of CTSB in these conditions inhibited apoptosis and increased cell viability. To our knowledge this is the first report uncovering the impact of redox environment on autophagy-apoptosis interplay in neuronal cells.
Collapse
Affiliation(s)
- Pandian Nagakannan
- Regenerative Medicine Program, Spinal Cord Research Centre, Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB, Canada
| | - Eftekhar Eftekharpour
- Regenerative Medicine Program, Spinal Cord Research Centre, Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
32
|
Weiss-Sadan T, Gotsman I, Blum G. Cysteine proteases in atherosclerosis. FEBS J 2017; 284:1455-1472. [PMID: 28207191 DOI: 10.1111/febs.14043] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 01/04/2017] [Accepted: 02/13/2017] [Indexed: 12/22/2022]
Abstract
Atherosclerosis predisposes patients to cardiovascular diseases, such as myocardial infarction and stroke. Instigation of vascular injury is triggered by retention of lipids and inflammatory cells in the vascular endothelium. Whereas these vascular lesions develop in young adults and are mostly considered harmless, over time persistent inflammatory and remodeling processes will ultimately damage the arterial wall and cause a thrombotic event due to exposure of tissue factors into the lumen. Evidence from human tissues and preclinical animal models has clearly established the role of cathepsin cysteine proteases in the development and progression of vascular lesions. Hence, understanding the function of cathepsins in atherosclerosis is important for developing novel therapeutic strategies and advanced point of care diagnostics. In this review we will describe the roles of cysteine cathepsins in different cellular process that become dysfunctional in atherosclerosis, such as lipid metabolism, inflammation and apoptosis, and how they contribute to arterial remodeling and atherogenesis. Finally, we will explore new horizons in protease molecular imaging, which may potentially become a surrogate marker to identify future cardiovascular events.
Collapse
Affiliation(s)
- Tommy Weiss-Sadan
- The Institute for Drug Research, School of Pharmacy, Faculty of Medicine, Hebrew University, Jerusalem, Israel
| | - Israel Gotsman
- Heart Institute, Hadassah University Hospital, Jerusalem, Israel
| | - Galia Blum
- The Institute for Drug Research, School of Pharmacy, Faculty of Medicine, Hebrew University, Jerusalem, Israel
| |
Collapse
|
33
|
Wang GQ, Tang T, Wang ZS, Liu YY, Wang L, Luo PF, Xia ZF. Overexpression of Hypo-Phosphorylated IκBβ at Ser313 Protects the Heart against Sepsis. PLoS One 2016; 11:e0160860. [PMID: 27508931 PMCID: PMC4979969 DOI: 10.1371/journal.pone.0160860] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 07/26/2016] [Indexed: 12/21/2022] Open
Abstract
IκBβis an inhibitor of nuclear factor kappa B(NF-κB) and participates in the cardiac response to sepsis. However, the role of the hypo-phosphorylated form of IκBβ at Ser313, which can be detected during sepsis, is unknown. Here, we examined the effects of IκBβ with a mutation at Ser313→Ala313 on cardiac damage induced by sepsis. Transgenic (Tg) mice were generated to overexpress IκBβ, in which Ser-313 is replaced with alanine ubiquitously, in order to mimic the hypo-phosphorylated form of IκBβ. Survival analysis showed that Tg mice exhibited decreased inflammatory cytokine levels and decreased rates of mortality in comparison to wild type (WT) mice, after sepsis in a cecal-ligation and puncture model (CLP). Compared to WT septic mice, sepsis in Tg mice resulted in improved cardiac functions, lower levels of troponin I and decreased rates of cardiomyocyte apoptosis, compared to WT mice. The increased formation of autophagicvacuoles detected with electron microscopy demonstrated the enhancement of cardiac autophagy. This phenomenon was further confirmed by the differential expression of genes related to autophagy, such as LC3, Atg5, Beclin-1, and p62. The increased expression of Cathepsin L(Ctsl), a specific marker for mitochondrial stress response, may be associated with the beneficial effects of the hypo-phosphorylated form of IκBβ. Our observations suggest that the hypo-phosphorylated form of IκBβ at Ser313 is beneficial to the heart in sepsis through inhibition of apoptosisand enhancement of autophagy in mutated IκBβ transgenic mice.
Collapse
Affiliation(s)
- Guang-Qing Wang
- Department of Burn Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Tao Tang
- Department of Surgery, 91528 Hospital of PLA, Shanghai, China
| | - Zhong-Shan Wang
- Department of Burn Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Ying-Ying Liu
- Department of Burn Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Li Wang
- Department of Burn Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Peng-Fei Luo
- Department of Burn Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Zhao-Fan Xia
- Department of Burn Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
- * E-mail:
| |
Collapse
|
34
|
Li L, Xu J, He L, Peng L, Zhong Q, Chen L, Jiang Z. The role of autophagy in cardiac hypertrophy. Acta Biochim Biophys Sin (Shanghai) 2016; 48:491-500. [PMID: 27084518 PMCID: PMC4913516 DOI: 10.1093/abbs/gmw025] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 01/25/2016] [Indexed: 12/12/2022] Open
Abstract
Autophagy is conserved in nature from lower eukaryotes to mammals and is an important self-cannibalizing, degradative process that contributes to the elimination of superfluous materials. Cardiac hypertrophy is primarily characterized by excess protein synthesis, increased cardiomyocyte size, and thickened ventricular walls and is a major risk factor that promotes arrhythmia and heart failure. In recent years, cardiomyocyte autophagy has been considered to play a role in controlling the hypertrophic response. However, the beneficial or aggravating role of cardiomyocyte autophagy in cardiac hypertrophy remains controversial. The exact mechanism of cardiomyocyte autophagy in cardiac hypertrophy requires further study. In this review, we summarize the controversies associated with autophagy in cardiac hypertrophy and provide insights into the role of autophagy in the development of cardiac hypertrophy. We conclude that future studies should emphasize the relationship between autophagy and the different stages of cardiac hypertrophy, as well as the autophagic flux and selective autophagy. Autophagy will be a potential therapeutic target for cardiac hypertrophy.
Collapse
Affiliation(s)
- Lanfang Li
- Post-Doctoral Mobile Stations for Basic Medicine, Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, University of South China, Hengyang 421001, China Hunan Children's Hospital and School of Pediatrics, University of South China, Changsha 410007, China
| | - Jin Xu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics, University of South China, Hengyang 421001, China
| | - Lu He
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics, University of South China, Hengyang 421001, China
| | - Lijun Peng
- Post-Doctoral Mobile Stations for Basic Medicine, Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, University of South China, Hengyang 421001, China Hunan Children's Hospital and School of Pediatrics, University of South China, Changsha 410007, China
| | - Qiaoqing Zhong
- Post-Doctoral Mobile Stations for Basic Medicine, Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, University of South China, Hengyang 421001, China
| | - Linxi Chen
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics, University of South China, Hengyang 421001, China
| | - Zhisheng Jiang
- Post-Doctoral Mobile Stations for Basic Medicine, Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, University of South China, Hengyang 421001, China
| |
Collapse
|
35
|
Olson OC, Joyce JA. Cysteine cathepsin proteases: regulators of cancer progression and therapeutic response. Nat Rev Cancer 2015; 15:712-29. [PMID: 26597527 DOI: 10.1038/nrc4027] [Citation(s) in RCA: 486] [Impact Index Per Article: 48.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cysteine cathepsin protease activity is frequently dysregulated in the context of neoplastic transformation. Increased activity and aberrant localization of proteases within the tumour microenvironment have a potent role in driving cancer progression, proliferation, invasion and metastasis. Recent studies have also uncovered functions for cathepsins in the suppression of the response to therapeutic intervention in various malignancies. However, cathepsins can be either tumour promoting or tumour suppressive depending on the context, which emphasizes the importance of rigorous in vivo analyses to ascertain function. Here, we review the basic research and clinical findings that underlie the roles of cathepsins in cancer, and provide a roadmap for the rational integration of cathepsin-targeting agents into clinical treatment.
Collapse
Affiliation(s)
- Oakley C Olson
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center
- Gerstner Sloan Kettering Graduate School of Biomedical Science, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Johanna A Joyce
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center
- Department of Oncology, University of Lausanne
- Ludwig Institute for Cancer Research, University of Lausanne, CH-1066 Lausanne, Switzerland
| |
Collapse
|
36
|
Sudhan DR, Siemann DW. Cathepsin L targeting in cancer treatment. Pharmacol Ther 2015; 155:105-16. [PMID: 26299995 DOI: 10.1016/j.pharmthera.2015.08.007] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 08/17/2015] [Indexed: 12/14/2022]
Abstract
Proteolytic enzymes may serve as promising targets for novel therapeutic treatment strategies seeking to impede cancer progression and metastasis. One such enzyme is cathepsin L (CTSL), a lysosomal cysteine protease. CTSL upregulation, a common occurrence in a variety of human cancers, has been widely correlated with metastatic aggressiveness and poor patient prognosis. In addition, CTSL has been implicated to contribute to cancer-associated osteolysis, a debilitating morbidity affecting both life expectancy and the quality of life. In this review, we highlight the mechanisms by which CTSL contributes to tumor progression and dissemination and discuss the therapeutic utility of CTSL intervention strategies aimed at impeding metastatic progression and bone resorption.
Collapse
Affiliation(s)
- Dhivya R Sudhan
- Department of Radiation Oncology, University of Florida Health Cancer Center, Gainesville, FL, USA
| | - Dietmar W Siemann
- Department of Radiation Oncology, University of Florida Health Cancer Center, Gainesville, FL, USA; Department of Pharmacology and Therapeutics, College of Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
37
|
Tholen M, Wolanski J, Stolze B, Chiabudini M, Gajda M, Bronsert P, Stickeler E, Rospert S, Reinheckel T. Stress-resistant Translation of Cathepsin L mRNA in Breast Cancer Progression. J Biol Chem 2015; 290:15758-15769. [PMID: 25957406 DOI: 10.1074/jbc.m114.624353] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Indexed: 11/06/2022] Open
Abstract
The cysteine protease cathepsin L (CTSL) is often thought to act as a tumor promoter by enhancing tumor progression and metastasis. This goes along with increased CTSL activity in various tumor entities; however, the mechanisms leading to high CTSL levels are incompletely understood. With the help of the polyoma middle T oncogene driven breast cancer mouse model expressing a human CTSL genomic transgene, we show that CTSL indeed promotes breast cancer metastasis to the lung. During tumor formation and progression high expression levels of CTSL are maintained by enduring translation of CTSL mRNA. Interestingly, human breast cancer specimens expressed the same pattern of 5' untranslated region (UTR) splice variants as the transgenic mice and the human cancer cell line MDA-MB 321. By polyribosome profiling of tumor tissues and human breast cancer cells, we observe an intrinsic resistance of CTSL to stress-induced shutdown of translation. This ability can be attributed to all 5' UTR variants of CTSL and is not dependent on a previously described internal ribosomal entry site motif. In conclusion, we provide in vivo functional evidence for overexpressed CTSL as a promoter of lung metastasis, whereas high CTSL levels are maintained during tumor progression due to stress-resistant mRNA translation.
Collapse
Affiliation(s)
- Martina Tholen
- Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; Faculty of Biology, Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany
| | - Julia Wolanski
- Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany
| | - Britta Stolze
- Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany
| | - Marco Chiabudini
- Institute of Biochemistry and Molecular Biology, Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; BIOSS Centre for Biological Signalling Studies, Albert-Ludwigs-University Freiburg, 79104 Freiburg
| | - Mieczyslaw Gajda
- Institute of Pathology, Friedrich-Schiller University, 07743 Jena, Germany
| | - Peter Bronsert
- Institute of Pathology, University Medical Center Freiburg, 79106 Freiburg; Comprehensive Cancer Center/German Cancer Consortium (DKTK), 79106 Freiburg
| | - Elmar Stickeler
- Comprehensive Cancer Center/German Cancer Consortium (DKTK), 79106 Freiburg; Clinic for Gynecology, University Medical Center Freiburg, 79106 Freiburg, Germany
| | - Sabine Rospert
- Institute of Biochemistry and Molecular Biology, Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; BIOSS Centre for Biological Signalling Studies, Albert-Ludwigs-University Freiburg, 79104 Freiburg
| | - Thomas Reinheckel
- Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; BIOSS Centre for Biological Signalling Studies, Albert-Ludwigs-University Freiburg, 79104 Freiburg; Comprehensive Cancer Center/German Cancer Consortium (DKTK), 79106 Freiburg.
| |
Collapse
|
38
|
A novel cysteine cathepsin inhibitor yields macrophage cell death and mammary tumor regression. Oncogene 2015; 34:6066-78. [PMID: 25798843 DOI: 10.1038/onc.2015.51] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2014] [Revised: 12/17/2014] [Accepted: 12/19/2014] [Indexed: 02/07/2023]
Abstract
Although cysteine cathepsins have been identified as key regulators of cancer growth, their specific role in tumor development remains unclear. Recent studies have shown that high activity levels of tumor cathepsins are primarily a result of increased cathepsin activity in cancer-promoting tumor-associated macrophages (TAMs). To further investigate the role of cysteine cathepsin activity in normal and polarized macrophages, we established in vitro and in vivo models of macrophage differentiation and polarization and used a novel cysteine cathepsin inhibitor, GB111-NH2, to block the activity of cathepsins B, L and S. Here we show that in vitro, cysteine cathepsin inhibition yields both apoptosis and proliferation of macrophages, owing to increased oxidative stress. Proteomic analysis of cathepsin- inhibited macrophages demonstrates inhibition of autophagy, suggesting a likely cause of elevated reactive oxygen species (ROS) levels. In vivo models of mammary cancer further show that cathepsin inhibition yields TAM death owing to increased ROS levels. Strikingly, apoptosis in TAMs yields a seemingly cell non-autonomous death of neighboring cancer cells, and regression of the primary growth. These results show that cysteine cathepsin inhibitors can specifically trigger macrophage cell death and may function as an effective anticancer therapy in tumors with high levels of TAMs.
Collapse
|
39
|
Kern U, Wischnewski V, Biniossek ML, Schilling O, Reinheckel T. Lysosomal protein turnover contributes to the acquisition of TGFβ-1 induced invasive properties of mammary cancer cells. Mol Cancer 2015; 14:39. [PMID: 25744631 PMCID: PMC4339013 DOI: 10.1186/s12943-015-0313-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 02/03/2015] [Indexed: 12/16/2022] Open
Abstract
Background Normal epithelial cells and carcinoma cells can acquire invasiveness by epithelial-to-mesenchymal transition (EMT), a process of considerable cellular remodeling. The endosomal/lysosomal compartment is a principal site of intracellular protein degradation. Lysosomal cathepsin proteases are secreted during cancer progression. The established pro-metastatic role of specific cysteine cathepsins has until now been ascribed to their contribution to extracellular matrix remodeling. We hypothesized that cysteine cathepsins affect transforming growth factor β-1 (TGFβ-1)-induced EMT of normal and malignant mammary epithelial cells. Methods The role of lysosomal proteolysis in TGFβ-1-induced EMT and invasion was investigated in a normal and a novel malignant murine mammary epithelial cell line. The contribution of cysteine cathepsins was determined by addition of the general cysteine cathepsin inhibitor E64d. Hallmarks of EMT were analyzed by molecular- and cell-biologic analyses including real-time cell migration/invasion assays. A quantitative proteome comparison using stable isotopic labeling with amino acids in culture (SILAC) showed the effect of E64d on TGFβ-1 induced proteome changes. Lysosomal patterning and junctional adhesion molecule A (Jam-a) localization and abundance were analyzed by immunofluorescence. Results We found increased lysosome activity during EMT of malignant mammary epithelial cells. Cysteine cathepsin inhibition had no effect on the induction of the TGFβ-1-induced EMT program on transcriptional level. Protease inhibition did not affect invasion of TGFβ-1 treated normal mammary epithelial cells, but reduced the invasion of murine breast cancer cells. Remarkably, reduced invasion was also evident if E64d was removed 24 h before the invasion assay in order to allow for recovery of cathepsin activity. Proteome analyses revealed a high abundance of lysosomal enzymes and lysosome-associated proteins in cancer cells treated with TGFβ-1 and E64d. An accumulation of those proteins and of lysosomal vesicles was further confirmed by independent methods. Interestingly, E64d caused lysosomal accumulation of Jam-a, a tight junction component facilitating epithelial cell-cell adhesion. Conclusion Our results demonstrate an important role of lysosomal proteolysis in cellular remodeling during EMT and a pivotal contribution of lysosomal cysteine cathepsins to TGFβ-1 induced acquisition of breast cancer cell invasiveness. These findings provide an additional rationale to use cathepsin inhibitors to stall tumor metastasis. Electronic supplementary material The online version of this article (doi:10.1186/s12943-015-0313-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ursula Kern
- Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University Freiburg, Stefan-Meier-Str. 17, Freiburg, D-79104, Germany. .,Spemann Graduate School of Biology and Medicine, Albert-Ludwigs-University Freiburg, Freiburg, Germany. .,Faculty of Biology, Albert-Ludwigs-University Freiburg, Freiburg, Germany.
| | - Vladimir Wischnewski
- Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University Freiburg, Stefan-Meier-Str. 17, Freiburg, D-79104, Germany.
| | - Martin L Biniossek
- Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University Freiburg, Stefan-Meier-Str. 17, Freiburg, D-79104, Germany.
| | - Oliver Schilling
- Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University Freiburg, Stefan-Meier-Str. 17, Freiburg, D-79104, Germany. .,BIOSS Centre for Biological Signalling Studies, Freiburg, Germany.
| | - Thomas Reinheckel
- Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University Freiburg, Stefan-Meier-Str. 17, Freiburg, D-79104, Germany. .,BIOSS Centre for Biological Signalling Studies, Freiburg, Germany. .,German Cancer Consortium (DKTK), Freiburg, Germany.
| |
Collapse
|
40
|
Abstract
Cardiovascular disease is the leading cause of death worldwide. As such, there is great interest in identifying novel mechanisms that govern the cardiovascular response to disease-related stress. First described in failing hearts, autophagy within the cardiovascular system has been widely characterized in cardiomyocytes, cardiac fibroblasts, endothelial cells, vascular smooth muscle cells, and macrophages. In all cases, a window of optimal autophagic activity appears to be critical to the maintenance of cardiovascular homeostasis and function; excessive or insufficient levels of autophagic flux can each contribute to heart disease pathogenesis. In this Review, we discuss the potential for targeting autophagy therapeutically and our vision for where this exciting biology may lead in the future.
Collapse
|
41
|
Out-of-frame start codons prevent translation of truncated nucleo-cytosolic cathepsin L in vivo. Nat Commun 2014; 5:4931. [PMID: 25222295 DOI: 10.1038/ncomms5931] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 08/07/2014] [Indexed: 11/08/2022] Open
Abstract
The lysosomal protease cathepsin L has been reported to cleave various functionally important cytosolic or nuclear proteins. To explain nucleo-cytosolic localization of cathepsin L, it has been hypothesized that skipping of the first start codon during translation initiation results in an N-terminally truncated protein lacking the endoplasmic reticulum-import signal. Here we demonstrate that out-of-frame AUGs prevent translation of truncated cathepsin L in cell culture as well as in a new knock-in mouse model. We further evaluate potential roles of nuclear cathepsin L during early embryonic development. Our analysis reveals normal epiblast development of cathepsin L-deficient embryos, but uncovers a pronounced lysosomal storage phenotype in the extra-embryonic tissue of the visceral endoderm. In conclusion, the phenotypes of cathepsin L deficiency can be fully assigned to lack of canonically targeted cathepsin L, while the biogenesis and functionality of nucleo-cytosolic cathepsin L remain elusive.
Collapse
|
42
|
Tholen S, Biniossek ML, Gansz M, Ahrens TD, Schlimpert M, Kizhakkedathu JN, Reinheckel T, Schilling O. Double deficiency of cathepsins B and L results in massive secretome alterations and suggests a degradative cathepsin-MMP axis. Cell Mol Life Sci 2014; 71:899-916. [PMID: 23811845 PMCID: PMC11113308 DOI: 10.1007/s00018-013-1406-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Revised: 06/06/2013] [Accepted: 06/10/2013] [Indexed: 12/31/2022]
Abstract
Endolysosomal cysteine cathepsins functionally cooperate. Cathepsin B (Ctsb) and L (Ctsl) double-knockout mice die 4 weeks after birth accompanied by (autophago-) lysosomal accumulations within neurons. Such accumulations are also observed in mouse embryonic fibroblasts (MEFs) deficient for Ctsb and Ctsl. Previous studies showed a strong impact of Ctsl on the MEF secretome. Here we show that Ctsb alone has only a mild influence on extracellular proteome composition. Protease cleavage sites dependent on Ctsb were identified by terminal amine isotopic labeling of substrates (TAILS), revealing a prominent yet mostly indirect impact on the extracellular proteolytic cleavages. To investigate the cooperation of Ctsb and Ctsl, we performed a quantitative secretome comparison of wild-type MEFs and Ctsb (-/-) Ctsl (-/-) MEFs. Deletion of both cathepsins led to drastic alterations in secretome composition, highlighting cooperative functionality. While many protein levels were decreased, immunodetection corroborated increased levels of matrix metalloproteinase (MMP)-2. Re-expression of Ctsl rescues MMP-2 abundance. Ctsl and to a much lesser extent Ctsb are able to degrade MMP-2 at acidic and neutral pH. Addition of active MMP-2 to the MEF secretome degrades proteins whose levels were also decreased by Ctsb and Ctsl double deficiency. These results suggest a degradative Ctsl-MMP-2 axis, resulting in increased MMP-2 levels upon cathepsin deficiency with subsequent degradation of secreted proteins such as collagen α-1 (I).
Collapse
Affiliation(s)
- Stefan Tholen
- Institute for Molecular Medicine and Cell Research, University of Freiburg, Stefan Meier Strasse 17, 79104 Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Martin L. Biniossek
- Institute for Molecular Medicine and Cell Research, University of Freiburg, Stefan Meier Strasse 17, 79104 Freiburg, Germany
| | - Martina Gansz
- Institute for Molecular Medicine and Cell Research, University of Freiburg, Stefan Meier Strasse 17, 79104 Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Theresa D. Ahrens
- Institute for Molecular Medicine and Cell Research, University of Freiburg, Stefan Meier Strasse 17, 79104 Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Manuel Schlimpert
- Institute for Molecular Medicine and Cell Research, University of Freiburg, Stefan Meier Strasse 17, 79104 Freiburg, Germany
| | - Jayachandran N. Kizhakkedathu
- Centre for Blood Research, University of British Columbia, Vancouver, BC V6T 1Z3 Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z3 Canada
- Department of Chemistry, University of British Columbia, Vancouver, BC V6T 1Z3 Canada
| | - Thomas Reinheckel
- Institute for Molecular Medicine and Cell Research, University of Freiburg, Stefan Meier Strasse 17, 79104 Freiburg, Germany
- BIOSS Centre for Biological Signaling Studies, University of Freiburg, 79104 Freiburg, Germany
| | - Oliver Schilling
- Institute for Molecular Medicine and Cell Research, University of Freiburg, Stefan Meier Strasse 17, 79104 Freiburg, Germany
- BIOSS Centre for Biological Signaling Studies, University of Freiburg, 79104 Freiburg, Germany
| |
Collapse
|
43
|
Collins-Hooper H, Sartori R, Macharia R, Visanuvimol K, Foster K, Matsakas A, Flasskamp H, Ray S, Dash PR, Sandri M, Patel K. Propeptide-Mediated Inhibition of Myostatin Increases Muscle Mass Through Inhibiting Proteolytic Pathways in Aged Mice. J Gerontol A Biol Sci Med Sci 2014; 69:1049-59. [DOI: 10.1093/gerona/glt170] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
44
|
Lockwood TD. Lysosomal metal, redox and proton cycles influencing the CysHis cathepsin reaction. Metallomics 2013; 5:110-24. [PMID: 23302864 DOI: 10.1039/c2mt20156a] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
In the 1930's pioneers discovered that maximal autolysis in tissue homogenates requires metal chelator, sulfhydryl reducing agent and acid pH. However, metals, reducing equivalents and protons (MR&P) have been overlooked as combined catalytic controls. Three categories of lysosomal machinery drive three distinguishable cycles importing and exporting MR&P. Zn(2+) preemptively inhibits CysHis catalysis under otherwise optimal protonation and reduction. Protein-bound cell Zn(2+) concentration is 200-2000 times the non-sequestered inhibitory concentration. Following autophagy, lysosomal proteolysis liberates much inhibitory Zn(2+). The vacuolar proton pump is the driving force for Zn(2+) export, as well as protonation of the peptidolytic mechanism. Other machinery of lysosomal cycles includes proton-driven Zn(2+) exporters (e.g. SLC11A1), Zn(2+) channels (e.g. TRPML-1), lysosomal thiol reductase, etc. The CysHis dyad is a sensor of the vacuolar environment of MR&P, an integrator of these simultaneous variables, and a catalytic responder. Rate-determination can shift between autophagic substrate acquisition (swallowing) and substrate degradation (digesting). Zn(2+) recycling from degraded proteins to new proteins is a fourth cycle that might pace lysosomal function under some conditions. Heritable insufficient or excess functions of CysHis cathepsins are associated with dysfunctional inflammation and immunity/auto-immunity, including diabetic pathogenesis.
Collapse
Affiliation(s)
- Thomas D Lockwood
- Dept. of Pharmacology, School of Medicine, Wright State University, Dayton, Ohio 45435, USA.
| |
Collapse
|
45
|
Aki T, Funakoshi T, Unuma K, Uemura K. Impairment of autophagy: from hereditary disorder to drug intoxication. Toxicology 2013; 311:205-15. [PMID: 23851159 DOI: 10.1016/j.tox.2013.07.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Revised: 06/30/2013] [Accepted: 07/01/2013] [Indexed: 12/11/2022]
Abstract
At first, the molecular mechanism of autophagy was unveiled in a unicellular organism Saccharomyces cerevisiae (budding yeast), followed by the discovery that the basic mechanism of autophagy is conserved in multicellular organisms including mammals. Although autophagy was considered to be a non-selective bulk protein degradation system to recycle amino acids during periods of nutrient starvation, it is also believed to be an essential mechanism for the selective elimination of proteins/organelles that are damaged under pathological conditions. Research advances made using autophagy-deficient animals have revealed that impairments of autophagy often underlie the pathogenesis of hereditary disorders such as Danon, Parkinson's, Alzheimer's, and Huntington's diseases, and amyotrophic lateral sclerosis. On the other hand, there are many reports that drugs and toxicants, including arsenic, cadmium, paraquat, methamphetamine, and ethanol, induce autophagy during the development of their toxicity on many organs including heart, brain, lung, kidney, and liver. Although the question as to whether autophagic machinery is involved in the execution of cell death or not remains controversial, the current view of the role of autophagy during cell/tissue injury is that it is an important, often essential, cytoprotective reaction; disturbances in cytoprotective autophagy aggravate cell/tissue injuries. The purpose of this review is to provide (1) a gross summarization of autophagy processes, which are becoming more important in the field of toxicology, and (2) examples of important studies reporting the involvement of perturbations in autophagy in cell/tissue injuries caused by acute as well as chronic intoxication.
Collapse
Affiliation(s)
- Toshihiko Aki
- Section of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8519, Japan.
| | | | | | | |
Collapse
|
46
|
Varma H, Gangadhar NM, Letso RR, Wolpaw AJ, Sriramaratnam R, Stockwell BR. Identification of a small molecule that induces ATG5-and-cathepsin-l-dependent cell death and modulates polyglutamine toxicity. Exp Cell Res 2013; 319:1759-1773. [PMID: 23588206 DOI: 10.1016/j.yexcr.2013.03.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 02/27/2013] [Accepted: 03/02/2013] [Indexed: 01/03/2023]
Abstract
Non-apoptotic cell death mechanisms are largely uncharacterized despite their importance in physiology and disease [1]. Here we sought to systematically identify non-apoptotic cell death pathways in mammalian cells. We screened 69,612 compounds for those that induce non-canonical cell death by counter screening in the presence of inhibitors of apoptosis and necrosis. We further selected compounds that require active protein synthesis for inducing cell death. Using this tiered approach, we identified NID-1 (Novel Inducer of Death-1), a small molecule that induces an active, energy-dependent cell death in diverse mammalian cell lines. NID-1-induced death required components of the autophagic machinery, including ATG5, and the lysosomal hydrolase cathepsin L, but was distinct from classical macroautophagy. Since macroautophagy can prevent cell death in several contexts, we tested and found that NID-1 suppressed cell death in a cell-based model of Huntington's disease, suggesting that NID-1 activates a specific pathway. Thus the discovery of NID-1 identifies a previously unexplored cell death pathway, and modulating this pathway may have therapeutic applications. Furthermore, these findings provide a proof-of-principle for using chemical screening to identify novel cell death paradigms.
Collapse
Affiliation(s)
- Hemant Varma
- Howard Hughes Medical Institute, Department of Biological Sciences, Columbia University, Northwest Corner Building, MC 4846, 550 West 120th Street, New York, NY 10027, United States; Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, United States
| | - Nidhi M Gangadhar
- Howard Hughes Medical Institute, Department of Biological Sciences, Columbia University, Northwest Corner Building, MC 4846, 550 West 120th Street, New York, NY 10027, United States
| | - Reka R Letso
- Howard Hughes Medical Institute, Department of Biological Sciences, Columbia University, Northwest Corner Building, MC 4846, 550 West 120th Street, New York, NY 10027, United States
| | - Adam J Wolpaw
- Howard Hughes Medical Institute, Department of Biological Sciences, Columbia University, Northwest Corner Building, MC 4846, 550 West 120th Street, New York, NY 10027, United States
| | - Rohitha Sriramaratnam
- Howard Hughes Medical Institute, Department of Biological Sciences, Columbia University, Northwest Corner Building, MC 4846, 550 West 120th Street, New York, NY 10027, United States
| | - Brent R Stockwell
- Howard Hughes Medical Institute, Department of Biological Sciences, Columbia University, Northwest Corner Building, MC 4846, 550 West 120th Street, New York, NY 10027, United States; Department of Chemistry, Columbia University, Northwest Corner Building, MC 4846, 550 West 120th Street, New York, NY 10027, United States.
| |
Collapse
|
47
|
Tholen S, Biniossek ML, Gansz M, Gomez-Auli A, Bengsch F, Noel A, Kizhakkedathu JN, Boerries M, Busch H, Reinheckel T, Schilling O. Deletion of cysteine cathepsins B or L yields differential impacts on murine skin proteome and degradome. Mol Cell Proteomics 2012; 12:611-25. [PMID: 23233448 DOI: 10.1074/mcp.m112.017962] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Numerous studies highlight the fact that concerted proteolysis is essential for skin morphology and function. The cysteine protease cathepsin L (Ctsl) has been implicated in epidermal proliferation and desquamation, as well as in hair cycle regulation. In stark contrast, mice deficient in cathepsin B (Ctsb) do not display an overt skin phenotype. To understand the systematic consequences of deleting Ctsb or Ctsl, we determined the protein abundances of >1300 proteins and proteolytic cleavage events in skin samples of wild-type, Ctsb(-/-), and Ctsl(-/-) mice via mass-spectrometry-based proteomics. Both protease deficiencies revealed distinct quantitative changes in proteome composition. Ctsl(-/-) skin revealed increased levels of the cysteine protease inhibitors cystatin B and cystatin M/E, increased cathepsin D, and an accumulation of the extracellular glycoprotein periostin. Immunohistochemistry located periostin predominantly in the hypodermal connective tissue of Ctsl(-/-) skin. The proteomic identification of proteolytic cleavage sites within skin proteins revealed numerous processing sites that are underrepresented in Ctsl(-/-) or Ctsb(-/-) samples. Notably, few of the affected cleavage sites shared the canonical Ctsl or Ctsb specificity, providing further evidence of a complex proteolytic network in the skin. Novel processing sites in proteins such as dermokine and Notch-1 were detected. Simultaneous analysis of acetylated protein N termini showed prototypical mammalian N-alpha acetylation. These results illustrate an influence of both Ctsb and Ctsl on the murine skin proteome and degradome, with the phenotypic consequences of the absence of either protease differing considerably.
Collapse
Affiliation(s)
- Stefan Tholen
- Institute of Molecular Medicine and Cell Research, University of Freiburg, Freiburg, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Wei DH, Jia XY, Liu YH, Guo FX, Tang ZH, Li XH, Wang Z, Liu LS, Wang GX, Jian ZS, Ruan CG. Cathepsin L stimulates autophagy and inhibits apoptosis of ox-LDL-induced endothelial cells: potential role in atherosclerosis. Int J Mol Med 2012; 31:400-6. [PMID: 23229094 DOI: 10.3892/ijmm.2012.1201] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Accepted: 11/07/2012] [Indexed: 11/06/2022] Open
Abstract
The activation of endothelial cells by oxidized low-density lipoprotein (ox-LDL) with subsequent increases in endothelial permeability occurs in the early stage of atherosclerosis. Cathepsin L (CATL) is one of the cysteine proteases and has been implicated in advanced atherosclerotic lesions and plaque instability. This study aimed to explore the role of CATL in ox-LDL-induced early atherosclerotic events and to delineate the underlying mechanism. Results showed that ox-LDL upregulated CATL protein levels and activation in human umbilical vein endothelial cells (ECs) in a concentration-dependent manner and stimulated EC autophagy and apoptosis and increased EC monolayer permeability. Concomitantly, VE-cadherin expression was decreased. When ECs were pretreated with a CATL inhibitor, ox-LDL-induced autophagy was inhibited while apoptosis was further increased. In addition, the VE-cadherin protein level was increased, and the EC monolayer permeability was reduced. Taken together, the present study showed that the upregulated expression and activation of CATL induced by ox-LDL, increased EC autophagy and antagonized EC apoptosis, which partly neutralized the effect of increased EC monolayer permeability mediated by the downregulation of VE-cadherin. Thus, the proatherogenic effect of CATL was partly neutralized by inducing autophagy and inhibiting apoptosis in early stages of atherosclerosis.
Collapse
Affiliation(s)
- Dang-Heng Wei
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, University of South China, Hengyang 421001, P.R. China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Lysosome vacuolation disrupts the completion of autophagy during norephedrine exposure in SH-SY5Y human neuroblastoma cells. Brain Res 2012; 1490:9-22. [PMID: 23123211 DOI: 10.1016/j.brainres.2012.10.056] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2012] [Revised: 09/29/2012] [Accepted: 10/27/2012] [Indexed: 11/22/2022]
Abstract
In our current study, we examined the mechanism underlying neuronal cell injuries caused by norephedrine in SH-SY5Y human neuroblastoma cells. Norephedrine was found to induce cytoplasmic vacuolation and a resultant loss of cell viability. In the cells treated with norephedrine also, an autophagic marker LC3 was converted to its LC3-II activated form, suggesting the induction of autophagy. In cells transfected with RFP-LC3 and GFP-LAMP1, a punctate patterning of LC3 expression and colocalization of LAMP1 with the formed vacuoles were observed, highlighting the lysosomal nature of the vacuoles and their association with autophagosomes. An autophagic flux assay using tfLC3 (mRFP-GFP-LC3) indicated the formation of autophagosomes and autolysosomes by norephedrine stimulation at an early timepoint (∼3 h). However, at a later timepoint (∼6 h), both the dilation of autolysosomes/lysosomes and the neutralization of the vacuolar pH were also observed. These results thus indicate that norephedrine induces autophagy at an early timepoint and cell death with lysosomal dysfunction and autophagy disruption at a later timepoint.
Collapse
|
50
|
Yu C, Huang X, Xu Y, LI H, Su J, Zhong J, Kang J, Liu Y, Sun L. Lysosome Dysfunction Enhances Oxidative Stress-Induced Apoptosis through Ubiquitinated Protein Accumulation in Hela Cells. Anat Rec (Hoboken) 2012; 296:31-9. [DOI: 10.1002/ar.22612] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Accepted: 08/23/2012] [Indexed: 12/16/2022]
|