1
|
Nixon RA, Rubinsztein DC. Mechanisms of autophagy-lysosome dysfunction in neurodegenerative diseases. Nat Rev Mol Cell Biol 2024; 25:926-946. [PMID: 39107446 DOI: 10.1038/s41580-024-00757-5] [Citation(s) in RCA: 51] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/17/2024] [Indexed: 08/15/2024]
Abstract
Autophagy is a lysosome-based degradative process used to recycle obsolete cellular constituents and eliminate damaged organelles and aggregate-prone proteins. Their postmitotic nature and extremely polarized morphologies make neurons particularly vulnerable to disruptions caused by autophagy-lysosomal defects, especially as the brain ages. Consequently, mutations in genes regulating autophagy and lysosomal functions cause a wide range of neurodegenerative diseases. Here, we review the role of autophagy and lysosomes in neurodegenerative diseases such as Alzheimer disease, Parkinson disease and frontotemporal dementia. We also consider the strong impact of cellular ageing on lysosomes and autophagy as a tipping point for the late-age emergence of related neurodegenerative disorders. Many of these diseases have primary defects in autophagy, for example affecting autophagosome formation, and in lysosomal functions, especially pH regulation and calcium homeostasis. We have aimed to provide an integrative framework for understanding the central importance of autophagic-lysosomal function in neuronal health and disease.
Collapse
Affiliation(s)
- Ralph A Nixon
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, New York, NY, USA.
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, USA.
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, USA.
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA.
| | - David C Rubinsztein
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge, UK
- UK Dementia Research Institute, University of Cambridge, Cambridge Institute for Medical Research, Cambridge, UK
| |
Collapse
|
2
|
Schneider J, Mitschke J, Bhat M, Vogele D, Schilling O, Reinheckel T, Heß L. Cathepsin D inhibition during neuronal differentiation selectively affects individual proteins instead of overall protein turnover. Biochimie 2024; 226:35-48. [PMID: 38552867 DOI: 10.1016/j.biochi.2024.03.013] [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: 12/20/2023] [Revised: 03/06/2024] [Accepted: 03/22/2024] [Indexed: 04/05/2024]
Abstract
Cathepsin D (CTSD) is a lysosomal aspartic protease and its inherited deficiency causes a severe pediatric neurodegenerative disease called neuronal ceroid lipofuscinosis (NCL) type 10. The lysosomal dysfunction in the affected patients leads to accumulation of undigested lysosomal cargo especially in none-dividing cells, such as neurons, resulting in death shortly after birth. To explore which proteins are mainly affected by the lysosomal dysfunction due to CTSD deficiency, Lund human mesencephalic (LUHMES) cells, capable of inducible dopaminergic neuronal differentiation, were treated with Pepstatin A. This inhibitor of "acidic" aspartic proteases caused accumulation of acidic intracellular vesicles in differentiating LUHMES cells. Pulse-chase experiments involving stable isotope labelling with amino acids in cell culture (SILAC) with subsequent mass-spectrometric protein identification and quantification were performed. By this approach, we studied the degradation and synthesis rates of 695 and 680 proteins during early and late neuronal LUHMES differentiation, respectively. Interestingly, lysosomal bulk proteolysis was not altered upon Pepstatin A treatment. Instead, the protease inhibitor selectively changed the turnover of individual proteins. Especially proteins belonging to the mitochondrial energy supply system were differentially degraded during early and late neuronal differentiation indicating a high energy demand as well as stress level in LUHMES cells treated with Pepstatin A.
Collapse
Affiliation(s)
- Johannes Schneider
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany; Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany
| | - Julia Mitschke
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany; German Cancer Consortium (DKTK), partner site Freiburg, 79104, Freiburg, Germany; German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Mahima Bhat
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
| | - Daniel Vogele
- Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany; Institute for Surgical Pathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany
| | - Oliver Schilling
- German Cancer Consortium (DKTK), partner site Freiburg, 79104, Freiburg, Germany; German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany; Institute for Surgical Pathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany; Centre for Biological Signalling Studies BIOSS, University of Freiburg, 79104, Freiburg, Germany
| | - Thomas Reinheckel
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany; German Cancer Consortium (DKTK), partner site Freiburg, 79104, Freiburg, Germany; German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany; Centre for Biological Signalling Studies BIOSS, University of Freiburg, 79104, Freiburg, Germany.
| | - Lisa Heß
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany
| |
Collapse
|
3
|
Cai Z, Xu S, Liu C. Cathepsin B in cardiovascular disease: Underlying mechanisms and therapeutic strategies. J Cell Mol Med 2024; 28:e70064. [PMID: 39248527 PMCID: PMC11382359 DOI: 10.1111/jcmm.70064] [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: 05/16/2024] [Revised: 08/01/2024] [Accepted: 08/22/2024] [Indexed: 09/10/2024] Open
Abstract
Cathepsin B (CTSB) is a member of the cysteine protease family, primarily responsible for degrading unnecessary organelles and proteins within the acidic milieu of lysosomes to facilitate recycling. Recent research has revealed that CTSB plays a multifaceted role beyond its function as a proteolytic enzyme in lysosomes. Importantly, recent data suggest that CTSB has significant impacts on different cardiac pathological conditions, such as atherosclerosis (AS), myocardial infarction, hypertension, heart failure and cardiomyopathy. Especially in the context of AS, preclinical models and clinical sample imaging data indicate that the cathepsin activity-based probe can reliably image CTSB activity in foam cells and atherosclerotic plaques; concurrently, it allows synchronous diagnostic and therapeutic interventions. However, our knowledge of CTSB in cardiovascular disease is still in the early stage. This paper aims to provide a comprehensive review of the significance of CTSB in cardiovascular physiology and pathology, with the objective of laying a theoretical groundwork for the development of drugs targeting CTSB.
Collapse
Affiliation(s)
- Zhulan Cai
- Department of Cardiology, Peking University Third Hospital, Beijing, P.R. China
| | - Shunyao Xu
- Department of Critical Care Medicine, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, P.R. China
| | - Chen Liu
- Department of Geriatrics, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, P.R. China
| |
Collapse
|
4
|
Calabrese L, Fiocco Z, Mellett M, Aoki R, Rubegni P, French LE, Satoh TK. Role of the NLRP1 inflammasome in skin cancer and inflammatory skin diseases. Br J Dermatol 2024; 190:305-315. [PMID: 37889986 DOI: 10.1093/bjd/ljad421] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/07/2023] [Accepted: 10/21/2023] [Indexed: 10/29/2023]
Abstract
Inflammasomes are cytoplasmic protein complexes that play a crucial role in protecting the host against pathogenic and sterile stressors by initiating inflammation. Upon activation, these complexes directly regulate the proteolytic processing and activation of proinflammatory cytokines interleukin (IL)-1β and IL-18 to induce a potent inflammatory response, and induce a programmed form of cell death called pyroptosis to expose intracellular pathogens to the surveillance of the immune system, thus perpetuating inflammation. There are various types of inflammasome complexes, with the NLRP1 (nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-1) inflammasome being the first one identified and currently recognized as the predominant inflammasome sensor protein in human keratinocytes. Human NLRP1 exhibits a unique domain structure, containing both an N-terminal pyrin (PYD) domain and an effector C-terminal caspase recruitment domain (CARD). It can be activated by diverse stimuli, such as viruses, ultraviolet B radiation and ribotoxic stress responses. Specific mutations in NLRP1 or related genes have been associated with rare monogenic skin disorders, such as multiple self-healing palmoplantar carcinoma; familial keratosis lichenoides chronica; autoinflammation with arthritis and dyskeratosis; and dipeptidyl peptidase 9 deficiency. Recent research breakthroughs have also highlighted the involvement of dysfunctions in the NLRP1 pathway in a handful of seemingly unrelated dermatological conditions. These range from monogenic autoinflammatory diseases to polygenic autoimmune diseases such as vitiligo, psoriasis, atopic dermatitis and skin cancer, including squamous cell carcinoma, melanoma and Kaposi sarcoma. Additionally, emerging evidence implicates NLRP1 in systemic lupus erythematosus, pemphigus vulgaris, Addison disease, Papillon-Lefèvre syndrome and leprosy. The aim of this review is to shed light on the implications of pathological dysregulation of the NLRP1 inflammasome in skin diseases and investigate the potential rationale for targeting this pathway as a future therapeutic approach.
Collapse
Affiliation(s)
- Laura Calabrese
- Dermatology Unit, Department of Medical, Surgical and Neurological Sciences, University of Siena, Siena, Italy
- Institute of Dermatology, Catholic University of the Sacred Heart, Rome, Italy
- Department of Dermatology and Allergy, University Hospital, LMU, Munich, Germany
| | - Zeno Fiocco
- Department of Dermatology and Allergy, University Hospital, LMU, Munich, Germany
| | - Mark Mellett
- Department of Dermatology, University Hospital Zürich, University of Zürich, Zürich, Switzerland
| | - Rui Aoki
- Department of Dermatology and Allergy, University Hospital, LMU, Munich, Germany
| | - Pietro Rubegni
- Dermatology Unit, Department of Medical, Surgical and Neurological Sciences, University of Siena, Siena, Italy
| | - Lars E French
- Department of Dermatology and Allergy, University Hospital, LMU, Munich, Germany
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Takashi K Satoh
- Department of Dermatology and Allergy, University Hospital, LMU, Munich, Germany
| |
Collapse
|
5
|
Stoka V, Vasiljeva O, Nakanishi H, Turk V. The Role of Cysteine Protease Cathepsins B, H, C, and X/Z in Neurodegenerative Diseases and Cancer. Int J Mol Sci 2023; 24:15613. [PMID: 37958596 PMCID: PMC10650516 DOI: 10.3390/ijms242115613] [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: 07/22/2023] [Revised: 10/17/2023] [Accepted: 10/18/2023] [Indexed: 11/15/2023] Open
Abstract
Papain-like cysteine proteases are composed of 11 human cysteine cathepsins, originally located in the lysosomes. They exhibit broad specificity and act as endopeptidases and/or exopeptidases. Among them, only cathepsins B, H, C, and X/Z exhibit exopeptidase activity. Recently, cysteine cathepsins have been found to be present outside the lysosomes and often participate in various pathological processes. Hence, they have been considered key signalling molecules. Their potentially hazardous proteolytic activities are tightly regulated. This review aims to discuss recent advances in understanding the structural aspects of these four cathepsins, mechanisms of their zymogen activation, regulation of their activities, and functional aspects of these enzymes in neurodegeneration and cancer. Neurodegenerative effects have been evaluated, particularly in Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, multiple sclerosis, and neuropsychiatric disorders. Cysteine cathepsins also participate in tumour progression and metastasis through the overexpression and secretion of proteases, which trigger extracellular matrix degradation. To our knowledge, this is the first review to provide an in-depth analysis regarding the roles of cysteine cathepsins B, H, C, and X in neurodegenerative diseases and cancer. Further advances in understanding the functions of cysteine cathepsins in these conditions will result in the development of novel, targeted therapeutic strategies.
Collapse
Affiliation(s)
- Veronika Stoka
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, SI-1000 Ljubljana, Slovenia;
- Jožef Stefan International Postgraduate School, SI-1000 Ljubljana, Slovenia
| | - Olga Vasiljeva
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, SI-1000 Ljubljana, Slovenia;
- CytomX Therapeutics, Inc., South San Francisco, CA 94080, USA
| | - Hiroshi Nakanishi
- Department of Pharmacology, Faculty of Pharmacy, Yasuda Women’s University, Hiroshima 731-0153, Japan;
| | - Vito Turk
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, SI-1000 Ljubljana, Slovenia;
- Jožef Stefan International Postgraduate School, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
6
|
Akiyama M. Cathepsin and cutaneous disorders of cornification and inflammation: their close links. Br J Dermatol 2023; 189:256-257. [PMID: 37287341 DOI: 10.1093/bjd/ljad190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 07/03/2023] [Indexed: 06/09/2023]
Abstract
As mentioned above, the cathepsin family is a group of important proteases that are deeply involved in the development of various skin disorders of cornification and inflammation, including AiKDs. The cathepsin family members are considered to play essential roles in the keratinocyte proliferation/differentiation and inflammatory pathways in the skin.
Collapse
Affiliation(s)
- Masashi Akiyama
- Department of Dermatology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| |
Collapse
|
7
|
Kordiš D, Turk V. Origin and Early Diversification of the Papain Family of Cysteine Peptidases. Int J Mol Sci 2023; 24:11761. [PMID: 37511529 PMCID: PMC10380794 DOI: 10.3390/ijms241411761] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/18/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023] Open
Abstract
Peptidases of the papain family play a key role in protein degradation, regulated proteolysis, and the host-pathogen arms race. Although the papain family has been the subject of many studies, knowledge about its diversity, origin, and evolution in Eukaryota, Bacteria, and Archaea is limited; thus, we aimed to address these long-standing knowledge gaps. We traced the origin and expansion of the papain family with a phylogenomic analysis, using sequence data from numerous prokaryotic and eukaryotic proteomes, transcriptomes, and genomes. We identified the full complement of the papain family in all prokaryotic and eukaryotic lineages. Analysis of the papain family provided strong evidence for its early diversification in the ancestor of eukaryotes. We found that the papain family has undergone complex and dynamic evolution through numerous gene duplications, which produced eight eukaryotic ancestral paralogous C1A lineages during eukaryogenesis. Different evolutionary forces operated on C1A peptidases, including gene duplication, horizontal gene transfer, and gene loss. This study challenges the current understanding of the origin and evolution of the papain family and provides valuable insights into their early diversification. The findings of this comprehensive study provide guidelines for future structural and functional studies of the papain family.
Collapse
Affiliation(s)
- Dušan Kordiš
- Department of Molecular and Biomedical Sciences, J. Stefan Institute, 1000 Ljubljana, Slovenia
| | - Vito Turk
- Department of Biochemistry, Molecular and Structural Biology, J. Stefan Institute, 1000 Ljubljana, Slovenia
- Jožef Stefan International Postgraduate School, Jamova 39, 1000 Ljubljana, Slovenia
| |
Collapse
|
8
|
Wang Y, Zhao J, Gu Y, Wang H, Jiang M, Zhao S, Qing H, Ni J. Cathepsin H: molecular characteristics and clues to function and mechanism. Biochem Pharmacol 2023; 212:115585. [PMID: 37148981 DOI: 10.1016/j.bcp.2023.115585] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 04/26/2023] [Accepted: 04/28/2023] [Indexed: 05/08/2023]
Abstract
Cathepsin H (CatH) is a lysosomal cysteine protease with a unique aminopeptidase activity that is extensively expressed in the lung, pancreas, thymus, kidney, liver, skin, and brain. Owing to its specific enzymatic activity, CatH has critical effects on the regulation of biological behaviours of cancer cells and pathological processes in brain diseases. Moreover, a neutral pH level is optimal for CatH activity, so it is expected to be active in the extra-lysosomal and extracellular space. In the present review, we describe the expression, maturation, and enzymatic properties of CatH, and summarize the available experimental evidence that mechanistically links CatH to various physiological and pathological processes. Finally, we discuss the challenges and potentials of CatH inhibitors in CatH-induced disease therapy.
Collapse
Affiliation(s)
- Yanfeng Wang
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Juan Zhao
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China; Aerospace Medical Center, Aerospace Center Hospital, Beijing, 100081, China
| | - Yebo Gu
- Department of Stomatology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Haiping Wang
- School of Pharmaceutical Science, Nanjing Tech University, Nanjing, China
| | - Muzhou Jiang
- Department of Periodontics, Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang, 110002, China
| | - Shuxuan Zhao
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Hong Qing
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China.
| | - Junjun Ni
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China.
| |
Collapse
|
9
|
Hald JD, Beck-Nielsen S, Gregersen PA, Gjørup H, Langdahl B. Pycnodysostosis in children and adults. Bone 2023; 169:116674. [PMID: 36646263 DOI: 10.1016/j.bone.2023.116674] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 01/02/2023] [Accepted: 01/05/2023] [Indexed: 01/14/2023]
Affiliation(s)
- Jannie Dahl Hald
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Denmark; Centre for Rare Diseases, Department of Pediatrics, Aarhus University Hospital, Denmark.
| | - Signe Beck-Nielsen
- Centre for Rare Diseases, Department of Pediatrics, Aarhus University Hospital, Denmark; Department of Clinical Medicine, Aarhus University, Denmark
| | - Pernille Axel Gregersen
- Centre for Rare Diseases, Department of Pediatrics, Aarhus University Hospital, Denmark; Department of Clinical Genetics, Aarhus University Hospital, Denmark
| | - Hans Gjørup
- Centre of Oral Health in Rare Diseases, Department of Maxillofacial Surgery, Aarhus University Hospital, Denmark
| | - Bente Langdahl
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Denmark; Department of Clinical Medicine, Aarhus University, Denmark
| |
Collapse
|
10
|
Bigot P, Chesseron S, Saidi A, Sizaret D, Parent C, Petit-Courty A, Courty Y, Lecaille F, Lalmanach G. Cleavage of Occludin by Cigarette Smoke-Elicited Cathepsin S Increases Permeability of Lung Epithelial Cells. Antioxidants (Basel) 2022; 12:antiox12010005. [PMID: 36670867 PMCID: PMC9854811 DOI: 10.3390/antiox12010005] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/05/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is an irreversible disease mainly caused by smoking. COPD is characterized by emphysema and chronic bronchitis associated with enhanced epithelial permeability. HYPOTHESIS Lung biopsies from smokers revealed a decreased expression level of occludin, which is a protein involved in the cohesion of epithelial tight junctions. Moreover, the occludin level correlated negatively with smoking history (pack-years), COPD grades, and cathepsin S (CatS) activity. Thus, we examined whether CatS could participate in the modulation of the integrity of human lung epithelial barriers. METHODS AND RESULTS Cigarette smoke extract (CSE) triggered the upregulation of CatS by THP-1 macrophages through the mTOR/TFEB signaling pathway. In a co-culture model, following the exposure of macrophages to CSE, an enhanced level of permeability of lung epithelial (16HBE and NHBE) cells towards FITC-Dextran was observed, which was associated with a decrease in occludin level. Similar results were obtained using 16HBE and NHBE cells cultured at the air-liquid interface. The treatment of THP-1 macrophages by CatS siRNAs or by a pharmacological inhibitor restored the barrier function of epithelial cells, suggesting that cigarette smoke-elicited CatS induced an alteration of epithelial integrity via the proteolytic injury of occludin. CONCLUSIONS Alongside its noteworthy resistance to oxidative stress induced by cigarette smoke oxidants and its deleterious elastin-degrading potency, CatS may also have a detrimental effect on the barrier function of epithelial cells through the cleavage of occludin. The obtained data emphasize the emerging role of CatS in smoking-related lung diseases and strengthen the relevance of targeting CatS in the treatment of emphysema and COPD.
Collapse
Affiliation(s)
- Paul Bigot
- Faculty of Medicine, University of Tours, 37000 Tours, France
- Team “Proteolytic Mechanisms in Inflammation”, INSERM, UMR1100, Research Center for Respiratory Diseases (CEPR), 37000 Tours, France
| | - Simon Chesseron
- Faculty of Medicine, University of Tours, 37000 Tours, France
- Team “Proteolytic Mechanisms in Inflammation”, INSERM, UMR1100, Research Center for Respiratory Diseases (CEPR), 37000 Tours, France
| | - Ahlame Saidi
- Faculty of Medicine, University of Tours, 37000 Tours, France
- Team “Proteolytic Mechanisms in Inflammation”, INSERM, UMR1100, Research Center for Respiratory Diseases (CEPR), 37000 Tours, France
| | - Damien Sizaret
- Faculty of Medicine, University of Tours, 37000 Tours, France
- Pathological Anatomy and Cytology, The University Hospital Center of Tours, 37000 Tours, France
| | - Christelle Parent
- Faculty of Medicine, University of Tours, 37000 Tours, France
- Team “Aerosol therapy and Biotherapeutics for Respiratory Diseases”, INSERM, UMR1100, Research Center for Respiratory Diseases (CEPR), 37000 Tours, France
| | - Agnès Petit-Courty
- Faculty of Medicine, University of Tours, 37000 Tours, France
- Team “Proteolytic Mechanisms in Inflammation”, INSERM, UMR1100, Research Center for Respiratory Diseases (CEPR), 37000 Tours, France
| | - Yves Courty
- Faculty of Medicine, University of Tours, 37000 Tours, France
- Team “Proteolytic Mechanisms in Inflammation”, INSERM, UMR1100, Research Center for Respiratory Diseases (CEPR), 37000 Tours, France
| | - Fabien Lecaille
- Faculty of Medicine, University of Tours, 37000 Tours, France
- Team “Proteolytic Mechanisms in Inflammation”, INSERM, UMR1100, Research Center for Respiratory Diseases (CEPR), 37000 Tours, France
| | - Gilles Lalmanach
- Faculty of Medicine, University of Tours, 37000 Tours, France
- Team “Proteolytic Mechanisms in Inflammation”, INSERM, UMR1100, Research Center for Respiratory Diseases (CEPR), 37000 Tours, France
- Correspondence: ; Tel.: +33-2-47-36-61-51
| |
Collapse
|
11
|
Biasizzo M, Javoršek U, Vidak E, Zarić M, Turk B. Cysteine cathepsins: A long and winding road towards clinics. Mol Aspects Med 2022; 88:101150. [PMID: 36283280 DOI: 10.1016/j.mam.2022.101150] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 12/03/2022]
Abstract
Biomedical research often focuses on properties that differentiate between diseased and healthy tissue; one of the current focuses is elevated expression and altered localisation of proteases. Among these proteases, dysregulation of cysteine cathepsins can frequently be observed in inflammation-associated diseases, which tips the functional balance from normal physiological to pathological manifestations. Their overexpression and secretion regularly exhibit a strong correlation with the development and progression of such diseases, making them attractive pharmacological targets. But beyond their mostly detrimental role in inflammation-associated diseases, cysteine cathepsins are physiologically highly important enzymes involved in various biological processes crucial for maintaining homeostasis and responding to different stimuli. Consequently, several challenges have emerged during the efforts made to translate basic research data into clinical applications. In this review, we present both physiological and pathological roles of cysteine cathepsins and discuss the clinical potential of cysteine cathepsin-targeting strategies for disease management and diagnosis.
Collapse
Affiliation(s)
- Monika Biasizzo
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000, Ljubljana, Slovenia; International Postgraduate School Jozef Stefan, Jamova 39, SI-1000, Ljubljana, Slovenia
| | - Urban Javoršek
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000, Ljubljana, Slovenia; International Postgraduate School Jozef Stefan, Jamova 39, SI-1000, Ljubljana, Slovenia
| | - Eva Vidak
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000, Ljubljana, Slovenia; International Postgraduate School Jozef Stefan, Jamova 39, SI-1000, Ljubljana, Slovenia
| | - Miki Zarić
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000, Ljubljana, Slovenia; International Postgraduate School Jozef Stefan, Jamova 39, SI-1000, Ljubljana, Slovenia
| | - Boris Turk
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000, Ljubljana, Slovenia; Faculty of Chemistry and Chemical Technology, University of Ljubljana, Vecna pot 113, SI-1000, Ljubljana, Slovenia.
| |
Collapse
|
12
|
Lecaille F, Chazeirat T, Saidi A, Lalmanach G. Cathepsin V: Molecular characteristics and significance in health and disease. Mol Aspects Med 2022; 88:101086. [PMID: 35305807 DOI: 10.1016/j.mam.2022.101086] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/23/2022] [Accepted: 02/26/2022] [Indexed: 12/31/2022]
Abstract
Human cysteine cathepsins form a family of eleven proteases (B, C, F, H, K, L, O, S, V, W, X/Z) that play important roles in a considerable number of biological and pathophysiological processes. Among them, cathepsin V, also known as cathepsin L2, is a lysosomal enzyme, which is mainly expressed in cornea, thymus, heart, brain, and skin. Cathepsin V is a multifunctional endopeptidase that is involved in both the release of antigenic peptides and the maturation of MHC class II molecules and participates in the turnover of elastin fibrils as well in the cleavage of intra- and extra-cellular substrates. Moreover, there is increasing evidence that cathepsin V may contribute to the progression of diverse diseases, due to the dysregulation of its expression and/or its activity. For instance, increased expression of cathepsin V is closely correlated with malignancies (breast cancer, squamous cell carcinoma, or colorectal cancer) as well vascular disorders (atherosclerosis, aortic aneurysm, hypertension) being the most prominent examples. This review aims to shed light on current knowledge on molecular aspects of cathepsin V (genomic organization, protein structure, substrate specificity), its regulation by protein and non-protein inhibitors as well to summarize its expression (tissue and cellular distribution). Then the core biological and pathophysiological roles of cathepsin V will be depicted, raising the question of its interest as a valuable target that can open up pioneering therapeutic avenues.
Collapse
Affiliation(s)
- Fabien Lecaille
- Université de Tours, Tours, France; INSERM, UMR 1100, Centre d'Etude des Pathologies Respiratoires (CEPR), Team "Mécanismes protéolytiques dans l'inflammation", Tours, France.
| | - Thibault Chazeirat
- Université de Tours, Tours, France; INSERM, UMR 1100, Centre d'Etude des Pathologies Respiratoires (CEPR), Team "Mécanismes protéolytiques dans l'inflammation", Tours, France
| | - Ahlame Saidi
- Université de Tours, Tours, France; INSERM, UMR 1100, Centre d'Etude des Pathologies Respiratoires (CEPR), Team "Mécanismes protéolytiques dans l'inflammation", Tours, France
| | - Gilles Lalmanach
- Université de Tours, Tours, France; INSERM, UMR 1100, Centre d'Etude des Pathologies Respiratoires (CEPR), Team "Mécanismes protéolytiques dans l'inflammation", Tours, France.
| |
Collapse
|
13
|
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: 38] [Impact Index Per Article: 12.7] [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
|
14
|
Hook G, Reinheckel T, Ni J, Wu Z, Kindy M, Peters C, Hook V. Cathepsin B Gene Knockout Improves Behavioral Deficits and Reduces Pathology in Models of Neurologic Disorders. Pharmacol Rev 2022; 74:600-629. [PMID: 35710131 PMCID: PMC9553114 DOI: 10.1124/pharmrev.121.000527] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cathepsin B (CTSB) is a powerful lysosomal protease. This review evaluated CTSB gene knockout (KO) outcomes for amelioration of brain dysfunctions in neurologic diseases and aging animal models. Deletion of the CTSB gene resulted in significant improvements in behavioral deficits, neuropathology, and/or biomarkers in traumatic brain injury, ischemia, inflammatory pain, opiate tolerance, epilepsy, aging, transgenic Alzheimer's disease (AD), and periodontitis AD models as shown in 12 studies. One study found beneficial effects for double CTSB and cathepsin S KO mice in a multiple sclerosis model. Transgenic AD models using amyloid precursor protein (APP) mimicking common sporadic AD in three studies showed that CTSB KO improved memory, neuropathology, and biomarkers; two studies used APP representing rare familial AD and found no CTSB KO effect, and two studies used highly engineered APP constructs and reported slight increases in a biomarker. In clinical studies, all reports found that CTSB enzyme was upregulated in diverse neurologic disorders, including AD in which elevated CTSB was positively correlated with cognitive dysfunction. In a wide range of neurologic animal models, CTSB was also upregulated and not downregulated. Further, human genetic mutation data provided precedence for CTSB upregulation causing disease. Thus, the consilience of data is that CTSB gene KO results in improved brain dysfunction and reduced pathology through blockade of CTSB enzyme upregulation that causes human neurologic disease phenotypes. The overall findings provide strong support for CTSB as a rational drug target and for CTSB inhibitors as therapeutic candidates for a wide range of neurologic disorders. SIGNIFICANCE STATEMENT: This review provides a comprehensive compilation of the extensive data on the effects of deleting the cathepsin B (CTSB) gene in neurological and aging mouse models of brain disorders. Mice lacking the CTSB gene display improved neurobehavioral deficits, reduced neuropathology, and amelioration of neuronal cell death and inflammatory biomarkers. The significance of the compelling CTSB evidence is that the data consilience validates CTSB as a drug target for discovery of CTSB inhibitors as potential therapeutics for treating numerous neurological diseases.
Collapse
Affiliation(s)
- Gregory Hook
- American Life Science Pharmaceuticals, La Jolla, California (G.H.); Institute of Molecular Medicine and Cell Research, Faculty of Medicine, Albert Ludwigs University, Freiburg, Germany (T.R.); German Cancer Consortium (DKTK) Partner Site Freiburg, Freiburg, Germany (T.R.); German Cancer Research Center (DKFZ), Heidelberg, Germany (T.R); Center for Biological Signaling Studies BIOSS, Albert Ludwigs University, Freiburg, Germany (T.R.); Key Laboratory of Molecular Medicine and Biotherapy, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, China (J.N.); Department of Aging Science and Pharmacology, OBT Research Center, Faculty of Dental Science, Kyushu University, Fukuoka, Japan (Z.W); Taneja College of Pharmacy, Department of Pharmaceutical Sciences, University of South Florida, Tampa, Florida (M.K.); James A Haley VAMC, Research Service, Tampa, Florida (M.K.); Institute of Molecular Medicine and Cell Research, Faculty of Biology, Albert Ludwigs University, Freiburg, Germany (C.P.); Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, La Jolla, CA (V.H.); and Department of Neuroscience and Department of Pharmacology, School of Medicine, University of California, La Jolla, CA (V.H.)
| | - Thomas Reinheckel
- American Life Science Pharmaceuticals, La Jolla, California (G.H.); Institute of Molecular Medicine and Cell Research, Faculty of Medicine, Albert Ludwigs University, Freiburg, Germany (T.R.); German Cancer Consortium (DKTK) Partner Site Freiburg, Freiburg, Germany (T.R.); German Cancer Research Center (DKFZ), Heidelberg, Germany (T.R); Center for Biological Signaling Studies BIOSS, Albert Ludwigs University, Freiburg, Germany (T.R.); Key Laboratory of Molecular Medicine and Biotherapy, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, China (J.N.); Department of Aging Science and Pharmacology, OBT Research Center, Faculty of Dental Science, Kyushu University, Fukuoka, Japan (Z.W); Taneja College of Pharmacy, Department of Pharmaceutical Sciences, University of South Florida, Tampa, Florida (M.K.); James A Haley VAMC, Research Service, Tampa, Florida (M.K.); Institute of Molecular Medicine and Cell Research, Faculty of Biology, Albert Ludwigs University, Freiburg, Germany (C.P.); Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, La Jolla, CA (V.H.); and Department of Neuroscience and Department of Pharmacology, School of Medicine, University of California, La Jolla, CA (V.H.)
| | - Junjun Ni
- American Life Science Pharmaceuticals, La Jolla, California (G.H.); Institute of Molecular Medicine and Cell Research, Faculty of Medicine, Albert Ludwigs University, Freiburg, Germany (T.R.); German Cancer Consortium (DKTK) Partner Site Freiburg, Freiburg, Germany (T.R.); German Cancer Research Center (DKFZ), Heidelberg, Germany (T.R); Center for Biological Signaling Studies BIOSS, Albert Ludwigs University, Freiburg, Germany (T.R.); Key Laboratory of Molecular Medicine and Biotherapy, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, China (J.N.); Department of Aging Science and Pharmacology, OBT Research Center, Faculty of Dental Science, Kyushu University, Fukuoka, Japan (Z.W); Taneja College of Pharmacy, Department of Pharmaceutical Sciences, University of South Florida, Tampa, Florida (M.K.); James A Haley VAMC, Research Service, Tampa, Florida (M.K.); Institute of Molecular Medicine and Cell Research, Faculty of Biology, Albert Ludwigs University, Freiburg, Germany (C.P.); Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, La Jolla, CA (V.H.); and Department of Neuroscience and Department of Pharmacology, School of Medicine, University of California, La Jolla, CA (V.H.)
| | - Zhou Wu
- American Life Science Pharmaceuticals, La Jolla, California (G.H.); Institute of Molecular Medicine and Cell Research, Faculty of Medicine, Albert Ludwigs University, Freiburg, Germany (T.R.); German Cancer Consortium (DKTK) Partner Site Freiburg, Freiburg, Germany (T.R.); German Cancer Research Center (DKFZ), Heidelberg, Germany (T.R); Center for Biological Signaling Studies BIOSS, Albert Ludwigs University, Freiburg, Germany (T.R.); Key Laboratory of Molecular Medicine and Biotherapy, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, China (J.N.); Department of Aging Science and Pharmacology, OBT Research Center, Faculty of Dental Science, Kyushu University, Fukuoka, Japan (Z.W); Taneja College of Pharmacy, Department of Pharmaceutical Sciences, University of South Florida, Tampa, Florida (M.K.); James A Haley VAMC, Research Service, Tampa, Florida (M.K.); Institute of Molecular Medicine and Cell Research, Faculty of Biology, Albert Ludwigs University, Freiburg, Germany (C.P.); Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, La Jolla, CA (V.H.); and Department of Neuroscience and Department of Pharmacology, School of Medicine, University of California, La Jolla, CA (V.H.)
| | - Mark Kindy
- American Life Science Pharmaceuticals, La Jolla, California (G.H.); Institute of Molecular Medicine and Cell Research, Faculty of Medicine, Albert Ludwigs University, Freiburg, Germany (T.R.); German Cancer Consortium (DKTK) Partner Site Freiburg, Freiburg, Germany (T.R.); German Cancer Research Center (DKFZ), Heidelberg, Germany (T.R); Center for Biological Signaling Studies BIOSS, Albert Ludwigs University, Freiburg, Germany (T.R.); Key Laboratory of Molecular Medicine and Biotherapy, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, China (J.N.); Department of Aging Science and Pharmacology, OBT Research Center, Faculty of Dental Science, Kyushu University, Fukuoka, Japan (Z.W); Taneja College of Pharmacy, Department of Pharmaceutical Sciences, University of South Florida, Tampa, Florida (M.K.); James A Haley VAMC, Research Service, Tampa, Florida (M.K.); Institute of Molecular Medicine and Cell Research, Faculty of Biology, Albert Ludwigs University, Freiburg, Germany (C.P.); Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, La Jolla, CA (V.H.); and Department of Neuroscience and Department of Pharmacology, School of Medicine, University of California, La Jolla, CA (V.H.)
| | - Christoph Peters
- American Life Science Pharmaceuticals, La Jolla, California (G.H.); Institute of Molecular Medicine and Cell Research, Faculty of Medicine, Albert Ludwigs University, Freiburg, Germany (T.R.); German Cancer Consortium (DKTK) Partner Site Freiburg, Freiburg, Germany (T.R.); German Cancer Research Center (DKFZ), Heidelberg, Germany (T.R); Center for Biological Signaling Studies BIOSS, Albert Ludwigs University, Freiburg, Germany (T.R.); Key Laboratory of Molecular Medicine and Biotherapy, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, China (J.N.); Department of Aging Science and Pharmacology, OBT Research Center, Faculty of Dental Science, Kyushu University, Fukuoka, Japan (Z.W); Taneja College of Pharmacy, Department of Pharmaceutical Sciences, University of South Florida, Tampa, Florida (M.K.); James A Haley VAMC, Research Service, Tampa, Florida (M.K.); Institute of Molecular Medicine and Cell Research, Faculty of Biology, Albert Ludwigs University, Freiburg, Germany (C.P.); Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, La Jolla, CA (V.H.); and Department of Neuroscience and Department of Pharmacology, School of Medicine, University of California, La Jolla, CA (V.H.)
| | - Vivian Hook
- American Life Science Pharmaceuticals, La Jolla, California (G.H.); Institute of Molecular Medicine and Cell Research, Faculty of Medicine, Albert Ludwigs University, Freiburg, Germany (T.R.); German Cancer Consortium (DKTK) Partner Site Freiburg, Freiburg, Germany (T.R.); German Cancer Research Center (DKFZ), Heidelberg, Germany (T.R); Center for Biological Signaling Studies BIOSS, Albert Ludwigs University, Freiburg, Germany (T.R.); Key Laboratory of Molecular Medicine and Biotherapy, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, China (J.N.); Department of Aging Science and Pharmacology, OBT Research Center, Faculty of Dental Science, Kyushu University, Fukuoka, Japan (Z.W); Taneja College of Pharmacy, Department of Pharmaceutical Sciences, University of South Florida, Tampa, Florida (M.K.); James A Haley VAMC, Research Service, Tampa, Florida (M.K.); Institute of Molecular Medicine and Cell Research, Faculty of Biology, Albert Ludwigs University, Freiburg, Germany (C.P.); Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, La Jolla, CA (V.H.); and Department of Neuroscience and Department of Pharmacology, School of Medicine, University of California, La Jolla, CA (V.H.)
| |
Collapse
|
15
|
Kolter J, Henneke P, Groß O, Kierdorf K, Prinz M, Graf L, Schwemmle M. Paradoxical immunodeficiencies-When failures of innate immunity cause immunopathology. Eur J Immunol 2022; 52:1419-1430. [PMID: 35551651 DOI: 10.1002/eji.202149531] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 05/05/2022] [Accepted: 05/10/2022] [Indexed: 11/06/2022]
Abstract
Innate immunity facilitates immediate defense against invading pathogens throughout all organs and tissues but also mediates tissue homeostasis and repair, thereby playing a key role in health and development. Recognition of pathogens is mediated by germline-encoded PRRs. Depending on the specific PRRs triggered, ligand binding leads to phagocytosis and pathogen killing and the controlled release of immune-modulatory factors such as IFNs, cytokines, or chemokines. PRR-mediated and other innate immune responses do not only prevent uncontrolled replication of intruding pathogens but also contribute to the tailoring of an effective adaptive immune response. Therefore, hereditary or acquired immunodeficiencies impairing innate responses may paradoxically cause severe immunopathology in patients. This can occur in the context of, but also independently of an increased microbial burden. It can include pathogen-dependent organ damage, autoinflammatory syndromes, and neurodevelopmental or neurodegenerative diseases. Here, we discuss the current state of research of several different such immune paradoxes. Understanding the underlying mechanisms causing immunopathology as a consequence of failures of innate immunity may help to prevent life-threatening disease.
Collapse
Affiliation(s)
- Julia Kolter
- Faculty of Medicine, Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), University of Freiburg, Freiburg, Germany
| | - Philipp Henneke
- Faculty of Medicine, Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), University of Freiburg, Freiburg, Germany.,Center for Pediatrics and Adolescent Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Olaf Groß
- Faculty of Medicine, Institute of Neuropathology, Medical Center, University of Freiburg, Freiburg, Germany.,Faculty of Medicine, Center for Basics in NeuroModulation (NeuroModulBasics), University of Freiburg, Freiburg, Germany.,CIBSS-Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | - Katrin Kierdorf
- Faculty of Medicine, Institute of Neuropathology, Medical Center, University of Freiburg, Freiburg, Germany.,Faculty of Medicine, Center for Basics in NeuroModulation (NeuroModulBasics), University of Freiburg, Freiburg, Germany.,CIBSS-Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | - Marco Prinz
- Faculty of Medicine, Institute of Neuropathology, Medical Center, University of Freiburg, Freiburg, Germany.,Faculty of Medicine, Center for Basics in NeuroModulation (NeuroModulBasics), University of Freiburg, Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Laura Graf
- Faculty of Medicine, Institute of Virology, University of Freiburg, Freiburg, Germany
| | - Martin Schwemmle
- Faculty of Medicine, Institute of Virology, University of Freiburg, Freiburg, Germany
| |
Collapse
|
16
|
Saudenova M, Promnitz J, Ohrenschall G, Himmerkus N, Böttner M, Kunke M, Bleich M, Theilig F. Behind every smile there's teeth: Cathepsin B's function in health and disease with a kidney view. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119190. [PMID: 34968578 DOI: 10.1016/j.bbamcr.2021.119190] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 12/10/2021] [Accepted: 12/13/2021] [Indexed: 06/14/2023]
Abstract
Cathepsin B (CatB) is a very abundant lysosomal protease with endo- and carboxydipeptidase activities and even ligase features. In this review, we will provide a general characterization of CatB and describe structure, structure-derived properties and location-dependent proteolytic actions. We depict CatB action within lysosome and its important roles in lysosomal biogenesis, lysosomal homeostasis and autophagy rendering this protease a key player in orchestrating lysosomal functions. Lysosomal leakage and subsequent escape of CatB into the cytosol lead to harmful actions, e.g. the role in activating the NLPR3 inflammasome, affecting immune responses and cell death. The second focus of this review addresses CatB functions in the kidney, i.e. the glomerulus, the proximal tubule and collecting duct with strong emphasis of its role in pathology of the respective segment. Finally, observations regarding CatB functions that need to be considered in cell culture will be discussed. In conclusion, CatB a physiologically important molecule may, upon aberrant expression in different cellular context, become a harmful player effectively showing its teeth behind its smile.
Collapse
Affiliation(s)
- Makhabbat Saudenova
- Institute of Anatomy, Department of Medicine, Christian-Albrechts-University Kiel, Germany
| | - Jessica Promnitz
- Institute of Anatomy, Department of Medicine, Christian-Albrechts-University Kiel, Germany
| | - Gerrit Ohrenschall
- Institute of Anatomy, Department of Medicine, Christian-Albrechts-University Kiel, Germany
| | - Nina Himmerkus
- Institute of Physiology, Department of Medicine, Christian-Albrechts-University Kiel, Germany
| | - Martina Böttner
- Institute of Anatomy, Department of Medicine, Christian-Albrechts-University Kiel, Germany
| | - Madlen Kunke
- Institute of Anatomy, Department of Medicine, Christian-Albrechts-University Kiel, Germany
| | - Markus Bleich
- Institute of Physiology, Department of Medicine, Christian-Albrechts-University Kiel, Germany
| | - Franziska Theilig
- Institute of Anatomy, Department of Medicine, Christian-Albrechts-University Kiel, Germany.
| |
Collapse
|
17
|
Romero A, Novoa B, Figueras A. Genomic and transcriptomic identification of the cathepsin superfamily in the Mediterranean mussel Mytilus galloprovincialis. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2022; 127:104286. [PMID: 34619173 DOI: 10.1016/j.dci.2021.104286] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 09/30/2021] [Accepted: 10/01/2021] [Indexed: 06/13/2023]
Abstract
Cathepsins are lysosomal enzymes that participate in important physiological processes, such as development, tissue remodelling, senescence and innate and adaptive immunity. The description of these proteins in molluscs is fragmented and incomplete. In the present work, we identified most of the cathepsin family members in the bivalve Mytilus galloprovincialis by screening published genomic and transcriptomic information. In this specie, the cathepsin family is composed of 41 proteins showing a high diversification of cathepsins D, L and F, not previously observed in other taxonomic groups. Specific set of cathepsins are constitutively expressed in the different mussel tissues. Transcriptomic analyses suggested coordinated activity of the different cathepsins and their sequential activation during larval development. Cathepsins also play an important role in the immune response of bivalves, and different immune pathways seem to be activated in response to Vibrio splendidus infection.
Collapse
Affiliation(s)
- Alejandro Romero
- Instituto de Investigaciones Marinas (CSIC), Eduardo Cabello 6, 36208, Vigo, Spain
| | - Beatriz Novoa
- Instituto de Investigaciones Marinas (CSIC), Eduardo Cabello 6, 36208, Vigo, Spain.
| | - Antonio Figueras
- Instituto de Investigaciones Marinas (CSIC), Eduardo Cabello 6, 36208, Vigo, Spain
| |
Collapse
|
18
|
Kos J, Mitrović A, Perišić Nanut M, Pišlar A. Lysosomal peptidases – Intriguing roles in cancer progression and neurodegeneration. FEBS Open Bio 2022; 12:708-738. [PMID: 35067006 PMCID: PMC8972049 DOI: 10.1002/2211-5463.13372] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 01/04/2022] [Accepted: 01/20/2022] [Indexed: 11/16/2022] Open
Abstract
Lysosomal peptidases are hydrolytic enzymes capable of digesting waste proteins that are targeted to lysosomes via endocytosis and autophagy. Besides intracellular protein catabolism, they play more specific roles in several other cellular processes and pathologies, either within lysosomes, upon secretion into the cell cytoplasm or extracellular space, or bound to the plasma membrane. In cancer, lysosomal peptidases are generally associated with disease progression, as they participate in crucial processes leading to changes in cell morphology, signaling, migration, and invasion, and finally metastasis. However, they can also enhance the mechanisms resulting in cancer regression, such as apoptosis of tumor cells or antitumor immune responses. Lysosomal peptidases have also been identified as hallmarks of aging and neurodegeneration, playing roles in oxidative stress, mitochondrial dysfunction, abnormal intercellular communication, dysregulated trafficking, and the deposition of protein aggregates in neuronal cells. Furthermore, deficiencies in lysosomal peptidases may result in other pathological states, such as lysosomal storage disease. The aim of this review was to highlight the role of lysosomal peptidases in particular pathological processes of cancer and neurodegeneration and to address the potential of lysosomal peptidases in diagnosing and treating patients.
Collapse
Affiliation(s)
- Janko Kos
- University of Ljubljana Faculty of Pharmacy Aškerčeva 7 1000 Ljubljana Slovenia
- Jožef Stefan Institute Department of Biotechnology Jamova 39 1000 Ljubljana Slovenia
| | - Ana Mitrović
- Jožef Stefan Institute Department of Biotechnology Jamova 39 1000 Ljubljana Slovenia
| | - Milica Perišić Nanut
- Jožef Stefan Institute Department of Biotechnology Jamova 39 1000 Ljubljana Slovenia
| | - Anja Pišlar
- University of Ljubljana Faculty of Pharmacy Aškerčeva 7 1000 Ljubljana Slovenia
| |
Collapse
|
19
|
Alquezar C, Schoch KM, Geier EG, Ramos EM, Scrivo A, Li KH, Argouarch AR, Mlynarski EE, Dombroski B, DeTure M, Dickson DW, Yokoyama JS, Cuervo AM, Burlingame AL, Schellenberg GD, Miller TM, Miller BL, Kao AW. TSC1 loss increases risk for tauopathy by inducing tau acetylation and preventing tau clearance via chaperone-mediated autophagy. SCIENCE ADVANCES 2021; 7:eabg3897. [PMID: 34739309 PMCID: PMC8570595 DOI: 10.1126/sciadv.abg3897] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 09/17/2021] [Indexed: 05/20/2023]
Abstract
Age-associated neurodegenerative disorders demonstrating tau-laden intracellular inclusions are known as tauopathies. We previously linked a loss-of-function mutation in the TSC1 gene to tau accumulation and frontotemporal lobar degeneration. Now, we have identified genetic variants in TSC1 that decrease TSC1/hamartin levels and predispose to tauopathies such as Alzheimer’s disease and progressive supranuclear palsy. Cellular and murine models of TSC1 haploinsufficiency, as well as human brains carrying a TSC1 risk variant, accumulated tau protein that exhibited aberrant acetylation. This acetylation hindered tau degradation via chaperone-mediated autophagy, thereby leading to its accumulation. Aberrant tau acetylation in TSC1 haploinsufficiency resulted from the dysregulation of both p300 acetyltransferase and SIRT1 deacetylase. Pharmacological modulation of either enzyme restored tau levels. This study substantiates TSC1 as a novel tauopathy risk gene and includes TSC1 haploinsufficiency as a genetic model for tauopathies. In addition, these findings promote tau acetylation as a rational target for tauopathy therapeutics and diagnostic.
Collapse
Affiliation(s)
- Carolina Alquezar
- UCSF Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA 94158, USA
| | - Kathleen M. Schoch
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ethan G. Geier
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94143, USA
| | - Eliana Marisa Ramos
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Aurora Scrivo
- Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Kathy H. Li
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158, USA
| | - Andrea R. Argouarch
- UCSF Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA 94158, USA
| | - Elisabeth E. Mlynarski
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-4238, USA
| | - Beth Dombroski
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-4238, USA
| | - Michael DeTure
- Department of Neuroscience, The Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| | - Dennis W. Dickson
- Department of Neuroscience, The Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| | - Jennifer S. Yokoyama
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94143, USA
| | - Ana M. Cuervo
- Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Alma L. Burlingame
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158, USA
| | - Gerard D. Schellenberg
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-4238, USA
| | - Timothy M. Miller
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Bruce L. Miller
- UCSF Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA 94158, USA
| | - Aimee W. Kao
- UCSF Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA 94158, USA
- Corresponding author.
| |
Collapse
|
20
|
Abstract
All living organisms depend on tightly regulated cellular networks to control biological functions. Proteolysis is an important irreversible post-translational modification that regulates most, if not all, cellular processes. Proteases are a large family of enzymes that perform hydrolysis of protein substrates, leading to protein activation or degradation. The 473 known and 90 putative human proteases are divided into 5 main mechanistic groups: metalloproteases, serine proteases, cysteine proteases, threonine proteases, and aspartic acid proteases. Proteases are fundamental to all biological systems, and when dysregulated they profoundly influence disease progression. Inhibiting proteases has led to effective therapies for viral infections, cardiovascular disorders, and blood coagulation just to name a few. Between 5 and 10% of all pharmaceutical targets are proteases, despite limited knowledge about their biological roles. More than 50% of all human proteases have no known substrates. We present here a comprehensive list of all current known human proteases. We also present current and novel biochemical tools to characterize protease functions in vitro, in vivo, and ex vivo. These tools make it achievable to define both beneficial and detrimental activities of proteases in health and disease.
Collapse
Affiliation(s)
- Longxiang Wang
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Kimberly Main
- Department of Physiology & Pharmacology, University of Calgary, Calgary, AB T2N 1N4, Canada.,McCaig Institute for Bone & Joint Health, University of Calgary, Calgary, AB T2N 1N4, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Henry Wang
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Olivier Julien
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Antoine Dufour
- Department of Physiology & Pharmacology, University of Calgary, Calgary, AB T2N 1N4, Canada.,McCaig Institute for Bone & Joint Health, University of Calgary, Calgary, AB T2N 1N4, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 1N4, Canada
| |
Collapse
|
21
|
Moghaddasi M, Ghassemi M, Shekari Yazdi M, Habibi SAH, Mohebi N, Goodarzi A. The first case report of Haim Munk disease with neurological manifestations and literature review. Clin Case Rep 2021; 9:e04802. [PMID: 34603725 PMCID: PMC8473953 DOI: 10.1002/ccr3.4802] [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: 06/26/2021] [Revised: 08/17/2021] [Accepted: 08/25/2021] [Indexed: 11/10/2022] Open
Abstract
HMS can have neurologic MS like manifestations. It is urgent to do more research and report probable unknown associations of HMS for its better management.
Collapse
Affiliation(s)
- Mehdi Moghaddasi
- Department of NeurologyRasool Akram Medical ComplexIran University of Medial Sciences (IUMS)TehranIran
| | - Mohammadreza Ghassemi
- Department of DermatologyRasool Akram Medical ComplexIran University of Medical Sciences (IUMS)TehranIran
| | - Mohammad Shekari Yazdi
- Department of DermatologyRasool Akram Medical ComplexIran University of Medical Sciences (IUMS)TehranIran
| | - Seyed Amir Hasan Habibi
- Department of NeurologyRasool Akram Medical ComplexIran University of Medial Sciences (IUMS)TehranIran
| | - Nafiseh Mohebi
- Department of NeurologyRasool Akram Medical ComplexIran University of Medial Sciences (IUMS)TehranIran
| | - Azadeh Goodarzi
- Department of DermatologyRasool Akram Medical ComplexIran University of Medical Sciences (IUMS)TehranIran
| |
Collapse
|
22
|
Wang Y, Zhang H, Feng S. Novel Compound Heterozygous Mutations in CTSC Gene in a Chinese Family with Papillon-Lefevre Syndrome. Ann Dermatol 2021; 33:369-372. [PMID: 34341640 PMCID: PMC8273319 DOI: 10.5021/ad.2021.33.4.369] [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: 01/03/2020] [Revised: 02/28/2020] [Accepted: 03/18/2020] [Indexed: 11/08/2022] Open
Abstract
Papillon-Lefevre syndrome (PLS) (OMIM: 245000) is a rare autosomal recessive disorder characterized by palmoplantar hyperkeratosis and early onset periodontitis, resulting in the premature loss of the deciduous and permanent teeth. PLS is caused by mutations in the cathepsin C (CTSC) gene (OMIM: 602365), which has been mapped to chromosome 11q14-q21. Genetic analysis can help early and rapid diagnosis of PLS. Here we report on a Chinese PLS pedigree with two affected siblings. We have identified two novel compound heterozygous mutations c.763T>C (p.C255R) and c.1015C>A (p.R339S) in the CTSC gene. The two mutations expand the spectrum of CTSC mutations in PLS.
Collapse
Affiliation(s)
- Yuan Wang
- Department of Dermatology, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Hanmei Zhang
- Department of Dermatology, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Suying Feng
- Department of Dermatology, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| |
Collapse
|
23
|
Juettner NE, Bogen JP, Bauer TA, Knapp S, Pfeifer F, Huettenhain SH, Meusinger R, Kraemer A, Fuchsbauer HL. Decoding the Papain Inhibitor from Streptomyces mobaraensis as Being Hydroxylated Chymostatin Derivatives: Purification, Structure Analysis, and Putative Biosynthetic Pathway. JOURNAL OF NATURAL PRODUCTS 2020; 83:2983-2995. [PMID: 32998509 DOI: 10.1021/acs.jnatprod.0c00201] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Streptomyces mobaraensis produces the papain inhibitor SPI consisting of a 12 kDa protein and small active compounds (SPIac). Purification of the papain inhibitory compounds resulted in four diverse chymostatin derivatives that were characterized by NMR and MS analysis. Chymostatins are hydrophobic tetrapeptide aldehydes from streptomycetes, e.g., S. lavendulae and S. hygroscopicus, that reverse chymosin-mediated angiotensin activation and inhibit other serine and cysteine proteases. Chymotrypsin and papain were both inhibited by the SPIac compounds in the low nanomolar range. SPIac differs from the characterized chymostatins by the exchange of phenylalanine for tyrosine. The crystal structure of one of these chymostatin variants confirmed its molecular structure and revealed a S-configured hemithioacetal bond with the catalytic Cys25 thiolate as well as close interactions with hydrophobic S1 and S2 subsite amino acids. A model for chymostatin biosynthesis is provided based on the discovery of clustered genes encoding several putative nonribosomal peptide synthetases; among them, there is the unusual CstF enzyme that accommodates two canonical amino acid activation domains as well as three peptide carrier protein domains.
Collapse
Affiliation(s)
- Norbert E Juettner
- The Department of Chemical Engineering and Biotechnology, University of Applied Sciences of Darmstadt, Stephanstraße 7, 64295 Darmstadt, Germany
- The Department of Biology, Technische Universität Darmstadt, Schnittspahnstraße 10, 64287 Darmstadt, Germany
| | - Jan P Bogen
- The Department of Chemical Engineering and Biotechnology, University of Applied Sciences of Darmstadt, Stephanstraße 7, 64295 Darmstadt, Germany
| | - Tobias A Bauer
- The Department of Chemical Engineering and Biotechnology, University of Applied Sciences of Darmstadt, Stephanstraße 7, 64295 Darmstadt, Germany
| | - Stefan Knapp
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Straße 9, 60438 Frankfurt am Main, Germany
- Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences (BMLS), Goethe University Frankfurt, Max-von-Laue-Straße 15, 60438 Frankfurt am Main, Germany
| | - Felicitas Pfeifer
- The Department of Biology, Technische Universität Darmstadt, Schnittspahnstraße 10, 64287 Darmstadt, Germany
| | - Stefan H Huettenhain
- The Department of Chemical Engineering and Biotechnology, University of Applied Sciences of Darmstadt, Stephanstraße 7, 64295 Darmstadt, Germany
| | - Reinhard Meusinger
- The Department of Chemistry, Technische Universität Darmstadt, Alarich-Weiss-Straße 8, 64287 Darmstadt, Germany
| | - Andreas Kraemer
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Straße 9, 60438 Frankfurt am Main, Germany
| | - Hans-Lothar Fuchsbauer
- The Department of Chemical Engineering and Biotechnology, University of Applied Sciences of Darmstadt, Stephanstraße 7, 64295 Darmstadt, Germany
| |
Collapse
|
24
|
Modelling frontotemporal dementia using patient-derived induced pluripotent stem cells. Mol Cell Neurosci 2020; 109:103553. [PMID: 32956830 DOI: 10.1016/j.mcn.2020.103553] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 08/27/2020] [Accepted: 09/12/2020] [Indexed: 12/12/2022] Open
Abstract
Frontotemporal dementia (FTD) describes a group of clinically heterogeneous conditions that frequently affect people under the age of 65 (Le Ber et al., 2013). There are multiple genetic causes of FTD, including coding or splice-site mutations in MAPT, GRN mutations that lead to haploinsufficiency of progranulin protein, and a hexanucleotide GGGGCC repeat expansion in C9ORF72. Pathologically, FTD is characterised by abnormal protein accumulations in neurons and glia. These aggregates can be composed of the microtubule-associated protein tau (observed in FTD with MAPT mutations), the DNA/RNA-binding protein TDP-43 (seen in FTD with mutations in GRN or C9ORF72 repeat expansions) or dipeptide proteins generated by repeat associated non-ATG translation of the C9ORF72 repeat expansion. There are currently no disease-modifying therapies for FTD and the availability of in vitro models that recapitulate pathologies in a disease-relevant cell type would accelerate the development of novel therapeutics. It is now possible to generate patient-specific stem cells through the reprogramming of somatic cells from a patient with a genotype/phenotype of interest into induced pluripotent stem cells (iPSCs). iPSCs can subsequently be differentiated into a plethora of cell types including neurons, astrocytes and microglia. Using this approach has allowed researchers to generate in vitro models of genetic FTD in human cell types that are largely inaccessible during life. In this review we explore the recent progress in the use of iPSCs to model FTD, and consider the merits, limitations and future prospects of this approach.
Collapse
|
25
|
Wang T, Gao X, Zhou K, Jiang T, Gao S, Liu P, Zuo X, Shi X. Role of ARID1A in epithelial‑mesenchymal transition in breast cancer and its effect on cell sensitivity to 5‑FU. Int J Mol Med 2020; 46:1683-1694. [PMID: 33000179 PMCID: PMC7521577 DOI: 10.3892/ijmm.2020.4727] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 07/21/2020] [Indexed: 01/01/2023] Open
Abstract
The loss of function mutation of AT‑rich interactive domain 1A (ARID1A) often occurs in patients with breast cancer. It has been found that ARID1A knockout can enhance both the migratory activity of renal carcinoma cells and their sensitivity to therapeutic drugs by promoting epithelial-mesenchymal transition (EMT); however, its mechanisms of action in breast cancer remain unclear. In the present study, immunohistochemistry and reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR) revealed that the expression of ARID1A in breast cancer tissues was significantly lower than that in paracancerous tissues, and patients with a low ARID1A expression had a lower survival rate. ARID1A was expressed at low levels in breast cancer cells. In addition, siRNA targeting ARID1A (siARID1A) and ARID1A overexpression vector were transfected into MCF7 and MDA‑MB‑231 cells, respectively. Proliferation assay revealed that ARID1A silencing increased cell viability and partially reversed the inhibitory effects of 5‑fluorouracil (5‑FU) on the MCF7 cells, while ARID1A overexpression exerted an opposite effect on the MDA‑MB‑231 cells. ARID1A silencing promoted proliferation, migration, invasion and angiogenesis, and partly reversed the inhibitory effects of 5‑FU on cell biological behaviors, while the overexpression of ARID1A further enhanced the inhibitory effect of 5‑FU on the cells. Furthermore, ARID1A regulated the migration and invasion of breast cancer cells through EMT. On the whole, the findings of the present study demonstrate that ARID1A exerts an antitumor effect on breast cancer, and its overexpression can enhance the sensitivity of breast cancer cells to 5‑FU.
Collapse
Affiliation(s)
- Tangshun Wang
- Department of General Surgery, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, P.R. China
| | - Xiang Gao
- Department of General Surgery, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, P.R. China
| | - Kexin Zhou
- Department of General Surgery, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, P.R. China
| | - Tao Jiang
- Department of General Surgery, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, P.R. China
| | - Shuang Gao
- Department of General Surgery, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, P.R. China
| | - Pengzhou Liu
- Department of General Surgery, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, P.R. China
| | - Ximeng Zuo
- Department of General Surgery, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, P.R. China
| | - Xiaoguang Shi
- Department of General Surgery, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, P.R. China
| |
Collapse
|
26
|
Mishra M. Evolutionary Aspects of the Structural Convergence and Functional Diversification of Kunitz-Domain Inhibitors. J Mol Evol 2020; 88:537-548. [PMID: 32696206 DOI: 10.1007/s00239-020-09959-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 07/04/2020] [Indexed: 11/28/2022]
Abstract
Kunitz-type domains are ubiquitously found in natural systems as serine protease inhibitors or animal toxins in venomous animals. Kunitz motif is a cysteine-rich peptide chain of ~ 60 amino acid residues with alpha and beta fold, stabilized by three conserved disulfide bridges. An extensive dataset of amino acid variations is found on sequence analysis of various Kunitz peptides. Kunitz peptides show diverse biological activities like inhibition of proteases of other classes and/or adopting a new function of blocking or modulating the ion channels. Based on the amino acid residues at the functional site of various Kunitz-type inhibitors, it is inferred that this 'flexibility within the structural rigidity' is responsible for multiple biological activities. Accelerated evolution of functional sites in response to the co-evolving molecular targets of the hosts of venomous animals or parasites, gene sharing, and gene duplication have been discussed as the most likely mechanisms responsible for the functional heterogeneity of Kunitz-domain inhibitors.
Collapse
Affiliation(s)
- Manasi Mishra
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Gautam Buddha Nagar, Uttar Pradesh, 201314, India.
| |
Collapse
|
27
|
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
|
28
|
Regensburger M, Minakaki G, Kettwig M, Huchzermeyer C, Eisenhut F, Haack TB, Kohl Z, Winkler J. Novel Biallelic
CTSD
Gene Variants Cause Late‐Onset Ataxia and Retinitis Pigmentosa. Mov Disord 2020; 35:1280-1282. [DOI: 10.1002/mds.28106] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 04/09/2020] [Indexed: 01/01/2023] Open
Affiliation(s)
- Martin Regensburger
- Department of Molecular NeurologyFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU) Erlangen Germany
- Department of NeurologyFAU Erlangen Germany
- Department of Stem Cell BiologyFAU Erlangen Germany
| | - Georgia Minakaki
- Department of Molecular NeurologyFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU) Erlangen Germany
| | - Matthias Kettwig
- Department of Pediatrics and Pediatric NeurologyUniversity Medical Center Göttingen, Georg August University Göttingen Göttingen Germany
| | | | | | - Tobias B. Haack
- Institute of Medical Genetics and Applied GenomicsUniversity of Tübingen Tübingen Germany
| | - Zacharias Kohl
- Department of Molecular NeurologyFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU) Erlangen Germany
- Department of NeurologyUniversity of Regensburg Regensburg Germany
| | - Jürgen Winkler
- Department of Molecular NeurologyFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU) Erlangen Germany
| |
Collapse
|
29
|
De Pasquale V, Moles A, Pavone LM. Cathepsins in the Pathophysiology of Mucopolysaccharidoses: New Perspectives for Therapy. Cells 2020; 9:cells9040979. [PMID: 32326609 PMCID: PMC7227001 DOI: 10.3390/cells9040979] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/13/2020] [Accepted: 04/14/2020] [Indexed: 02/07/2023] Open
Abstract
Cathepsins (CTSs) are ubiquitously expressed proteases normally found in the endolysosomal compartment where they mediate protein degradation and turnover. However, CTSs are also found in the cytoplasm, nucleus, and extracellular matrix where they actively participate in cell signaling, protein processing, and trafficking through the plasma and nuclear membranes and between intracellular organelles. Dysregulation in CTS expression and/or activity disrupts cellular homeostasis, thus contributing to many human diseases, including inflammatory and cardiovascular diseases, neurodegenerative disorders, diabetes, obesity, cancer, kidney dysfunction, and others. This review aimed to highlight the involvement of CTSs in inherited lysosomal storage disorders, with a primary focus to the emerging evidence on the role of CTSs in the pathophysiology of Mucopolysaccharidoses (MPSs). These latter diseases are characterized by severe neurological, skeletal and cardiovascular phenotypes, and no effective cure exists to date. The advance in the knowledge of the molecular mechanisms underlying the activity of CTSs in MPSs may open a new challenge for the development of novel therapeutic approaches for the cure of such intractable diseases.
Collapse
Affiliation(s)
- Valeria De Pasquale
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131 Naples, Italy;
| | - Anna Moles
- Institute of Biomedical Research of Barcelona, Spanish Research Council, 08036 Barcelona, Spain;
| | - Luigi Michele Pavone
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131 Naples, Italy;
- Correspondence: ; Tel.: +39-081-7463043
| |
Collapse
|
30
|
Bojarski KK, Karczyńska AS, Samsonov SA. Role of Glycosaminoglycans in Procathepsin B Maturation: Molecular Mechanism Elucidated by a Computational Study. J Chem Inf Model 2020; 60:2247-2256. [PMID: 32155059 PMCID: PMC7588040 DOI: 10.1021/acs.jcim.0c00023] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
![]()
Procathepsins
are an inactive, immature form of cathepsins, predominantly
cysteine proteases present in the extracellular matrix (ECM) and in
lysosomes that play a key role in various biological processes such
as bone resorption or intracellular proteolysis. The enzymatic activity
of cathepsins can be mediated by glycosaminoglycans (GAGs), long unbranched
periodic negatively charged polysaccharides found in ECM that take
part in many biological processes such as anticoagulation, angiogenesis,
and tissue regeneration. In addition to the known effects on mature
cathepsins, GAGs can mediate the maturation process of procathepsins,
in particular, procathepsin B. However, the detailed mechanism of
this mediation at the molecular level is still unknown. In this study,
for the first time, we aimed to unravel the role of GAGs in this process
using computational approaches. We rigorously analyzed procathepsin
B–GAG complexes in terms of their dynamics, energetics, and
potential allosteric regulation. We revealed that GAGs can stabilize
the conformation of the procathepsin B structure with the active site
accessible for the substrate and concluded that GAGs most probably
bind to procathepsin B once the zymogen adopts the enzymatically active
conformation. Our data provided a novel mechanistic view of the maturation
process of procathepsin B, while the approaches elaborated here might
be useful to study other procathepsins. Furthermore, our data can
serve as a rational guide for experimental work on procathepsin–GAG
systems that are not characterized in vivo and in vitro yet.
Collapse
Affiliation(s)
- Krzysztof K Bojarski
- Faculty of Chemistry, University of Gdańsk, Wita Stwosza 63, 80-308 Gdańsk, Poland
| | | | - Sergey A Samsonov
- Faculty of Chemistry, University of Gdańsk, Wita Stwosza 63, 80-308 Gdańsk, Poland
| |
Collapse
|
31
|
Eftekhariyazdi M, Meshkani M, Moslem A, Hakimi P, Safari S, Khaligh A, Zare-Abdollahi D. Ellis-van Creveld syndrome: Report of a case and recurrent variant. J Gene Med 2020; 22:e3175. [PMID: 32072716 DOI: 10.1002/jgm.3175] [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] [Received: 05/16/2019] [Revised: 01/06/2020] [Accepted: 01/28/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Ellis-van Creveld syndrome (EvCS) is a rare autosomal recessive skeletal dysplasia that is characterized by short stature, short limbs, short ribs, polydactyly and structural heart defect. Despite locus heterogeneity, in the majority of the cases, the disorder segregates with mutations in the EVC and EVC2 genes, notably mutations with truncating protein as a final sequence. In the present study, we report the prenatal findings and genetic analysis of a terminated pregnancy affected by severe thoracic and skeletal dysplasia. METHODS After detailed physical and clinical examination, whole exome sequencing (WES) was performed and the variant was confirmed by Sanger sequencing. RESULTS One homozygote variant in EVC2 gene was identified in the fetus (NM_147127, c.942G>A, p.W314X). The EVC2 gene is strongly associated with EvCS, which is consistent with the sonographic findings of the fetus. CONCLUSIONS The homozygous p.W314X mutation found in this family was recently reported to be segregated in a consanguineous family originating from Pakistan. The occurrence of the p.W314X mutation in two unrelated families (Iranian and Pakistani) may be the result of an old founder effect or arose because of a mutational hotspot and is supporting evidence for the pathogenicity of this variant. Because skeletal dysplasia belongs to a broad spectrum of syndromes and therefore exhibits considerable background locus and allelic heterogeneity, our report highlights the need for appropriate genetic counseling and supports the feasibility of WES to determine an accurate diagnosis, as well as precise recurrence risk prediction.
Collapse
Affiliation(s)
- Mitra Eftekhariyazdi
- Department of Obstetrics and Gynecology, School of Medicine, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Mahshid Meshkani
- Department of Genetics, Faculty of Biological sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Pooria Hakimi
- Department of Biology, Faculty of science, Islamic azad University, Neyshabour, Iran
| | - Shamsi Safari
- Department of Medical Genetics, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Khaligh
- Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Davood Zare-Abdollahi
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| |
Collapse
|
32
|
Foucher JR, Gawlik M, Roth JN, de Crespin de Billy C, Jeanjean LC, Obrecht A, Mainberger O, Clauss JME, Elowe J, Weibel S, Schorr B, Cetkovich M, Morra C, Rebok F, Ban TA, Bollmann B, Roser MM, Hanke MS, Jabs BE, Franzek EJ, Berna F, Pfuhlmann B. Wernicke-Kleist-Leonhard phenotypes
of endogenous psychoses: a review of their validity
. DIALOGUES IN CLINICAL NEUROSCIENCE 2020; 22:37-49. [PMID: 32699504 PMCID: PMC7365293 DOI: 10.31887/dcns.2020.22.1/jfoucher] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
While the ICD-DSM paradigm has been a major advance in clinical psychiatry, its usefulness for biological psychiatry is debated. By defining consensus-based disorders rather than empirically driven phenotypes, consensus classifications were not an implementation of the biomedical paradigm. In the field of endogenous psychoses, the Wernicke-Kleist-Leonhard (WKL) pathway has optimized the descriptions of 35 major phenotypes using common medical heuristics on lifelong diachronic observations. Regarding their construct validity, WKL phenotypes have good reliability and predictive and face validity. WKL phenotypes come with remarkable evidence for differential validity on age of onset, familiality, pregnancy complications, precipitating factors, and treatment response. Most impressive is the replicated separation of high- and low-familiality phenotypes. Created in the purest tradition of the biomedical paradigm, the WKL phenotypes deserve to be contrasted as credible alternatives with other approaches currently under discussion.
.
Collapse
Affiliation(s)
- Jack R Foucher
- ICube - CNRS UMR 7357, neurophysiology, FMTS, University of Strasbourg, France ; CEMNIS - Noninvasive Neuromodulation Center, University Hospital Strasbourg, France
| | - Micha Gawlik
- Department of Psychiatry and Psychotherapy, University of Würzburg, Würzburg, Germany
| | - Julian N Roth
- Department of Psychiatry and Psychotherapy, University of Würzburg, Würzburg, Germany
| | - Clément de Crespin de Billy
- ICube - CNRS UMR 7357, neurophysiology, FMTS, University of Strasbourg, France; CEMNIS - Noninvasive Neuromodulation Center, University Hospital Strasbourg, France
| | - Ludovic C Jeanjean
- IICube - CNRS UMR 7357, neurophysiology, FMTS, University of Strasbourg, France; CEMNIS - Noninvasive Neuromodulation Center, University Hospital Strasbourg, France
| | - Alexandre Obrecht
- ICube - CNRS UMR 7357, neurophysiology, FMTS, University of Strasbourg, France; Pôle de Psychiatrie, Santé Mentale et Addictologie, University Hospital Strasbourg, France
| | - Olivier Mainberger
- ICube - CNRS UMR 7357, neurophysiology, FMTS, University of Strasbourg, France. CEMNIS - Noninvasive Neuromodulation Center, University Hospital Strasbourg, France
| | - Julie M E Clauss
- Pôle de Psychiatrie, Santé Mentale et Addictologie, University Hospital Strasbourg, France. SAGE - CNRS UMR 7363, FMTS, University of Strasbourg, France
| | - Julien Elowe
- Department of Psychiatry, Prangins Psychiatric Hospital (CHUV), Route de Benex, Prangins, Switzerland
| | - Sébastien Weibel
- IPôle de Psychiatrie, Santé Mentale et Addictologie, University Hospital Strasbourg, France; Physiopathologie et Psychopathologie Cognitive de la Schizophrénie - INSERM 1114, FMTS, University of Strasbourg, France
| | - Benoit Schorr
- Pôle de Psychiatrie, Santé Mentale et Addictologie, University Hospital Strasbourg, France; Physiopathologie et Psychopathologie Cognitive de la Schizophrénie - INSERM 1114, FMTS, University of Strasbourg, France
| | - Marcelo Cetkovich
- Institute of Translational and Cognitive Neuroscience (INCyT), INECO Foundation, Favaloro University, Buenos Aires, Argentina; National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| | - Carlos Morra
- ICube - CNRS UMR 7357, neurophysiology, FMTS, University of StInstitute of Translational and Cognitive Neuroscience (INCyT), INECO Foundation, Favaloro University, Buenos Aires, Argentina; National Scientific and Technical Research Council (CONICET), Sanatorio Morra, Córdoba, Argentina
| | - Federico Rebok
- "Servicio de Emergencia", Acute Inpatient Unit, Moyano Neuropsychiatric Hospital, Buenos Aires, Argentina
| | - Thomas A Ban
- International Network for the History of Neuropsychopharmacology (INHN), Córdoba, Argentina
| | - Barbara Bollmann
- Klinik für Psychiatrie, Psychotherapie und Psychosomatik, Berlin, Germany
| | - Mathilde M Roser
- Department of Psychiatry, Mondor Hospital France, Creteil, France
| | - Markus S Hanke
- Universitäre psychiatrische Dienste Bern, Spiez, Switzerland
| | - Burkhard E Jabs
- Klinik für Psychiatrie and Psychotherapie, Städtisches Klinikum Dresden, Dresden, Germany
| | - Ernst J Franzek
- Yes We Can Clinics, Department of Research and Development, Eindhoven, The Netherlands
| | - Fabrice Berna
- Department of Psychiatry and Psychotherapy, University of Würzburg, Würzburg, Germany; Department of Psychiatry, Prangins Psychiatric Hospital (CHUV), Route de Benex, Prangins, Switzerland
| | - Bruno Pfuhlmann
- IKlinik für Psychiatrie and Psychotherapie, Städtisches Klinikum Dresden, Dresden, Germany
| |
Collapse
|
33
|
Mitschke J, Burk UC, Reinheckel T. The role of proteases in epithelial-to-mesenchymal cell transitions in cancer. Cancer Metastasis Rev 2020; 38:431-444. [PMID: 31482486 DOI: 10.1007/s10555-019-09808-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Changing the characteristics of cells from epithelial states to mesenchymal properties is a key process involved in developmental and physiological processes as well as in many diseases with cancer as the most prominent example. Nowadays, a great deal of work and literature concerns the understanding of the process of epithelial-to-mesenchymal transition (EMT) in terms of its molecular regulation and its implications for cancer. Similar statements can certainly be made regarding the investigation of the more than 500 proteases typically encoded by a mammalian genome. Specifically, the impact of proteases on tumor biology has been a long-standing topic of interest. However, although EMT actively regulates expression of many proteases and proteolytic enzymes are clearly involved in survival, division, differentiation, and movements of cells, information on the diverse roles of proteases in EMT has been rarely compiled. Here we aim to conceptually connect the scientific areas of "EMT" and "protease" research by describing how several important classes of proteolytic enzymes are regulated by EMT and how they are involved in initiation and execution of the EMT program. To do so, we briefly introduce the evolving key features of EMT and its regulation followed by discussion of protease involvement in this process.
Collapse
Affiliation(s)
- Julia Mitschke
- Institute of Molecular Medicine and Cell Research, University of Freiburg, 79104, Freiburg, Germany
| | - Ulrike C Burk
- Institute of Molecular Medicine and Cell Research, University of Freiburg, 79104, Freiburg, Germany
| | - Thomas Reinheckel
- Institute of Molecular Medicine and Cell Research, University of Freiburg, 79104, Freiburg, Germany. .,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) Heidelberg, partner site Freiburg, 79106, Freiburg, Germany. .,BIOSS Centre for Biological Signalling Studies, University of Freiburg, 79104, Freiburg, Germany.
| |
Collapse
|
34
|
Ding L, Goossens GH, Oligschlaeger Y, Houben T, Blaak EE, Shiri-Sverdlov R. Plasma cathepsin D activity is negatively associated with hepatic insulin sensitivity in overweight and obese humans. Diabetologia 2020; 63:374-384. [PMID: 31690989 PMCID: PMC6946744 DOI: 10.1007/s00125-019-05025-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 09/13/2019] [Indexed: 01/19/2023]
Abstract
AIMS/HYPOTHESIS Insulin resistance in skeletal muscle and liver plays a major role in the pathophysiology of type 2 diabetes. The hyperinsulinaemic-euglycaemic clamp is considered the gold standard for assessing peripheral and hepatic insulin sensitivity, yet it is a costly and labour-intensive procedure. Therefore, easy-to-measure, cost-effective approaches to determine insulin sensitivity are needed to enable organ-specific interventions. Recently, evidence emerged that plasma cathepsin D (CTSD) is associated with insulin sensitivity and hepatic inflammation. Here, we aimed to investigate whether plasma CTSD is associated with hepatic and/or peripheral insulin sensitivity in humans. METHODS As part of two large clinical trials (one designed to investigate the effects of antibiotics, and the other to investigate polyphenol supplementation, on insulin sensitivity), 94 overweight and obese adults (BMI 25-35 kg/m2) previously underwent a two-step hyperinsulinaemic-euglycaemic clamp (using [6,6-2H2]glucose) to assess hepatic and peripheral insulin sensitivity (per cent suppression of endogenous glucose output during the low-insulin-infusion step, and the rate of glucose disappearance during high-insulin infusion [40 mU/(m2 × min)], respectively). In this secondary analysis, plasma CTSD levels, CTSD activity and plasma inflammatory cytokines were measured. RESULTS Plasma CTSD levels were positively associated with the proinflammatory cytokines IL-8 and TNF-α (IL-8: standardised β = 0.495, p < 0.001; TNF-α: standardised β = 0.264, p = 0.012). Plasma CTSD activity was negatively associated with hepatic insulin sensitivity (standardised β = -0.206, p = 0.043), independent of age, sex, BMI and waist circumference, but it was not associated with peripheral insulin sensitivity. However, plasma IL-8 and TNF-α were not significantly correlated with hepatic insulin sensitivity. CONCLUSIONS/INTERPRETATION We demonstrate that plasma CTSD activity, but not systemic inflammation, is inversely related to hepatic insulin sensitivity, suggesting that plasma CTSD activity may be used as a non-invasive marker for hepatic insulin sensitivity in humans.
Collapse
Affiliation(s)
- Lingling Ding
- Department of Molecular Genetics, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands
| | - Gijs H Goossens
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands
| | - Yvonne Oligschlaeger
- Department of Molecular Genetics, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands
| | - Tom Houben
- Department of Molecular Genetics, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands
| | - Ellen E Blaak
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands.
| | - Ronit Shiri-Sverdlov
- Department of Molecular Genetics, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands.
| |
Collapse
|
35
|
Hepp N, Frederiksen AL, Dunø M, Jørgensen NR, Langdahl B, Vedtofte P, Hove HB, Hindsø K, Jensen JEB. Multiple Fractures and Impaired Bone Fracture Healing in a Patient with Pycnodysostosis and Hypophosphatasia. Calcif Tissue Int 2019; 105:681-686. [PMID: 31489468 DOI: 10.1007/s00223-019-00605-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 08/21/2019] [Indexed: 02/07/2023]
Abstract
Pycnodysostosis (PYCD) is a rare recessive inherited skeletal disease, characterized by short stature, brittle bones, and recurrent fractures, caused by variants in the Cathepsin K encoding gene that leads to impaired osteoclast-mediated bone resorption. Hypophosphatasia (HPP) is a dominant or recessive inherited condition representing a heterogeneous phenotype with dental symptoms, recurrent fractures, and musculoskeletal problems. The disease results from mutation(s) in the tissue non-specific alkaline phosphate encoding gene with reduced activity of alkaline phosphatase and secondarily defective mineralization of bone and teeth. Here, we present the first report of a patient with the coexistence of PYCD and HPP. This patient presented typical clinical findings of PYCD, including short stature, maxillary hypoplasia, and sleep apnoea. However, the burden of disease was caused by over 30 fractures, whereupon most showed delayed healing and non-union. Biochemical analysis revealed suppressed bone resorption and low bone formation capacity. We suggest that the coexistence of impaired bone resorption and mineralization may explain the severe bone phenotype with poor fracture healing.
Collapse
Affiliation(s)
- Nicola Hepp
- Department of Endocrinology, Hvidovre University Hospital Copenhagen, Kettegård Alle 30, 2650, Hvidovre, Denmark.
| | - Anja Lisbeth Frederiksen
- Department of Clinical Genetics, Odense University Hospital, Winsløws Vej 4, 5000, Odense C, Denmark
- Department of Clinical Research, Faculty of Health, University of Southern Denmark, Winsløwparken 19. 3, 5000, Odense C, Denmark
| | - Morten Dunø
- Department of Clinical Genetics, University Hospital Copenhagen Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Niklas Rye Jørgensen
- Department of Clinical Biochemistry, Rigshospitalet, Valdemar Hansens Vej 13, 2600, Glostrup, Denmark
- OPEN, Odense Patient Data Explorative Network, Odense University Hospital/Institute of Clinical Research, University of Southern Denmark, J.B.Winsløws Vej 9, 5000, Odense C, Denmark
| | - Bente Langdahl
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Palle Juul Jensens Boulevard 99, G317, 8200, Aarhus N, Denmark
| | - Poul Vedtofte
- Department of Oral and Maxillofacial Surgery, University Hospital Copenhagen Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Hanne B Hove
- Center for Rare Diseases, Department of Pediatrics, University Hospital Copenhagen, Juliane Maries Vej 6, 2100, Copenhagen, Denmark
| | - Klaus Hindsø
- Paediatric Section, Department of Orthopedic Surgery, University Hospital Copenhagen Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Jens-Erik Beck Jensen
- Department of Endocrinology, Hvidovre University Hospital Copenhagen, Kettegård Alle 30, 2650, Hvidovre, Denmark
- Department of Clinical Medicine, University of Copenhagen, Blegdamsvej 9, 2100, Copenhagen, Denmark
| |
Collapse
|
36
|
Wallings RL, Humble SW, Ward ME, Wade-Martins R. Lysosomal Dysfunction at the Centre of Parkinson's Disease and Frontotemporal Dementia/Amyotrophic Lateral Sclerosis. Trends Neurosci 2019; 42:899-912. [PMID: 31704179 DOI: 10.1016/j.tins.2019.10.002] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 09/25/2019] [Accepted: 10/04/2019] [Indexed: 12/14/2022]
Abstract
Parkinson's disease (PD) and frontotemporal dementia/amyotrophic lateral sclerosis (FTD/ALS) are insidious and incurable neurodegenerative diseases that represent a significant burden to affected individuals, caregivers, and an ageing population. Both PD and FTD/ALS are defined at post mortem by the presence of protein aggregates and the loss of specific subsets of neurons. We examine here the crucial role of lysosome dysfunction in these diseases and discuss recent evidence for converging mechanisms. This review draws upon multiple lines of evidence from genetic studies, human tissue, induced pluripotent stem cells (iPSCs), and animal models to argue that lysosomal failure is a primary mechanism of disease, rather than merely reflecting association with protein aggregate end-points. This review provides compelling rationale for targeting lysosomes in future therapeutics for both PD and FTD/ALS.
Collapse
Affiliation(s)
- Rebecca L Wallings
- Department of Physiology, Emory University, Decatur, GA, USA; Current address: Department of Neuroscience, Center for Translational Research and Neurodegenerative Disease, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Stewart W Humble
- Oxford Parkinson's Disease Centre, Department of Physiology Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, OX1 3QX, UK; National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Michael E Ward
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Richard Wade-Martins
- Oxford Parkinson's Disease Centre, Department of Physiology Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, OX1 3QX, UK.
| |
Collapse
|
37
|
Impact of proteolysis on cancer stem cell functions. Biochimie 2019; 166:214-222. [DOI: 10.1016/j.biochi.2019.03.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 03/07/2019] [Indexed: 12/14/2022]
|
38
|
Flanagan-Steet H, Christian C, Lu PN, Aarnio-Peterson M, Sanman L, Archer-Hartmann S, Azadi P, Bogyo M, Steet RA. TGF-ß Regulates Cathepsin Activation during Normal and Pathogenic Development. Cell Rep 2019. [PMID: 29539424 PMCID: PMC6247414 DOI: 10.1016/j.celrep.2018.02.066] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Cysteine cathepsins play roles during development and disease beyond their function in lysosomal protein turnover. Here, we leverage a fluorescent activity-based probe (ABP), BMV109, to track cysteine cathepsins in normal and diseased zebrafish embryos. Using this probe in a model of mucolipidosis II, we show that loss of carbohydrate-dependent lysosomal sorting alters the activity of several cathepsin proteases. The data support a pathogenic mechanism where TGF-β signals enhance the proteolytic processing of pro-Ctsk by modulating the expression of chondroitin 4-sulfate (C4-S). In MLII, elevated C4-S corresponds with TGF-β-mediated increases in chst11 expression. Inhibiting chst11 impairs the proteolytic activation of Ctsk and alleviates the MLII phenotypes. These findings uncover a regulatory loop between TGF-β signaling and Ctsk activation that is altered in the context of lysosomal disease. This work highlights the power of ABPs to identify mechanisms underlying pathogenic development in living animals. Chondroitin sulfate is a known regulator of cathepsin protease activity. Flanagan-Steet et al. identify a positive feedback mechanism whereby cathepsins secreted from chondrocytes upon loss of lysosomal targeting activate TGF-β signaling in developing cartilage. This increased signaling, in turn, stimulates chondroitin-4 sulfation and enhances cathepsin activity.
Collapse
Affiliation(s)
| | - Courtney Christian
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30606, USA
| | - Po-Nien Lu
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30606, USA
| | | | - Laura Sanman
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305-5324, USA
| | | | - Parastoo Azadi
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30606, USA
| | - Matthew Bogyo
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305-5324, USA
| | - Richard A Steet
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30606, USA
| |
Collapse
|
39
|
Association between Lysosomal Dysfunction and Obesity-Related Pathology: A Key Knowledge to Prevent Metabolic Syndrome. Int J Mol Sci 2019; 20:ijms20153688. [PMID: 31357643 PMCID: PMC6696452 DOI: 10.3390/ijms20153688] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 07/25/2019] [Accepted: 07/26/2019] [Indexed: 02/06/2023] Open
Abstract
Obesity causes various health problems, such as type 2 diabetes, non-alcoholic fatty liver disease, and cardio- and cerebrovascular diseases. Metabolic organs, particularly white adipose tissue (WAT) and liver, are deeply involved in obesity. WAT contains many adipocytes with energy storage capacity and secretes adipokines depending on the obesity state, while liver plays pivotal roles in glucose and lipid metabolism. This review outlines and underscores the relationship between obesity and lysosomal functions, including lysosome biogenesis, maturation and activity of lysosomal proteases in WAT and liver. It has been revealed that obesity-induced abnormalities of lysosomal proteases contribute to inflammation and cellular senescence in adipocytes. Previous reports have demonstrated obesity-induced ectopic lipid accumulation in liver is associated with abnormality of lysosomal proteases as well as other lysosomal enzymes. These studies demonstrate that lysosomal dysfunction in WAT and liver underlies part of the obesity-related pathology, raising the possibility that strategies to modulate lysosomal function may be effective in preventing or treating the metabolic syndrome.
Collapse
|
40
|
Marques ARA, Di Spiezio A, Thießen N, Schmidt L, Grötzinger J, Lüllmann-Rauch R, Damme M, Storck SE, Pietrzik CU, Fogh J, Bär J, Mikhaylova M, Glatzel M, Bassal M, Bartsch U, Saftig P. Enzyme replacement therapy with recombinant pro-CTSD (cathepsin D) corrects defective proteolysis and autophagy in neuronal ceroid lipofuscinosis. Autophagy 2019; 16:811-825. [PMID: 31282275 DOI: 10.1080/15548627.2019.1637200] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
CTSD (cathepsin D) is one of the major lysosomal proteases indispensable for the maintenance of cellular proteostasis by turning over substrates of endocytosis, phagocytosis and autophagy. Consequently, CTSD deficiency leads to a strong impairment of the lysosomal-autophagy machinery. In mice and humans CTSD dysfunction underlies the congenital variant (CLN10) of neuronal ceroid lipofuscinosis (NCL). NCLs are distinct lysosomal storage disorders (LSDs) sharing various hallmarks, namely accumulation of protein aggregates and ceroid lipofuscin leading to neurodegeneration and blindness. The most established and clinically approved approach to treat LSDs is enzyme replacement therapy (ERT) aiming to replace the defective hydrolase with an exogenously applied recombinant protein. Here we reveal that recombinant human pro-CTSD produced in a mammalian expression system can be efficiently taken up by a variety of cell models, is correctly targeted to lysosomes and processed to the active mature form of the protease. In proof-of-principle experiments we provide evidence that recombinant human CTSD (rhCTSD) can improve the biochemical phenotype of CTSD-deficient hippocampal slice cultures in vitro and retinal cells in vivo. Furthermore, we demonstrate that dosing of rhCTSD in the murine CLN10 model leads to a correction of lysosomal hypertrophy, storage accumulation and impaired autophagic flux in the viscera and central nervous system (CNS). We establish that direct delivery of the recombinant protease to the CNS is required for improvement of neuropathology and lifespan extension. Together these data support the continuation of the pre-clinical studies for the application of rhCTSD in the treatment of NCL.Abbreviations: AIF1/IBA1: allograft inflammatory factor 1; BBB: blood brain barrier; CNS: central nervous system; CTSB: cathepsin B; CTSD: cathepsin D; CTSL: cathepsin L; ERT: enzyme replacement therapy; GFAP: glial fibrillary acidic protein; INL: inner nuclear layer; LAMP1: lysosomal-associated membrane protein 1; LAMP2: lysosomal-associated membrane protein 2; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; LDL: low-density lipoprotein; LRP1: low density lipoprotein receptor-related protein 1; LSD: lysosomal storage disorder; MEFs: mouse embryonic fibroblasts; M6P: mannose 6-phosphate; mCTSD: mature CTSD; NCL: neuronal ceroid lipofuscinosis; ONL: outer nuclear layer; PB: phosphate buffer; proCTSD: pro-cathepsin D; LRPAP1: low density lipoprotein receptor-related protein associated protein 1; rhCTSD: human recombinant CTSD; SAPC: saposin C; SAPD: saposin D; ATP5G1: ATP synthase, H+ transporting, mitochondrial F0 complex, subunit C1 (subunit 9); SQSTM1/p62: sequestosome 1; TPP1: tripeptidyl peptidase I.
Collapse
Affiliation(s)
- André R A Marques
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| | | | - Niklas Thießen
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Lina Schmidt
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Joachim Grötzinger
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| | | | - Markus Damme
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Steffen E Storck
- Institute for Pathobiochemistry, University Medical Center of Johannes Gutenberg University Mainz, Mainz, Germany
| | - Claus U Pietrzik
- Institute for Pathobiochemistry, University Medical Center of Johannes Gutenberg University Mainz, Mainz, Germany
| | | | - Julia Bär
- Center for Molecular Neurobiology, Emmy-Noether Group "Neuronal Protein Transport", ZMNH, University Medical Center, Hamburg, Germany
| | - Marina Mikhaylova
- Center for Molecular Neurobiology, Emmy-Noether Group "Neuronal Protein Transport", ZMNH, University Medical Center, Hamburg, Germany
| | - Markus Glatzel
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Mahmoud Bassal
- Department of Ophthalmology, Experimental Ophthalmology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Udo Bartsch
- Department of Ophthalmology, Experimental Ophthalmology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Paul Saftig
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| |
Collapse
|
41
|
Dahlgren C, Karlsson A, Bylund J. Intracellular Neutrophil Oxidants: From Laboratory Curiosity to Clinical Reality. THE JOURNAL OF IMMUNOLOGY 2019; 202:3127-3134. [DOI: 10.4049/jimmunol.1900235] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 03/25/2019] [Indexed: 12/13/2022]
|
42
|
Grewal S, Kilic Ö, Savci-Heijink CD, Kloen P. Disturbed remodeling and delayed fracture healing in pediatric pycnodysostosis patients. J Orthop 2019; 16:373-377. [PMID: 31048950 DOI: 10.1016/j.jor.2019.03.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 03/31/2019] [Indexed: 11/18/2022] Open
Abstract
Pycnodysostosis is an autosomal recessive disease caused by a gene mutation leading cathepsin K deficiency. Pathological fractures of the long bones are common, but guidelines on fracture treatment in these patients are still lacking. We have treated 5 fractures in 2 pediatric pycnodysostosis patients. We hypothesize that pycnodysostosis patients have an incomplete remodeling process in fracture healing because of cathepsin K deficiency. Therefore, to minimize the role of endochondral bone formation (indirect) after a fracture, it seems prudent to strive for direct bone healing (intramembranous) instead of indirect bone healing. Open reduction with internal fixation should be the goal.
Collapse
Affiliation(s)
- Simran Grewal
- Department of Orthopaedic Surgery, Amsterdam University Medical Centers, Amsterdam Movement Sciences, the Netherlands
| | - Özgür Kilic
- Department of Orthopaedic Surgery, Amsterdam University Medical Centers, Amsterdam Movement Sciences, the Netherlands
| | | | - Peter Kloen
- Department of Orthopaedic Surgery, Amsterdam University Medical Centers, Amsterdam Movement Sciences, the Netherlands
| |
Collapse
|
43
|
Biguanide is a modifiable pharmacophore for recruitment of endogenous Zn 2+ to inhibit cysteinyl cathepsins: review and implications. Biometals 2019; 32:575-593. [PMID: 31044334 PMCID: PMC6647370 DOI: 10.1007/s10534-019-00197-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 04/13/2019] [Indexed: 01/28/2023]
Abstract
Excessive activities of cysteinyl cathepsins (CysCts) contribute to the progress of many diseases; however, therapeutic inhibition has been problematic. Zn2+ is a natural inhibitor of proteases with CysHis dyads or CysHis(Xaa) triads. Biguanide forms bidentate metal complexes through the two imino nitrogens. Here, it is discussed that phenformin (phenylethyl biguanide) is a model for recruitment of endogenous Zn2+ to inhibit CysHis/CysHis(X) peptidolysis. Phenformin is a Zn2+-interactive, anti-proteolytic agent in bioassay of living tissue. Benzoyl-L-arginine amide (BAA) is a classical substrate of papain-like proteases; the amide bond is scissile. In this review, the structures of BAA and the phenformin-Zn2+ complex were compared in silico. Their chemistry and dimensions are discussed in light of the active sites of papain-like proteases. The phenyl moieties of both structures bind to the "S2" substrate-binding site that is typical of many proteases. When the phenyl moiety of BAA binds to S2, then the scissile amide bond is directed to the position of the thiolate-imidazolium ion pair, and is then hydrolyzed. However, when the phenyl moiety of phenformin binds to S2, then the coordinated Zn2+ is directed to the identical position; and catalysis is inhibited. Phenformin stabilizes a "Zn2+ sandwich" between the drug and protease active site. Hundreds of biguanide derivatives have been synthesized at the 1 and 5 nitrogen positions; many more are conceivable. Various substituent moieties can register with various arrays of substrate-binding sites so as to align coordinated Zn2+ with catalytic partners of diverse proteases. Biguanide is identified here as a modifiable pharmacophore for synthesis of therapeutic CysCt inhibitors with a wide range of potencies and specificities. Phenformin-Zn2+ Complex.
Collapse
|
44
|
Wartenberg M, Saidi A, Galibert M, Joulin-Giet A, Burlaud-Gaillard J, Lecaille F, Scott CJ, Aucagne V, Delmas AF, Lalmanach G. Imaging of extracellular cathepsin S activity by a selective near infrared fluorescence substrate-based probe. Biochimie 2019; 166:84-93. [PMID: 30914255 DOI: 10.1016/j.biochi.2019.03.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 03/19/2019] [Indexed: 02/05/2023]
Abstract
We designed a near-infrared fluorescent substrate-based probe (SBP), termed MG101, for monitoring extracellular cathepsin S (CatS) activity. We conceived a fused peptide hairpin loop-structure, combining a CatS recognition domain, an electrostatic zipper (with complementary charges of a polyanionic (D-Glu)5 segment and a polycationic (D-Arg)5 motif, as well as a N and C terminal Förster resonance energy transfer pair (donor: AlexaFluor680; quencher: BHQ3) to facilitate activity-dependent imaging. MG101 showed excellent stability since no fluorescence release corresponding to a self-dequenching was observed in the presence of either 2 M NaCl or after incubation at a broad range of pH (2.2-8.2). Cathepsins B, D, G, H, and K, neutrophil elastase and proteinase 3 did not cleave MG101, while CatS, and to a lesser extent CatL, hydrolysed MG101 at pH 5.5. However MG101 was fully selective for CatS at pH 7.4 (kcat/Km = 140,000 M-1 s-1) and sensitive to low concentration of CatS (<1 nM). The selectivity of MG101 was successfully endorsed ex vivo, as it was hydrolysed in cell lysates derived from wild-type but not knockout CatS murine spleen. Furthermore, application of the SBP probe with confocal microscopy confirmed the secretion of active CatS from THP-1 macrophages, which could be abrogated by pharmacological CatS inhibitors. Taken together, present data highlight MG101 as a novel near-infrared fluorescent SBP for the visualization of extracellular active CatS from macrophages and other cell types.
Collapse
Affiliation(s)
- Mylène Wartenberg
- Université de Tours, Tours, France; INSERM, UMR 1100, Research Center for Respiratory Diseases (CEPR), Team: "Proteolytic Mechanisms in Inflammation", Tours, France
| | - Ahlame Saidi
- Université de Tours, Tours, France; INSERM, UMR 1100, Research Center for Respiratory Diseases (CEPR), Team: "Proteolytic Mechanisms in Inflammation", Tours, France
| | - Mathieu Galibert
- CNRS UPR 4301, Center for Molecular Biophysics (CBM), Team: "Molecular, Structural and Chemical Biology", Orléans, France
| | - Alix Joulin-Giet
- Université de Tours, Tours, France; INSERM, UMR 1100, Research Center for Respiratory Diseases (CEPR), Team: "Proteolytic Mechanisms in Inflammation", Tours, France
| | - Julien Burlaud-Gaillard
- Université de Tours, Tours, France; Plateforme IBiSA de Microscopie Electronique, Université de Tours, Tours, France
| | - Fabien Lecaille
- Université de Tours, Tours, France; INSERM, UMR 1100, Research Center for Respiratory Diseases (CEPR), Team: "Proteolytic Mechanisms in Inflammation", Tours, France
| | - Christopher J Scott
- Centre for Cancer Research and Cell Biology, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Vincent Aucagne
- CNRS UPR 4301, Center for Molecular Biophysics (CBM), Team: "Molecular, Structural and Chemical Biology", Orléans, France
| | - Agnès F Delmas
- CNRS UPR 4301, Center for Molecular Biophysics (CBM), Team: "Molecular, Structural and Chemical Biology", Orléans, France
| | - Gilles Lalmanach
- Université de Tours, Tours, France; INSERM, UMR 1100, Research Center for Respiratory Diseases (CEPR), Team: "Proteolytic Mechanisms in Inflammation", Tours, France.
| |
Collapse
|
45
|
Parzych KR, Klionsky DJ. Vacuolar hydrolysis and efflux: current knowledge and unanswered questions. Autophagy 2018; 15:212-227. [PMID: 30422029 DOI: 10.1080/15548627.2018.1545821] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Hydrolysis within the vacuole in yeast and the lysosome in mammals is required for the degradation and recycling of a multitude of substrates, many of which are delivered to the vacuole/lysosome by autophagy. In humans, defects in lysosomal hydrolysis and efflux can have devastating consequences, and contribute to a class of diseases referred to as lysosomal storage disorders. Despite the importance of these processes, many of the proteins and regulatory mechanisms involved in hydrolysis and efflux are poorly understood. In this review, we describe our current knowledge of the vacuolar/lysosomal degradation and efflux of a vast array of substrates, focusing primarily on what is known in the yeast Saccharomyces cerevisiae. We also highlight many unanswered questions, the answers to which may lead to new advances in the treatment of lysosomal storage disorders. Abbreviations: Ams1: α-mannosidase; Ape1: aminopeptidase I; Ape3: aminopeptidase Y; Ape4: aspartyl aminopeptidase; Atg: autophagy related; Cps1: carboxypeptidase S; CTNS: cystinosin, lysosomal cystine transporter; CTSA: cathepsin A; CTSD: cathepsin D; Cvt: cytoplasm-to-vacuole targeting; Dap2: dipeptidyl aminopeptidase B; GS-bimane: glutathione-S-bimane; GSH: glutathione; LDs: lipid droplets; MVB: multivesicular body; PAS: phagophore assembly site; Pep4: proteinase A; PolyP: polyphosphate; Prb1: proteinase B; Prc1: carboxypeptidase Y; V-ATPase: vacuolar-type proton-translocating ATPase; VTC: vacuolar transporter chaperone.
Collapse
Affiliation(s)
- Katherine R Parzych
- a Life Sciences Institute, and Department of Molecular, Cellular and Developmental Biology , University of Michigan , Ann Arbor , MI , USA
| | - Daniel J Klionsky
- a Life Sciences Institute, and Department of Molecular, Cellular and Developmental Biology , University of Michigan , Ann Arbor , MI , USA
| |
Collapse
|
46
|
Yamazaki N, Kanazawa K, Kimura M, Ike H, Shinomiya M, Tanaka S, Shinohara Y, Minakawa N, Itoh K, Takiguchi Y. Use of modified U1 small nuclear RNA for rescue from exon 7 skipping caused by 5′-splice site mutation of human cathepsin A gene. Gene 2018; 677:41-48. [DOI: 10.1016/j.gene.2018.07.030] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Revised: 06/30/2018] [Accepted: 07/11/2018] [Indexed: 01/04/2023]
|
47
|
Korkmaz B, Caughey GH, Chapple I, Gauthier F, Hirschfeld J, Jenne DE, Kettritz R, Lalmanach G, Lamort AS, Lauritzen C, Łȩgowska M, Lesner A, Marchand-Adam S, McKaig SJ, Moss C, Pedersen J, Roberts H, Schreiber A, Seren S, Thakker NS. Therapeutic targeting of cathepsin C: from pathophysiology to treatment. Pharmacol Ther 2018; 190:202-236. [DOI: 10.1016/j.pharmthera.2018.05.011] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
48
|
Roy S, Das Chakraborty S, Biswas S. Not all pycnodysostosis-related mutants of human cathepsin K are inactive - crystal structure and biochemical studies of an active mutant I249T. FEBS J 2018; 285:4265-4280. [PMID: 30199612 DOI: 10.1111/febs.14655] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 08/21/2018] [Accepted: 09/07/2018] [Indexed: 12/18/2022]
Abstract
Human cathepsin K (CTSK) is a collagenolytic lysosomal cysteine protease that plays an important role in bone turnover. Mutation in CTSK gene is associated with loss of collagenolytic activity of CTSK leading to an autosomal recessive bone disorder called pycnodysostosis. Although a number of pycnodysostotic missense mutations have been reported, underlying mechanism of the disease is not clear. In this study, we investigated in vitro six recombinant pycnodysostosis-related mutants of human CTSK (G79E, I249T, G243E, G303E, G319C and Q187P). While all the mutants, like wild-type, show similar high levels of expression in Escherichia coli, four of them (G79E, G303E, G319C and Q187P) are inactive, unstable and spontaneously degrade during purification process. In contrast, proteolytic/collagenolytic activity, zymogen activation kinetics and stability of G243E and I249T mutants are nominally affected. Crystal structure of I249T at 1.92 Å resolution shows that the mutation in R-domain causes conformational changes of a surface loop in the L-domain although the catalytic cleft remains unaltered. Molecular simulation, normal mode analysis and fluorescence lifetime measurement eliminated the possibility that the change in L-domain surface loop orientation is a crystallization artefact. CD-based thermal melting profile indicates that stability of I249T is significantly higher than wild-type. Our studies first time reports that pycnodysostosis-related mutations do not always lead to complete loss of general proteolytic activity or specific collagenolytic activity of CTSK. The first crystal structure of a pycnodysostotic mutant (I249T) provides critical information that may pave new avenues towards understanding the disease at molecular level. DATABASE: The atomic co-ordinates and structure factors for I249T mutant of human CTSK (codes 5Z5O) have been deposited in the Protein Data Bank (http://wwpdb.org/).
Collapse
Affiliation(s)
- Sumana Roy
- Crystallography and Molecular Biology Division, Saha Institute of Nuclear Physics, Kolkata, India
| | | | - Sampa Biswas
- Crystallography and Molecular Biology Division, Saha Institute of Nuclear Physics, Kolkata, India.,Homi Bhaba National Institute, Mumbai, India
| |
Collapse
|
49
|
Araujo TF, Cordeiro AV, Vasconcelos DAA, Vitzel KF, Silva VRR. The role of cathepsin B in autophagy during obesity: A systematic review. Life Sci 2018; 209:274-281. [PMID: 30107168 DOI: 10.1016/j.lfs.2018.08.024] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 08/02/2018] [Accepted: 08/10/2018] [Indexed: 12/31/2022]
Abstract
White adipose tissue (WAT) regulates energy homeostasis by releasing adipokines and modulating cell maintenance. Nutrient excess affects adipocyte hypertrophy directly in WAT by increasing excessively the activity of autophagy systems, generating proinflammatory markers and increasing infiltration of macrophages, causing metabolic diseases such as obesity and diabetes. Evidences suggest that cathepsin B (CTSB), a papain-like cysteine peptidase protein, can modulate autophagy processes in adipocytes. This review will focus on the role of CTSB in autophagy under conditions of obesity.
Collapse
Affiliation(s)
- Thaís F Araujo
- University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | | | | | - Kaio F Vitzel
- School of Health Sciences, College of Health, Massey University, Auckland, New Zealand
| | | |
Collapse
|
50
|
Otaify GA, Abdel-Hamid MS, Mehrez MI, Aboul-Ezz E, Zaki MS, Aglan MS, Temtamy SA. Genetic study of eight Egyptian patients with pycnodysostosis: identification of novel CTSK mutations and founder effect. Osteoporos Int 2018; 29:1833-1841. [PMID: 29796728 DOI: 10.1007/s00198-018-4555-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 04/27/2018] [Indexed: 12/13/2022]
Abstract
UNLABELLED This is the first Egyptian study with detailed clinical and orodental evaluation of eight patients with pycnodysostosis and identification of four mutations in CTSK gene with two novel ones and a founder effect. INTRODUCTION Pycnodysostosis is a rare autosomal recessive skeletal dysplasia due to mutations in the CTSK gene encoding for cathepsin K, a lysosomal cysteine protease. METHODS We report on the clinical, orodental, radiological, and molecular findings of eight patients, from seven unrelated Egyptian families with pycnodysostosis. RESULTS All patients were offspring of consanguineous parents and presented with the typical clinical picture of the disorder including short stature, delayed closure of fontanels, hypoplastic premaxilla, obtuse mandibular angle, and drum stick terminal phalanges with dysplastic nails. Their radiological findings showed increased bone density, acro-osteolysis, and open cranial sutures. Mutational analysis of CTSK gene revealed four distinct homozygous missense mutations including two novel ones, c.164A>C (p. K55T) and c.433G>A (p.V145M). The c.164A>C (p. K55T) mutation was recurrent in three unrelated patients who also shared similar haplotype, suggesting a founder effect. CONCLUSION Our findings expand the mutational spectrum of CTSK gene and emphasize the importance of full clinical examination of all body systems including thorough orodental evaluation in patients with pycnodysostosis.
Collapse
Affiliation(s)
- G A Otaify
- Clinical Genetics Department, Human Genetics and Genome Research Division, Center of Excellence for Human Genetics, National Research Centre, Cairo, Egypt.
| | - M S Abdel-Hamid
- Medical Molecular Genetics Department, Human Genetics and Genome Research Division, National Research Centre, Cairo, Egypt
| | - M I Mehrez
- Orodental Genetics Department, Human Genetics and Genome Research Division, Center of Excellence for Human Genetics, National Research Centre, Cairo, Egypt
| | - E Aboul-Ezz
- Dental Basic Science Department, Dental and Oral Research Division, National Research Centre, Cairo, Egypt
| | - M S Zaki
- Clinical Genetics Department, Human Genetics and Genome Research Division, Center of Excellence for Human Genetics, National Research Centre, Cairo, Egypt
| | - M S Aglan
- Clinical Genetics Department, Human Genetics and Genome Research Division, Center of Excellence for Human Genetics, National Research Centre, Cairo, Egypt
| | - S A Temtamy
- Clinical Genetics Department, Human Genetics and Genome Research Division, Center of Excellence for Human Genetics, National Research Centre, Cairo, Egypt
| |
Collapse
|