1
|
El-Sabawi B, Tanriverdi K, Gajjar P, Nayor M, Landman JM, Below JE, Haff M, Long M, Ezpeleta M, Freedman JE, Varady K, Shah R, Perry AS. Circulating Proteomics Identifies a Dynamic Profile of Hepatic Steatosis During Metabolic Intervention. J Am Heart Assoc 2025; 14:e037100. [PMID: 40371575 DOI: 10.1161/jaha.124.037100] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 03/05/2025] [Indexed: 05/16/2025]
Abstract
BACKGROUND Weight reduction through lifestyle, activity, and dietary interventions are the mainstay of initial therapy for metabolic dysfunction associated steatotic liver disease. Data on the relative effectiveness and metabolic pathways linking weight loss and decreased hepatic steatosis are lacking. We sought to identify coordinated changes between the circulating proteome and hepatic steatosis within a randomized clinical trial of alternate day fasting and exercise and prioritize proteins relevant to hepatic steatosis within a broader context using a community cohort. METHODS AND RESULTS We quantified a broad cardiometabolic proteome (>300 proteins) in 67 individuals randomized in a 2×2 factorial design to alternate day fasting and exercise before and after the 3-month intervention to identify proteomic signatures of hepatic steatosis (measured by magnetic resonance imaging proton density fat fraction). Then, we analyzed the cross-sectional relationship of overlapping proteins (≈170) with hepatic attenuation (a computed tomographic technique linked to steatosis) in 707 participants from a community cohort. Principal component analysis demonstrated a proteomic signature associated with intrahepatic triglyceride content (Spearman rho=0.55, P<0.001) and insulin resistance (homeostatic model assessment for insulin resistance, Spearman rho=0.39, P=0.001). Changes in this proteomic signature were associated with changes in intrahepatic triglyceride content over the intervention period (beta=0.12, P<0.001). Moreover, cross-sectional analysis of overlapping proteins with hepatic attenuation in the community cohort showed generally, directionally consistent associations with hepatic steatosis. CONCLUSIONS These findings highlight the potential for broad proteomic profiling in small nutritional interventional studies with serial phenotyping alongside confirmatory large cohort epidemiology to prioritize targets of hepatic steatosis and cardiometabolic risk for mechanistic study.
Collapse
Affiliation(s)
- Bassim El-Sabawi
- Vanderbilt Translational and Clinical Cardiovascular Research Center Vanderbilt University School of Medicine Nashville TN USA
| | - Kahraman Tanriverdi
- Vanderbilt Translational and Clinical Cardiovascular Research Center Vanderbilt University School of Medicine Nashville TN USA
| | - Priya Gajjar
- Sections of Cardiovascular Medicine and Preventive Medicine and Epidemiology, Department of Medicine Boston University School of Medicine Boston MA USA
| | - Matthew Nayor
- Sections of Cardiovascular Medicine and Preventive Medicine and Epidemiology, Department of Medicine Boston University School of Medicine Boston MA USA
| | - Joshua M Landman
- Vanderbilt Genetics Institute Vanderbilt University Medical Center Nashville TN USA
| | - Jennifer E Below
- Vanderbilt Genetics Institute Vanderbilt University Medical Center Nashville TN USA
| | - Madeleine Haff
- Sections of Gastroenterology and Preventive Medicine and Epidemiology, Department of Medicine Boston University School of Medicine Boston MA USA
| | - Michelle Long
- Sections of Gastroenterology and Preventive Medicine and Epidemiology, Department of Medicine Boston University School of Medicine Boston MA USA
| | | | - Jane E Freedman
- Vanderbilt Translational and Clinical Cardiovascular Research Center Vanderbilt University School of Medicine Nashville TN USA
| | | | - Ravi Shah
- Vanderbilt Translational and Clinical Cardiovascular Research Center Vanderbilt University School of Medicine Nashville TN USA
| | - Andrew S Perry
- Vanderbilt Translational and Clinical Cardiovascular Research Center Vanderbilt University School of Medicine Nashville TN USA
| |
Collapse
|
2
|
Nelson S, Gaza J, Ajayebi S, Masse R, Pho R, Scutero C, Martinusen S, Long L, Menezes A, Perez A, Denard C. PERRC: Protease Engineering with Reactant Residence Time Control. ACS Synth Biol 2025. [PMID: 40388903 DOI: 10.1021/acssynbio.5c00154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/21/2025]
Abstract
Proteases with engineered specificity hold great potential for targeted therapeutics, protein circuit construction, and biotechnology applications. However, many proteases exhibit broad substrate specificity, limiting their use in such applications. Engineering protease specificity remains challenging because evolving a protease to recognize a new substrate, without counterselecting against its native substrate, often results in high residual activity on the original substrate. To address this, we developed Protease Engineering with Reactant Residence Time Control (PERRC), a platform that exploits the correlation between endoplasmic reticulum (ER) retention sequence strength and ER residence time. PERRC allows precise control over the stringency of protease evolution by adjusting counterselection to selection substrate ratios. Using PERRC, we evolved an orthogonal tobacco etch virus protease variant, TEVESNp, that selectively cleaves a substrate (ENLYFES) that differs by only one amino acid from its parent sequence (ENLYFQS). TEVESNp exhibits a remarkable 65-fold preference for the evolved substrate, marking the first example of an engineered orthogonal protease driven by such a slight difference in substrate recognition. Furthermore, TEVESNp functions as a competent protease for constructing orthogonal protein circuits in bacteria, and molecular dynamics simulations analysis reveals subtle yet functionally significant active site rearrangements. PERRC is a modular dual-substrate display system that facilitates precise engineering of protease specificity.
Collapse
Affiliation(s)
- Sage Nelson
- Department of Chemical Engineering, University of Florida, Gainesville 32611, United States
| | - Jokent Gaza
- Department of Chemistry, University of Florida, Gainesville 32611, United States
| | - Seyednima Ajayebi
- Department of Chemical Engineering, University of Florida, Gainesville 32611, United States
| | - Ronald Masse
- Genetics Institute, University of Florida, Gainesville 32611, United States
| | - Raymond Pho
- Department of Chemical Engineering, University of Florida, Gainesville 32611, United States
| | - Cianna Scutero
- Department of Chemical Engineering, University of Florida, Gainesville 32611, United States
| | - Samantha Martinusen
- Department of Chemical Engineering, University of Florida, Gainesville 32611, United States
| | - Lawton Long
- Department of Chemical Engineering, University of Florida, Gainesville 32611, United States
| | - Amor Menezes
- Genetics Institute, University of Florida, Gainesville 32611, United States
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville 32611, United States
| | - Alberto Perez
- Department of Chemistry, University of Florida, Gainesville 32611, United States
| | - Carl Denard
- Department of Chemical Engineering, University of Florida, Gainesville 32611, United States
- UF Health Cancer Center, University of Florida, Gainesville 32611, United States
| |
Collapse
|
3
|
Liao HC, Kuo LM, Chen WT, Huang YL, Sethy B, Dhandabani GK, Hsieh PW. Isolation, synthesis and structure-activity relationships of gallotannin derivatives as cathepsin C inhibitor. Bioorg Med Chem Lett 2025; 120:130133. [PMID: 39933615 DOI: 10.1016/j.bmcl.2025.130133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/16/2025] [Accepted: 02/08/2025] [Indexed: 02/13/2025]
Abstract
Cathepsin C (CatC), a key enzyme in neutrophil serine protease activation, is a promising target for treating neutrophilic inflammatory diseases like acute lung injury, ARDS, and COVID-19. Despite its therapeutic potential, no CatC inhibitors are currently available. In this study, a series of gallotannin derivatives were isolated from the traditional Chinese medicine Rhois Galla. Among these, 1,2,3,6-tetra-O-galloyl-β-d-glucose (1) inhibited CatC with an IC50 of 32.69 ± 2.95 nM. Subsequently, fifteen derivatives of 1 were synthesized and evaluated, revealing key structure-activity relationships. Compound 1 emerged as a potent and selective CatC inhibitor, while a novel synthetic derivative, 15, demonstrated dual inhibitory effects on CatC and cathepsin L. Structural features, including O-galloyl groups at positions 1, 2, and 6 of β-glucose and a hydrogen donor at position 4, were identified as favorable for CatC inhibition. These findings provide valuable insights for developing novel CatC inhibitors.
Collapse
Affiliation(s)
- Hung-Chen Liao
- Graduate Institute of Natural Products School of Traditional Chinese Medicine College of Medicine Chang Gung University Taoyuan Taiwan.
| | - Liang-Mou Kuo
- Department of General Surgery Chang Gung Memorial Hospital Chiayi Taiwan
| | - Wei-Ting Chen
- Graduate Institute of Natural Products School of Traditional Chinese Medicine College of Medicine Chang Gung University Taoyuan Taiwan
| | - Yu-Ling Huang
- Graduate Institute of Natural Products School of Traditional Chinese Medicine College of Medicine Chang Gung University Taoyuan Taiwan
| | - Bidyadhar Sethy
- Graduate Institute of Natural Products School of Traditional Chinese Medicine College of Medicine Chang Gung University Taoyuan Taiwan
| | - Ganesh Kumar Dhandabani
- Graduate Institute of Natural Products School of Traditional Chinese Medicine College of Medicine Chang Gung University Taoyuan Taiwan
| | - Pei-Wen Hsieh
- Graduate Institute of Natural Products School of Traditional Chinese Medicine College of Medicine Chang Gung University Taoyuan Taiwan; Department of General Surgery Chang Gung Memorial Hospital Chiayi Taiwan; Graduate Institute of Biomedical Sciences College of Medicine Chang Gung University Taoyuan Taiwan; Center for Drug Research and Development Chang Gung University of Science and Technology Taoyuan Taiwan.
| |
Collapse
|
4
|
Kwon N, Weng H, Rajora MA, Zheng G. Activatable Photosensitizers: From Fundamental Principles to Advanced Designs. Angew Chem Int Ed Engl 2025; 64:e202423348. [PMID: 39899458 PMCID: PMC11976215 DOI: 10.1002/anie.202423348] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/23/2025] [Accepted: 01/28/2025] [Indexed: 02/05/2025]
Abstract
Photodynamic therapy (PDT) is a promising treatment that uses light to excite photosensitizers in target tissue, producing reactive oxygen species and localized cell death. It is recognized as a minimally invasive, clinically approved cancer therapy with additional preclinical applications in arthritis, atherosclerosis, and infection control. A hallmark of ideal PDT is delivering disease-specific cytotoxicity while sparing healthy tissue. However, conventional photosensitizers often suffer from non-specific photoactivation, causing off-target toxicity. Activatable photosensitizers (aPS) have emerged as more precise alternatives, offering controlled activation. Unlike traditional photosensitizers, they remain inert and photoinactive during circulation and off-target accumulation, minimizing collateral damage. These photosensitizers are designed to "turn on" in response to disease-specific biostimuli, enhancing therapeutic selectivity and reducing off-target effects. This review explores the principles of aPS, including quenching mechanisms stemming from activatable fluorescent probes and applied to activatable photosensitizers (RET, PeT, ICT, ACQ, AIE), as well as pathological biostimuli (pH, enzymes, redox conditions, cellular internalization), and bioresponsive constructs enabling quenching and activation. We also provide a critical assessment of unresolved challenges in aPS development, including limitations in targeting precision, selectivity under real-world conditions, and potential solutions to persistent issues (dual-lock, targeting moieties, biorthogonal chemistry and artificial receptors). Additionally, it provides an in-depth discussion of essential research design considerations needed to develop translationally relevant aPS with improved therapeutic outcomes and specificity.
Collapse
Affiliation(s)
- Nahyun Kwon
- Princess Margaret Cancer CentreUniversity Health Network101 College Street, PMCRT 5–354Toronto, ONM5G1L7Canada
| | - Hanyi Weng
- Princess Margaret Cancer CentreUniversity Health Network101 College Street, PMCRT 5–354Toronto, ONM5G1L7Canada
- Department of Medical BiophysicsUniversity of TorontoToronto, ONCanada
| | - Maneesha A. Rajora
- Princess Margaret Cancer CentreUniversity Health Network101 College Street, PMCRT 5–354Toronto, ONM5G1L7Canada
- Department of MedicineUniversity of TorontoToronto, ONCanada
| | - Gang Zheng
- Princess Margaret Cancer CentreUniversity Health Network101 College Street, PMCRT 5–354Toronto, ONM5G1L7Canada
- Department of Medical BiophysicsUniversity of TorontoToronto, ONCanada
| |
Collapse
|
5
|
Vermeulen I, Li M, van Mourik H, Yadati T, Eijkel G, Balluff B, Godschalk R, Temmerman L, Biessen EAL, Kulkarni A, Theys J, Houben T, Cillero‐Pastor B, Shiri‐Sverdlov R. Inhibition of intracellular versus extracellular cathepsin D differentially alters the liver lipidome of mice with metabolic dysfunction-associated steatohepatitis. FEBS J 2025; 292:1781-1797. [PMID: 39726152 PMCID: PMC11970712 DOI: 10.1111/febs.17358] [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: 03/14/2024] [Revised: 08/09/2024] [Accepted: 12/03/2024] [Indexed: 12/28/2024]
Abstract
The prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) progressing to metabolic dysfunction-associated steatohepatitis (MASH), characterized by hepatic inflammation, has significantly increased in recent years due to unhealthy dietary practices and sedentary lifestyles. Cathepsin D (CTSD), a lysosomal protease involved in lipid homeostasis, is linked to abnormal lipid metabolism and inflammation in MASH. Although primarily intracellular, CTSD can be secreted extracellularly. Our previous proteomics research has shown that inhibition of extracellular CTSD results in more anti-inflammatory effects and fewer potential side effects compared to intracellular CTSD inhibition. However, the correlation between reduced side effects and alterations in the hepatic lipid composition remains unknown. This study aims to investigate the correlation between intra- and extracellular CTSD inhibition and potential alterations in the hepatic lipid composition in MASH. Low-density lipoprotein receptor knockout (Ldlr-/-) mice were fed a high-fat diet for 10 weeks and received subcutaneous injections every 2 days of vehicle, intracellular CTSD inhibitor (GA-12), or extracellular CTSD inhibitor (CTD-002). Matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) was used to visualize and compare the lipid composition in liver tissues. Hepatic phosphatidylcholine remodeling was observed with both inhibitors, suggesting their therapeutic potential in treating MASH. Treatment with an intracellular CTSD inhibitor resulted in elevated levels of cardiolipin, reactive oxygen species, phosphatidylinositol, phosphatidylethanolamine, and lipids that are linked to mitochondrial dysfunction and inflammation, and induced more oxidative stress. The observed modifications in lipid composition demonstrate the clinical advantages of extracellular CTSD inhibition as a potentially beneficial therapeutic approach for MASH.
Collapse
Affiliation(s)
- Isabeau Vermeulen
- Maastricht Multimodal Molecular Imaging Institute (M4i)University of MaastrichtThe Netherlands
| | - Mengying Li
- Department of Genetics and Cell Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM)Maastricht UniversityThe Netherlands
| | - Hester van Mourik
- Department of Genetics and Cell Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM)Maastricht UniversityThe Netherlands
- Department of Precision Medicine, Institute for Oncology and Reproduction (GROW)Maastricht UniversityThe Netherlands
| | - Tulasi Yadati
- Department of Genetics and Cell Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM)Maastricht UniversityThe Netherlands
| | - Gert Eijkel
- Maastricht Multimodal Molecular Imaging Institute (M4i)University of MaastrichtThe Netherlands
| | - Benjamin Balluff
- Maastricht Multimodal Molecular Imaging Institute (M4i)University of MaastrichtThe Netherlands
| | - Roger Godschalk
- Department of Pharmacology and Toxicology, Institute for Nutrition and Translational Research in Metabolism (NUTRIM)Maastricht UniversityThe Netherlands
| | - Lieve Temmerman
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM)Maastricht University Medical Center (UMC)The Netherlands
| | - Erik A. L. Biessen
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM)Maastricht University Medical Center (UMC)The Netherlands
- Institute for Molecular Cardiovascular ResearchRheinisch‐Westfälische Technische Hochschule (RWTH) Aachen UniversityGermany
| | | | - Jan Theys
- Department of Precision Medicine, Institute for Oncology and Reproduction (GROW)Maastricht UniversityThe Netherlands
| | - Tom Houben
- Department of Genetics and Cell Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM)Maastricht UniversityThe Netherlands
| | - Berta Cillero‐Pastor
- Maastricht Multimodal Molecular Imaging Institute (M4i)University of MaastrichtThe Netherlands
- Cell Biology‐Inspired Tissue Engineering (cBITE), MERLNMaastricht UniversityThe Netherlands
| | - Ronit Shiri‐Sverdlov
- Department of Genetics and Cell Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM)Maastricht UniversityThe Netherlands
| |
Collapse
|
6
|
Zhao Y, Zhuang Y, Shi J, Fan H, Lv Q, Guo X. Cathepsin B induces kidney diseases through different types of programmed cell death. Front Immunol 2025; 16:1535313. [PMID: 40129990 PMCID: PMC11930809 DOI: 10.3389/fimmu.2025.1535313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 02/25/2025] [Indexed: 03/26/2025] Open
Abstract
Cathepsin B (CTSB), a key cysteine protease, plays essential roles in physiological and pathological processes. As research progresses, interest in how CTSB triggers different types of programmed cell death (PCD) to induce the onset and development of diseases is increasing. Several recent studies suggest that different types of PCD mediated by CTSB play key roles in kidney diseases. In this review, we outline the fundamental mechanisms by which CTSB triggers different types of PCD in several kidney diseases and discuss the function of CTSB in various segments of the kidney. Moreover, we explore the possibilities and prospects of using CTSB as a therapeutic target for kidney diseases.
Collapse
Affiliation(s)
- Yunlong Zhao
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Key Laboratory for Disaster Medicine Technology, Tianjin University, Tianjin, China
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, China
| | - Yong Zhuang
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Key Laboratory for Disaster Medicine Technology, Tianjin University, Tianjin, China
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, China
| | - Jie Shi
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Key Laboratory for Disaster Medicine Technology, Tianjin University, Tianjin, China
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, China
| | - Haojun Fan
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Key Laboratory for Disaster Medicine Technology, Tianjin University, Tianjin, China
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, China
| | - Qi Lv
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Key Laboratory for Disaster Medicine Technology, Tianjin University, Tianjin, China
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, China
| | - Xiaoqin Guo
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Key Laboratory for Disaster Medicine Technology, Tianjin University, Tianjin, China
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, China
| |
Collapse
|
7
|
Wyżewski Z, Gregorczyk-Zboroch KP, Mielcarska MB, Świtlik W, Niedzielska A. Bid Protein: A Participant in the Apoptotic Network with Roles in Viral Infections. Int J Mol Sci 2025; 26:2385. [PMID: 40141030 PMCID: PMC11942203 DOI: 10.3390/ijms26062385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 03/01/2025] [Accepted: 03/04/2025] [Indexed: 03/28/2025] Open
Abstract
The BH3-interacting domain death agonist (Bid), a proapoptotic signaling molecule of the B-cell lymphoma 2 (Bcl-2) family, is a key regulator of mitochondrial outer membrane (MOM) permeability. Uniquely positioned at the intersection of extrinsic and intrinsic apoptosis pathways, Bid links death receptor signaling to the mitochondria-dependent cascade and can also be activated by endoplasmic reticulum (ER) stress. In its active forms, cleaved Bid (cBid) and truncated Bid (tBid), it disrupts MOM integrity via Bax/Bak-dependent and independent mechanisms. Apoptosis plays a dual role in viral infections, either promoting or counteracting viral propagation. Consequently, viruses modulate Bid signaling to favor their replication. The deregulation of Bid activity contributes to oncogenic transformation, inflammation, immunosuppression, neurotoxicity, and pathogen propagation during various viral infections. In this work, we explore Bid's structure, function, activation processes, and mitochondrial targeting. We describe its role in apoptosis induction and its involvement in infections with multiple viruses. Additionally, we discuss the therapeutic potential of Bid in antiviral strategies. Understanding Bid's signaling pathways offers valuable insights into host-virus interactions and the pathogenesis of infections. This knowledge may facilitate the development of novel therapeutic approaches to combat virus-associated diseases effectively.
Collapse
Affiliation(s)
- Zbigniew Wyżewski
- Institute of Biological Sciences, Cardinal Stefan Wyszynski University in Warsaw, Dewajtis 5, 01-815 Warsaw, Poland
| | - Karolina Paulina Gregorczyk-Zboroch
- Division of Immunology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences—SGGW, Ciszewskiego 8, 02-786 Warsaw, Poland; (K.P.G.-Z.); (M.B.M.); (A.N.)
| | - Matylda Barbara Mielcarska
- Division of Immunology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences—SGGW, Ciszewskiego 8, 02-786 Warsaw, Poland; (K.P.G.-Z.); (M.B.M.); (A.N.)
| | - Weronika Świtlik
- Centre for Advanced Materials and Technologies, Warsaw University of Technology, Poleczki 19, 02-822 Warsaw, Poland;
| | - Adrianna Niedzielska
- Division of Immunology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences—SGGW, Ciszewskiego 8, 02-786 Warsaw, Poland; (K.P.G.-Z.); (M.B.M.); (A.N.)
| |
Collapse
|
8
|
Nelson S, Gaza J, Ajayebi S, Masse R, Pho R, Scutero C, Martinusen S, Long L, Menezes A, Perez A, Denard C. PERRC: Protease Engineering with Reactant Residence Time Control. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.02.641063. [PMID: 40093119 PMCID: PMC11908129 DOI: 10.1101/2025.03.02.641063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Proteases with engineered specificity hold great potential for targeted therapeutics, protein circuit construction, and biotechnology applications. However, many proteases exhibit broad substrate specificity, limiting their applications. Engineering protease specificity remains challenging because evolving a protease to recognize a new substrate, without counterselecting against its native substrate, often results in high residual activity on the original substrate. To address this, we developed Protease Engineering with Reactant Residence Time Control (PERRC), a platform that exploits the correlation between endoplasmic reticulum (ER) retention sequence strength and ER residence time. PERRC allows precise control over the stringency of protease evolution by adjusting counterselection to selection substrate ratios. Using PERRC, we evolved an orthogonal tobacco etch virus protease variant, TEVESNp, that selectively cleaves a substrate (ENLYFES) that differs by only one amino acid from its parent sequence (ENLYFQS). TEVESNp exhibits a remarkable 65-fold preference for the evolved substrate, marking the first example of an engineered orthogonal protease driven by such a slight difference in substrate recognition. Furthermore, TEVESNp functions as a competent protease for constructing orthogonal protein circuits in bacteria, and molecular dynamic simulations analysis reveals subtle yet functionally significant active site rearrangements. PERRC is a modular dual-substrate display system that facilitates precise engineering of protease specificity.
Collapse
Affiliation(s)
- Sage Nelson
- Department of Chemical Engineering, University of Florida, Gainesville, 32611, USA
| | - Jokent Gaza
- Department of Chemistry, University of Florida, Gainesville, 32611, USA
| | - Seyednima Ajayebi
- Department of Chemical Engineering, University of Florida, Gainesville, 32611, USA
| | - Ronald Masse
- Genetics Institute, University of Florida, Gainesville, 32611, USA
| | - Raymond Pho
- Department of Chemical Engineering, University of Florida, Gainesville, 32611, USA
| | - Cianna Scutero
- Department of Chemical Engineering, University of Florida, Gainesville, 32611, USA
| | - Samantha Martinusen
- Department of Chemical Engineering, University of Florida, Gainesville, 32611, USA
| | - Lawton Long
- Department of Chemical Engineering, University of Florida, Gainesville, 32611, USA
| | - Amor Menezes
- Genetics Institute, University of Florida, Gainesville, 32611, USA
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, 32611, USA
| | - Alberto Perez
- Department of Chemistry, University of Florida, Gainesville, 32611, USA
| | - Carl Denard
- Department of Chemical Engineering, University of Florida, Gainesville, 32611, USA
- UF Health Cancer Center, University of Florida, Gainesville, 32611, USA
| |
Collapse
|
9
|
Lago-Baameiro N, Camino T, Vazquez-Durán A, Sueiro A, Couto I, Santos F, Baltar J, Falcón-Pérez JM, Pardo M. Intra and inter-organ communication through extracellular vesicles in obesity: functional role of obesesomes and steatosomes. J Transl Med 2025; 23:207. [PMID: 39979938 PMCID: PMC11844161 DOI: 10.1186/s12967-024-06024-7] [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: 09/11/2024] [Accepted: 12/22/2024] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND Extracellular vesicles (EVs) represent a sophisticated mechanism of intercellular communication that is implicated in health and disease. Specifically, the role of EVs in metabolic regulation and their implications in metabolic pathologies, such as obesity and its comorbidities, remain unclear. METHODS Extracellular vesicles (EVs) were isolated through serial ultracentrifugation from murine adipocytes treated with palmitate or oleic acid, whole visceral and subcutaneous adipose tissue (obesesomes) of bariatric surgery obese donors, and human hepatocytes under steatosis (steatosomes) for functional in vitro experiments. Functional effects on inflammation and glucose and lipid metabolism of target cells (human and murine macrophages and hepatocytes) were assessed using ELISA, RT-PCR, and immunodetection. Isolated EVs from human steatotic (steatosomes) and control hepatocytes (hepatosomes) were characterized for quantity, size, and tetraspanin profile by NTA and Single Particle Interferometric Reflectance Imaging Sensor (SP-IRIS), and their protein cargo analyzed by qualitative (DDA) and quantitative (DIA-SWATH) proteomics using LC-MS/MS. Proteins identified by proteomics were validated by capturing EVs on functionalized chips by SP-IRIS. RESULTS AND CONCLUSIONS In this study, we investigated the role of EVs in the local communication between obese adipocytes and immune cells within adipose tissue, and the interaction of steatotic and healthy hepatocytes in the context of fatty liver disease progression. Furthermore, we analyzed obese adipose tissue-to-liver interactions through EV-obesesomes to elucidate their role in obesity-associated hepatic metabolic dysregulation. Our findings reveal that obesesomes promote inflammation and the secretion of pro-inflammatory cytokines upon interaction with macrophages, exerting a significant impact on reducing insulin resistance and altering lipid and glucose metabolism upon interaction with hepatocytes; in both cases, EVs from palmitate-loaded adipocytes and obesesomes from human visceral adipose depots demonstrated the most deleterious effect. Additionally, EVs secreted by steatotic hepatocytes (steatosomes) induced insulin resistance and altered lipid and glucose metabolism in healthy hepatocytes, suggesting their involvement in MASLD development. Proteomic analysis of steatosomes revealed that these vesicles contain liver disease-associated proteins, rendering them significant repositories of real-time biomarkers for the early stages and progression of MASLD.
Collapse
Affiliation(s)
- N Lago-Baameiro
- Grupo Obesidómica, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela/SERGAS, Santiago de Compostela, Travesía da Choupana s/n, 15706, Santiago de Compostela, A Coruña, Spain
| | - T Camino
- Grupo Obesidómica, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela/SERGAS, Santiago de Compostela, Travesía da Choupana s/n, 15706, Santiago de Compostela, A Coruña, Spain
| | - A Vazquez-Durán
- Grupo Obesidómica, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela/SERGAS, Santiago de Compostela, Travesía da Choupana s/n, 15706, Santiago de Compostela, A Coruña, Spain
| | - A Sueiro
- Grupo Endocrinología Molecular y Celular, Instituto de Investigación Sanitaria de Santiago (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela/SERGAS, Santiago de Compostela, Spain
| | - I Couto
- Grupo Obesidómica, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela/SERGAS, Santiago de Compostela, Travesía da Choupana s/n, 15706, Santiago de Compostela, A Coruña, Spain
- Servicio de Cirugía Plástica y Reparadora, Complexo Hospitalario Universitario de Santiago de Compostela/SERGAS, Santiago de Compostela, Spain
| | - F Santos
- Grupo Obesidómica, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela/SERGAS, Santiago de Compostela, Travesía da Choupana s/n, 15706, Santiago de Compostela, A Coruña, Spain
- Servicio de Cirugía General, Complexo Hospitalario Universitario de Santiago de Compostela/SERGAS, Santiago de Compostela, Spain
| | - J Baltar
- Grupo Obesidómica, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela/SERGAS, Santiago de Compostela, Travesía da Choupana s/n, 15706, Santiago de Compostela, A Coruña, Spain
- Servicio de Cirugía General, Complexo Hospitalario Universitario de Santiago de Compostela/SERGAS, Santiago de Compostela, Spain
| | - J M Falcón-Pérez
- Exosomes Laboratory and Metabolomics Platform, CIC bioGUNE-BRTA, CIBERehd, Derio, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - M Pardo
- Grupo Obesidómica, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela/SERGAS, Santiago de Compostela, Travesía da Choupana s/n, 15706, Santiago de Compostela, A Coruña, Spain.
- CIBER Fisiopatología Obesidad y Nutrición, Instituto de Salud Carlos III, Santiago de Compostela, Spain.
| |
Collapse
|
10
|
Qian J, Shao X, Bao H, Fang Y, Guo W, Li C, Li A, Hua H, Fan X. Identification and characterization of cell niches in tissue from spatial omics data at single-cell resolution. Nat Commun 2025; 16:1693. [PMID: 39956823 PMCID: PMC11830827 DOI: 10.1038/s41467-025-57029-9] [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: 08/12/2024] [Accepted: 02/03/2025] [Indexed: 02/18/2025] Open
Abstract
Deciphering the features, structure, and functions of the cell niche in tissues remains a major challenge. Here, we present scNiche, a computational framework to identify and characterize cell niches from spatial omics data at single-cell resolution. We benchmark scNiche with both simulated and biological datasets, and demonstrate that scNiche can effectively and robustly identify cell niches while outperforming other existing methods. In spatial proteomics data from human triple-negative breast cancer, scNiche reveals the influence of the microenvironment on cellular phenotypes, and further dissects patient-specific niches with distinct cellular compositions or phenotypic characteristics. By analyzing mouse liver spatial transcriptomics data across normal and early-onset liver failure donors, scNiche uncovers disease-specific liver injury niches, and further delineates the niche remodeling from normal liver to liver failure. Overall, scNiche enables decoding the cellular microenvironment in tissues from single-cell spatial omics data.
Collapse
Affiliation(s)
- Jingyang Qian
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- State Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314102, China
| | - Xin Shao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- State Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314102, China.
- Zhejiang Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314100, China.
| | - Hudong Bao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yin Fang
- College of Computer Science and Technology, Zhejiang University, Hangzhou, 310013, China
| | - Wenbo Guo
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- State Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314102, China
- Zhejiang Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314100, China
| | - Chengyu Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- State Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314102, China
| | - Anyao Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- State Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314102, China
| | - Hua Hua
- Translational Chinese Medicine Key Laboratory of Sichuan Province, SiChuan Institute for Translational Chinese Medicine, Chengdu, 610041, China.
| | - Xiaohui Fan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- State Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314102, China.
- Zhejiang Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314100, China.
- Zhejiang Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, 310006, Hangzhou, China.
| |
Collapse
|
11
|
Du J, Zhang K, Miao J, Yang Y, Tian Y, Wu T, Tao C, Wang Y, Yang S. Molecular pathological characteristics and mechanisms of the liver in metabolic disease-susceptible transgenic pigs. Life Sci 2025; 362:123337. [PMID: 39734013 DOI: 10.1016/j.lfs.2024.123337] [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: 08/18/2024] [Revised: 11/05/2024] [Accepted: 12/19/2024] [Indexed: 12/31/2024]
Abstract
AIMS This study aimed to explore the molecular pathological mechanisms of the liver in metabolic disease-susceptible transgenic pigs via multiomics analysis. MATERIALS AND METHODS The triple-transgenic (PNPLA3I148M-GIPRdn-hIAPP) pig model (TG pig) was successfully constructed in our laboratory via the CRISPR/Cas9 technique previously described. Wild-type (WT) pigs and TG pigs after 2 or 12 months of high-fat and high-sucrose diet (HFHSD) induction (WT2, TG2, WT12, and TG12 groups, respectively) were used as materials. The transcriptome, metabolome, and lipidome were used to investigate the molecular mechanisms of the liver in pigs. KEY FINDINGS The TG2 pigs presented mild metaflammation and insulin resistance (IR) which was similar to WT12 pigs. Compared with the other three groups, the TG12 pigs presented severe hepatocyte ballooning, fat deposition, and portal area fibrosis. The transcriptome data suggested that the TG2 pigs presented upregulated gene expression in the extracellular matrix (ECM). The TG12 pigs presented more severe metaflammation and exhibited imbalanced glycolipid metabolism. Interestingly, genes such as ETNPPL, GABBR2, and BMP8B might be key regulatory targets for liver injury. The metabolome and lipidome suggested that long-chain polyunsaturated fatty acids (LCPUFAs) and phospholipids with corresponding LCPUFAs were remodelled. Importantly, bis(monoacylglycerol) phosphates (BMPs) and sulfatides (SLs) could be the key regulatory metabolites in liver injury. SIGNIFICANCE ETNPPL, GABBR2, and BMP8B might be potential therapeutic targets for liver injury. BMPs and SLs might be biomarkers for the diagnosis and treatment of liver diseases.
Collapse
Affiliation(s)
- Juan Du
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China
| | - Kaiyi Zhang
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China
| | - Jiakun Miao
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China
| | - Yu Yang
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China
| | - Yuying Tian
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China
| | - Tianwen Wu
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China
| | - Cong Tao
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China
| | - Yanfang Wang
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China.
| | - Shulin Yang
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China.
| |
Collapse
|
12
|
Zhou P, Yang L, Li H, Zeng L, Zhang Y, Zhong Z, Li R, Yin Y, Tao K, Zhang P. IRG1/Itaconate inhibits hepatic stellate cells ferroptosis and attenuates TAA-induced liver fibrosis by regulating SLC39A14 expression. Int Immunopharmacol 2025; 146:113945. [PMID: 39724735 DOI: 10.1016/j.intimp.2024.113945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 12/13/2024] [Accepted: 12/22/2024] [Indexed: 12/28/2024]
Abstract
This study aimed to elucidate the protective roles of Immune Response Gene-1 (IRG1) and exogenous itaconate in murine models of hepatic fibrosis and to delineate the underlying mechanistic pathways using both wild-type and IRG1-deficient (IRG1-/-) mice. Primary murine stellate cells (mHSC) and bone marrow-derived macrophages (BMDM) were isolated and cocultured. Hepatocellular fibrosis was induced in vitro using Transforming Growth Factor-beta (TGF-β) to evaluate the protective efficacy of IRG1/itaconate. Histopathological damage in the hepatic tissues was assessed using Hematoxylin and Eosin (H&E), Masson's trichrome, and Sirius red staining, followed by hepatic fibrosis scoring. The levels of released inflammatory cytokines were quantified using enzyme-linked immunosorbent assay (ELISA) kits. Immunohistochemistry was used to detect 4-Hydroxynonenal (4-HNE) levels and Perls staining was used to assess ferroptosis. RNA sequencing and gene enrichment analyses were performed to identify implicated molecular entities and signaling pathways. IRG1 and SLC39A14 knockdown and overexpression cell lines were generated. Quantitative real-time PCR (qRT-PCR) and western blotting (WB) were used to measure the mRNA and protein expression levels in hepatic tissues and cells. Kits were used to assess reactive oxygen species (ROS) and malondialdehyde (MDA) levels, and the concentrations of liver enzymes, iron, GSH, and GSSG within hepatic tissues and cells.4-octyl itaconate (4-OI) significantly attenuated the histopathological damage in hepatic tissues, preserved the normal hepatic function, effectively reduced the release of inflammatory cytokines, and mitigated oxidative stress markers such as ROS and MDA in Thioacetamide (TAA)-induced fibrotic mice. Notably, this study is the first to reveal the pivotal role of SLC39A14 in the pathogenesis of hepatic fibrosis in murine models and elucidate how IRG1/itaconate mediates downstream ferroptosis-related signaling pathways by targeting SLC39A14, thereby inhibiting ferroptosis-induced hepatic fibrosis. IRG1/itaconate can alleviate the TAA-induced hepatic fibrosis in mice by regulating the expression of SLC39A14, consequently suppressing hepatic stellate cell ferroptosis.
Collapse
Affiliation(s)
- Pei Zhou
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lei Yang
- Department of General Surgery, The Second Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Hang Li
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Liwu Zeng
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yizhuo Zhang
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ziyou Zhong
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ruidong Li
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yuping Yin
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Kaixiong Tao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Peng Zhang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
13
|
Lalmanach G, Rigoux B, David A, Tahri-Joutey M, Lecaille F, Marchand-Adam S, Saidi A. Human cystatin C in fibrotic diseases. Clin Chim Acta 2025; 565:120016. [PMID: 39461496 DOI: 10.1016/j.cca.2024.120016] [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: 06/11/2024] [Revised: 10/22/2024] [Accepted: 10/22/2024] [Indexed: 10/29/2024]
Abstract
Human cystatin C (hCC), which has a pervasive distribution within body fluids and is ubiquitously expressed by numerous cells and tissues, is a highly potent extracellular inhibitor of cysteine proteases. Besides measurement of serum creatinine, which is the most widely used technique for appraising glomerular filtration rate (GFR), hCC has emerged as a relevant GFR biomarker, because its quantification in serum is less sensitive to interferences with factors such as age, muscle mass or diet. Moreover, there are growing body of evidence that hCC overexpression and/or oversecretion, which is primarily driven by TGF-β1, occur during fibrogenesis (cardiac, liver, oral, and lung fibrosis). Even though molecular mechanisms and signaling pathways governing the regulation of hCC remain to be deciphered more acutely, current data sustain that hCC expression relates to myofibrogenesis and that hCC could be a specific and valuable biomarker of fibrotic disease.
Collapse
Affiliation(s)
- Gilles Lalmanach
- University of Tours, Tours, France; INSERM, UMR1100, Research Center for Respiratory Diseases (CEPR), Team "Proteolytic Enzymes and Their Pharmacological Targeting in Lung Diseases", Tours, France.
| | - Baptiste Rigoux
- University of Tours, Tours, France; INSERM, UMR1100, Research Center for Respiratory Diseases (CEPR), Team "Proteolytic Enzymes and Their Pharmacological Targeting in Lung Diseases", Tours, France
| | - Alexis David
- University of Tours, Tours, France; INSERM, UMR1100, Research Center for Respiratory Diseases (CEPR), Team "Proteolytic Enzymes and Their Pharmacological Targeting in Lung Diseases", Tours, France
| | - Mounia Tahri-Joutey
- University of Tours, Tours, France; INSERM, UMR1100, Research Center for Respiratory Diseases (CEPR), Team "Proteolytic Enzymes and Their Pharmacological Targeting in Lung Diseases", Tours, France
| | - Fabien Lecaille
- University of Tours, Tours, France; INSERM, UMR1100, Research Center for Respiratory Diseases (CEPR), Team "Proteolytic Enzymes and Their Pharmacological Targeting in Lung Diseases", Tours, France
| | - Sylvain Marchand-Adam
- University of Tours, Tours, France; INSERM, UMR1100, Research Center for Respiratory Diseases (CEPR), Team "Proteolytic Enzymes and Their Pharmacological Targeting in Lung Diseases", Tours, France; The University Hospital Center of Tours (CHRU Tours), Pulmonology Department, Tours, France
| | - Ahlame Saidi
- University of Tours, Tours, France; INSERM, UMR1100, Research Center for Respiratory Diseases (CEPR), Team "Proteolytic Enzymes and Their Pharmacological Targeting in Lung Diseases", Tours, France
| |
Collapse
|
14
|
Jiang Y, Fan W, Li Y, Xue H. Genetic Insights Into the Role of Cathepsins in Alzheimer's Disease, Parkinson's Disease, and Amyotrophic Lateral Sclerosis: Evidence From Mendelian Randomization Study. Brain Behav 2025; 15:e70207. [PMID: 39740768 DOI: 10.1002/brb3.70207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 11/17/2024] [Accepted: 12/07/2024] [Indexed: 01/02/2025] Open
Abstract
BACKGROUND Previous studies have confirmed the significant role of cathepsins in the development of neurodegenerative diseases. We aimed to determine whether genetically predicted 10 cathepsins may have a causal effect on Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS). METHODS We conducted a two-sample bidirectional Mendelian randomization (MR) study using publicly available data from genome-wide association study (GWAS) to assess the causal associations between 10 cathepsins and three neurodegenerative diseases, including AD, PD, and ALS. We employed the following methods, including inverse variance weighting (IVW), MR-Egger, and weighted median (WM). The results were further validated using sensitivity analysis. RESULTS The forward MR analysis results indicate that elevated cathepsin H levels increase the risk of AD (p = 0.005, odds ratio [OR] = 1.040, 95% confidence interval [CI] = 1.011-1.069), elevated cathepsin B levels decrease the risk of PD (p < 0.001, OR = 0.890, 95% CI = 0.831-0.954), and no significant association was found between cathepsin levels and ALS. Reverse MR analysis suggests that there is no causal association between 10 cathepsins and three neurodegenerative diseases. CONCLUSION Our study provides new genetic insights into the role of cathepsin H in AD and cathepsin B in PD. However, our findings need to be further validated in a wider population, and future research should explore the potential mechanisms of cathepsins in these diseases in order to provide a basis for the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Yanhong Jiang
- Department of Neurology, Sichuan Taikang Hospital, Chengdu, Sichuan, China
| | - Wenhui Fan
- Department of Neurology, Sichuan Taikang Hospital, Chengdu, Sichuan, China
| | - Yaxin Li
- Department of Neurology, Sichuan Taikang Hospital, Chengdu, Sichuan, China
| | - Hua Xue
- Department of Neurology, Sichuan Taikang Hospital, Chengdu, Sichuan, China
| |
Collapse
|
15
|
Leblond AL, Helmchen B, Ankavay M, Lenggenhager D, Jetzer J, Helmchen F, Yurtsever H, Parrotta R, Healy ME, Pöschel A, Markkanen E, Semmo N, Ferrié M, Cocquerel L, Seeger H, Hopfer H, Müllhaupt B, Gouttenoire J, Moradpour D, Gaspert A, Weber A. HEV ORF2 protein-antibody complex deposits are associated with glomerulonephritis in hepatitis E with reduced immune status. Nat Commun 2024; 15:8849. [PMID: 39397005 PMCID: PMC11471813 DOI: 10.1038/s41467-024-53072-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 09/27/2024] [Indexed: 10/15/2024] Open
Abstract
Hepatitis E virus (HEV) infection, one of the most common forms of hepatitis worldwide, is often associated with extrahepatic, particularly renal, manifestations. However, the underlying mechanisms are incompletely understood. Here, we report the development of a de novo immune complex-mediated glomerulonephritis (GN) in a kidney transplant recipient with chronic hepatitis E. Applying immunostaining, electron microscopy, and mass spectrometry after laser-capture microdissection, we show that GN develops in parallel with increasing glomerular deposition of a non-infectious, genome-free and non-glycosylated HEV open reading frame 2 (ORF2) capsid protein. No productive HEV infection of kidney cells is detected. Patients with acute hepatitis E display similar but less pronounced deposits. Our results establish a link between the production of HEV ORF2 protein and the development of hepatitis E-associated GN in the immunocompromised state. The formation of glomerular IgG-HEV ORF2 immune complexes discovered here provides a potential mechanistic explanation of how the hepatotropic HEV can cause variable renal manifestations. These findings directly provide a tool for etiology-based diagnosis of hepatitis E-associated GN as a distinct entity and suggest therapeutic implications.
Collapse
Affiliation(s)
- Anne-Laure Leblond
- Department of Pathology and Molecular Pathology, University of Zurich (UZH) and University Hospital Zurich (USZ), Zurich, Switzerland
| | - Birgit Helmchen
- Department of Pathology and Molecular Pathology, University of Zurich (UZH) and University Hospital Zurich (USZ), Zurich, Switzerland
| | - Maliki Ankavay
- Division of Gastroenterology and Hepatology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Daniela Lenggenhager
- Department of Pathology and Molecular Pathology, University of Zurich (UZH) and University Hospital Zurich (USZ), Zurich, Switzerland
| | - Jasna Jetzer
- Department of Pathology and Molecular Pathology, University of Zurich (UZH) and University Hospital Zurich (USZ), Zurich, Switzerland
| | - Fritjof Helmchen
- Brain Research Institute, University of Zurich, Zurich, Switzerland
| | | | - Rossella Parrotta
- Department of Pathology and Molecular Pathology, University of Zurich (UZH) and University Hospital Zurich (USZ), Zurich, Switzerland
| | - Marc E Healy
- Department of Pathology and Molecular Pathology, University of Zurich (UZH) and University Hospital Zurich (USZ), Zurich, Switzerland
| | - Amiskwia Pöschel
- Institute of Veterinary Pharmacology and Toxicology, University of Zurich - Vetsuisse Faculty, Zürich, Switzerland
| | - Enni Markkanen
- Institute of Veterinary Pharmacology and Toxicology, University of Zurich - Vetsuisse Faculty, Zürich, Switzerland
| | - Nasser Semmo
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Martin Ferrié
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Laurence Cocquerel
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Harald Seeger
- Clinic of Nephrology, University Hospital Zurich, Zurich, Switzerland
| | - Helmut Hopfer
- Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Beat Müllhaupt
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Jérôme Gouttenoire
- Division of Gastroenterology and Hepatology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Darius Moradpour
- Division of Gastroenterology and Hepatology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Ariana Gaspert
- Department of Pathology and Molecular Pathology, University of Zurich (UZH) and University Hospital Zurich (USZ), Zurich, Switzerland
| | - Achim Weber
- Department of Pathology and Molecular Pathology, University of Zurich (UZH) and University Hospital Zurich (USZ), Zurich, Switzerland.
- Institute of Molecular Cancer Research (IMCR), University of Zurich (UZH), Zurich, Switzerland.
| |
Collapse
|
16
|
Hardesty JE, Warner JB, Wilkey DW, Phinney BS, Salemi MR, Merchant ML, McClain CJ, Warner DR, Kirpich IA. Hepatic Proteomic Changes Associated with Liver Injury Caused by Alcohol Consumption in Fpr2-/- Mice. Int J Mol Sci 2024; 25:9807. [PMID: 39337294 PMCID: PMC11432144 DOI: 10.3390/ijms25189807] [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: 08/19/2024] [Revised: 09/05/2024] [Accepted: 09/07/2024] [Indexed: 09/30/2024] Open
Abstract
Alcohol-associated liver disease (ALD) is a prevalent medical problem with limited effective treatment strategies. Although many biological processes contributing to ALD have been elucidated, a complete understanding of the underlying mechanisms is still lacking. The current study employed a proteomic approach to identify hepatic changes resulting from ethanol (EtOH) consumption and the genetic ablation of the formyl peptide receptor 2 (FPR2), a G-protein coupled receptor known to regulate multiple signaling pathways and biological processes, in a mouse model of ALD. Since previous research from our team demonstrated a notable reduction in hepatic FPR2 protein levels in patients with alcohol-associated hepatitis (AH), the proteomic changes in the livers of Fpr2-/- EtOH mice were compared to those observed in patients with AH in order to identify common hepatic proteomic alterations. Several pathways linked to exacerbated ALD in Fpr2-/- EtOH mice, as well as hepatic protein changes resembling those found in patients suffering from AH, were identified. These alterations included decreased levels of coagulation factors F2 and F9, as well as reduced hepatic levels of glutamate-cysteine ligase catalytic subunit (GCLC) and total glutathione in Fpr2-/- EtOH compared to WT EtOH mice. In conclusion, the data suggest that FPR2 may play a regulatory role in hepatic blood coagulation and the antioxidant system, both in a pre-clinical model of ALD and in human AH, however further experiments are required to validate these findings.
Collapse
Affiliation(s)
- Josiah E. Hardesty
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Louisville, Louisville, KY 40202, USA; (J.E.H.); (J.B.W.); (C.J.M.); (D.R.W.)
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Louisville, KY 40202, USA;
| | - Jeffrey B. Warner
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Louisville, Louisville, KY 40202, USA; (J.E.H.); (J.B.W.); (C.J.M.); (D.R.W.)
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Louisville, KY 40202, USA;
| | - Daniel W. Wilkey
- The Proteomics Core, School of Medicine, University of Louisville, Louisville, KY 40202, USA;
| | - Brett S. Phinney
- Proteomics Core Facility, University of California Davis, Davis, CA 95616, USA; (B.S.P.); (M.R.S.)
| | - Michelle R. Salemi
- Proteomics Core Facility, University of California Davis, Davis, CA 95616, USA; (B.S.P.); (M.R.S.)
| | - Michael L. Merchant
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Louisville, KY 40202, USA;
- The Proteomics Core, School of Medicine, University of Louisville, Louisville, KY 40202, USA;
| | - Craig J. McClain
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Louisville, Louisville, KY 40202, USA; (J.E.H.); (J.B.W.); (C.J.M.); (D.R.W.)
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Louisville, KY 40202, USA;
- Robley Rex Veterans Medical Center, Louisville, KY 40202, USA
- Alcohol Research Center, School of Medicine, University of Louisville, Louisville, KY 40202, USA
- Hepatobiology & Toxicology Center, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Dennis R. Warner
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Louisville, Louisville, KY 40202, USA; (J.E.H.); (J.B.W.); (C.J.M.); (D.R.W.)
| | - Irina A. Kirpich
- Alcohol Research Center, School of Medicine, University of Louisville, Louisville, KY 40202, USA
- Hepatobiology & Toxicology Center, School of Medicine, University of Louisville, Louisville, KY 40202, USA
- Department of Microbiology and Immunology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| |
Collapse
|
17
|
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
|
18
|
Ruiz-Blázquez P, Fernández-Fernández M, Pistorio V, Martinez-Sanchez C, Costanzo M, Iruzubieta P, Zhuravleva E, Cacho-Pujol J, Ariño S, Del Castillo-Cruz A, Núñez S, Andersen JB, Ruoppolo M, Crespo J, García-Ruiz C, Pavone LM, Reinheckel T, Sancho-Bru P, Coll M, Fernández-Checa JC, Moles A. Cathepsin D is essential for the degradomic shift of macrophages required to resolve liver fibrosis. Mol Metab 2024; 87:101989. [PMID: 39019115 PMCID: PMC11327474 DOI: 10.1016/j.molmet.2024.101989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 07/03/2024] [Accepted: 07/08/2024] [Indexed: 07/19/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Fibrosis contributes to 45% of deaths in industrialized nations and is characterized by an abnormal accumulation of extracellular matrix (ECM). There are no specific anti-fibrotic treatments for liver fibrosis, and previous unsuccessful attempts at drug development have focused on preventing ECM deposition. Because liver fibrosis is largely acknowledged to be reversible, regulating fibrosis resolution could offer novel therapeutical options. However, little is known about the mechanisms controlling ECM remodeling during resolution. Changes in proteolytic activity are essential for ECM homeostasis and macrophages are an important source of proteases. Herein, in this study we evaluate the role of macrophage-derived cathepsin D (CtsD) during liver fibrosis. METHODS CtsD expression and associated pathways were characterized in single-cell RNA sequencing and transcriptomic datasets in human cirrhosis. Liver fibrosis progression, reversion and functional characterization were assessed in novel myeloid-CtsD and hepatocyte-CtsD knock-out mice. RESULTS Analysis of single-cell RNA sequencing datasets demonstrated CtsD was expressed in macrophages and hepatocytes in human cirrhosis. Liver fibrosis progression, reversion and functional characterization were assessed in novel myeloid-CtsD (CtsDΔMyel) and hepatocyte-CtsD knock-out mice. CtsD deletion in macrophages, but not in hepatocytes, resulted in enhanced liver fibrosis. Both inflammatory and matrisome proteomic signatures were enriched in fibrotic CtsDΔMyel livers. Besides, CtsDΔMyel liver macrophages displayed functional, phenotypical and secretomic changes, which resulted in a degradomic phenotypical shift, responsible for the defective proteolytic processing of collagen I in vitro and impaired collagen remodeling during fibrosis resolution in vivo. Finally, CtsD-expressing mononuclear phagocytes of cirrhotic human livers were enriched in lysosomal and ECM degradative signaling pathways. CONCLUSIONS Our work describes for the first-time CtsD-driven lysosomal activity as a central hub for restorative macrophage function during fibrosis resolution and opens new avenues to explore their degradome landscape to inform drug development.
Collapse
Affiliation(s)
- Paloma Ruiz-Blázquez
- Institute of Biomedical Research of Barcelona, Spanish National Research Council, Barcelona, Spain; CiberEHD, Spain; University of Barcelona, Barcelona, Spain; IDIBAPS, Barcelona, Spain
| | - María Fernández-Fernández
- Institute of Biomedical Research of Barcelona, Spanish National Research Council, Barcelona, Spain; CiberEHD, Spain; University of Barcelona, Barcelona, Spain; IDIBAPS, Barcelona, Spain
| | - Valeria Pistorio
- Institute of Biomedical Research of Barcelona, Spanish National Research Council, Barcelona, Spain; Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy; Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine (CRSA), Paris, France
| | | | - Michele Costanzo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy; CEINGE-Biotecnologie Avanzate Franco Salvatore s.c.ar.l., Naples, Italy
| | - Paula Iruzubieta
- Department of Gastroenterology and Hepatology, Marqués de Valdecilla University Hospital, Research Institute Marqués de Valdecilla (IDIVAL), Santander, Spain
| | - Ekaterina Zhuravleva
- Biotech Research and Innovation Centre (BRIC), Department of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; LEO Foundation Skin Immunology Research Center (SIC), Department of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Júlia Cacho-Pujol
- Institute of Biomedical Research of Barcelona, Spanish National Research Council, Barcelona, Spain; University of Barcelona, Barcelona, Spain; IDIBAPS, Barcelona, Spain
| | - Silvia Ariño
- CiberEHD, Spain; University of Barcelona, Barcelona, Spain; IDIBAPS, Barcelona, Spain
| | | | | | - Jesper B Andersen
- Biotech Research and Innovation Centre (BRIC), Department of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Margherita Ruoppolo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy; CEINGE-Biotecnologie Avanzate Franco Salvatore s.c.ar.l., Naples, Italy
| | - Javier Crespo
- Department of Gastroenterology and Hepatology, Marqués de Valdecilla University Hospital, Research Institute Marqués de Valdecilla (IDIVAL), Santander, Spain
| | - Carmen García-Ruiz
- Institute of Biomedical Research of Barcelona, Spanish National Research Council, Barcelona, Spain; CiberEHD, Spain; IDIBAPS, Barcelona, Spain; USC Research Center for ALPD, Los Angeles, United States; Associated Unit IIBB-IMIM, Barcelona, Spain
| | - Luigi Michele Pavone
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Thomas Reinheckel
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, Albert-Ludwigs-University, Freiburg, Germany; German Cancer Consortium (DKTK), DKFZ Partner Site Freiburg, Germany; Center for Biological Signaling Studies BIOSS, University of Freiburg, Germany
| | - Pau Sancho-Bru
- CiberEHD, Spain; University of Barcelona, Barcelona, Spain; IDIBAPS, Barcelona, Spain
| | - Mar Coll
- CiberEHD, Spain; University of Barcelona, Barcelona, Spain; IDIBAPS, Barcelona, Spain; Medicine Department, Faculty of Medicine, University of Barcelona, Spain
| | - José C Fernández-Checa
- Institute of Biomedical Research of Barcelona, Spanish National Research Council, Barcelona, Spain; CiberEHD, Spain; IDIBAPS, Barcelona, Spain; USC Research Center for ALPD, Los Angeles, United States; Associated Unit IIBB-IMIM, Barcelona, Spain
| | - Anna Moles
- Institute of Biomedical Research of Barcelona, Spanish National Research Council, Barcelona, Spain; CiberEHD, Spain; IDIBAPS, Barcelona, Spain; Associated Unit IIBB-IMIM, Barcelona, Spain.
| |
Collapse
|
19
|
Karnawat K, Parthasarathy R, Sakhrie M, Karthik H, Krishna KV, Balachander GM. Building in vitro models for mechanistic understanding of liver regeneration in chronic liver diseases. J Mater Chem B 2024; 12:7669-7691. [PMID: 38973693 DOI: 10.1039/d4tb00738g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/09/2024]
Abstract
The liver has excellent regeneration potential and attains complete functional recovery from partial hepatectomy. The regenerative mechanisms malfunction in chronic liver diseases (CLDs), which fuels disease progression. CLDs account for 2 million deaths per year worldwide. Pathophysiological studies with clinical correlation have shown evidence of deviation of normal regenerative mechanisms and its contribution to fueling fibrosis and disease progression. However, we lack realistic in vitro models that can allow experimental manipulation for mechanistic understanding of liver regeneration in CLDs and testing of candidate drugs. In this review, we aim to provide the framework for building appropriate organotypic models for dissecting regenerative responses in CLDs, with the focus on non-alcoholic steatohepatitis (NASH). By drawing parallels with development and hepatectomy, we explain the selection of critical components such as cells, signaling, and, substrate-driven biophysical cues to build an appropriate CLD model. We highlight the organoid-based organotypic models available for NASH disease modeling, including organ-on-a-chip and 3D bioprinted models. With the focus on bioprinting as a fabrication method, we prescribe building in vitro CLD models and testing schemes for exploring the regenerative responses in the bioprinted model.
Collapse
Affiliation(s)
- Khushi Karnawat
- School of Biomedical Engineering, Indian Institute of Technology (BHU) Varanasi, Varanasi-221005, India.
| | - Rithika Parthasarathy
- School of Biomedical Engineering, Indian Institute of Technology (BHU) Varanasi, Varanasi-221005, India.
| | - Mesevilhou Sakhrie
- School of Biomedical Engineering, Indian Institute of Technology (BHU) Varanasi, Varanasi-221005, India.
| | - Harikeshav Karthik
- School of Biomedical Engineering, Indian Institute of Technology (BHU) Varanasi, Varanasi-221005, India.
| | - Konatala Vibhuvan Krishna
- School of Biomedical Engineering, Indian Institute of Technology (BHU) Varanasi, Varanasi-221005, India.
| | - Gowri Manohari Balachander
- School of Biomedical Engineering, Indian Institute of Technology (BHU) Varanasi, Varanasi-221005, India.
| |
Collapse
|
20
|
Vashisth C, Kaushik T, Vashisth N, Raghav N. Cinnamaldehyde hydrazone derivatives as potential cathepsin B inhibitors: parallel in-vitro investigation in liver and cerebrospinal fluid. Int J Biol Macromol 2024; 272:132684. [PMID: 38810845 DOI: 10.1016/j.ijbiomac.2024.132684] [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: 02/19/2024] [Revised: 05/14/2024] [Accepted: 05/24/2024] [Indexed: 05/31/2024]
Abstract
The emergence of cathepsins as a potential target for anticancer drugs has led to extensive research in the development of their inhibitors. In the present study, we designed, synthesized, and characterized several cinnamaldehyde schiff bases employing diverse hydrazines, as potential cathepsin B inhibitors. The parallel studies on cathepsin B isolated from liver and cerebrospinal fluid unveiled the significance of the synthesized compounds as cathepsin B inhibitors at nanomolar concentrations. The compound, 7 exhibited the highest inhibition of 83.48 % and 82.96 % with an IC50 value of 0.06 nM and 0.09 nM for liver and cerebrospinal fluid respectively. The inhibitory potential of synthesized compounds has been extremely effective in comparison to previous reports. With the help of molecular docking studies using iGEMDOCK software, we found that the active site -CH2SH group is involved in the case of α-N-benzoyl-D, l-arginine-b-naphthylamide (BANA), curcumin 2, 3, 6, and 7. For toxicity prediction, ADMET studies were conducted and the synthesized compounds emerged to be non-toxic. The results obtained from the in vitro studies were supported with in silico studies. The synthesized cinnamaldehyde schiff bases can be considered promising drug candidates in conditions with elevated cathepsin B levels.
Collapse
Affiliation(s)
- Chanchal Vashisth
- Department of Chemistry, Kurukshetra University, Kurukshetra, Haryana 136119, India
| | - Tushar Kaushik
- Lala Lajpat Rai Memorial Medical College (LLRM), Meerut, Uttar Pradesh 250004, India
| | - Naman Vashisth
- Mahatma Gandhi Memorial Medical College, Indore, Madhya Pradesh 452001, India
| | - Neera Raghav
- Department of Chemistry, Kurukshetra University, Kurukshetra, Haryana 136119, India.
| |
Collapse
|
21
|
Deng T, Lu X, Jia X, Du J, Wang L, Cao B, Yang M, Yin Y, Liu F. Cathepsins and cancer risk: a Mendelian randomization study. Front Endocrinol (Lausanne) 2024; 15:1428433. [PMID: 38883596 PMCID: PMC11176415 DOI: 10.3389/fendo.2024.1428433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 05/17/2024] [Indexed: 06/18/2024] Open
Abstract
Background Previous observational epidemiological studies reported an association between cathepsins and cancer, however, a causal relationship is uncertain. This study evaluated the causal relationship between cathepsins and cancer using Mendelian randomization (MR) analysis. Methods We used publicly available genome-wide association study (GWAS) data for bidirectional MR analysis. Inverse variance weighting (IVW) was used as the primary MR method of MR analysis. Results After correction for the False Discovery Rate (FDR), two cathepsins were found to be significantly associated with cancer risk: cathepsin H (CTSH) levels increased the risk of lung cancer (OR = 1.070, 95% CI = 1.027-1.114, P = 0.001, PFDR = 0.009), and CTSH levels decreased the risk of basal cell carcinoma (OR = 0.947, 95% CI = 0.919-0.975, P = 0.0002, P FDR = 0.002). In addition, there was no statistically significant effect of the 20 cancers on the nine cathepsins. Some unadjusted low P-value phenotypes are worth mentioning, including a positive correlation between cathepsin O (CTSO) and breast cancer (OR = 1.012, 95% CI = 1.001-1.025, P = 0.041), cathepsin S (CTSS) and pharyngeal cancer (OR = 1.017, 95% CI = 1.001-1.034, P = 0.043), and CTSS and endometrial cancer (OR = 1.055, 95% CI = 1.012-1.101, P = 0.012); and there was a negative correlation between cathepsin Z and ovarian cancer (CTSZ) (OR = 0.970, 95% CI = 0.949-0.991, P = 0.006), CTSS and prostate cancer (OR = 0.947, 95% CI = 0.902-0.944, P = 0.028), and cathepsin E (CTSE) and pancreatic cancer (OR = 0.963, 95% CI = 0.938-0.990, P = 0.006). Conclusion Our MR analyses showed a causal relationship between cathepsins and cancers and may help provide new insights for further mechanistic and clinical studies of cathepsin-mediated cancer.
Collapse
Affiliation(s)
- Tingting Deng
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xixue Lu
- Bone Biomechanics Engineering Laboratory of Shandong Province, Shandong Medicinal Biotechnology Center (School of Biomedical Sciences), Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Xuemin Jia
- Bone Biomechanics Engineering Laboratory of Shandong Province, Shandong Medicinal Biotechnology Center (School of Biomedical Sciences), Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Jinxin Du
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lijuan Wang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Bone Biomechanics Engineering Laboratory of Shandong Province, Shandong Medicinal Biotechnology Center (School of Biomedical Sciences), Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Baorui Cao
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Meina Yang
- National Health Commission (NHC) Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Biomedical Sciences College, Shandong First Medical University, Jinan, China
- Department of Endocrinology, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Ying Yin
- Department of Acupuncture, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Fanjie Liu
- Bone Biomechanics Engineering Laboratory of Shandong Province, Shandong Medicinal Biotechnology Center (School of Biomedical Sciences), Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
22
|
Meroni M, De Caro E, Chiappori F, Longo M, Paolini E, Mosca E, Merelli I, Lombardi R, Badiali S, Maggioni M, Orro A, Mezzelani A, Valenti L, Fracanzani AL, Dongiovanni P. Hepatic and adipose tissue transcriptome analysis highlights a commonly deregulated autophagic pathway in severe MASLD. Obesity (Silver Spring) 2024; 32:923-937. [PMID: 38439203 DOI: 10.1002/oby.23996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/21/2023] [Accepted: 01/02/2024] [Indexed: 03/06/2024]
Abstract
OBJECTIVE The incidence of metabolic dysfunction-associated steatotic liver disease (MASLD) is rapidly ramping up due to the spread of obesity, which is characterized by expanded and dysfunctional visceral adipose tissue (VAT). Previous studies have investigated the hepatic transcriptome across MASLD, whereas few studies have focused on VAT. METHODS We performed RNA sequencing in 167 hepatic samples from patients with obesity and in a subset of 79 matched VAT samples. Circulating cathepsin D (CTSD), a lysosomal protease, was measured by ELISA, whereas the autophagy-lysosomal pathway was assessed by Western blot in hepatic and VAT samples (n = 20). RESULTS Inflammation, extracellular matrix remodeling, and mitochondrial dysfunction were upregulated in severe MASLD in both tissues, whereas autophagy and oxidative phosphorylation were reduced. Tissue comparative analysis revealed 13 deregulated genes, including CTSD, which showed the most robust diagnostic accuracy in discriminating mild and severe MASLD. CTSD expression correlated with circulating protein, whose increase was further validated in 432 histologically characterized MASLD patients, showing a high accuracy in foreseeing severe liver injury. In addition, the assessment of serum CTSD increased the performance of fibrosis 4 in diagnosing advanced disease. CONCLUSIONS By comparing the hepatic and VAT transcriptome during MASLD, we refined the concept by which CTSD may represent a potential biomarker of severe disease.
Collapse
Affiliation(s)
- Marica Meroni
- Medicine and Metabolic Diseases, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Emilia De Caro
- Life and Medical Sciences Institute (LIMES), University of Bonn, Germany/System Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Federica Chiappori
- Institute for Biomedical Technologies, National Research Council (ITB-CNR), Segrate, Italy
| | - Miriam Longo
- Medicine and Metabolic Diseases, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Erika Paolini
- Medicine and Metabolic Diseases, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Ettore Mosca
- Institute for Biomedical Technologies, National Research Council (ITB-CNR), Segrate, Italy
| | - Ivan Merelli
- Institute for Biomedical Technologies, National Research Council (ITB-CNR), Segrate, Italy
| | - Rosa Lombardi
- Medicine and Metabolic Diseases, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Sara Badiali
- Department of Surgery, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Marco Maggioni
- Department of Pathology, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Alessandro Orro
- Institute for Biomedical Technologies, National Research Council (ITB-CNR), Segrate, Italy
| | - Alessandra Mezzelani
- Institute for Biomedical Technologies, National Research Council (ITB-CNR), Segrate, Italy
| | - Luca Valenti
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
- Precision Medicine Lab, Biological Resource Center, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Anna Ludovica Fracanzani
- Medicine and Metabolic Diseases, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Paola Dongiovanni
- Medicine and Metabolic Diseases, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
23
|
Pawłowska M, Mila-Kierzenkowska C. Effect of Alpha-1 Antitrypsin and Irisin on Post-Exercise Inflammatory Response: A Narrative Review. IRANIAN JOURNAL OF MEDICAL SCIENCES 2024; 49:205-218. [PMID: 38680225 PMCID: PMC11053258 DOI: 10.30476/ijms.2023.97480.2925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/12/2023] [Accepted: 02/16/2023] [Indexed: 05/01/2024]
Abstract
Physical activity has a positive effect on human health and emotional well-being. However, in both amateur and professional athletes, training poses a risk of acute or chronic injury through repetitive overloading of bones, joints, and muscles. Inflammation can be an adverse effect of intense exercise caused by several factors including oxidative stress. The present narrative review summarizes current knowledge on inflammatory markers induced by physical exercise. Post-exercise recovery may reduce inflammatory responses and is key to effective training and adaptation of muscle tissues to sustained physical exertion.
Collapse
Affiliation(s)
- Marta Pawłowska
- Department of Medical Biology and Biochemistry, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Poland
| | - Celestyna Mila-Kierzenkowska
- Department of Medical Biology and Biochemistry, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Poland
| |
Collapse
|
24
|
Huang X, Deng H, Zhang B, Wang K, Qu Y, Li T, Liu T. The causal relationship between cathepsins and digestive system tumors: a Mendelian randomization study. Front Oncol 2024; 14:1365138. [PMID: 38590662 PMCID: PMC10999587 DOI: 10.3389/fonc.2024.1365138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 03/14/2024] [Indexed: 04/10/2024] Open
Abstract
Background Multiple studies have confirmed the significant role of cathepsins in the development and progression of digestive system tumors. However, further investigation is needed to determine the causal relationships. Methods We conducted a two-sample bidirectional Mendelian randomization (MR) study using pooled data from a genome-wide association study (GWAS) to assess the causal associations between nine cathepsins (cathepsin B, E, F, G, H, L2, O, S, and Z) and six types of digestive system tumors, including hepatocellular carcinoma (HCC), pancreatic cancer (PCa), biliary tract cancer (BTC), colorectal cancer (CRC), gastric carcinoma (GC), and esophageal cancer (EC). We employed the following methods including inverse variance weighting (IVW), MR-Egger, weighted median (WM), Cochran's Q, MR-PRESSO, MR-Egger intercept test and leave-one-out sensitivity analysis. The STROBE-MR checklist for the reporting of MR studies was used in this study. Results The risk of HCC increased with high levels of cathepsin G (IVW: p = 0.029, odds ratio (OR) = 1.369, 95% confidence interval (CI) = 1.033-1.814). Similarly, BTC was associated with elevated cathepsin B levels (IVW: p = 0.025, OR = 1.693, 95% CI = 1.070-2.681). Conversely, a reduction in PCa risk was associated with increased cathepsin H levels (IVW: p = 0.027, OR = 0.896, 95% CI = 0.812-0.988). Lastly, high levels of cathepsin L2 were found to lower the risk of CRC (IVW: p = 0.034, OR = 0.814, 95% CI = 0.674-0.985). Conclusion Our findings confirm the causal relationship between cathepsins and digestive system tumors, which can offer valuable insights for the diagnosis and treatment of digestive system tumors.
Collapse
Affiliation(s)
- Xupeng Huang
- Graduate School, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Houbo Deng
- Department of Hepatology, First Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Bo Zhang
- Graduate School, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Kuisong Wang
- Graduate School, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Yi Qu
- Graduate School, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Ting Li
- Department of Hepatology, First Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Tiejun Liu
- Department of Hepatology, First Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
25
|
Kulik U, Moesta C, Spanel R, Borlak J. Dysfunctional Cori and Krebs cycle and inhibition of lactate transporters constitute a mechanism of primary nonfunction of fatty liver allografts. Transl Res 2024; 264:33-65. [PMID: 37722450 DOI: 10.1016/j.trsl.2023.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 09/06/2023] [Accepted: 09/06/2023] [Indexed: 09/20/2023]
Abstract
Orthotopic liver transplantation (OLT) is a lifesaving procedure. However, grafts may fail due to primary nonfunction (PNF). In the past, we demonstrated PNFs to be mainly associated with fatty allografts, and given its unpredictable nature, the development of a disease model is urgently needed. In an effort to investigate mechanism of fatty allograft-associated PNFs, we induced fatty liver disease in donor animals by feeding rats a diet deficient in methionine and choline (MCD). We performed OLT with allografts of different grades of hepatic steatosis and compared the results to healthy ones. We assessed liver function by considering serum biochemistries, and investigated genome wide responses following OLT of healthy and fatty allograft-associated PNFs. Furthermore, we performed immunohistochemistry to evaluate markers of oxidative stress and reperfusion injury, inflammation, glycolysis and gluconeogenesis, lactate transport, and its utilization as part of the Cori cycle. Strikingly, PNFs are strictly lipid content dependent. Nonetheless, a fat content of ≤17% and an increase in the size of hepatocytes of ≤11% (ballooning) greatly improved outcome of OLTs and the hepatic microcirculation. Mechanistically, PNFs arise from a dysfunctional Cori cycle with complete ablation of the lactate transporter SLC16A1. Thus, lipid-laden hepatocytes fail to perform gluconeogenesis via lactate reutilization, and the resultant hyperlactatemia and lactic acidosis causes cardiac arrhythmogenicity and death. Furthermore, the genomic and immunohistochemistry investigations underscore a dysfunctional Krebs cycle with impaired energy metabolism in lipid-burdened mitochondria. Together, we show fatty allografts to be highly vulnerable towards ischemia/reperfusion-injury, and stabilizing the Cori cycle is of critical importance to avert PNFs.
Collapse
Affiliation(s)
- Ulf Kulik
- Department of General, Visceral- and Transplantation Surgery, Hannover Medical School, Hannover, Germany
| | - Caroline Moesta
- Centre for Pharmacology and Toxicology, Hannover Medical School, Hannover, Germany
| | - Reinhard Spanel
- Centre for Pharmacology and Toxicology, Hannover Medical School, Hannover, Germany
| | - Jürgen Borlak
- Centre for Pharmacology and Toxicology, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
26
|
Dai Q, Xie L, Ren E, Liu G. Cathepsin B Responsive Peptide-Purpurin Conjugates Assembly-Initiated in Situ Self-Aggregation for Cancer Sonotheranostics. NANO LETTERS 2024; 24:950-957. [PMID: 38198622 DOI: 10.1021/acs.nanolett.3c04371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2024]
Abstract
Sonodynamic therapy (SDT) was hampered by the sonosensitizers with low bioavailability, tumor accumulation, and therapeutic efficiency. In situ responsive sonosensitizer self-assembly strategy may provide a promising route for cancer sonotheranositics. Herein, an intelligent sonotheranostic peptide-purpurin conjugate (P18-P) is developed that can self-assemble into supramolecular structures via self-aggregation triggered by rich enzyme cathepsin B (CTSB). After intravenous injection, the versatile probe could achieve deep tissue penetration because of the penetration sequence of P18-P. More importantly, CTSB-triggered self-assembly strongly prolonged retention time, amplified photoacoustic imaging signal for sensitive CTSB detection, and boosted reactive oxygen species for advanced SDT, evoking specific CTSB responsive sonotheranostics. This peptide-purpurin conjugate may serve as an efficient sonotheranostic platform for the early diagnosis of CTSB activity and effective cancer therapy.
Collapse
Affiliation(s)
- Qixuan Dai
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Lisi Xie
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, China
| | - En Ren
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Gang Liu
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| |
Collapse
|
27
|
Kasera H, Shekhawat RS, Yadav P, Singh P. Gene expression profiling and protein-protein network analysis revealed prognostic hub biomarkers linking cancer risk in type 2 diabetic patients. Sci Rep 2023; 13:22605. [PMID: 38114687 PMCID: PMC10730526 DOI: 10.1038/s41598-023-49715-9] [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: 07/27/2023] [Accepted: 12/11/2023] [Indexed: 12/21/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) and cancer are highly prevalent diseases imposing major health burden globally. Several epidemiological studies indicate increased susceptibility to cancer in T2DM patients. However, genetic factors linking T2DM with cancer have been poorly studied. In this study, we followed computational approaches using the raw gene expression data of peripheral blood mononuclear cells of T2DM and cancer patients available in the gene expression omnibus (GEO) database. Our analysis identified shared differentially expressed genes (DEGs) in T2DM and three common cancer types, namely, pancreatic cancer (PC), liver cancer (LC), and breast cancer (BC). The functional and pathway enrichment analysis of identified common DEGs highlighted the involvement of critical biological pathways, including cell cycle events, immune system processes, cell morphogenesis, gene expression, and metabolism. We retrieved the protein-protein interaction network for the top DEGs to deduce molecular-level interactions. The network analysis found 7, 6, and 5 common hub genes in T2DM vs. PC, T2DM vs. LC, and T2DM vs. BC comparisons, respectively. Overall, our analysis identified important genetic markers potentially able to predict the chances of PC, LC, and BC onset in T2DM patients.
Collapse
Affiliation(s)
- Harshita Kasera
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, NH 62, Nagaur Road, Karwar, Jodhpur, Rajasthan, 342037, India
| | - Rajveer Singh Shekhawat
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, NH 62, Nagaur Road, Karwar, Jodhpur, Rajasthan, 342037, India
| | - Pankaj Yadav
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, NH 62, Nagaur Road, Karwar, Jodhpur, Rajasthan, 342037, India.
| | - Priyanka Singh
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, NH 62, Nagaur Road, Karwar, Jodhpur, Rajasthan, 342037, India.
| |
Collapse
|
28
|
Zhang C, Zhang D, Huang H, Lu X, Shi H, Liu K, Guo X, Zhang R, Wang H. Cathepsin D mediates prenatal caffeine exposure-caused NAFLD susceptibility in male rat offspring by regulating autophagy. Free Radic Biol Med 2023; 208:684-699. [PMID: 37743032 DOI: 10.1016/j.freeradbiomed.2023.09.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 09/16/2023] [Accepted: 09/22/2023] [Indexed: 09/26/2023]
Abstract
Epidemiological evidence has revealed that non-alcoholic fatty liver disease (NAFLD) harbors an intrauterine origin. Autophagy is known to be involved in the protective mechanism in the development of adult NAFLD, but whether it engages in the occurrence of fetal-originated NAFLD remains unclear. In this study, a rat model of fetal-originated NAFLD was established by giving a high-fat diet or chronic stress after birth on prenatal caffeine exposure (PCE) male offspring. The alterations of liver morphologic analysis, lipid metabolism, and autophagy before and after birth were determined to confirm autophagy mechanism, NAFLD susceptibility, and intrauterine origin in PCE male adult offspring. Our results revealed that PCE-induced intrauterine high concentration of corticosterone exposure blocked autophagic flux by inhibiting cathepsin D expression in hepatocytes, leading to β-oxidation inhibition and lipid accumulation in the liver. Moreover, high concentration of corticosterone upregulated miR-665 by activating the glucocorticoid receptor to suppress cathepsin D, thus causing lysosomal degradation dysfunction and autophagy flux blockade. Notably, hepatic overexpression of cathepsin D could reverse PCE-induced postnatal NAFLD susceptibility in male rat offspring. This study elucidates the epigenetic programming mechanism of intrauterine autophagy-related fetal-originated NAFLD susceptibility, and identifies cathepsin D as its early intervention target, providing an experimental basis for exploring early prevention and treatment strategies for fetal-originated NAFLD.
Collapse
Affiliation(s)
- Cong Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Dingmei Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Hegui Huang
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China; Wuhan No.1 Hospital, Wuhan, 430022, China
| | - Xiaoqian Lu
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Huasong Shi
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Kexin Liu
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Xiaoling Guo
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Rui Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Hui Wang
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China.
| |
Collapse
|
29
|
Dipali SS, King CD, Rose JP, Burdette JE, Campisi J, Schilling B, Duncan FE. Proteomic quantification of native and ECM-enriched mouse ovaries reveals an age-dependent fibro-inflammatory signature. Aging (Albany NY) 2023; 15:10821-10855. [PMID: 37899138 PMCID: PMC10637783 DOI: 10.18632/aging.205190] [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: 07/17/2023] [Accepted: 10/02/2023] [Indexed: 10/31/2023]
Abstract
The ovarian microenvironment becomes fibrotic and stiff with age, in part due to increased collagen and decreased hyaluronan. However, the extracellular matrix (ECM) is a complex network of hundreds of proteins, glycoproteins, and glycans which are highly tissue specific and undergo pronounced changes with age. To obtain an unbiased and comprehensive profile of age-associated alterations to the murine ovarian proteome and ECM, we used a label-free quantitative proteomic methodology. We validated conditions to enrich for the ECM prior to proteomic analysis. Following analysis by data-independent acquisition (DIA) and quantitative data processing, we observed that both native and ECM-enriched ovaries clustered separately based on age, indicating distinct age-dependent proteomic signatures. We identified a total of 4,721 proteins from both native and ECM-enriched ovaries, of which 383 proteins were significantly altered with advanced age, including 58 ECM proteins. Several ECM proteins upregulated with age have been associated with fibrosis in other organs, but to date their roles in ovarian fibrosis are unknown. Pathways regulating DNA metabolism and translation were downregulated with age, whereas pathways involved in ECM remodeling and immune response were upregulated. Interestingly, immune-related pathways were upregulated with age even in ECM-enriched ovaries, suggesting a novel interplay between the ECM and the immune system. Moreover, we identified putative markers of unique immune cell populations present in the ovary with age. These findings provide evidence from a proteomic perspective that the aging ovary provides a fibroinflammatory milieu, and our study suggests target proteins which may drive these age-associated phenotypes for future investigation.
Collapse
Affiliation(s)
- Shweta S. Dipali
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | | | - Jacob P. Rose
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Joanna E. Burdette
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Judith Campisi
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | | | - Francesca E. Duncan
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| |
Collapse
|
30
|
Ji ZZ, Chan MKK, Chan ASW, Leung KT, Jiang X, To KF, Wu Y, Tang PMK. Tumour-associated macrophages: versatile players in the tumour microenvironment. Front Cell Dev Biol 2023; 11:1261749. [PMID: 37965573 PMCID: PMC10641386 DOI: 10.3389/fcell.2023.1261749] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 10/12/2023] [Indexed: 11/16/2023] Open
Abstract
Tumour-Associated Macrophages (TAMs) are one of the pivotal components of the tumour microenvironment. Their roles in the cancer immunity are complicated, both pro-tumour and anti-cancer activities are reported, including not only angiogenesis, extracellular matrix remodeling, immunosuppression, drug resistance but also phagocytosis and tumour regression. Interestingly, TAMs are highly dynamic and versatile in solid tumours. They show anti-cancer or pro-tumour activities, and interplay between the tumour microenvironment and cancer stem cells and under specific conditions. In addition to the classic M1/M2 phenotypes, a number of novel dedifferentiation phenomena of TAMs are discovered due to the advanced single-cell technology, e.g., macrophage-myofibroblast transition (MMT) and macrophage-neuron transition (MNT). More importantly, emerging information demonstrated the potential of TAMs on cancer immunotherapy, suggesting by the therapeutic efficiency of the checkpoint inhibitors and chimeric antigen receptor engineered cells based on macrophages. Here, we summarized the latest discoveries of TAMs from basic and translational research and discussed their clinical relevance and therapeutic potential for solid cancers.
Collapse
Affiliation(s)
- Zoey Zeyuan Ji
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Max Kam-Kwan Chan
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Alex Siu-Wing Chan
- Department of Applied Social Sciences, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Kam-Tong Leung
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Xiaohua Jiang
- Key Laboratory for Regenerative Medicine of the Ministry of Education of China, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Ka-Fai To
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Yi Wu
- MOE Key Laboratory of Environment and Genes Related to Diseases, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, China
| | - Patrick Ming-Kuen Tang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| |
Collapse
|
31
|
Zhao S, Jiang M, Qing H, Ni J. Cathepsins and SARS-CoV-2 infection: From pathogenic factors to potential therapeutic targets. Br J Pharmacol 2023; 180:2455-2481. [PMID: 37403614 DOI: 10.1111/bph.16187] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 05/04/2023] [Accepted: 06/20/2023] [Indexed: 07/06/2023] Open
Abstract
Coronavirus disease-19 (COVID-19) is caused by severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) infection. The COVID-19 pandemic began in March 2020 and has wrought havoc on health and economic systems worldwide. Efficacious treatment for COVID-19 is lacking: Only preventive measures as well as symptomatic and supportive care are available. Preclinical and clinical studies have indicated that lysosomal cathepsins might contribute to the pathogenesis and disease outcome of COVID-19. Here, we discuss cutting-edge evidence on the pathological roles of cathepsins in SARS-CoV-2 infection, host immune dysregulations, and the possible underlying mechanisms. Cathepsins are attractive drug targets because of their defined substrate-binding pockets, which can be exploited as binding sites for pharmaceutical enzyme inhibitors. Accordingly, the potential modulatory strategies of cathepsin activity are discussed. These insights could shed light on the development of cathepsin-based interventions for COVID-19.
Collapse
Affiliation(s)
- Shuxuan Zhao
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Muzhou Jiang
- Department of Periodontics, Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang, China
| | - Hong Qing
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Junjun Ni
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
| |
Collapse
|
32
|
Li Y, Xu M, Xiang BL, Li X, Zhang DF, Zhao H, Bi R, Yao YG. Functional genomics identify causal variant underlying the protective CTSH locus for Alzheimer's disease. Neuropsychopharmacology 2023; 48:1555-1566. [PMID: 36739351 PMCID: PMC10516988 DOI: 10.1038/s41386-023-01542-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 12/30/2022] [Accepted: 01/25/2023] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD) is the most prevalent age-related neurodegenerative disease, which has a high heritability of up to 79%. Exploring the genetic basis is essential for understanding the pathogenic mechanisms underlying AD development. Recent genome-wide association studies (GWASs) reported an AD-associated signal in the Cathepsin H (CTSH) gene in European populations. However, the exact functional/causal variant(s), and the genetic regulating mechanism of CTSH in AD remain to be determined. In this study, we carried out a comprehensive study to characterize the role of CTSH variants in the pathogenesis of AD. We identified rs2289702 in CTSH as the most significant functional variant that is associated with a protective effect against AD. The genetic association between rs2289702 and AD was validated in independent cohorts of the Han Chinese population. The CTSH mRNA expression level was significantly increased in AD patients and AD animal models, and the protective allele T of rs2289702 was associated with a decreased expression level of CTSH through the disruption of the binding affinity of transcription factors. Human microglia cells with CTSH knockout showed a significantly increased phagocytosis of Aβ peptides. Our study identified CTSH as being involved in AD genetic susceptibility and uncovered the genetic regulating mechanism of CTSH in pathogenesis of AD.
Collapse
Affiliation(s)
- Yu Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650204, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204, Yunnan, China
| | - Min Xu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650204, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204, Yunnan, China
| | - Bo-Lin Xiang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650204, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204, Yunnan, China
| | - Xiao Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650204, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204, Yunnan, China
| | - Deng-Feng Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650204, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204, Yunnan, China
| | - Hui Zhao
- KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Yunnan, 650204, Kunming, China
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Hong Kong Branch of CAS Center for Excellence in Animal Evolution and Genetics, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Rui Bi
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650204, Yunnan, China.
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204, Yunnan, China.
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Yong-Gang Yao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650204, Yunnan, China.
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204, Yunnan, China.
- KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Yunnan, 650204, Kunming, China.
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
33
|
Zuo T, Xie Q, Liu J, Yang J, Shi J, Kong D, Wang Y, Zhang Z, Gao H, Zeng DB, Wang X, Tao P, Wei W, Wang J, Li Y, Long Q, Li C, Chang L, Ning H, Li Y, Cui C, Ge X, Wu J, Li G, Hong X, Yang X, Dai E, He F, Wu J, Ruan Y, Lu S, Xu P. Macrophage-Derived Cathepsin S Remodels the Extracellular Matrix to Promote Liver Fibrogenesis. Gastroenterology 2023; 165:746-761.e16. [PMID: 37263311 DOI: 10.1053/j.gastro.2023.05.039] [Citation(s) in RCA: 4] [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: 08/18/2022] [Revised: 05/20/2023] [Accepted: 05/25/2023] [Indexed: 06/03/2023]
Abstract
BACKGROUND & AIMS Liver fibrosis is an intrinsic wound-healing response to chronic injury and the major cause of liver-related morbidity and mortality worldwide. However, no effective diagnostic or therapeutic strategies are available, owing to its poorly characterized molecular etiology. We aimed to elucidate the mechanisms underlying liver fibrogenesis. METHODS We performed a quantitative proteomic analysis of clinical fibrotic liver samples to identify dysregulated proteins. Further analyses were performed on the sera of 164 patients with liver fibrosis. Two fibrosis mouse models and several biochemical experiments were used to elucidate liver fibrogenesis. RESULTS We identified cathepsin S (CTSS) up-regulation as a central node for extracellular matrix remodeling in the human fibrotic liver by proteomic screening. Increased serum CTSS levels efficiently predicted liver fibrosis, even at an early stage. Secreted CTSS cleaved collagen 18A1 at its C-terminus, releasing endostatin peptide, which directly bound to and activated hepatic stellate cells via integrin α5β1 signaling, whereas genetic ablation of Ctss remarkably suppressed liver fibrogenesis via endostatin reduction in vivo. Further studies identified macrophages as the main source of hepatic CTSS, and splenectomy effectively attenuated macrophage infiltration and CTSS expression in the fibrotic liver. Pharmacologic inhibition of CTSS ameliorated liver fibrosis progression in the mouse models. CONCLUSIONS CTSS functions as a novel profibrotic factor by remodeling extracellular matrix proteins and may represent a promising target for the diagnosis and treatment of liver fibrosis.
Collapse
Affiliation(s)
- Tao Zuo
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics and Research and Development of New Drug, Research Unit of Proteomics Driven Cancer Precision Medicine, Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, China
| | - Qi Xie
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics and Research and Development of New Drug, Research Unit of Proteomics Driven Cancer Precision Medicine, Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, China; TaiKang Medical School (School of Basic Medical Sciences), Key Laboratory of Combinatorial Biosynthesis and Drug Discovery of Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan, China; Department of Neurology, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jinfang Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics and Research and Development of New Drug, Research Unit of Proteomics Driven Cancer Precision Medicine, Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, China; TaiKang Medical School (School of Basic Medical Sciences), Key Laboratory of Combinatorial Biosynthesis and Drug Discovery of Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| | - Jing Yang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics and Research and Development of New Drug, Research Unit of Proteomics Driven Cancer Precision Medicine, Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, China; TaiKang Medical School (School of Basic Medical Sciences), Key Laboratory of Combinatorial Biosynthesis and Drug Discovery of Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| | - Jiahui Shi
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics and Research and Development of New Drug, Research Unit of Proteomics Driven Cancer Precision Medicine, Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, China
| | - Degang Kong
- Faculty of Hepato-Pancreato-Biliary Surgery, Institute of Hepatobiliary Surgery, Key Laboratory of Digital Hepatobiliary Surgery, Chinese People's Liberation Army Medical School, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yin Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics and Research and Development of New Drug, Research Unit of Proteomics Driven Cancer Precision Medicine, Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, China; TaiKang Medical School (School of Basic Medical Sciences), Key Laboratory of Combinatorial Biosynthesis and Drug Discovery of Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| | - Zhenpeng Zhang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics and Research and Development of New Drug, Research Unit of Proteomics Driven Cancer Precision Medicine, Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, China
| | - Huixia Gao
- Second Department of Internal Medicine, Shijiazhuang Fifth Hospital, Shijiazhuang, China
| | - Dao-Bing Zeng
- Bejing You-An Hospital, Capital Medical University, Beijing, China
| | - Xinxin Wang
- Bejing You-An Hospital, Capital Medical University, Beijing, China
| | - Ping Tao
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics and Research and Development of New Drug, Research Unit of Proteomics Driven Cancer Precision Medicine, Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, China; Bejing You-An Hospital, Capital Medical University, Beijing, China
| | - Wei Wei
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics and Research and Development of New Drug, Research Unit of Proteomics Driven Cancer Precision Medicine, Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, China
| | - Jun Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics and Research and Development of New Drug, Research Unit of Proteomics Driven Cancer Precision Medicine, Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, China
| | - Yuan Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics and Research and Development of New Drug, Research Unit of Proteomics Driven Cancer Precision Medicine, Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qi Long
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics and Research and Development of New Drug, Research Unit of Proteomics Driven Cancer Precision Medicine, Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, China
| | - Chonghui Li
- Faculty of Hepato-Pancreato-Biliary Surgery, Institute of Hepatobiliary Surgery, Key Laboratory of Digital Hepatobiliary Surgery, Chinese People's Liberation Army Medical School, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Lei Chang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics and Research and Development of New Drug, Research Unit of Proteomics Driven Cancer Precision Medicine, Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, China
| | - Huimin Ning
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics and Research and Development of New Drug, Research Unit of Proteomics Driven Cancer Precision Medicine, Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, China
| | - Yanchang Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics and Research and Development of New Drug, Research Unit of Proteomics Driven Cancer Precision Medicine, Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, China
| | - Chunping Cui
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics and Research and Development of New Drug, Research Unit of Proteomics Driven Cancer Precision Medicine, Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, China
| | - Xinlan Ge
- Faculty of Hepato-Pancreato-Biliary Surgery, Institute of Hepatobiliary Surgery, Key Laboratory of Digital Hepatobiliary Surgery, Chinese People's Liberation Army Medical School, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Jushan Wu
- Bejing You-An Hospital, Capital Medical University, Beijing, China
| | - Guangming Li
- Bejing You-An Hospital, Capital Medical University, Beijing, China
| | - Xuechuan Hong
- TaiKang Medical School (School of Basic Medical Sciences), Key Laboratory of Combinatorial Biosynthesis and Drug Discovery of Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| | - Xiao Yang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics and Research and Development of New Drug, Research Unit of Proteomics Driven Cancer Precision Medicine, Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, China
| | - Erhei Dai
- Second Department of Internal Medicine, Shijiazhuang Fifth Hospital, Shijiazhuang, China
| | - Fuchu He
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics and Research and Development of New Drug, Research Unit of Proteomics Driven Cancer Precision Medicine, Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, China
| | - Junzhu Wu
- TaiKang Medical School (School of Basic Medical Sciences), Key Laboratory of Combinatorial Biosynthesis and Drug Discovery of Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan, China.
| | - Yuanyuan Ruan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China.
| | - Shichun Lu
- Faculty of Hepato-Pancreato-Biliary Surgery, Institute of Hepatobiliary Surgery, Key Laboratory of Digital Hepatobiliary Surgery, Chinese People's Liberation Army Medical School, Chinese People's Liberation Army General Hospital, Beijing, China.
| | - Ping Xu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics and Research and Development of New Drug, Research Unit of Proteomics Driven Cancer Precision Medicine, Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, China; TaiKang Medical School (School of Basic Medical Sciences), Key Laboratory of Combinatorial Biosynthesis and Drug Discovery of Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Guizhou University, School of Medicine, Guiyang, China.
| |
Collapse
|
34
|
Bradić I, Liesinger L, Kuentzel KB, Vujić N, Trauner M, Birner-Gruenberger R, Kratky D. Metabolic changes and propensity for inflammation, fibrosis, and cancer in livers of mice lacking lysosomal acid lipase. J Lipid Res 2023; 64:100427. [PMID: 37595802 PMCID: PMC10482749 DOI: 10.1016/j.jlr.2023.100427] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/20/2023] [Accepted: 08/10/2023] [Indexed: 08/20/2023] Open
Abstract
Lysosomal acid lipase (LAL) is the sole lysosomal enzyme responsible for the degradation of cholesteryl esters and triacylglycerols at acidic pH. Impaired LAL activity leads to LAL deficiency (LAL-D), a severe and fatal disease characterized by ectopic lysosomal lipid accumulation. Reduced LAL activity also contributes to the development and progression of non-alcoholic fatty liver disease (NAFLD). To advance our understanding of LAL-related liver pathologies, we performed comprehensive proteomic profiling of livers from mice with systemic genetic loss of LAL (Lal-/-) and from mice with hepatocyte-specific LAL-D (hepLal-/-). Lal-/- mice exhibited drastic proteome alterations, including dysregulation of multiple proteins related to metabolism, inflammation, liver fibrosis, and cancer. Global loss of LAL activity impaired both acidic and neutral lipase activities and resulted in hepatic lipid accumulation, indicating a complete metabolic shift in Lal-/- livers. Hepatic inflammation and immune cell infiltration were evident, with numerous upregulated inflammation-related gene ontology biological process terms. In contrast, both young and mature hepLal-/- mice displayed only minor changes in the liver proteome, suggesting that loss of LAL solely in hepatocytes does not phenocopy metabolic alterations observed in mice globally lacking LAL. These findings provide valuable insights into the mechanisms underlying liver dysfunction in LAL-D and may help in understanding why decreased LAL activity contributes to NAFLD. Our study highlights the importance of LAL in maintaining liver homeostasis and demonstrates the drastic consequences of its global deficiency on the liver proteome and liver function.
Collapse
Affiliation(s)
- Ivan Bradić
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Laura Liesinger
- Institute of Chemical Technologies and Analytics, TU Wien, Vienna, Austria
| | - Katharina B Kuentzel
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Nemanja Vujić
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Michael Trauner
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Ruth Birner-Gruenberger
- Institute of Chemical Technologies and Analytics, TU Wien, Vienna, Austria; BioTechMed-Graz, Graz, Austria; Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria.
| | - Dagmar Kratky
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria.
| |
Collapse
|
35
|
Lin SZ, Wu WJ, Cheng YQ, Zhang JB, Jiang DX, Ren TY, Ding WJ, Liu M, Chen YW, Fan JG. Prolyl endopeptidase remodels macrophage function as a novel transcriptional coregulator and inhibits fibrosis. Exp Mol Med 2023; 55:1437-1450. [PMID: 37394591 PMCID: PMC10394032 DOI: 10.1038/s12276-023-01027-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 03/28/2023] [Accepted: 04/06/2023] [Indexed: 07/04/2023] Open
Abstract
Macrophages are immune cells crucial for host defense and homeostasis maintenance, and their dysregulation is involved in multiple pathological conditions, such as liver fibrosis. The transcriptional regulation in macrophage is indispensable for fine-tuning of macrophage functions, but the details have not been fully elucidated. Prolyl endopeptidase (PREP) is a dipeptidyl peptidase with both proteolytic and non-proteolytic functions. In this study, we found that Prep knockout significantly contributed to transcriptomic alterations in quiescent and M1/M2-polarized bone marrow-derived macrophages (BMDMs), as well as aggravated fibrosis in an experimental nonalcoholic steatohepatitis (NASH) model. Mechanistically, PREP predominantly localized to the macrophage nuclei and functioned as a transcriptional coregulator. Using CUT&Tag and co-immunoprecipitation, we found that PREP was mainly distributed in active cis-regulatory genomic regions and physically interacted with the transcription factor PU.1. Among PREP-regulated downstream genes, genes encoding profibrotic cathepsin B and D were overexpressed in BMDMs and fibrotic liver tissue. Our results indicate that PREP in macrophages functions as a transcriptional coregulator that finely tunes macrophage functions, and plays a protective role against liver fibrosis pathogenesis.
Collapse
Affiliation(s)
- Shuang-Zhe Lin
- Department of Gastroenterology, Xin Hua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Wei-Jie Wu
- Department of Gastroenterology, Xin Hua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Gastrointestinal Endoscopy Center, Fujian Provincial Hospital South Branch, Fuzhou, 350003, Fujian, China
| | - Yu-Qing Cheng
- Department of Gastroenterology, Xin Hua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Jian-Bin Zhang
- Department of Gastroenterology, Xin Hua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Dai-Xi Jiang
- Department of Gastroenterology, Xin Hua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Tian-Yi Ren
- Department of Gastroenterology, Xin Hua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, 200092, China
| | - Wen-Jin Ding
- Department of Gastroenterology, Xin Hua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Mingxi Liu
- State Key Laboratory of Reproductive Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Nanjing, 211166, Jiangsu, China.
| | - Yuan-Wen Chen
- Department of Gastroenterology, Huadong Hospital Affiliated to Fudan University, Shanghai, 200040, China.
- Department of Gerontology, Huadong Hospital Affiliated to Fudan University, Shanghai, 200040, China.
| | - Jian-Gao Fan
- Department of Gastroenterology, Xin Hua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, 200092, China.
| |
Collapse
|
36
|
Park SH, Lee JH, Yang SB, Lee DN, Kang TB, Park J. Development of a Peptide-Based Nano-Sized Cathepsin B Inhibitor for Anticancer Therapy. Pharmaceutics 2023; 15:pharmaceutics15041131. [PMID: 37111617 PMCID: PMC10141979 DOI: 10.3390/pharmaceutics15041131] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/17/2023] [Accepted: 03/29/2023] [Indexed: 04/07/2023] Open
Abstract
Numerous cathepsin B inhibitors have been developed and are under investigation as potential cancer treatments. They have been evaluated for their ability to inhibit cathepsin B activity and reduce tumor growth. However, they have shown critical limitations, including low anticancer efficacy and high toxicity, due to their low selectivity and delivery problems. In this study, we developed a novel peptide and drug conjugate (PDC)-based cathepsin B inhibitor using cathepsin-B-specific peptide (RR) and bile acid (BA). Interestingly, this RR and BA conjugate (RR–BA) was able to self-assemble in an aqueous solution, and as a result, it formed stable nanoparticles. The nano-sized RR–BA conjugate showed significant cathepsin B inhibitory effects and anticancer effects against mouse colorectal cancer (CT26) cells. Its therapeutic effect and low toxicity were also confirmed in CT26 tumor-bearing mice after intravenous injection. Therefore, based on these results, the RR–BA conjugate could be developed as an effective anticancer drug candidate for inhibiting cathepsin B in anticancer therapy.
Collapse
Affiliation(s)
- So-Hyeon Park
- Department of Applied Life Science, BK21 Program, Graduate School, Konkuk University, Chungju 27478, Republic of Korea
| | - Jun-Hyuck Lee
- Department of Applied Life Science, BK21 Program, Graduate School, Konkuk University, Chungju 27478, Republic of Korea
| | - Seong-Bin Yang
- Department of Applied Life Science, BK21 Program, Graduate School, Konkuk University, Chungju 27478, Republic of Korea
| | - Dong-Nyeong Lee
- Department of Applied Life Science, BK21 Program, Graduate School, Konkuk University, Chungju 27478, Republic of Korea
| | - Tae-Bong Kang
- Department of Applied Life Science, BK21 Program, Graduate School, Konkuk University, Chungju 27478, Republic of Korea
| | - Jooho Park
- Department of Applied Life Science, BK21 Program, Graduate School, Konkuk University, Chungju 27478, Republic of Korea
- Center for Metabolic Diseases, Konkuk University, Chungju 27478, Republic of Korea
| |
Collapse
|
37
|
Mechanistic Insights into the Role of OPN in Mediating Brain Damage via Triggering Lysosomal Damage in Microglia/Macrophage. Cells 2023; 12:cells12060854. [PMID: 36980197 PMCID: PMC10046941 DOI: 10.3390/cells12060854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 03/02/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
We previously found that osteopontin (OPN) played a role in hypoxia–ischemia (HI) brain damage. However, its underlying mechanism is still unknown. Bioinformatics analysis revealed that the OPN protein was linked to the lysosomal cathepsin B (CTSB) and galectin-3 (GAL-3) proteins after HI exposure. In the present study, we tested the hypothesis that OPN was able to play a critical role in the lysosomal damage of microglia/macrophages following HI insult in neonatal mice. The results showed that OPN expression was enhanced, especially in microglia/macrophages, and colocalized with lysosomal-associated membrane protein 1 (LAMP1) and GAL-3; this was accompanied by increased LAMP1 and GAL-3 expression, CTSB leakage, as well as impairment of autophagic flux in the early stage of the HI process. In addition, the knockdown of OPN expression markedly restored lysosomal function with significant improvements in the autophagic flux after HI insult. Interestingly, cleavage of OPN was observed in the ipsilateral cortex following HI. The wild-type OPN and C-terminal OPN (Leu152-Asn294), rather than N-terminal OPN (Met1-Gly151), interacted with GAL-3 to induce lysosomal damage. Furthermore, the secreted OPN stimulated lysosomal damage by binding to CD44 in microglia in vitro. Collectively, this study demonstrated that upregulated OPN in microglia/macrophages and its cleavage product was able to interact with GAL-3, and secreted OPN combined with CD44, leading to lysosomal damage and exacerbating autophagosome accumulation after HI exposure.
Collapse
|
38
|
Olianas A, Guadalupi G, Cabras T, Contini C, Serrao S, Iavarone F, Castagnola M, Messana I, Onali S, Chessa L, Diaz G, Manconi B. Top-Down Proteomics Detection of Potential Salivary Biomarkers for Autoimmune Liver Diseases Classification. Int J Mol Sci 2023; 24:959. [PMID: 36674470 PMCID: PMC9866740 DOI: 10.3390/ijms24020959] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/23/2022] [Accepted: 12/27/2022] [Indexed: 01/06/2023] Open
Abstract
(1) Autoimmune hepatitis (AIH) and primary biliary cholangitis (PBC) are autoimmune liver diseases characterized by chronic hepatic inflammation and progressive liver fibrosis. The possible use of saliva as a diagnostic tool has been explored in several oral and systemic diseases. The use of proteomics for personalized medicine is a rapidly emerging field. (2) Salivary proteomic data of 36 healthy controls (HCs), 36 AIH and 36 PBC patients, obtained by liquid chromatography/mass spectrometry top-down pipeline, were analyzed by multiple Mann-Whitney test, Kendall correlation, Random Forest (RF) analysis and Linear Discriminant Analysis (LDA); (3) Mann-Whitney tests provided indications on the panel of differentially expressed salivary proteins and peptides, namely cystatin A, statherin, histatin 3, histatin 5 and histatin 6, which were elevated in AIH patients with respect to both HCs and PBC patients, while S100A12, S100A9 short, cystatin S1, S2, SN and C showed varied levels in PBC with respect to HCs and/or AIH patients. RF analysis evidenced a panel of salivary proteins/peptides able to classify with good accuracy PBC vs. HCs (83.3%), AIH vs. HCs (79.9%) and PBC vs. AIH (80.2%); (4) RF appears to be an attractive machine-learning tool suited for classification of AIH and PBC based on their different salivary proteomic profiles.
Collapse
Affiliation(s)
- Alessandra Olianas
- Dipartimento di Scienze della Vita e dell’Ambiente, Università di Cagliari, 09042 Cagliari, Italy
| | - Giulia Guadalupi
- Dipartimento di Scienze della Vita e dell’Ambiente, Università di Cagliari, 09042 Cagliari, Italy
| | - Tiziana Cabras
- Dipartimento di Scienze della Vita e dell’Ambiente, Università di Cagliari, 09042 Cagliari, Italy
| | - Cristina Contini
- Dipartimento di Scienze della Vita e dell’Ambiente, Università di Cagliari, 09042 Cagliari, Italy
| | - Simone Serrao
- Dipartimento di Scienze della Vita e dell’Ambiente, Università di Cagliari, 09042 Cagliari, Italy
| | - Federica Iavarone
- Fondazione Policlinico Universitario “A. Gemelli”—IRCCS, 00168 Rome, Italy
| | - Massimo Castagnola
- Laboratorio di Proteomica, Centro Europeo di Ricerca sul Cervello, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy
| | - Irene Messana
- Istituto di Scienze e Tecnologie Chimiche “Giulio Natta”, Consiglio Nazionale delle Ricerche, 00168 Rome, Italy
| | - Simona Onali
- Liver Unit, University Hospital of Cagliari, 09042 Cagliari, Italy
| | - Luchino Chessa
- Liver Unit, University Hospital of Cagliari, 09042 Cagliari, Italy
- Dipartimento di Scienze Mediche e Sanità Pubblica, Università di Cagliari, 09042 Cagliari, Italy
| | - Giacomo Diaz
- Dipartimento di Scienze Biomediche, Università di Cagliari, 09042 Cagliari, Italy
| | - Barbara Manconi
- Dipartimento di Scienze della Vita e dell’Ambiente, Università di Cagliari, 09042 Cagliari, Italy
| |
Collapse
|
39
|
Norén Å, Oltean M, Friman S, Molinaro A, Mölne J, Sihlbom C, Herlenius G, Thorsell A. Liver Graft Proteomics Reveals Potential Incipient Mechanisms behind Early Renal Dysfunction after Liver Transplantation. Int J Mol Sci 2022; 23:ijms231911929. [PMID: 36233231 PMCID: PMC9569532 DOI: 10.3390/ijms231911929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 09/30/2022] [Accepted: 10/04/2022] [Indexed: 12/04/2022] Open
Abstract
Acute kidney injury (AKI) is frequent after liver transplantation (LT) and correlates with later development of chronic kidney disease. Its etiology is multifactorial and combines pre-, intra-, and postoperative factors. Additionally, the liver graft itself seems an important element in the development of AKI, yet the detailed mechanisms remain unclear. We hypothesized that grafts of LT recipients developing significant early AKI may show distinct proteomic alterations, and we set out to identify proteome differences between LT recipients developing moderate or severe AKI (n = 7) and LT recipients without early renal injury (n = 7). Liver biopsies obtained one hour after reperfusion were assessed histologically and using quantitative proteomics. Several cytokines and serum amyloid A2 (SAA2) were analyzed in serum samples obtained preoperatively, 2−4 h, and 20−24 h after graft reperfusion, respectively. LT induced mild histological alterations without significant differences between groups but uniformly altered liver function tests peaking on postoperative day 1, with a trend towards more severe alterations in patients developing AKI. Global quantitative proteomic analysis revealed 136 proteins differing significantly in their expression levels (p < 0.05, FC 20%): 80 proteins had higher and 56 had lower levels in the AKI group. Most of these proteins were related to immune and inflammatory responses, host defense, and neutrophil degranulation. No differences between the studied pro- and anti-inflammatory cytokines or SAA2 between groups were found at any moment. Our results suggest that grafts of LT patients who develop early AKI reveal a distinct proteome dominated by an early yet prominent activation of the innate immunity. These findings support the hypothesis that AKI after LT may be favored by certain graft characteristics.
Collapse
Affiliation(s)
- Åsa Norén
- The Transplant Institute, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, 41345 Gothenburg, Sweden
| | - Mihai Oltean
- The Transplant Institute, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, 41345 Gothenburg, Sweden
- Correspondence:
| | - Styrbjörn Friman
- The Transplant Institute, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, 41345 Gothenburg, Sweden
| | - Antonio Molinaro
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Sahlgrenska Academy at the University of Gothenburg, 41345 Gothenburg, Sweden
| | - Johan Mölne
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at the University of Gothenburg, 40530 Gothenburg, Sweden
| | - Carina Sihlbom
- Proteomics Core Facility, Sahlgrenska Academy at the University of Gothenburg, Medicinaregatan 5, 41390 Gothenburg, Sweden
| | - Gustaf Herlenius
- The Transplant Institute, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, 41345 Gothenburg, Sweden
| | - Annika Thorsell
- Proteomics Core Facility, Sahlgrenska Academy at the University of Gothenburg, Medicinaregatan 5, 41390 Gothenburg, Sweden
| |
Collapse
|
40
|
All Roads Lead to Cathepsins: The Role of Cathepsins in Non-Alcoholic Steatohepatitis-Induced Hepatocellular Carcinoma. Biomedicines 2022; 10:biomedicines10102351. [PMID: 36289617 PMCID: PMC9598942 DOI: 10.3390/biomedicines10102351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/16/2022] [Accepted: 09/16/2022] [Indexed: 11/17/2022] Open
Abstract
Cathepsins are lysosomal proteases that are essential to maintain cellular physiological homeostasis and are involved in multiple processes, such as immune and energy regulation. Predominantly, cathepsins reside in the lysosomal compartment; however, they can also be secreted by cells and enter the extracellular space. Extracellular cathepsins have been linked to several pathologies, including non-alcoholic steatohepatitis (NASH) and hepatocellular carcinoma (HCC). NASH is an increasingly important risk factor for the development of HCC, which is the third leading cause of cancer-related deaths and poses a great medical and economic burden. While information regarding the involvement of cathepsins in NASH-induced HCC (NASH-HCC) is limited, data to support the role of cathepsins in either NASH or HCC is accumulating. Since cathepsins play a role in both NASH and HCC, it is likely that the role of cathepsins is more significant in NASH-HCC compared to HCC derived from other etiologies. In the current review, we provide an overview on the available data regarding cathepsins in NASH and HCC, argue that cathepsins play a key role in the transition from NASH to HCC, and shed light on therapeutic options in this context.
Collapse
|
41
|
Arsenic-Induced Injury of Mouse Hepatocytes through Lysosome and Mitochondria: An In Vitro Study. Int J Hepatol 2022; 2022:1546297. [PMID: 36117518 PMCID: PMC9477643 DOI: 10.1155/2022/1546297] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 08/19/2022] [Accepted: 08/26/2022] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND AND AIMS The cellular mechanism of liver injury related to arsenic toxicity is ill defined. It is thought that oxidative stress and mitochondrial dysfunction may play some role in arsenic-induced liver damage. In this study, we evaluated subcellular events within the primary cultured mouse hepatocytes when exposed to inorganic arsenic. METHODS Primary cultured mouse hepatocytes were treated with 10 μM arsenic for different time periods. Reactive oxygen species (ROS) formation, functional changes of the lysosome and mitochondria, and mode of hepatocytes death were studied by laser confocal microscopy, fluorescence spectroscopy, and flow cytometry. Expression of proapoptotic member of the BCL-2 family of genes BAX and antiapoptotic BCL-2 mRNA expression were studied by real-time PCR. Cytochrome c expression was studied by Western blotting. RESULTS Fluorescence spectroscopy as well as flow cytometric analysis revealed that arsenic-induced formation of ROS was time dependent. Confocal microscopy showed initiation of ROS formation from periphery of the hepatocytes at 30 min of arsenic exposure that progressed to central part of the hepatocytes at 3 h of arsenic exposure. The ROS formation was found to be NADPH oxidase (NOX) dependent. This low level of intracellular ROS induced lysosomal membrane permeabilization (LMP) and subsequently released cathepsin B to the cytosol. The LMP further increased intracellular ROS which in turn triggered induction of mitochondrial permeability transition (MPT). Pretreatment of hepatocytes with LMP inhibitor bafilomycin A (BafA) significantly decreased, and LMP inducer chloroquine (ChQ) significantly increased the production of ROS suggesting that LMP preceded enhanced ROS generation in response to arsenic. MPT was accompanied with increase in BAX : BCL2 mRNA ratio resulting in upregulation of caspase 3 and increased hepatocyte apoptosis. CONCLUSION Although arsenic-related oxidative liver injury is well established, neither the site of origin of ROS nor the early sequence of events in arsenic toxicity due to ROS is known. We believe that our study provides evidences elucidating the early sequence of events that culminates in the death of the mouse hepatocytes during arsenic exposure.
Collapse
|
42
|
Balboa E, Marín T, Oyarzún JE, Contreras PS, Hardt R, van den Bosch T, Alvarez AR, Rebolledo-Jaramillo B, Klein AD, Winter D, Zanlungo S. Proteomic Analysis of Niemann-Pick Type C Hepatocytes Reveals Potential Therapeutic Targets for Liver Damage. Cells 2021; 10:cells10082159. [PMID: 34440927 PMCID: PMC8392304 DOI: 10.3390/cells10082159] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/16/2021] [Accepted: 08/18/2021] [Indexed: 12/21/2022] Open
Abstract
Niemann-Pick type C disease (NPCD) is a lysosomal storage disorder caused by mutations in the NPC1 gene. The most affected tissues are the central nervous system and liver, and while significant efforts have been made to understand its neurological component, the pathophysiology of the liver damage remains unclear. In this study, hepatocytes derived from wild type and Npc1-/- mice were analyzed by mass spectrometry (MS)-based proteomics in conjunction with bioinformatic analysis. We identified 3832 proteins: 416 proteins had a p-value smaller than 0.05, of which 37% (n = 155) were considered differentially expressed proteins (DEPs), 149 of them were considered upregulated, and 6 were considered downregulated. We focused the analysis on pathways related to NPC pathogenic mechanisms, finding that the most significant changes in expression levels occur in proteins that function in the pathways of liver damage, lipid metabolism, and inflammation. Moreover, in the group of DEPs, 30% (n = 47) were identified as lysosomal proteins and 7% (n = 10) were identified as mitochondrial proteins. Importantly, we found that lysosomal DEPs, including CTSB/D/Z, LIPA, DPP7 and GLMP, and mitocondrial DEPs, AKR1B10, and VAT1 had been connected with liver fibrosis, damage, and steatosis in previous studies, validiting our dataset. Our study found potential therapeutic targets for the treatment of liver damage in NPCD.
Collapse
Affiliation(s)
- Elisa Balboa
- Departamento de Gastroenterología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
| | - Tamara Marín
- Departamento de Gastroenterología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
| | - Juan Esteban Oyarzún
- Departamento de Gastroenterología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
| | - Pablo S Contreras
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892-8018, USA
| | - Robert Hardt
- Institute for Biochemistry and Molecular Biology, Medical Faculty, University of Bonn, 53115 Bonn, Germany
| | - Thea van den Bosch
- Institute for Biochemistry and Molecular Biology, Medical Faculty, University of Bonn, 53115 Bonn, Germany
| | - Alejandra R Alvarez
- Laboratory of Cell Signaling, Department of Cellular and Molecular Biology, Biological Sciences Faculty, CARE UC, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
| | - Boris Rebolledo-Jaramillo
- Centro de Genética y Genómica, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago 7710162, Chile
| | - Andres D Klein
- Centro de Genética y Genómica, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago 7710162, Chile
| | - Dominic Winter
- Institute for Biochemistry and Molecular Biology, Medical Faculty, University of Bonn, 53115 Bonn, Germany
| | - Silvana Zanlungo
- Departamento de Gastroenterología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
| |
Collapse
|