1
|
Zhang Y, Rao Y, Lu J, Wang J, Ker DFE, Zhou J, Wang DM. The influence of biophysical niche on tumor-associated macrophages in liver cancer. Hepatol Commun 2024; 8:e0569. [PMID: 39470328 PMCID: PMC11524744 DOI: 10.1097/hc9.0000000000000569] [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: 03/04/2024] [Accepted: 08/30/2024] [Indexed: 10/30/2024] Open
Abstract
HCC, the most common type of primary liver cancer, is a leading cause of cancer-related mortality worldwide. Although the advancement of immunotherapies by immune checkpoint inhibitors (ICIs) that target programmed cell death 1 or programmed cell death 1-ligand 1 has revolutionized the treatment for HCC, the majority is still not beneficial. Accumulating evidence has pointed out that the potent immunosuppressive tumor microenvironment in HCC poses a great challenge to ICI therapeutic efficacy. As a key component in tumor microenvironment, tumor-associated macrophages (TAMs) play vital roles in HCC development, progression, and ICI low responsiveness. Mechanistically, TAM can promote cancer invasion and metastasis, angiogenesis, epithelial-mesenchymal transition, maintenance of stemness, and most importantly, immunosuppression. Targeting TAMs, therefore, represents an opportunity to enhance the ICI therapeutic efficacy in patients with HCC. While previous research has primarily focused on biochemical cues influencing macrophages, emerging evidence highlights the critical role of biophysical signals, such as substrate stiffness, topography, and external forces. In this review, we summarize the influence of biophysical characteristics within the tumor microenvironment that regulate the phenotype and function of TAMs in HCC pathogenesis and progression. We also explore the possible mechanisms and discuss the potential of manipulating biophysical cues in regulating TAM for HCC therapy. By gaining a deeper understanding of how macrophages sense and respond to mechanical forces, we may potentially usher in a path toward a curative approach for combinatory cancer immunotherapies.
Collapse
Affiliation(s)
- Ying Zhang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
- Institute of Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
- Institute of Precision Medicine, Jining Medical University, Jining, China
| | - Ying Rao
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
- Institute of Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
| | - Jiahuan Lu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
| | - Jiyu Wang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
- Institute of Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
| | - Dai Fei Elmer Ker
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Sha Tin, Hong Kong, SAR, China
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, SAR, China
| | - Jingying Zhou
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
| | - Dan Michelle Wang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
- Institute of Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Sha Tin, Hong Kong, SAR, China
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
| |
Collapse
|
2
|
Ye J, Li F, Hua T, Ma K, Wang J, Zhao Z, Yang Z, Luo C, Jia R, Li Y, Hao M, Wu J, Lu M, Yuan Z, Zhang J, Chen J. Liver mechanosignaling as a natural anti-hepatitis B virus mechanism. Nat Commun 2024; 15:8375. [PMID: 39333106 PMCID: PMC11437074 DOI: 10.1038/s41467-024-52718-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 09/18/2024] [Indexed: 09/29/2024] Open
Abstract
The mechanisms underlying the natural control of hepatitis B virus (HBV) infection have long been an intriguing question. Given the wide physiological range of liver stiffness and the growing attention to the role of mechanical microenvironment in homeostasis and diseases, we investigated how physical matrix cues impact HBV replication. High matrix stiffness significantly inhibited HBV replication and activated YAP in primary hepatocyte culture system, a key molecule in mechanosignaling. YAP activation notably suppressed HBV transcription and antigen expression. Several YAP-induced genes exhibited strong anti-HBV effects. Single-cell analysis of liver tissue from male individuals with active HBV replication revealed a strong significant negative correlation between YAP signature activation and HBV transcript levels. Intraperitoneal administration of YAP small molecule agonist potently controls HBV in male mouse models. These findings unveil a mechanism that involves the mechanical environment of hepatocytes and YAP to clear hepatotropic viral infection in the liver, providing new perspectives for HBV cure studies and antiviral development.
Collapse
Affiliation(s)
- Jianyu Ye
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Huashan Hospital, Shanghai Medical College Fudan University, Shanghai, China
- Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Institute of Infectious Diseases and Biosecurity, Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Fudan University, Shanghai, China
| | - Fahong Li
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Huashan Hospital, Shanghai Medical College Fudan University, Shanghai, China
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Ting Hua
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Huashan Hospital, Shanghai Medical College Fudan University, Shanghai, China
- Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Institute of Infectious Diseases and Biosecurity, Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Fudan University, Shanghai, China
| | - Kewei Ma
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Huashan Hospital, Shanghai Medical College Fudan University, Shanghai, China
| | - Jinyu Wang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Huashan Hospital, Shanghai Medical College Fudan University, Shanghai, China
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Zixin Zhao
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Huashan Hospital, Shanghai Medical College Fudan University, Shanghai, China
| | - Zhongning Yang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Huashan Hospital, Shanghai Medical College Fudan University, Shanghai, China
- Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Institute of Infectious Diseases and Biosecurity, Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Fudan University, Shanghai, China
| | - Chen Luo
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Huashan Hospital, Shanghai Medical College Fudan University, Shanghai, China
- Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Institute of Infectious Diseases and Biosecurity, Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Fudan University, Shanghai, China
| | - Ruohan Jia
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Huashan Hospital, Shanghai Medical College Fudan University, Shanghai, China
- Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Institute of Infectious Diseases and Biosecurity, Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Fudan University, Shanghai, China
| | - Yaming Li
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Huashan Hospital, Shanghai Medical College Fudan University, Shanghai, China
- Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Institute of Infectious Diseases and Biosecurity, Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Fudan University, Shanghai, China
| | - Menghan Hao
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Huashan Hospital, Shanghai Medical College Fudan University, Shanghai, China
- Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Institute of Infectious Diseases and Biosecurity, Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Fudan University, Shanghai, China
| | - Jian Wu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Huashan Hospital, Shanghai Medical College Fudan University, Shanghai, China
| | - Mengji Lu
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Zhenghong Yuan
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Huashan Hospital, Shanghai Medical College Fudan University, Shanghai, China.
- Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Institute of Infectious Diseases and Biosecurity, Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Fudan University, Shanghai, China.
| | - Jiming Zhang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Huashan Hospital, Shanghai Medical College Fudan University, Shanghai, China.
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China.
| | - Jieliang Chen
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Huashan Hospital, Shanghai Medical College Fudan University, Shanghai, China.
- Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Institute of Infectious Diseases and Biosecurity, Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Fudan University, Shanghai, China.
| |
Collapse
|
3
|
Khanmohammadi M, Danish H, Sekar NC, Suarez SA, Chheang C, Peter K, Khoshmanesh K, Baratchi S. Cyclic stretch enhances neutrophil extracellular trap formation. BMC Biol 2024; 22:209. [PMID: 39289752 PMCID: PMC11409804 DOI: 10.1186/s12915-024-02009-6] [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: 05/24/2024] [Accepted: 09/05/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND Neutrophils, the most abundant leukocytes circulating in blood, contribute to host defense and play a significant role in chronic inflammatory disorders. They can release their DNA in the form of extracellular traps (NETs), which serve as scaffolds for capturing bacteria and various blood cells. However, uncontrolled formation of NETs (NETosis) can lead to excessive activation of coagulation pathways and thrombosis. Once neutrophils are migrated to infected or injured tissues, they become exposed to mechanical forces from their surrounding environment. However, the impact of transient changes in tissue mechanics due to the natural process of aging, infection, tissue injury, and cancer on neutrophils remains unknown. To address this gap, we explored the interactive effects of changes in substrate stiffness and cyclic stretch on NETosis. Primary neutrophils were cultured on a silicon-based substrate with stiffness levels of 30 and 300 kPa for at least 3 h under static conditions or cyclic stretch levels of 5% and 10%, mirroring the biomechanics of aged and young arteries. RESULTS Using this approach, we found that neutrophils are sensitive to cyclic stretch and that increases in stretch intensity and substrate stiffness enhance nuclei decondensation and histone H3 citrullination (CitH3). In addition, stretch intensity and substrate stiffness promote the response of neutrophils to the NET-inducing agents phorbol 12-myristate 13-acetate (PMA), adenosine triphosphate (ATP), and lipopolysaccharides (LPS). Stretch-induced activation of neutrophils was dependent on calpain activity, the phosphatidylinositol 3-kinase (PI3K)/focal adhesion kinase (FAK) signalling and actin polymerization. CONCLUSIONS In summary, these results demonstrate that the mechanical forces originating from the surrounding tissue influence NETosis, an important neutrophil function, and thus identify a potential novel therapeutic target.
Collapse
Affiliation(s)
- Manijeh Khanmohammadi
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Habiba Danish
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Nadia Chandra Sekar
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | | | - Chanly Chheang
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Karlheinz Peter
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Cardiometabolic Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Khashayar Khoshmanesh
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- School of Engineering, RMIT University, Melbourne, VIC, Australia
| | - Sara Baratchi
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia.
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.
- Department of Cardiometabolic Health, The University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
4
|
Sun Z, Chen G. Impact of heterogeneity in liver matrix and intrahepatic cells on the progression of hepatic fibrosis. Tissue Cell 2024; 91:102559. [PMID: 39293139 DOI: 10.1016/j.tice.2024.102559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 08/05/2024] [Accepted: 09/09/2024] [Indexed: 09/20/2024]
Abstract
Liver fibrosis is a disease with a high prevalence worldwide. The development of hepatic fibrosis results from a combination of factors within the liver, such as extracellular matrix (ECM) deposition, hepatic stellate cells (HSCs) activation, collagen cross-linking, and inflammatory response. Heterogeneity in fibrotic liver is the result of a combination of heterogeneity in the intrahepatic microenvironment as well as heterogeneous expression of fibrosis-associated enzymes and cells, complicating the study of the mechanisms underlying the progression of liver fibrosis. The role of this heterogeneity on the crosstalk between cells and matrix and on the fibrotic process is worth exploring. In this paper, we will describe the phenomenon and mechanism of heterogeneity of liver matrix and intrahepatic cells in the process of hepatic fibrosis and discuss the crosstalk between heterogeneous factors on the development of fibrosis. The elucidation of heterogeneity is important for a deeper understanding of the pathological mechanisms of liver fibrosis as well as for clinical diagnosis and targeted therapies.
Collapse
Affiliation(s)
- Zhongtao Sun
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Guobao Chen
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China.
| |
Collapse
|
5
|
Son DO, Benitez R, Diao L, Hinz B. How to Keep Myofibroblasts under Control: Culture of Mouse Skin Fibroblasts on Soft Substrates. J Invest Dermatol 2024; 144:1923-1934. [PMID: 39078357 DOI: 10.1016/j.jid.2024.05.033] [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: 02/12/2024] [Revised: 05/03/2024] [Accepted: 05/21/2024] [Indexed: 07/31/2024]
Abstract
During the physiological healing of skin wounds, fibroblasts recruited from the uninjured adjacent dermis and deeper subcutaneous fascia layers are transiently activated into myofibroblasts to first secrete and then contract collagen-rich extracellular matrix into a mechanically resistant scar. Scar tissue restores skin integrity after damage but comes at the expense of poor esthetics and loss of tissue function. Stiff scar matrix also mechanically activates various precursor cells into myofibroblasts in a positive feedback loop. Persistent myofibroblast activation results in pathologic accumulation of fibrous collagen and hypertrophic scarring, called fibrosis. Consequently, the mechanisms of fibroblast-to-myofibroblast activation and persistence are studied to develop antifibrotic and prohealing treatments. Mechanistic understanding often starts in a plastic cell culture dish. This can be problematic because contact of fibroblasts with tissue culture plastic or glass surfaces invariably generates myofibroblast phenotypes in standard culture. We describe a straight-forward method to produce soft cell culture surfaces for fibroblast isolation and continued culture and highlight key advantages and limitations of the approach. Adding a layer of elastic silicone polymer tunable to the softness of normal skin and the stiffness of pathologic scars allows to control mechanical fibroblast activation while preserving the simplicity of conventional 2-dimensional cell culture.
Collapse
Affiliation(s)
- Dong Ok Son
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, Canada
| | - Raquel Benitez
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, Canada
| | - Li Diao
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, Canada
| | - Boris Hinz
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, Canada; Faculty of Dentistry, University of Toronto, Toronto, Canada.
| |
Collapse
|
6
|
Younesi FS, Miller AE, Barker TH, Rossi FMV, Hinz B. Fibroblast and myofibroblast activation in normal tissue repair and fibrosis. Nat Rev Mol Cell Biol 2024; 25:617-638. [PMID: 38589640 DOI: 10.1038/s41580-024-00716-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2024] [Indexed: 04/10/2024]
Abstract
The term 'fibroblast' often serves as a catch-all for a diverse array of mesenchymal cells, including perivascular cells, stromal progenitor cells and bona fide fibroblasts. Although phenotypically similar, these subpopulations are functionally distinct, maintaining tissue integrity and serving as local progenitor reservoirs. In response to tissue injury, these cells undergo a dynamic fibroblast-myofibroblast transition, marked by extracellular matrix secretion and contraction of actomyosin-based stress fibres. Importantly, whereas transient activation into myofibroblasts aids in tissue repair, persistent activation triggers pathological fibrosis. In this Review, we discuss the roles of mechanical cues, such as tissue stiffness and strain, alongside cell signalling pathways and extracellular matrix ligands in modulating myofibroblast activation and survival. We also highlight the role of epigenetic modifications and myofibroblast memory in physiological and pathological processes. Finally, we discuss potential strategies for therapeutically interfering with these factors and the associated signal transduction pathways to improve the outcome of dysregulated healing.
Collapse
Affiliation(s)
- Fereshteh Sadat Younesi
- Keenan Research Institute for Biomedical Science of the St. Michael's Hospital, Toronto, Ontario, Canada
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Andrew E Miller
- Department of Biomedical Engineering, School of Engineering and Applied Science, University of Virginia, Charlottesville, VA, USA
| | - Thomas H Barker
- Department of Biomedical Engineering, School of Engineering and Applied Science, University of Virginia, Charlottesville, VA, USA
| | - Fabio M V Rossi
- School of Biomedical Engineering and Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Boris Hinz
- Keenan Research Institute for Biomedical Science of the St. Michael's Hospital, Toronto, Ontario, Canada.
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
7
|
Ramkissoon R, Cao S, Shah VH. The Pathophysiology of Portal Hypertension. Clin Liver Dis 2024; 28:369-381. [PMID: 38945632 DOI: 10.1016/j.cld.2024.03.001] [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] [Indexed: 07/02/2024]
Abstract
This article reviews the pathophysiology of portal hypertension that includes multiple mechanisms internal and external to the liver. This article starts with a review of literature describing the cellular and molecular mechanisms of portal hypertension, microvascular thrombosis, sinusoidal venous congestion, portal angiogenesis, vascular hypocontractility, and hyperdynamic circulation. Mechanotransduction and the gut-liver axis, which are newer areas of research, are reviewed. Dysfunction of this axis contributes to chronic liver injury, inflammation, fibrosis, and portal hypertension. Sequelae of portal hypertension are discussed in subsequent studies.
Collapse
Affiliation(s)
- Resham Ramkissoon
- Department of Gastroenterology & Hepatology, Mayo Clinic, 200 1st Street, SW, Rochester, MN 55902, USA
| | - Sheng Cao
- Mayo College of Medicine, Mayo Clinic, 200 1st Street, SW, Rochester, MN 55902, USA
| | - Vijay H Shah
- Department of Gastroenterology & Hepatology, Mayo Clinic, 200 1st Street, SW, Rochester, MN 55902, USA; Department of Internal Medicine, Mayo Clinic, 200 1st Street, SW, Rochester, MN 55902, USA.
| |
Collapse
|
8
|
Suffredini G, Gao WD, Dodd-O JM. Ultrasound Shear Wave Elastography Evaluation of the Liver and Implications for Perioperative Medicine. J Clin Med 2024; 13:3633. [PMID: 38999199 PMCID: PMC11242192 DOI: 10.3390/jcm13133633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 06/10/2024] [Accepted: 06/18/2024] [Indexed: 07/14/2024] Open
Abstract
Ultrasound shear wave elastography (SWE) is a non-invasive, low risk technology allowing the assessment of tissue stiffness. Used clinically for nearly two decades to diagnose and stage liver fibrosis and cirrhosis, it has recently been appreciated for its ability to differentiate between more subtle forms of liver dysfunction. In this review, we will discuss the principle of ultrasound shear wave elastography, its traditional utilization in grading liver cirrhosis, as well as its evolving role in identifying more subtle degrees of liver injury. Finally, we will show how this capacity to distinguish nuanced changes may provide an opportunity for its use in perioperative risk stratification.
Collapse
Affiliation(s)
- Giancarlo Suffredini
- Department of Anesthesiology and Critical Care Medicine, Division of Cardiac Anesthesia, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Wei Dong Gao
- Department of Anesthesiology and Critical Care Medicine, Division of Cardiac Anesthesia, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jeffrey M Dodd-O
- Department of Anesthesiology and Critical Care Medicine, Division of Cardiac Anesthesia, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
9
|
He Y, Wang Y, Wang L, Jiang W, Wilhelm S. Understanding nanoparticle-liver interactions in nanomedicine. Expert Opin Drug Deliv 2024; 21:829-843. [PMID: 38946471 PMCID: PMC11281865 DOI: 10.1080/17425247.2024.2375400] [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: 01/19/2024] [Accepted: 06/28/2024] [Indexed: 07/02/2024]
Abstract
INTRODUCTION Understanding the interactions between administered nanoparticles and the liver is crucial for developing safe and effective nanomedicines. As the liver can sequester up to 99% of these particles due to its major phagocytic role, understanding these interactions is vital for clinical translation. AREAS COVERED This review highlights recent studies on nanoparticle-liver interactions, including the influence of nanoparticle physicochemical properties on delivery, strategies to enhance delivery efficiency by modulating liver Kupffer cells, and their potential for treating certain hepatic diseases. Additionally, we discuss how aging impacts the liver's phagocytic functions. EXPERT OPINION While liver accumulation can hinder nanomedicine safety and effectiveness, it also presents opportunities for treating certain liver diseases. A thorough understanding of nanoparticle-liver interactions is essential for advancing the clinical application of nanomedicines.
Collapse
Affiliation(s)
- Yuxin He
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Yifan Wang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lin Wang
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Wen Jiang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Stefan Wilhelm
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
- Institute for Biomedical Engineering, Science, and Technology (IBEST), Norman, OK, USA
- Stephenson Cancer Center, Oklahoma City, OK, USA
| |
Collapse
|
10
|
Bansal S, Taddei T, Wells R, Serper M, Bittermann T, Mahmud N, Kaplan DE. Transjugular Intrahepatic Portosystemic Shunt Linked to Increased Risk of Hepatocellular Carcinoma: A VA Matched Cohort Study. J Clin Transl Hepatol 2024; 12:534-538. [PMID: 38779520 PMCID: PMC11106346 DOI: 10.14218/jcth.2023.00554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 02/01/2024] [Accepted: 02/09/2024] [Indexed: 05/25/2024] Open
Affiliation(s)
- Shalini Bansal
- Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, USA
| | - Tamar Taddei
- Division of Gastroenterology and Hepatology Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
| | - Rebecca Wells
- Division of Gastroenterology and Hepatology Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
| | - Marina Serper
- Division of Gastroenterology and Hepatology Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
| | - Theresa Bittermann
- Division of Gastroenterology and Hepatology Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
| | - Nadim Mahmud
- Division of Gastroenterology and Hepatology Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Division of Gastroenterology and Hepatology, Yale University School of Medicine, New Haven, CT, USA
- VA Connecticut Healthcare System, West Haven, CT, USA
| | - David E. Kaplan
- Division of Gastroenterology and Hepatology Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
| |
Collapse
|
11
|
Qi L, Duan B, Wang H, Liu Y, Han H, Han M, Xing L, Jiang H, Pandol SJ, Li L. Reactive Oxygen Species-Responsive Nanoparticles Toward Extracellular Matrix Normalization for Pancreatic Fibrosis Regression. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401254. [PMID: 38483920 PMCID: PMC11109658 DOI: 10.1002/advs.202401254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Indexed: 05/23/2024]
Abstract
Pancreatic fibrosis (PF) is primarily characterized by aberrant production and degradation modes of extracellular matrix (ECM) components, resulting from the activation of pancreatic stellate cells (PSCs) and the pathological cross-linking of ECM mediated by lysyl oxidase (LOX) family members. The excessively deposited ECM increases matrix stiffness, and the over-accumulated reactive oxygen species (ROS) induces oxidative stress, which further stimulates the continuous activation of PSCs and advancing PF; challenging the strategy toward normalizing ECM homeostasis for the regression of PF. Herein, ROS-responsive and Vitamin A (VA) decorated micelles (named LR-SSVA) to reverse the imbalanced ECM homeostasis for ameliorating PF are designed and synthesized. Specifically, LR-SSVA selectively targets PSCs via VA, thereby effectively delivering siLOXL1 and resveratrol (RES) into the pancreas. The ROS-responsive released RES inhibits the overproduction of ECM by eliminating ROS and inactivating PSCs, meanwhile, the decreased expression of LOXL1 ameliorates the cross-linked collagen for easier degradation by collagenase which jointly normalizes ECM homeostasis and alleviates PF. This research shows that LR-SSVA is a safe and efficient ROS-response and PSC-targeted drug-delivery system for ECM normalization, which will propose an innovative and ideal platform for the reversal of PF.
Collapse
Affiliation(s)
- Liang Qi
- Department of EndocrinologyZhongda HospitalSchool of MedicineSoutheast UniversityNanjing210009China
| | - Bo‐Wen Duan
- State Key Laboratory of Natural MedicinesDepartment of PharmaceuticsChina Pharmaceutical UniversityNanjing210009China
| | - Hui Wang
- Department of EndocrinologyZhongda HospitalSchool of MedicineSoutheast UniversityNanjing210009China
| | - Yan‐Jun Liu
- State Key Laboratory of Natural MedicinesDepartment of PharmaceuticsChina Pharmaceutical UniversityNanjing210009China
| | - Han Han
- State Key Laboratory of Natural MedicinesDepartment of PharmaceuticsChina Pharmaceutical UniversityNanjing210009China
| | - Meng‐Meng Han
- State Key Laboratory of Natural MedicinesDepartment of PharmaceuticsChina Pharmaceutical UniversityNanjing210009China
| | - Lei Xing
- State Key Laboratory of Natural MedicinesDepartment of PharmaceuticsChina Pharmaceutical UniversityNanjing210009China
- Jiangsu Key Laboratory of Druggability of BiopharmaceuticalsChina Pharmaceutical UniversityNanjing210009China
| | - Hu‐Lin Jiang
- State Key Laboratory of Natural MedicinesDepartment of PharmaceuticsChina Pharmaceutical UniversityNanjing210009China
- Jiangsu Key Laboratory of Druggability of BiopharmaceuticalsChina Pharmaceutical UniversityNanjing210009China
| | - Stephen J. Pandol
- Division of GastroenterologyDepartment of MedicineCedars‐Sinai Medical CenterLos AngelesCA90048USA
- Basic and Translational Pancreatic ResearchCedars‐Sinai Medical CenterLos AngelesCA90048USA
| | - Ling Li
- Department of EndocrinologyZhongda HospitalSchool of MedicineSoutheast UniversityNanjing210009China
- Institute of Glucose and Lipid MetabolismSoutheast UniversityNanjing210009China
- Department of Clinical Science and ResearchZhongda HospitalSchool of MedicineSoutheast UniversityNanjing210009China
| |
Collapse
|
12
|
Reiss AB, Jacob B, Zubair A, Srivastava A, Johnson M, De Leon J. Fibrosis in Chronic Kidney Disease: Pathophysiology and Therapeutic Targets. J Clin Med 2024; 13:1881. [PMID: 38610646 PMCID: PMC11012936 DOI: 10.3390/jcm13071881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 03/18/2024] [Accepted: 03/20/2024] [Indexed: 04/14/2024] Open
Abstract
Chronic kidney disease (CKD) is a slowly progressive condition characterized by decreased kidney function, tubular injury, oxidative stress, and inflammation. CKD is a leading global health burden that is asymptomatic in early stages but can ultimately cause kidney failure. Its etiology is complex and involves dysregulated signaling pathways that lead to fibrosis. Transforming growth factor (TGF)-β is a central mediator in promoting transdifferentiation of polarized renal tubular epithelial cells into mesenchymal cells, resulting in irreversible kidney injury. While current therapies are limited, the search for more effective diagnostic and treatment modalities is intensive. Although biopsy with histology is the most accurate method of diagnosis and staging, imaging techniques such as diffusion-weighted magnetic resonance imaging and shear wave elastography ultrasound are less invasive ways to stage fibrosis. Current therapies such as renin-angiotensin blockers, mineralocorticoid receptor antagonists, and sodium/glucose cotransporter 2 inhibitors aim to delay progression. Newer antifibrotic agents that suppress the downstream inflammatory mediators involved in the fibrotic process are in clinical trials, and potential therapeutic targets that interfere with TGF-β signaling are being explored. Small interfering RNAs and stem cell-based therapeutics are also being evaluated. Further research and clinical studies are necessary in order to avoid dialysis and kidney transplantation.
Collapse
Affiliation(s)
- Allison B. Reiss
- Department of Medicine and Biomedical Research Institute, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA; (B.J.); (A.Z.); (A.S.); (M.J.); (J.D.L.)
| | | | | | | | | | | |
Collapse
|
13
|
Zhang X, Li P, Zhou J, Zhang Z, Wu H, Shu X, Li W, Wu Y, Du Y, Lü D, Lü S, Li N, Long M. FAK-p38 signaling serves as a potential target for reverting matrix stiffness-modulated liver sinusoidal endothelial cell defenestration. Biomaterials 2024; 305:122462. [PMID: 38171118 DOI: 10.1016/j.biomaterials.2023.122462] [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: 10/17/2023] [Revised: 12/26/2023] [Accepted: 12/31/2023] [Indexed: 01/05/2024]
Abstract
Liver sinusoidal endothelial cells (LSECs) are highly specific endothelial cells which play an essential role in the maintenance of liver homeostasis. During the progression of liver fibrosis, matrix stiffening promotes LSEC defenestration, however, the underlying mechanotransduction mechanism remains poorly understood. Here, we applied stiffness-tunable hydrogels to assess the matrix stiffening-induced phenotypic changes in primary mouse LSECs. Results indicated that increased stiffness promoted LSEC defenestration through cytoskeletal reorganization. LSECs sensed the increased matrix stiffness via focal adhesion kinase (FAK), leading to the activation of p38-mitogen activated protein kinase activated protein kinase 2 (MK2) pathway, thereby inducing actin remodeling via LIM Kinase 1 (LIMK1) and Cofilin. Interestingly, inhibition of FAK or p38-MK2 pathway was able to effectively restore the fenestrae to a certain degree in LSECs isolated from early to late stages of liver fibrosis mice. Thus, this study highlights the impact of mechanotransduction in LSEC defenestration, and provides novel insights for potential therapeutic interventions for liver fibrosis.
Collapse
Affiliation(s)
- Xiaoyu Zhang
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China; School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Peiwen Li
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China
| | - Jin Zhou
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China
| | - Ziliang Zhang
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Huan Wu
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China
| | - Xinyu Shu
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China; School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Wang Li
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China; School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yi Wu
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China; School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yu Du
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China
| | - Dongyuan Lü
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China; School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shouqin Lü
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China; School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ning Li
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China; School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Mian Long
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China; School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
14
|
Mittelheisser V, Gensbittel V, Bonati L, Li W, Tang L, Goetz JG. Evidence and therapeutic implications of biomechanically regulated immunosurveillance in cancer and other diseases. NATURE NANOTECHNOLOGY 2024; 19:281-297. [PMID: 38286876 DOI: 10.1038/s41565-023-01535-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 09/26/2023] [Indexed: 01/31/2024]
Abstract
Disease progression is usually accompanied by changes in the biochemical composition of cells and tissues and their biophysical properties. For instance, hallmarks of cancer include the stiffening of tissues caused by extracellular matrix remodelling and the softening of individual cancer cells. In this context, accumulating evidence has shown that immune cells sense and respond to mechanical signals from the environment. However, the mechanisms regulating these mechanical aspects of immune surveillance remain partially understood. The growing appreciation for the 'mechano-immunology' field has urged researchers to investigate how immune cells sense and respond to mechanical cues in various disease settings, paving the way for the development of novel engineering strategies that aim at mechanically modulating and potentiating immune cells for enhanced immunotherapies. Recent pioneer developments in this direction have laid the foundations for leveraging 'mechanical immunoengineering' strategies to treat various diseases. This Review first outlines the mechanical changes occurring during pathological progression in several diseases, including cancer, fibrosis and infection. We next highlight the mechanosensitive nature of immune cells and how mechanical forces govern the immune responses in different diseases. Finally, we discuss how targeting the biomechanical features of the disease milieu and immune cells is a promising strategy for manipulating therapeutic outcomes.
Collapse
Affiliation(s)
- Vincent Mittelheisser
- Tumor Biomechanics, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France
| | - Valentin Gensbittel
- Tumor Biomechanics, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France
| | - Lucia Bonati
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Weilin Li
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Li Tang
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
- Institute of Materials Science and Engineering, EPFL, Lausanne, Switzerland.
| | - Jacky G Goetz
- Tumor Biomechanics, INSERM UMR_S1109, Strasbourg, France.
- Université de Strasbourg, Strasbourg, France.
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France.
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France.
| |
Collapse
|
15
|
Wendong Y, Jiali J, Qiaomei F, Yayun W, Xianze X, Zheng S, Wei H. Biomechanical forces and force-triggered drug delivery in tumor neovascularization. Biomed Pharmacother 2024; 171:116117. [PMID: 38171243 DOI: 10.1016/j.biopha.2023.116117] [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: 11/02/2023] [Revised: 12/25/2023] [Accepted: 12/29/2023] [Indexed: 01/05/2024] Open
Abstract
Tumor angiogenesis is one of the typical hallmarks of tumor occurrence and development, and tumor neovascularization also exhibits distinct characteristics from normal blood vessels. As the number of cells and matrix inside the tumor increases, the biomechanical force is enhanced, specifically manifested as solid stress, fluid stress, stiffness, and topology. This mechanical microenvironment also provides shelter for tumors and intensifies angiogenesis, providing oxygen and nutritional support for tumor progression. During tumor development, the biomechanical microenvironment also emerges, which in turn feeds back to regulate the tumor progression, including tumor angiogenesis, and biochemical and biomechanical signals can regulate tumor angiogenesis. Blood vessels possess inherent sensitivity to mechanical stimuli, but compared to the extensive research on biochemical signal regulation, the study of the regulation of tumor neovascularization by biomechanical signals remains relatively scarce. Biomechanical forces can affect the phenotypic characteristics and mechanical signaling pathways of tumor blood vessels, directly regulating angiogenesis. Meanwhile, they can indirectly regulate tumor angiogenesis by causing an imbalance in angiogenesis signals and affecting stromal cell function. Understanding the regulatory mechanism of biomechanical forces in tumor angiogenesis is beneficial for better identifying and even taming the mechanical forces involved in angiogenesis, providing new therapeutic targets for tumor vascular normalization. Therefore, we summarized the composition of biomechanical forces and their direct or indirect regulation of tumor neovascularization. In addition, this review discussed the use of biomechanical forces in combination with anti-angiogenic therapies for the treatment of tumors, and biomechanical forces triggered delivery systems.
Collapse
Affiliation(s)
- Yao Wendong
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310005, China
| | - Jiang Jiali
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310005, China
| | - Fan Qiaomei
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310005, China
| | - Weng Yayun
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310005, China
| | - Xie Xianze
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310005, China
| | - Shi Zheng
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310005, China.
| | - Huang Wei
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310005, China.
| |
Collapse
|
16
|
Kim WS, Min S, Kim SK, Kang S, An S, Criado-Hidalgo E, Davis H, Bar-Zion A, Malounda D, Kim YH, Lee JH, Bae SH, Lee JG, Kwak M, Cho SW, Shapiro MG, Cheon J. Magneto-acoustic protein nanostructures for non-invasive imaging of tissue mechanics in vivo. NATURE MATERIALS 2024; 23:290-300. [PMID: 37845321 PMCID: PMC10837075 DOI: 10.1038/s41563-023-01688-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 09/12/2023] [Indexed: 10/18/2023]
Abstract
Measuring cellular and tissue mechanics inside intact living organisms is essential for interrogating the roles of force in physiological and disease processes. Current agents for studying the mechanobiology of intact, living organisms are limited by poor light penetration and material stability. Magnetomotive ultrasound is an emerging modality for real-time in vivo imaging of tissue mechanics. Nonetheless, it has poor sensitivity and spatiotemporal resolution. Here we describe magneto-gas vesicles (MGVs), protein nanostructures based on gas vesicles and magnetic nanoparticles that produce differential ultrasound signals in response to varying mechanical properties of surrounding tissues. These hybrid nanomaterials significantly improve signal strength and detection sensitivity. Furthermore, MGVs enable non-invasive, long-term and quantitative measurements of mechanical properties within three-dimensional tissues and in vivo fibrosis models. Using MGVs as novel contrast agents, we demonstrate their potential for non-invasive imaging of tissue elasticity, offering insights into mechanobiology and its application to disease diagnosis and treatment.
Collapse
Affiliation(s)
- Whee-Soo Kim
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA
- Department of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea
| | - Sungjin Min
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Su Kyeom Kim
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Sunghwi Kang
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
- Department of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea
- Department of Chemistry, Yonsei University, Seoul, Republic of Korea
| | - Soohwan An
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Ernesto Criado-Hidalgo
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Hunter Davis
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Avinoam Bar-Zion
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Dina Malounda
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Yu Heun Kim
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Jae-Hyun Lee
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
- Department of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea
| | - Soo Han Bae
- Severance Biomedical Science Institute, Yonsei Biomedical Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
- Severance Biomedical Science Institute, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jin Gu Lee
- Department of Thoracic and Cardiovascular Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Minsuk Kwak
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
- Department of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea
| | - Seung-Woo Cho
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea.
- Department of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea.
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea.
| | - Mikhail G Shapiro
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea.
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA.
- Department of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea.
- Andrew and Peggy Cherng Department of Medical Engineering, California Institute of Technology, Pasadena, CA, USA.
- Howard Hughes Medical Institute, Pasadena, CA, USA.
| | - Jinwoo Cheon
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea.
- Department of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea.
- Department of Chemistry, Yonsei University, Seoul, Republic of Korea.
| |
Collapse
|
17
|
Martinez-Vidal L, Testi C, Pontecorvo E, Pederzoli F, Alchera E, Locatelli I, Venegoni C, Spinelli A, Lucianò R, Salonia A, Podestà A, Ruocco G, Alfano M. Progressive alteration of murine bladder elasticity in actinic cystitis detected by Brillouin microscopy. Sci Rep 2024; 14:484. [PMID: 38177637 PMCID: PMC10766652 DOI: 10.1038/s41598-023-51006-2] [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: 07/13/2023] [Accepted: 12/29/2023] [Indexed: 01/06/2024] Open
Abstract
Bladder mechanical properties are critical for organ function and tissue homeostasis. Therefore, alterations of tissue mechanics are linked to disease onset and progression. This study aims to characterize the tissue elasticity of the murine bladder wall considering its different anatomical components, both in healthy conditions and in actinic cystitis, a state characterized by tissue fibrosis. Here, we exploit Brillouin microscopy, an emerging technique in the mechanobiology field that allows mapping tissue mechanics at the microscale, in non-contact mode and free of labeling. We show that Brillouin imaging of bladder tissues is able to recognize the different anatomical components of the bladder wall, confirmed by histopathological analysis, showing different tissue mechanical properties of the physiological bladder, as well as a significant alteration in the presence of tissue fibrosis. Our results point out the potential use of Brillouin imaging on clinically relevant samples as a complementary technique to histopathological analysis, deciphering complex mechanical alteration of each tissue layer of an organ that strongly relies on mechanical properties to perform its function.
Collapse
Affiliation(s)
- Laura Martinez-Vidal
- Division of Experimental Oncology/Unit of Urology, IRCCS Ospedale San Raffaele, 20132, Milan, Italy.
- Università Vita-Salute San Raffaele, Via Olgettina, 60, 20132, Milan, Italy.
| | - Claudia Testi
- Center for Life Nano- and Neuro-Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161, Roma, Italy.
| | - Emanuele Pontecorvo
- Center for Life Nano- and Neuro-Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161, Roma, Italy
- CrestOptics S.p.A., Via Di Torre Rossa, 66, 00165, Roma, Italy
| | - Filippo Pederzoli
- Division of Experimental Oncology/Unit of Urology, IRCCS Ospedale San Raffaele, 20132, Milan, Italy
- Università Vita-Salute San Raffaele, Via Olgettina, 60, 20132, Milan, Italy
| | - Elisa Alchera
- Division of Experimental Oncology/Unit of Urology, IRCCS Ospedale San Raffaele, 20132, Milan, Italy
| | - Irene Locatelli
- Division of Experimental Oncology/Unit of Urology, IRCCS Ospedale San Raffaele, 20132, Milan, Italy
| | - Chiara Venegoni
- Division of Experimental Oncology/Unit of Urology, IRCCS Ospedale San Raffaele, 20132, Milan, Italy
| | - Antonello Spinelli
- Experimental Imaging Centre, San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Roberta Lucianò
- Pathology Unit, IRCCS Ospedale San Raffaele, 20132, Milan, Italy
| | - Andrea Salonia
- Division of Experimental Oncology/Unit of Urology, IRCCS Ospedale San Raffaele, 20132, Milan, Italy
- Università Vita-Salute San Raffaele, Via Olgettina, 60, 20132, Milan, Italy
| | - Alessandro Podestà
- Dipartimento Di Fisica "Aldo Pontremoli" and CIMAINA, Università Degli Studi Di Milano, 20133, Milan, Italy
| | - Giancarlo Ruocco
- Center for Life Nano- and Neuro-Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161, Roma, Italy
- Dipartimento Di Fisica, Universitá Di Roma "La Sapienza", Piazzale Aldo Moro, 5, 00185, Roma, Italy
| | - Massimo Alfano
- Division of Experimental Oncology/Unit of Urology, IRCCS Ospedale San Raffaele, 20132, Milan, Italy
| |
Collapse
|
18
|
Sumey JL, Harrell AM, Johnston PC, Caliari SR. Serial Passaging Affects Stromal Cell Mechanosensitivity on Hyaluronic Acid Hydrogels. Macromol Biosci 2024; 24:e2300110. [PMID: 37747449 DOI: 10.1002/mabi.202300110] [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: 03/16/2023] [Revised: 09/15/2023] [Indexed: 09/26/2023]
Abstract
There is a tremendous interest in developing hydrogels as tunable in vitro cell culture platforms to study cell response to mechanical cues in a controlled manner. However, little is known about how common cell culture techniques, such as serial expansion on tissue culture plastic, affect subsequent cell behavior when cultured on hydrogels. In this work, a methacrylated hyaluronic acid hydrogel platform is leveraged to study stromal cell mechanotransduction. Hydrogels are first formed through thiol-Michael addition to model normal soft tissue (e.g., lung) stiffness (E ≈ 1 kPa). Secondary cross-linking via radical photopolymerization of unconsumed methacrylates allows matching of early- (E ≈ 6 kPa) and late-stage fibrotic tissue (E ≈ 50 kPa). Early passage (P1) human bone marrow mesenchymal stromal cells (hMSCs) display increased spreading, myocardin-related transcription factor-A (MRTF-A) nuclear localization, and focal adhesion size with increasing hydrogel stiffness. However, late passage (P5) hMSCs show reduced sensitivity to substrate mechanics with lower MRTF-A nuclear translocation and smaller focal adhesions on stiffer hydrogels compared to early passage hMSCs. Similar trends are observed in an immortalized human lung fibroblast line. Overall, this work highlights the implications of standard cell culture practices on investigating cell response to mechanical signals using in vitro hydrogel models.
Collapse
Affiliation(s)
- Jenna L Sumey
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA, 22903, USA
| | - Abigail M Harrell
- Department of Chemistry, University of Virginia, Charlottesville, VA, 22903, USA
| | - Peyton C Johnston
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, 22903, USA
| | - Steven R Caliari
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA, 22903, USA
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, 22903, USA
| |
Collapse
|
19
|
Ward NA, Hanley S, Tarpey R, Schreiber LHJ, O'Dwyer J, Roche ET, Duffy GP, Dolan EB. Intermittent actuation attenuates fibrotic behaviour of myofibroblasts. Acta Biomater 2024; 173:80-92. [PMID: 37967693 DOI: 10.1016/j.actbio.2023.11.017] [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: 06/22/2023] [Revised: 10/31/2023] [Accepted: 11/09/2023] [Indexed: 11/17/2023]
Abstract
The foreign body response (FBR) to implanted materials culminates in the deposition of a hypo-permeable, collagen rich fibrotic capsule by myofibroblast cells at the implant site. The fibrotic capsule can be deleterious to the function of some medical implants as it can isolate the implant from the host environment. Modulation of fibrotic capsule formation has been achieved using intermittent actuation of drug delivery implants, however the mechanisms underlying this response are not well understood. Here, we use analytical, computational, and in vitro models to understand the response of human myofibroblasts (WPMY-1 stromal cell line) to intermittent actuation using soft robotics and investigate how actuation can alter the secretion of collagen and pro/anti-inflammatory cytokines by these cells. Our findings suggest that there is a mechanical loading threshold that can modulate the fibrotic behaviour of myofibroblasts, by reducing the secretion of soluble collagen, transforming growth factor beta-1 and interleukin 1-beta, and upregulating the anti-inflammatory interleukin-10. By improving our understanding of how cells involved in the FBR respond to mechanical actuation, we can harness this technology to improve functional outcomes for a wide range of implanted medical device applications including drug delivery and cell encapsulation platforms. STATEMENT OF SIGNIFICANCE: A major barrier to the successful clinical translation of many implantable medical devices is the foreign body response (FBR) and resultant deposition of a hypo-permeable fibrotic capsule (FC) around the implant. Perturbation of the implant site using intermittent actuation (IA) of soft-robotic implants has previously been shown to modulate the FBR and reduce FC thickness. However, the mechanisms of action underlying this response were largely unknown. Here, we investigate how IA can alter the activity of myofibroblast cells, and ultimately suggest that there is a mechanical loading threshold within which their fibrotic behaviour can be modulated. These findings can be harnessed to improve functional outcomes for a wide range of medical implants, particularly drug delivery and cell encapsulation devices.
Collapse
Affiliation(s)
- Niamh A Ward
- Biomedical Engineering, School of Engineering, College of Science and Engineering, University of Galway, Galway, Ireland
| | - Shirley Hanley
- Flow Cytometry Core Facility, University of Galway, Galway, Ireland
| | - Ruth Tarpey
- Biomedical Engineering, School of Engineering, College of Science and Engineering, University of Galway, Galway, Ireland; Anatomy and Regenerative Medicine Institute (REMEDI), School of Medicine, University of Galway, Galway, Ireland
| | - Lucien H J Schreiber
- Biomedical Engineering, School of Engineering, College of Science and Engineering, University of Galway, Galway, Ireland; Anatomy and Regenerative Medicine Institute (REMEDI), School of Medicine, University of Galway, Galway, Ireland
| | - Joanne O'Dwyer
- Anatomy and Regenerative Medicine Institute (REMEDI), School of Medicine, University of Galway, Galway, Ireland
| | - Ellen T Roche
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA; Harvard-MIT Program in Health Sciences and Technology, Cambridge, MA, USA
| | - Garry P Duffy
- Anatomy and Regenerative Medicine Institute (REMEDI), School of Medicine, University of Galway, Galway, Ireland; Advanced Materials and BioEngineering Research Centre (AMBER), Trinity College Dublin, Dublin, Ireland; CÚRAM, Centre for Research in Medical Devices, University of Galway, Galway, Ireland
| | - Eimear B Dolan
- Biomedical Engineering, School of Engineering, College of Science and Engineering, University of Galway, Galway, Ireland; CÚRAM, Centre for Research in Medical Devices, University of Galway, Galway, Ireland.
| |
Collapse
|
20
|
Wells RG. Liver fibrosis: Our evolving understanding. Clin Liver Dis (Hoboken) 2024; 23:e0243. [PMID: 38961878 PMCID: PMC11221862 DOI: 10.1097/cld.0000000000000243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 03/29/2024] [Indexed: 07/05/2024] Open
|
21
|
Khalfallah M, Doblas S, Hammoutene A, Julea F, Postic C, Valla D, Paradis V, Garteiser P, Van Beers BE. Visco-Elastic Parameters at Three-Dimensional MR Elastography for Diagnosing Non-Alcoholic Steatohepatitis and Substantial Fibrosis in Mice. J Magn Reson Imaging 2024; 59:97-107. [PMID: 37158252 DOI: 10.1002/jmri.28765] [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: 01/08/2023] [Revised: 04/12/2023] [Accepted: 04/13/2023] [Indexed: 05/10/2023] Open
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) is increasing worldwide and is a growing cause of liver cirrhosis and cancer. The performance of the magnetic resonance elastography (MRE) visco-elastic parameters in diagnosing progressive forms of NAFLD, including nonalcoholic steatohepatitis (NASH) and substantial fibrosis (F ≥ 2), needs to be clarified. PURPOSE To assess the value of three-dimensional MRE visco-elastic parameters as markers of NASH and substantial fibrosis in mice with NAFLD. STUDY TYPE Prospective. ANIMAL MODEL Two mouse models of NAFLD were induced by feeding with high fat diet or high fat, choline-deficient, amino acid-defined diet. FIELD STRENGTH/SEQUENCE 7T/multi-slice multi-echo spin-echo MRE at 400 Hz with motion encoding in the three spatial directions. ASSESSMENT Hepatic storage and loss moduli were calculated. Histological analysis was based on the NASH Clinical Research Network criteria. STATISTICAL TESTS Mann-Whitney, Kruskal-Wallis tests, Spearman rank correlations and multiple regressions were used. Diagnostic performance was assessed with areas under the receiver operating characteristic curves (AUCs). P value <0.05 was considered significant. RESULTS Among the 59 mice with NAFLD, 21 had NASH and 20 had substantial fibrosis (including 8 mice without and 12 mice with NASH). The storage and loss moduli had similar moderate accuracy for diagnosing NASH with AUCs of 0.67 and 0.66, respectively. For diagnosing substantial fibrosis, the AUC of the storage modulus was 0.73 and the AUC of the loss modulus was 0.81, indicating good diagnostic performance. Using Spearman correlations, histological fibrosis, inflammation and steatosis, but not ballooning, were significantly correlated with the visco-elastic parameters. Using multiple regression, fibrosis was the only histological feature independently associated with the visco-elastic parameters. CONCLUSION MRE in mice with NAFLD suggests that the storage and loss moduli have good diagnostic performance for detecting progressive NAFLD defined as substantial fibrosis rather than NASH. EVIDENCE LEVEL 1 TECHNICAL EFFICACY STAGE: 2.
Collapse
Affiliation(s)
- Meryem Khalfallah
- Laboratory of Imaging Biomarkers, Center for Research on Inflammation, Université Paris Cité, UMR 1149 Inserm, Paris, France
| | - Sabrina Doblas
- Laboratory of Imaging Biomarkers, Center for Research on Inflammation, Université Paris Cité, UMR 1149 Inserm, Paris, France
| | - Adel Hammoutene
- Team "From Inflammation to Cancer in Digestive Disease", Center for Research on Inflammation, Université Paris Cité, UMR 1149 Inserm, Paris, France
| | - Felicia Julea
- Laboratory of Imaging Biomarkers, Center for Research on Inflammation, Université Paris Cité, UMR 1149 Inserm, Paris, France
| | - Catherine Postic
- Team "Glucose Sensing, Insulin Signaling and Glucotoxicity", Institut Cochin, Université Paris Cité, CNRS, Inserm, Paris, France
| | - Dominique Valla
- Department of Hepatology, Beaujon University Hospital Paris Nord, AP-HP, Clichy, France
| | - Valérie Paradis
- Team "From Inflammation to Cancer in Digestive Disease", Center for Research on Inflammation, Université Paris Cité, UMR 1149 Inserm, Paris, France
- Department of Pathology, Beaujon University Hospital Paris Nord, AP-HP, Clichy, France
| | - Philippe Garteiser
- Laboratory of Imaging Biomarkers, Center for Research on Inflammation, Université Paris Cité, UMR 1149 Inserm, Paris, France
| | - Bernard E Van Beers
- Laboratory of Imaging Biomarkers, Center for Research on Inflammation, Université Paris Cité, UMR 1149 Inserm, Paris, France
- Department of Radiology, Beaujon University Hospital Paris Nord, AP-HP, Clichy, France
| |
Collapse
|
22
|
Bonnevie ED, Scanzello CR, Mauck RL. Modulating mechanobiology as a therapeutic target for synovial fibrosis to restore joint lubrication. Osteoarthritis Cartilage 2024; 32:41-51. [PMID: 37866546 PMCID: PMC10880438 DOI: 10.1016/j.joca.2023.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 09/10/2023] [Accepted: 09/12/2023] [Indexed: 10/24/2023]
Abstract
OBJECTIVES Fibroses are disorders linked to persistence of myofibroblasts due to biochemical (e.g., Transforming growth factor-β) and biophysical cues (e.g., a stiff microenvironment). In the context of osteoarthritis, fibrotic changes in the joint-lining synovium have been linked with disease progression. The objective of this study was to probe synovial fibroblast mechanobiology and how essential functions (i.e., lubrication) are altered in fibrotic environments. DESIGN Both ex vivo and in vitro synovium models were assessed for fibrotic and lubrication biomarkers to better understand the role of mechanobiology and lubrication. Additionally, in vitro, work on small molecules targeting mechanobiology was assessed. RESULTS Our results indicated that modulating mechanobiology could rescue the fibrotic phenotype instigated by stiffening microenvironment that resulted in altered lubricant expression. A small molecule therapeutic, fasudil, blocked ROCK-mediated contractility and this inhibition of the fibrotic mechano-response of synovial fibroblasts restored proper lubrication function, providing insight into mechanisms of disease progression as well as a new avenue for therapeutic development. CONCLUSION This study identifies synovial fibrosis as a condition that potentially has joint-wide deficits through inhibiting lubrication. Additionally, modulating mechanobiology (i.e., ROCK-mediated contractility) may pose a potential target for small molecule therapies that can be delivered to the joint space. CLASSIFICATION Applied Biological Sciences.
Collapse
Affiliation(s)
- Edward D Bonnevie
- Translational Musculoskeletal Research Center, CMC VA Medical Center, United States; McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, University of Pennsylvania, United States.
| | - Carla R Scanzello
- Translational Musculoskeletal Research Center, CMC VA Medical Center, United States; Division of Rheumatology, Perelman School of Medicine, University of Pennsylvania, United States
| | - Robert L Mauck
- Translational Musculoskeletal Research Center, CMC VA Medical Center, United States; McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, University of Pennsylvania, United States; Department of Bioengineering, University of Pennsylvania, United States.
| |
Collapse
|
23
|
Carnicer-Lombarte A, Barone DG, Wronowski F, Malliaras GG, Fawcett JW, Franze K. Regenerative capacity of neural tissue scales with changes in tissue mechanics post injury. Biomaterials 2023; 303:122393. [PMID: 37977006 DOI: 10.1016/j.biomaterials.2023.122393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 10/23/2023] [Accepted: 11/05/2023] [Indexed: 11/19/2023]
Abstract
Spinal cord injuries have devastating consequences for humans, as mammalian neurons of the central nervous system (CNS) cannot regenerate. In the peripheral nervous system (PNS), however, neurons may regenerate to restore lost function following injury. While mammalian CNS tissue softens after injury, how PNS tissue mechanics changes in response to mechanical trauma is currently poorly understood. Here we characterised mechanical rat nerve tissue properties before and after in vivo crush and transection injuries using atomic force microscopy-based indentation measurements. Unlike CNS tissue, PNS tissue significantly stiffened after both types of tissue damage. This nerve tissue stiffening strongly correlated with an increase in collagen I levels. Schwann cells, which crucially support PNS regeneration, became more motile and proliferative on stiffer substrates in vitro, suggesting that changes in tissue stiffness may play a key role in facilitating or impeding nervous system regeneration.
Collapse
Affiliation(s)
- Alejandro Carnicer-Lombarte
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0PY, UK; Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3DY, UK; Electrical Engineering Division, Department of Engineering, University of Cambridge, Cambridge, CB3 0FA, UK.
| | - Damiano G Barone
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0PY, UK
| | - Filip Wronowski
- Electrical Engineering Division, Department of Engineering, University of Cambridge, Cambridge, CB3 0FA, UK
| | - George G Malliaras
- Electrical Engineering Division, Department of Engineering, University of Cambridge, Cambridge, CB3 0FA, UK
| | - James W Fawcett
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0PY, UK; Centre for Reconstructive Neuroscience, Institute for Experimental Medicine CAS, Prague, Czech Republic
| | - Kristian Franze
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3DY, UK; Institute of Medical Physics and Micro-Tissue Engineering, Friedrich-Alexander Universität Erlangen-Nürnberg, 91052, Erlangen, Germany; Max-Planck-Zentrum für Physik und Medizin, 91054, Erlangen, Germany.
| |
Collapse
|
24
|
Felli E, Selicean S, Guixé-Muntet S, Wang C, Bosch J, Berzigotti A, Gracia-Sancho J. Mechanobiology of portal hypertension. JHEP Rep 2023; 5:100869. [PMID: 37841641 PMCID: PMC10568428 DOI: 10.1016/j.jhepr.2023.100869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 06/28/2023] [Accepted: 07/03/2023] [Indexed: 10/17/2023] Open
Abstract
The interplay between mechanical stimuli and cellular mechanobiology orchestrates the physiology of tissues and organs in a dynamic balance characterized by constant remodelling and adaptative processes. Environmental mechanical properties can be interpreted as a complex set of information and instructions that cells read continuously, and to which they respond. In cirrhosis, chronic inflammation and injury drive liver cells dysfunction, leading to excessive extracellular matrix deposition, sinusoidal pseudocapillarization, vascular occlusion and parenchymal extinction. These pathological events result in marked remodelling of the liver microarchitecture, which is cause and result of abnormal environmental mechanical forces, triggering and sustaining the long-standing and progressive process of liver fibrosis. Multiple mechanical forces such as strain, shear stress, and hydrostatic pressure can converge at different stages of the disease until reaching a point of no return where the fibrosis is considered non-reversible. Thereafter, reciprocal communication between cells and their niches becomes the driving force for disease progression. Accumulating evidence supports the idea that, rather than being a passive consequence of fibrosis and portal hypertension (PH), mechanical force-mediated pathways could themselves represent strategic targets for novel therapeutic approaches. In this manuscript, we aim to provide a comprehensive review of the mechanobiology of PH, by furnishing an introduction on the most important mechanisms, integrating these concepts into a discussion on the pathogenesis of PH, and exploring potential therapeutic strategies.
Collapse
Affiliation(s)
- Eric Felli
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Switzerland
- Department for BioMedical Research, Visceral Surgery and Medicine, University of Bern, Switzerland
| | - Sonia Selicean
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Switzerland
- Department for BioMedical Research, Visceral Surgery and Medicine, University of Bern, Switzerland
| | - Sergi Guixé-Muntet
- Liver Vascular Biology Research Group, IDIBAPS Biomedical Research Institute, CIBEREHD, Spain
| | - Cong Wang
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Switzerland
- Department for BioMedical Research, Visceral Surgery and Medicine, University of Bern, Switzerland
| | - Jaume Bosch
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Switzerland
- Department for BioMedical Research, Visceral Surgery and Medicine, University of Bern, Switzerland
- Liver Vascular Biology Research Group, IDIBAPS Biomedical Research Institute, CIBEREHD, Spain
| | - Annalisa Berzigotti
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Switzerland
- Department for BioMedical Research, Visceral Surgery and Medicine, University of Bern, Switzerland
| | - Jordi Gracia-Sancho
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Switzerland
- Department for BioMedical Research, Visceral Surgery and Medicine, University of Bern, Switzerland
- Liver Vascular Biology Research Group, IDIBAPS Biomedical Research Institute, CIBEREHD, Spain
| |
Collapse
|
25
|
Bogahawaththa S, Kawamura T, Bandaranayake U, Hirakawa T, Yamada G, Ishino H, Hirohashi T, Kawaguchi SI, Wijesundera KK, Wijayagunawardane MPB, Ishimaru K, Kodithuwakku SP, Tsujita T. Identification and mechanistic investigation of ellagitannins from Osbeckia octandra that attenuate liver fibrosis via the TGF-β/SMAD signaling pathway. Biosci Biotechnol Biochem 2023; 87:1295-1309. [PMID: 37580142 DOI: 10.1093/bbb/zbad114] [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: 06/20/2023] [Accepted: 08/04/2023] [Indexed: 08/16/2023]
Abstract
Fibrosis is a major problem in chronic liver disease with limited treatment options due to its complex nature. Herbal medicines are often used as an alternative. The aim of this study was to investigate the therapeutic potential of Osbeckia octandra and to identify its active compounds and regulatory pathways. The effects of crude leaf suspension and boiled leaf extract were investigated in an animal model, and the extract was found to be the more effective treatment. Three major bioactive compounds, pedunculagin, casuarinin, and gallic acid, were isolated from the extract using the hepatic stellate cell line, LX-2-based antifibrotic effect evaluation system. The results showed that all these compounds ameliorated LX-2 in fibrotic state. This inhibitory mechanism was confirmed through the TGF-β/SMAD signaling pathway. Collectively, the presence of these compounds in O. octandra suggests its potential as a treatment for liver fibrosis.
Collapse
Affiliation(s)
- Sudarma Bogahawaththa
- The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima, 890-0065, Japan
- Laboratory of Biochemistry, Department of Advanced Lifesciences and Food Chemistry, Faculty of Agriculture, Saga University, Saga, 840-8502, Japan
- Department of Animal Science, Faculty of Agriculture, University of Peradeniya, Peradeniya, 20400, Sri Lanka
| | - Tomoaki Kawamura
- Laboratory of Biochemistry, Department of Advanced Lifesciences and Food Chemistry, Faculty of Agriculture, Saga University, Saga, 840-8502, Japan
| | - Udari Bandaranayake
- Department of Animal Science, Faculty of Agriculture, University of Peradeniya, Peradeniya, 20400, Sri Lanka
- Department of Functional Material Science, Graduate School of Science and Engineering, Saitama University, Shimo-Okubo 225, Sakura-ku, Saitama, 338-8570, Japan
| | - Tomoaki Hirakawa
- The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima, 890-0065, Japan
- Laboratory of Biochemistry, Department of Advanced Lifesciences and Food Chemistry, Faculty of Agriculture, Saga University, Saga, 840-8502, Japan
| | - Goki Yamada
- The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima, 890-0065, Japan
- Laboratory of Biochemistry, Department of Advanced Lifesciences and Food Chemistry, Faculty of Agriculture, Saga University, Saga, 840-8502, Japan
| | - Hana Ishino
- Department of Biological Resource Sciences, Faculty of Agriculture, Saga University, 1 Honjo, Saga, 840-8502, Japan
| | - Tsuzumi Hirohashi
- Department of Biological Resource Sciences, Faculty of Agriculture, Saga University, 1 Honjo, Saga, 840-8502, Japan
| | - Shin-Ichi Kawaguchi
- The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima, 890-0065, Japan
- Center for Education and Research in Agricultural Innovation, Faculty of Agriculture, Saga University, Karatsu, Saga, 847-0021, Japan
| | - Kavindra K Wijesundera
- Department of Veterinary Pathobiology, Faculty of Veterinary Medicine and Animal Science, University of Peradeniya, Peradeniya, 20400, Sri Lanka
| | | | - Kanji Ishimaru
- Department of Biological Resource Sciences, Faculty of Agriculture, Saga University, 1 Honjo, Saga, 840-8502, Japan
| | - Suranga P Kodithuwakku
- Department of Animal Science, Faculty of Agriculture, University of Peradeniya, Peradeniya, 20400, Sri Lanka
- ERA Chair COMBIVET, Institute of veterinary Medicine and Animal Science, Estonian University of Life Science, Tartu, 51014, Estonia
| | - Tadayuki Tsujita
- The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima, 890-0065, Japan
- Laboratory of Biochemistry, Department of Advanced Lifesciences and Food Chemistry, Faculty of Agriculture, Saga University, Saga, 840-8502, Japan
| |
Collapse
|
26
|
Phogat S, Thiam F, Al Yazeedi S, Abokor FA, Osei ET. 3D in vitro hydrogel models to study the human lung extracellular matrix and fibroblast function. Respir Res 2023; 24:242. [PMID: 37798767 PMCID: PMC10552248 DOI: 10.1186/s12931-023-02548-6] [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: 06/26/2023] [Accepted: 09/25/2023] [Indexed: 10/07/2023] Open
Abstract
The pulmonary extracellular matrix (ECM) is a macromolecular structure that provides mechanical support, stability and elastic recoil for different pulmonary cells including the lung fibroblasts. The ECM plays an important role in lung development, remodeling, repair, and the maintenance of tissue homeostasis. Biomechanical and biochemical signals produced by the ECM regulate the phenotype and function of various cells including fibroblasts in the lungs. Fibroblasts are important lung structural cells responsible for the production and repair of different ECM proteins (e.g., collagen and fibronectin). During lung injury and in chronic lung diseases such as asthma, idiopathic pulmonary fibrosis (IPF) and chronic obstructive pulmonary disease (COPD), an abnormal feedback between fibroblasts and the altered ECM disrupts tissue homeostasis and leads to a vicious cycle of fibrotic changes resulting in tissue remodeling. In line with this, using 3D hydrogel culture models with embedded lung fibroblasts have enabled the assessment of the various mechanisms involved in driving defective (fibrotic) fibroblast function in the lung's 3D ECM environment. In this review, we provide a summary of various studies that used these 3D hydrogel models to assess the regulation of the ECM on lung fibroblast phenotype and function in altered lung ECM homeostasis in health and in chronic respiratory disease.
Collapse
Affiliation(s)
- Sakshi Phogat
- Department of Biology, Okanagan Campus, University of British Columbia, 3187 University Way, ASC366, Kelowna, BC, V1V1V7, Canada
| | - Fama Thiam
- Department of Biology, Okanagan Campus, University of British Columbia, 3187 University Way, ASC366, Kelowna, BC, V1V1V7, Canada
| | - Safiya Al Yazeedi
- Department of Biology, Okanagan Campus, University of British Columbia, 3187 University Way, ASC366, Kelowna, BC, V1V1V7, Canada
| | - Filsan Ahmed Abokor
- Department of Biology, Okanagan Campus, University of British Columbia, 3187 University Way, ASC366, Kelowna, BC, V1V1V7, Canada
| | - Emmanuel Twumasi Osei
- Department of Biology, Okanagan Campus, University of British Columbia, 3187 University Way, ASC366, Kelowna, BC, V1V1V7, Canada.
- Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, V6Z 1Y6, Canada.
| |
Collapse
|
27
|
Wang S, Friedman SL. Found in translation-Fibrosis in metabolic dysfunction-associated steatohepatitis (MASH). Sci Transl Med 2023; 15:eadi0759. [PMID: 37792957 PMCID: PMC10671253 DOI: 10.1126/scitranslmed.adi0759] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 09/15/2023] [Indexed: 10/06/2023]
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) is a severe form of liver disease that poses a global health threat because of its potential to progress to advanced fibrosis, leading to cirrhosis and liver cancer. Recent advances in single-cell methodologies, refined disease models, and genetic and epigenetic insights have provided a nuanced understanding of MASH fibrogenesis, with substantial cellular heterogeneity in MASH livers providing potentially targetable cell-cell interactions and behavior. Unlike fibrogenesis, mechanisms underlying fibrosis regression in MASH are still inadequately understood, although antifibrotic targets have been recently identified. A refined antifibrotic treatment framework could lead to noninvasive assessment and targeted therapies that preserve hepatocellular function and restore the liver's architectural integrity.
Collapse
Affiliation(s)
- Shuang Wang
- Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Scott L. Friedman
- Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| |
Collapse
|
28
|
Ezzo M, Hinz B. Novel approaches to target fibroblast mechanotransduction in fibroproliferative diseases. Pharmacol Ther 2023; 250:108528. [PMID: 37708995 DOI: 10.1016/j.pharmthera.2023.108528] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/09/2023] [Accepted: 09/07/2023] [Indexed: 09/16/2023]
Abstract
The ability of cells to sense and respond to changes in mechanical environment is vital in conditions of organ injury when the architecture of normal tissues is disturbed or lost. Among the various cellular players that respond to injury, fibroblasts take center stage in re-establishing tissue integrity by secreting and organizing extracellular matrix into stabilizing scar tissue. Activation, activity, survival, and death of scar-forming fibroblasts are tightly controlled by mechanical environment and proper mechanotransduction ensures that fibroblast activities cease after completion of the tissue repair process. Conversely, dysregulated mechanotransduction often results in fibroblast over-activation or persistence beyond the state of normal repair. The resulting pathological accumulation of extracellular matrix is called fibrosis, a condition that has been associated with over 40% of all deaths in the industrialized countries. Consequently, elements in fibroblast mechanotransduction are scrutinized for their suitability as anti-fibrotic therapeutic targets. We review the current knowledge on mechanically relevant factors in the fibroblast extracellular environment, cell-matrix and cell-cell adhesion structures, stretch-activated membrane channels, stress-regulated cytoskeletal structures, and co-transcription factors. We critically discuss the targetability of these elements in therapeutic approaches and their progress in pre-clinical and/or clinical trials to treat organ fibrosis.
Collapse
Affiliation(s)
- Maya Ezzo
- Keenan Research Institute for Biomedical Science of the St. Michael's Hospital, and Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Boris Hinz
- Keenan Research Institute for Biomedical Science of the St. Michael's Hospital, and Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
29
|
Jun M, Lee YL, Zhou T, Maric M, Burke B, Park S, Low BC, Chiam KH. Subcellular Force Imbalance in Actin Bundles Induces Nuclear Repositioning and Durotaxis. ACS APPLIED MATERIALS & INTERFACES 2023; 15:43387-43402. [PMID: 37674326 DOI: 10.1021/acsami.3c07546] [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: 09/08/2023]
Abstract
Durotaxis is a phenomenon in which cells migrate toward substrates of increasing stiffness. However, how cells assimilate substrate stiffness as a directional cue remains poorly understood. In this study, we experimentally show that mouse embryonic fibroblasts can discriminate between different substrate stiffnesses and develop higher traction forces at regions of the cell adhering to the stiffer pillars. In this way, the cells generate a force imbalance between adhesion sites. It is this traction force imbalance that drives durotaxis by providing directionality for cell migration. Significantly, we found that traction forces are transmitted via LINC complexes to the cell nucleus, which serves to maintain the global force imbalance. In this way, LINC complexes play an essential role in anterograde nuclear movement and durotaxis. This conclusion is supported by the fact that LINC complex-deficient cells are incapable of durotaxis and instead migrate randomly on substrates featuring a stiffness gradient.
Collapse
Affiliation(s)
- Myeongjun Jun
- Bioinformatics institute, A*STAR, Singapore 138671, Singapore
- Department of Biological Science, National University of Singapore, Singapore 117558, Singapore
- School of Mechanical Engineering, Sungkyunkwan University, Suwon 16419, Korea
| | - Yin Loon Lee
- A*STAR Skin Research Laboratories, A*STAR, Singapore 138648, Singapore
| | - Tianxun Zhou
- Bioinformatics institute, A*STAR, Singapore 138671, Singapore
| | - Martina Maric
- A*STAR Skin Research Laboratories, A*STAR, Singapore 138648, Singapore
| | - Brian Burke
- A*STAR Skin Research Laboratories, A*STAR, Singapore 138648, Singapore
| | - Sungsu Park
- School of Mechanical Engineering, Sungkyunkwan University, Suwon 16419, Korea
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon 16419, Korea
| | - Boon Chuan Low
- Department of Biological Science, National University of Singapore, Singapore 117558, Singapore
- Mechanobiology Institute, National University of Singapore, Singapore 117558, Singapore
- NUS college, National University of Singapore, Singapore 117558, Singapore
| | - Keng-Hwee Chiam
- Bioinformatics institute, A*STAR, Singapore 138671, Singapore
| |
Collapse
|
30
|
Poe A, Martinez Yus M, Wang H, Santhanam L. Lysyl oxidase like-2 in fibrosis and cardiovascular disease. Am J Physiol Cell Physiol 2023; 325:C694-C707. [PMID: 37458436 PMCID: PMC10635644 DOI: 10.1152/ajpcell.00176.2023] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 07/12/2023] [Accepted: 07/12/2023] [Indexed: 09/01/2023]
Abstract
Fibrosis is an important and essential reparative response to injury that, if left uncontrolled, results in the excessive synthesis, deposition, remodeling, and stiffening of the extracellular matrix, which is deleterious to organ function. Thus, the sustained activation of enzymes that catalyze matrix remodeling and cross linking is a fundamental step in the pathology of fibrotic diseases. Recent studies have implicated the amine oxidase lysyl oxidase like-2 (LOXL2) in this process and established significantly elevated expression of LOXL2 as a key component of profibrotic conditions in several organ systems. Understanding the relationship between LOXL2 and fibrosis as well as the mechanisms behind these relationships can offer significant insights for developing novel therapies. Here, we summarize the key findings that demonstrate the link between LOXL2 and fibrosis and inflammation, examine current therapeutics targeting LOXL2 for the treatment of fibrosis, and discuss future directions for experiments and biomedical engineering.
Collapse
Affiliation(s)
- Alan Poe
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, United States
| | - Marta Martinez Yus
- Department of Anesthesiology and CCM, Johns Hopkins University, Baltimore, Maryland, United States
| | - Huilei Wang
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, United States
| | - Lakshmi Santhanam
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, United States
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, United States
- Department of Anesthesiology and CCM, Johns Hopkins University, Baltimore, Maryland, United States
| |
Collapse
|
31
|
Luangmonkong T, Parichatikanond W, Olinga P. Targeting collagen homeostasis for the treatment of liver fibrosis: Opportunities and challenges. Biochem Pharmacol 2023; 215:115740. [PMID: 37567319 DOI: 10.1016/j.bcp.2023.115740] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 07/24/2023] [Accepted: 08/08/2023] [Indexed: 08/13/2023]
Abstract
Liver fibrosis is an excessive production, aberrant deposition, and deficit degradation of extracellular matrix (ECM). Patients with unresolved fibrosis ultimately undergo end-stage liver diseases. To date, the effective and safe strategy to cease fibrosis progression remains an unmet clinical need. Since collagens are the most abundant ECM protein which play an essential role in fibrogenesis, the suitable regulation of collagen homeostasis could be an effective strategy for the treatment of liver fibrosis. Therefore, this review provides a brief overview on the dysregulation of ECM homeostasis, focusing on collagens, in the pathogenesis of liver fibrosis. Most importantly, promising therapeutic mechanisms related to biosynthesis, deposition and extracellular interactions, and degradation of collagens, together with preclinical and clinical antifibrotic evidence of drugs affecting each target are orderly criticized. In addition, challenges for targeting collagen homeostasis in the treatment of liver fibrosis are discussed.
Collapse
Affiliation(s)
- Theerut Luangmonkong
- Department of Pharmacology, Faculty of Pharmacy, Mahidol University, Thailand; Centre of Biopharmaceutical Science for Healthy Ageing (BSHA), Faculty of Pharmacy, Mahidol University, Bangkok, Thailand.
| | - Warisara Parichatikanond
- Department of Pharmacology, Faculty of Pharmacy, Mahidol University, Thailand; Centre of Biopharmaceutical Science for Healthy Ageing (BSHA), Faculty of Pharmacy, Mahidol University, Bangkok, Thailand
| | - Peter Olinga
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, The Netherlands
| |
Collapse
|
32
|
Llewellyn J, Fede C, Loneker AE, Friday CS, Hast MW, Theise ND, Furth EE, Guido M, Stecco C, Wells RG. Glisson's capsule matrix structure and function is altered in patients with cirrhosis irrespective of aetiology. JHEP Rep 2023; 5:100760. [PMID: 37534230 PMCID: PMC10393548 DOI: 10.1016/j.jhepr.2023.100760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 03/14/2023] [Accepted: 03/31/2023] [Indexed: 08/04/2023] Open
Abstract
Background & Aims Glisson's capsule is the interstitial connective tissue that surrounds the liver. As part of its normal physiology, it withstands significant daily changes in liver size. The pathophysiology of the capsule in disease is not well understood. The aim of this study was to characterise the changes in capsule matrix, cellular composition, and mechanical properties that occur in liver disease and to determine whether these correlate with disease severity or aetiology. Methods Samples from ten control patients, and six with steatosis, seven with moderate fibrosis, and 37 with cirrhosis were collected from autopsies, intraoperative biopsies, and liver explants. Matrix proteins and cell markers were assessed by staining and second harmonic generation imaging. Mechanical tensile testing was performed on a test frame. Results Capsule thickness was significantly increased in cirrhotic samples compared with normal controls irrespective of disease aetiology (70.12 ± 14.16 μm and 231.58 ± 21.82 μm, respectively), whereas steatosis and moderate fibrosis had no effect on thickness (90.91 ± 11.40 μm). Changes in cirrhosis included an increase in cell number (fibroblasts, vascular cells, infiltrating immune cells, and biliary epithelial cells). Key matrix components (collagens 1 and 3, hyaluronan, versican, and elastin) were all deposited in the lower capsule, although only the relative amounts per area of hyaluronan and versican were increased. Organisational features, including crimping and alignment of collagen fibres, were also altered in cirrhosis. Unexpectedly, capsules from cirrhotic livers had decreased resistance to loading compared with controls. Conclusions The liver capsule, similar to the parenchyma, is an active site of disease, demonstrating changes in matrix and cell composition as well as mechanical properties. Impact and implications We assessed the changes in composition and response to stretching of the liver outer sheath, the capsule, in human liver disease. We found an increase in key structural components and numbers of cells as well as a change in matrix organisation of the capsule during the later stages of disease. This allows the diseased capsule to stretch more under any given force, suggesting that it is less stiff than healthy tissue.
Collapse
Affiliation(s)
- Jessica Llewellyn
- Department of Gastroenterology and Hepatology, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Engineering MechanoBiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Caterina Fede
- Department of Neuroscience, University of Padova, Padova, Italy
| | - Abigail E. Loneker
- Center for Engineering MechanoBiology, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Chet S. Friday
- Department of Orthopaedic Surgery, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael W. Hast
- Department of Orthopaedic Surgery, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Neil D. Theise
- Department of Pathology, New York University, School of Medicine, New York, NY, USA
| | - Emma E. Furth
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Maria Guido
- Department of Pathology, University of Padova, Italy
| | - Carla Stecco
- Department of Neuroscience, University of Padova, Padova, Italy
| | - Rebecca G. Wells
- Department of Gastroenterology and Hepatology, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Engineering MechanoBiology, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
33
|
Li K, Ding K, Zhu Q, Han F, He X, Tan S, Wu Z, Zheng Z, Tang Z, Liu Y. Extracellular matrix stiffness aggravates urethral stricture through Igfbp3/Smad pathway. Sci Rep 2023; 13:14315. [PMID: 37653219 PMCID: PMC10471624 DOI: 10.1038/s41598-023-41584-6] [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: 05/15/2023] [Accepted: 08/29/2023] [Indexed: 09/02/2023] Open
Abstract
Urethral stricture refers to the narrowing of the urethral lumen. While previous studies have hinted at inflammation as the initial driver of this condition, the reasons and mechanisms behind its progression remain largely unknown. By Atomic force microscope (AFM), researchers measured the matrix stiffness of urethra to be 5.23 ± 0.37 kPa for normal tissue and 41.59 ± 2.48 kPa for stricture urethral scar. Similar results were observed in rat urethral stricture models, where the matrix stiffness of normal urethra was 4.29 ± 0.82 kPa, while 32.94 ± 7.12 kPa for urethral stricture scar. Notably, the matrix stiffness increased in rat models over time. To further investigate, polyacrylamide hydrogels were employed to mimic different levels of stiffness for normal and stricture condition. Interestingly, higher matrix stiffness led to an increased fibroblast-to-myofibroblast transition (FMT) in rat urethral fibroblasts, indicated by enhanced expression of α-SMA and Collagen I, as well as changing in the morphology of fibroblast. RNA-seq analysis suggested that Igfbp3/Smads might regulate the progressive FMT in urethral stricture. In the experiment where the expression of Igfbp3 was inhibited, increasing matrix stiffness lose the potential to stimulate FMT progression and the expression of p-Smad2/3 decreased. On the contrary, overexpression of Igfbp3 promoted the process of FMT in urethral fibroblasts. In conclusion, Igfbp3/Smad pathway appeared to be involved in the progression of urethral fibrosis. This finding suggested that Igfbp3/Smad might be an promising target for future research and treatment in this filed.
Collapse
Affiliation(s)
- Kaixuan Li
- Department of Cardiac Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Department of Urology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- Provincial Laboratory for Diagnosis and Treatment of Genitourinary System Disease, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Ke Ding
- Department of Urology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- Provincial Laboratory for Diagnosis and Treatment of Genitourinary System Disease, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Quan Zhu
- Department of Urology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- Provincial Laboratory for Diagnosis and Treatment of Genitourinary System Disease, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Feng Han
- Department of Urology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- Provincial Laboratory for Diagnosis and Treatment of Genitourinary System Disease, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Xi He
- Department of Orthopedics, The First Affiliated Hospital, Medical College of Zhejiang University, Hangzhou, 310003, Zhejiang, China
| | - Shuo Tan
- Provincial Laboratory for Diagnosis and Treatment of Genitourinary System Disease, 87 Xiangya Road, Changsha, 410008, Hunan, China
- Department of Urology, Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Ziqiang Wu
- Department of Urology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- Provincial Laboratory for Diagnosis and Treatment of Genitourinary System Disease, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Zhihuan Zheng
- Department of Urology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- Provincial Laboratory for Diagnosis and Treatment of Genitourinary System Disease, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Zhengyan Tang
- Department of Urology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China.
- Provincial Laboratory for Diagnosis and Treatment of Genitourinary System Disease, 87 Xiangya Road, Changsha, 410008, Hunan, China.
| | - Yanling Liu
- Department of Urology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China.
- Provincial Laboratory for Diagnosis and Treatment of Genitourinary System Disease, 87 Xiangya Road, Changsha, 410008, Hunan, China.
| |
Collapse
|
34
|
Bybee G, Moeun Y, Wang W, Kharbanda KK, Poluektova LY, Kidambi S, Osna NA, Ganesan M. Increased liver stiffness promotes hepatitis B progression by impairing innate immunity in CCl4-induced fibrotic HBV + transgenic mice. Front Immunol 2023; 14:1166171. [PMID: 37600826 PMCID: PMC10435739 DOI: 10.3389/fimmu.2023.1166171] [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: 02/14/2023] [Accepted: 07/17/2023] [Indexed: 08/22/2023] Open
Abstract
Background Hepatitis B virus (HBV) infection develops as an acute or chronic liver disease, which progresses from steatosis, hepatitis, and fibrosis to end-stage liver diseases such as cirrhosis and hepatocellular carcinoma (HCC). An increased stromal stiffness accompanies fibrosis in chronic liver diseases and is considered a strong predictor for disease progression. The goal of this study was to establish the mechanisms by which enhanced liver stiffness regulates HBV infectivity in the fibrotic liver tissue. Methods For in vitro studies, HBV-transfected HepG2.2.15 cells were cultured on polydimethylsiloxane gels coated by polyelectrolyte multilayer films of 2 kPa (soft) or 24 kPa (stiff) rigidity mimicking the stiffness of the healthy or fibrotic liver. For in vivo studies, hepatic fibrosis was induced in C57Bl/6 parental and HBV+ transgenic (HBVTg) mice by injecting CCl4 twice a week for 6 weeks. Results We found higher levels of HBV markers in stiff gel-attached hepatocytes accompanied by up-regulated OPN content in cell supernatants as well as suppression of anti-viral interferon-stimulated genes (ISGs). This indicates that pre-requisite "fibrotic" stiffness increases osteopontin (OPN) content and releases and suppresses anti-viral innate immunity, causing a subsequent rise in HBV markers expression in hepatocytes. In vitro results were corroborated by data from HBVTg mice administered CCl4 (HBVTg CCl4). These mice showed higher HBV RNA, DNA, HBV core antigen (HBcAg), and HBV surface antigen (HBsAg) levels after liver fibrosis induction as judged by a rise in Col1a1, SMA, MMPs, and TIMPs mRNAs and by increased liver stiffness. Importantly, CCl4-induced the pro-fibrotic activation of liver cells, and liver stiffness was higher in HBVTg mice compared with control mice. Elevation of HBV markers and OPN levels corresponded to decreased ISG activation in HBVTg CCl4 mice vs HBVTg control mice. Conclusion Based on our data, we conclude that liver stiffness enhances OPN levels to limit anti-viral ISG activation in hepatocytes and promote an increase in HBV infectivity, thereby contributing to end-stage liver disease progression.
Collapse
Affiliation(s)
- Grace Bybee
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Youra Moeun
- Department of Chemical and Biomolecular Engineering, University of Nebraska at Lincoln, Lincoln, NE, United States
| | - Weimin Wang
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| | - Kusum K. Kharbanda
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Larisa Y. Poluektova
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| | - Srivatsan Kidambi
- Department of Chemical and Biomolecular Engineering, University of Nebraska at Lincoln, Lincoln, NE, United States
| | - Natalia A. Osna
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Murali Ganesan
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
35
|
Liu X, Li J, Yang X, Li X, Kong J, Qi D, Zhang F, Sun B, Liu Y, Liu T. Carcinoma-associated fibroblast-derived lysyl oxidase-rich extracellular vesicles mediate collagen crosslinking and promote epithelial-mesenchymal transition via p-FAK/p-paxillin/YAP signaling. Int J Oral Sci 2023; 15:32. [PMID: 37532712 PMCID: PMC10397209 DOI: 10.1038/s41368-023-00236-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 07/05/2023] [Accepted: 07/05/2023] [Indexed: 08/04/2023] Open
Abstract
Carcinoma-associated fibroblasts (CAFs) are the main cellular components of the tumor microenvironment and promote cancer progression by modifying the extracellular matrix (ECM). The tumor-associated ECM is characterized by collagen crosslinking catalyzed by lysyl oxidase (LOX). Small extracellular vesicles (sEVs) mediate cell-cell communication. However, the interactions between sEVs and the ECM remain unclear. Here, we demonstrated that sEVs released from oral squamous cell carcinoma (OSCC)-derived CAFs induce collagen crosslinking, thereby promoting epithelial-mesenchymal transition (EMT). CAF sEVs preferably bound to the ECM rather than being taken up by fibroblasts and induced collagen crosslinking, and a LOX inhibitor or blocking antibody suppressed this effect. Active LOX (αLOX), but not the LOX precursor, was enriched in CAF sEVs and interacted with periostin, fibronectin, and bone morphogenetic protein-1 on the surface of sEVs. CAF sEV-associated integrin α2β1 mediated the binding of CAF sEVs to collagen I, and blocking integrin α2β1 inhibited collagen crosslinking by interfering with CAF sEV binding to collagen I. CAF sEV-induced collagen crosslinking promoted the EMT of OSCC through FAK/paxillin/YAP pathway. Taken together, these findings reveal a novel role of CAF sEVs in tumor ECM remodeling, suggesting a critical mechanism for CAF-induced EMT of cancer cells.
Collapse
Affiliation(s)
- Xue Liu
- Department of Oral Pathology, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China
- Department of Orthodontics, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, China
- School of Stomatology, Dalian Medical University, Dalian, China
| | - Jiao Li
- Department of Oral Pathology, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China
- Department of Orthodontics, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, China
- School of Stomatology, Dalian Medical University, Dalian, China
| | - Xuesong Yang
- Department of Biochemistry and Molecular Biology, Liaoning Provincial Core Lab of Glycobiology and Glycoengineering, Dalian Medical University, Dalian, China
| | - Xiaojie Li
- School of Stomatology, Dalian Medical University, Dalian, China
| | - Jing Kong
- School of Stomatology, Dalian Medical University, Dalian, China
| | - Dongyuan Qi
- Department of Oral Surgery, the First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Fuyin Zhang
- Department of Oral Surgery, the Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Bo Sun
- Department of Oral Surgery, the Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yuehua Liu
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China.
- Department of Orthodontics, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, China.
| | - Tingjiao Liu
- Department of Oral Pathology, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, China.
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China.
| |
Collapse
|
36
|
Sapudom J, Karaman S, Quartey BC, Mohamed WKE, Mahtani N, Garcia-Sabaté A, Teo J. Collagen Fibril Orientation Instructs Fibroblast Differentiation Via Cell Contractility. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301353. [PMID: 37249413 PMCID: PMC10401101 DOI: 10.1002/advs.202301353] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/03/2023] [Indexed: 05/31/2023]
Abstract
Collagen alignment is one of the key microarchitectural signatures of many pathological conditions, including scarring and fibrosis. Investigating how collagen alignment modulates cellular functions will pave the way for understanding tissue scarring and regeneration and new therapeutic strategies. However, current approaches for the fabrication of three-dimensional (3D) aligned collagen matrices are low-throughput and require special devices. To overcome these limitations, a simple approach to reconstitute homogeneous 3D collagen matrices with adjustable degree of fibril alignment using 3D printed inclined surfaces is developed. By characterizing the mechanical properties of reconstituted matrices, it is found that the elastic modulus of collagen matrices is enhanced with an increase in the alignment degree. The reconstituted matrices are used to study fibroblast behavior to reveal the progression of scar formation where a gradual enhancement of collagen alignment can be observed. It is found that matrices with aligned fibrils trigger fibroblast differentiation into myofibroblasts via cell contractility, while collagen stiffening through a crosslinker does not. The results suggest the impact of collagen fibril organization on the regulation of fibroblast differentiation. Overall, this approach to reconstitute 3D collagen matrices with fibril alignment opens opportunities for biomimetic pathological-relevant tissue in vitro, which can be applied for other biomedical research.
Collapse
Affiliation(s)
- Jiranuwat Sapudom
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, 129188, UAE
| | - Shaza Karaman
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, 129188, UAE
| | - Brian Chesney Quartey
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, 129188, UAE
| | - Walaa Kamal Eldin Mohamed
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, 129188, UAE
| | - Nick Mahtani
- School of Engineering, Ecole Polytechnique Federale de Lausanne, Lausanne, 1015, Switzerland
| | - Anna Garcia-Sabaté
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, 129188, UAE
| | - Jeremy Teo
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, 129188, UAE
- Department of Mechanical and Biomedical Engineering, Tandon School of Engineering, New York University, New York, 11201, USA
| |
Collapse
|
37
|
Di X, Gao X, Peng L, Ai J, Jin X, Qi S, Li H, Wang K, Luo D. Cellular mechanotransduction in health and diseases: from molecular mechanism to therapeutic targets. Signal Transduct Target Ther 2023; 8:282. [PMID: 37518181 PMCID: PMC10387486 DOI: 10.1038/s41392-023-01501-9] [Citation(s) in RCA: 46] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 05/10/2023] [Accepted: 05/11/2023] [Indexed: 08/01/2023] Open
Abstract
Cellular mechanotransduction, a critical regulator of numerous biological processes, is the conversion from mechanical signals to biochemical signals regarding cell activities and metabolism. Typical mechanical cues in organisms include hydrostatic pressure, fluid shear stress, tensile force, extracellular matrix stiffness or tissue elasticity, and extracellular fluid viscosity. Mechanotransduction has been expected to trigger multiple biological processes, such as embryonic development, tissue repair and regeneration. However, prolonged excessive mechanical stimulation can result in pathological processes, such as multi-organ fibrosis, tumorigenesis, and cancer immunotherapy resistance. Although the associations between mechanical cues and normal tissue homeostasis or diseases have been identified, the regulatory mechanisms among different mechanical cues are not yet comprehensively illustrated, and no effective therapies are currently available targeting mechanical cue-related signaling. This review systematically summarizes the characteristics and regulatory mechanisms of typical mechanical cues in normal conditions and diseases with the updated evidence. The key effectors responding to mechanical stimulations are listed, such as Piezo channels, integrins, Yes-associated protein (YAP) /transcriptional coactivator with PDZ-binding motif (TAZ), and transient receptor potential vanilloid 4 (TRPV4). We also reviewed the key signaling pathways, therapeutic targets and cutting-edge clinical applications of diseases related to mechanical cues.
Collapse
Affiliation(s)
- Xingpeng Di
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Xiaoshuai Gao
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Liao Peng
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Jianzhong Ai
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Xi Jin
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Shiqian Qi
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Hong Li
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Kunjie Wang
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China.
| | - Deyi Luo
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China.
| |
Collapse
|
38
|
Li D, Janmey PA, Wells RG. Local fat content determines global and local stiffness in livers with simple steatosis. FASEB Bioadv 2023; 5:251-261. [PMID: 37287868 PMCID: PMC10242205 DOI: 10.1096/fba.2022-00134] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 03/09/2023] [Accepted: 04/04/2023] [Indexed: 06/09/2023] Open
Abstract
Fat accumulation during liver steatosis precedes inflammation and fibrosis in fatty liver diseases, and is associated with disease progression. Despite a large body of evidence that liver mechanics play a major role in liver disease progression, the effect of fat accumulation by itself on liver mechanics remains unclear. Thus, we conducted ex vivo studies of liver mechanics in rodent models of simple steatosis to isolate and examine the mechanical effects of intrahepatic fat accumulation, and found that fat accumulation softens the liver. Using a novel adaptation of microindentation to permit association of local mechanics with microarchitectural features, we found evidence that the softening of fatty liver results from local softening of fatty regions rather than uniform softening of the liver. These results suggest that fat accumulation itself exerts a softening effect on liver tissue. This, along with the localized heterogeneity of softening within the liver, has implications in what mechanical mechanisms are involved in the progression of liver steatosis to more severe pathologies and disease. Finally, the ability to examine and associate local mechanics with microarchitectural features is potentially applicable to the study of the role of heterogeneous mechanical microenvironments in both other liver pathologies and other organ systems.
Collapse
Affiliation(s)
- David Li
- Division of Gastroenterology and HepatologyDepartment of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- NSF Science and Technology Center for Engineering MechanoBiologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Paul A. Janmey
- NSF Science and Technology Center for Engineering MechanoBiologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of BioengineeringUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Institute for Medicine and EngineeringUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of PhysiologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Rebecca G. Wells
- Division of Gastroenterology and HepatologyDepartment of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- NSF Science and Technology Center for Engineering MechanoBiologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of BioengineeringUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
39
|
Fan Y, Na SY, Jung YS, Radhakrishnan K, Choi HS. Estrogen-related receptor γ (ERRγ) is a key regulator of lysyl oxidase gene expression in mouse hepatocytes. Steroids 2023; 194:109226. [PMID: 36948345 DOI: 10.1016/j.steroids.2023.109226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 03/17/2023] [Accepted: 03/18/2023] [Indexed: 03/24/2023]
Abstract
Lysyl oxidase (LOX), the copper-dependent extracellular enzyme, plays a critical role in the regulation of protein cross-linking in the extracellular matrix (ECM). It is also involved in liver regeneration and liver fibrosis. However, the mechanism of LOX regulation in mouse hepatocytes is still unclear. Here, we identify a molecular mechanism showing that orphan nuclear receptor estrogen-related receptor γ (ERRγ) regulates LOX gene expression in the presence of the pro-inflammatory cytokine, interleukin 6 (IL6). IL6 significantly stimulated the expression of ERRγ and LOX in mouse hepatocytes. Overexpression of ERRγ increased LOX mRNA and protein levels. Moreover, knockdown of ERRγ attenuated IL6-mediated LOX gene expression at mRNA and protein levels. Overexpression of ERRγ or IL6 treatment upregulated LOX gene promoter activity, while knockdown of ERRγ decreased the IL6-induced LOX promoter activity. Furthermore, GSK5182, a specific ERRγ inverse agonist, inhibited the induction effect of IL6 on LOX promoter activity and gene expression in mouse hepatocytes. Overall, our study elucidates the mechanism involved in the LOX gene regulation by nuclear receptor ERRγ in response to IL6 in mouse hepatocytes, suggesting that, in conditions such as chronic inflammation, IL6 may contribute to liver fibrosis via inducing LOX gene expression. Thus, LOX gene regulation by the inverse agonist of ERRγ can be applied to improve liver fibrosis.
Collapse
Affiliation(s)
- Yiwen Fan
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Soon-Young Na
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Yoon Seok Jung
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Kamalakannan Radhakrishnan
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Hueng-Sik Choi
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea.
| |
Collapse
|
40
|
Sumey JL, Harrell AM, Johnston PC, Caliari SR. Serial passaging affects stromal cell mechanosensitivity on hyaluronic acid hydrogels. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.16.532853. [PMID: 36993247 PMCID: PMC10055097 DOI: 10.1101/2023.03.16.532853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
There is tremendous interest in developing hydrogels as tunable in vitro cell culture platforms to study cell response to mechanical cues in a controlled manner. However, little is known about how common cell culture techniques, such as serial expansion on tissue culture plastic, affect subsequent cell behavior when cultured on hydrogels. In this work we leverage a methacrylated hyaluronic acid hydrogel platform to study stromal cell mechanotransduction. Hydrogels are first formed through thiol-Michael addition to model normal soft tissue (e.g., lung) stiffness ( E ~ 1 kPa). Secondary crosslinking via radical photopolymerization of unconsumed methacrylates allows matching of early- ( E ~ 6 kPa) and late-stage fibrotic tissue ( E ~ 50 kPa). Early passage (P1) primary human mesenchymal stromal cells (hMSCs) display increased spreading, myocardin-related transcription factor-A (MRTF-A) nuclear localization, and focal adhesion size with increasing hydrogel stiffness. However, late passage (P5) hMSCs show reduced sensitivity to substrate mechanics with lower MRTF-A nuclear translocation and smaller focal adhesions on stiffer hydrogels compared to early passage hMSCs. Similar trends are observed in an immortalized human lung fibroblast line. Overall, this work highlights the implications of standard cell culture practices on investigating cell response to mechanical signals using in vitro hydrogel models.
Collapse
Affiliation(s)
- Jenna L. Sumey
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia 22903
| | - Abigail M. Harrell
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22903
| | - Peyton C. Johnston
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22903
| | - Steven R. Caliari
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia 22903
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22903
| |
Collapse
|
41
|
Wu B, Liu DA, Guan L, Myint PK, Chin L, Dang H, Xu Y, Ren J, Li T, Yu Z, Jabban S, Mills GB, Nukpezah J, Chen YH, Furth EE, Gimotty PA, Wells RG, Weaver VM, Radhakrishnan R, Wang XW, Guo W. Stiff matrix induces exosome secretion to promote tumour growth. Nat Cell Biol 2023; 25:415-424. [PMID: 36797475 PMCID: PMC10351222 DOI: 10.1038/s41556-023-01092-1] [Citation(s) in RCA: 75] [Impact Index Per Article: 75.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 01/12/2023] [Indexed: 02/18/2023]
Abstract
Tissue fibrosis and extracellular matrix (ECM) stiffening promote tumour progression. The mechanisms by which ECM regulates its contacting cells have been extensively studied. However, how stiffness influences intercellular communications in the microenvironment for tumour progression remains unknown. Here we report that stiff ECM stimulates the release of exosomes from cancer cells. We delineate a molecular pathway that links stiff ECM to activation of Akt, which in turn promotes GTP loading to Rab8 that drives exosome secretion. We further show that exosomes generated from cells grown on stiff ECM effectively promote tumour growth. Proteomic analysis revealed that the Notch signalling pathway is activated in cells treated with exosomes derived from tumour cells grown on stiff ECM, consistent with our gene expression analysis of liver tissues from patients. Our study reveals a molecular mechanism that regulates exosome secretion and provides insight into how mechanical properties of the ECM control the tumour microenvironment for tumour growth.
Collapse
Affiliation(s)
- Bin Wu
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Di-Ao Liu
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Lei Guan
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Phyoe Kyawe Myint
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - LiKang Chin
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hien Dang
- Department of Surgery, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Ye Xu
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Jinqi Ren
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Ting Li
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ziyan Yu
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Sophie Jabban
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Gordon B Mills
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Jonathan Nukpezah
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Youhai H Chen
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Emma E Furth
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Phyllis A Gimotty
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Rebecca G Wells
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Valerie M Weaver
- Department of Surgery, Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA, USA
| | - Ravi Radhakrishnan
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Xin Wei Wang
- Laboratory of Human Carcinogenesis and Liver Cancer Program, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Wei Guo
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
42
|
Júnior C, Ulldemolins A, Narciso M, Almendros I, Farré R, Navajas D, López J, Eroles M, Rico F, Gavara N. Multi-Step Extracellular Matrix Remodelling and Stiffening in the Development of Idiopathic Pulmonary Fibrosis. Int J Mol Sci 2023; 24:ijms24021708. [PMID: 36675222 PMCID: PMC9865994 DOI: 10.3390/ijms24021708] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/04/2023] [Accepted: 01/05/2023] [Indexed: 01/18/2023] Open
Abstract
The extracellular matrix (ECM) of the lung is a filamentous network composed mainly of collagens, elastin, and proteoglycans that provides structural and physical support to its populating cells. Proliferation, migration and overall behaviour of those cells is greatly determined by micromechanical queues provided by the ECM. Lung fibrosis displays an aberrant increased deposition of ECM which likely changes filament organization and stiffens the ECM, thus upregulating the profibrotic profile of pulmonary cells. We have previously used AFM to assess changes in the Young's Modulus (E) of the ECM in the lung. Here, we perform further ECM topographical, mechanical and viscoelastic analysis at the micro- and nano-scale throughout fibrosis development. Furthermore, we provide nanoscale correlations between topographical and elastic properties of the ECM fibres. Firstly, we identify a softening of the ECM after rats are instilled with media associated with recovery of mechanical homeostasis, which is hindered in bleomycin-instilled lungs. Moreover, we find opposite correlations between fibre stiffness and roughness in PBS- vs bleomycin-treated lung. Our findings suggest that changes in ECM nanoscale organization take place at different stages of fibrosis, with the potential to help identify pharmacological targets to hinder its progression.
Collapse
Affiliation(s)
- Constança Júnior
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain
- The Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - Anna Ulldemolins
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain
- CIBER de Enfermedades Respiratorias, 28029 Madrid, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, 08036 Barcelona, Spain
| | - Maria Narciso
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain
- The Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - Isaac Almendros
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain
- CIBER de Enfermedades Respiratorias, 28029 Madrid, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, 08036 Barcelona, Spain
| | - Ramon Farré
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain
- CIBER de Enfermedades Respiratorias, 28029 Madrid, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, 08036 Barcelona, Spain
| | - Daniel Navajas
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain
- The Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
- CIBER de Enfermedades Respiratorias, 28029 Madrid, Spain
| | - Javier López
- Institut Pasteur de Lille, U1019-UMR9017-CIIL-Centre d’Infection et d’Immunité de Lille, Université de Lille, CNRS, Inserm, CHU Lille, 59000 Lille, France
| | - Mar Eroles
- Aix-Marseille, CNRS, INSERM, LAI, Centuri Centre for Living Systems, 13009 Marseille, France
| | - Felix Rico
- Aix-Marseille, CNRS, INSERM, LAI, Centuri Centre for Living Systems, 13009 Marseille, France
| | - Núria Gavara
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain
- The Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
- Correspondence:
| |
Collapse
|
43
|
Guo T, Wantono C, Tan Y, Deng F, Duan T, Liu D. Regulators, functions, and mechanotransduction pathways of matrix stiffness in hepatic disease. Front Physiol 2023; 14:1098129. [PMID: 36711017 PMCID: PMC9878334 DOI: 10.3389/fphys.2023.1098129] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 01/06/2023] [Indexed: 01/15/2023] Open
Abstract
The extracellular matrix (ECM) provides physical support and imparts significant biochemical and mechanical cues to cells. Matrix stiffening is a hallmark of liver fibrosis and is associated with many hepatic diseases, especially liver cirrhosis and carcinoma. Increased matrix stiffness is not only a consequence of liver fibrosis but is also recognized as an active driver in the progression of fibrotic hepatic disease. In this article, we provide a comprehensive view of the role of matrix stiffness in the pathological progression of hepatic disease. The regulators that modulate matrix stiffness including ECM components, MMPs, and crosslinking modifications are discussed. The latest advances of the research on the matrix mechanics in regulating intercellular signaling and cell phenotype are classified, especially for hepatic stellate cells, hepatocytes, and immunocytes. The molecular mechanism that sensing and transducing mechanical signaling is highlighted. The current progress of ECM stiffness's role in hepatic cirrhosis and liver cancer is introduced and summarized. Finally, the recent trials targeting ECM stiffness for the treatment of liver disease are detailed.
Collapse
Affiliation(s)
- Ting Guo
- Department of Gastroenterology, The Second Xiangya Hospital of Central South University, Changsha, China,Research Center of Digestive Disease, Central South University, Changsha, China
| | - Cindy Wantono
- Department of Gastroenterology, The Second Xiangya Hospital of Central South University, Changsha, China,Research Center of Digestive Disease, Central South University, Changsha, China
| | - Yuyong Tan
- Department of Gastroenterology, The Second Xiangya Hospital of Central South University, Changsha, China,Research Center of Digestive Disease, Central South University, Changsha, China
| | - Feihong Deng
- Department of Gastroenterology, The Second Xiangya Hospital of Central South University, Changsha, China,Research Center of Digestive Disease, Central South University, Changsha, China
| | - Tianying Duan
- Department of Gastroenterology, The Second Xiangya Hospital of Central South University, Changsha, China,Research Center of Digestive Disease, Central South University, Changsha, China
| | - Deliang Liu
- Department of Gastroenterology, The Second Xiangya Hospital of Central South University, Changsha, China,Research Center of Digestive Disease, Central South University, Changsha, China,*Correspondence: Deliang Liu,
| |
Collapse
|
44
|
Mukade Y, Kobayashi S, Nishijima Y, Kimura K, Watanabe A, Ikota H, Shirabe K, Yokoo H, Saio M. Phosphotungstic Acid-treated Picrosirius Red Staining Improves Whole-slide Quantitative Analysis of Collagen in Histological Specimens. J Histochem Cytochem 2023; 71:11-26. [PMID: 36433833 PMCID: PMC9912349 DOI: 10.1369/00221554221141140] [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: 09/16/2022] [Accepted: 11/02/2022] [Indexed: 11/28/2022] Open
Abstract
We tried to prevent nonspecific nuclear staining (NS-NS) of picrosirius red (PSR) staining by treating the specimens with one of the heteropoly acids phosphotungstic acid (PTA). We analyzed a total of 35 cases of non-cancerous liver tissue for fibrosis and NS-NS under PSR-alone, phosphomolybdic acid (PMA)-pretreated PSR (PMA + PSR), or PTA-pretreated PSR (PTA + PSR) condition. In addition, we analyzed the photosensitivity of PMA or PTA single stain specimens. PTA + PSR significantly suppressed NS-NS compared with PSR. The color of the specimens did not change into blue by 30 times the exposure to whole slide scanner (WSS) light. The PTA + PSR condition showed the highest correlation with the Ishak score (pathological evaluation of liver fibrosis) compared with other conditions. Furthermore, Sirius Red-positive percentage (SRP%) in PSR was increased in the NS-NS observed cases. SRP% in PMA + PSR was significantly affected by WSS light exposure time. Moreover, the deposition of non-polarized PSR-stained substances (NP-PSR+S) clinging to the collagen fibers potentially explains why SRP% seemed bigger under PSR than PTA + PSR. Our protocol enabled us to analyze the whole slide image of PSR staining by high magnification, which would contribute to the accurate analysis of collagen amount in the tissue sections.
Collapse
Affiliation(s)
- Yui Mukade
- Laboratory of Histopathology and Cytopathology,
Department of Laboratory Sciences, Gunma University Graduate School of
Health Sciences, Maebashi, Japan
| | - Sayaka Kobayashi
- Laboratory of Histopathology and Cytopathology,
Department of Laboratory Sciences, Gunma University Graduate School of
Health Sciences, Maebashi, Japan
| | - Yoshimi Nishijima
- Laboratory of Histopathology and Cytopathology,
Department of Laboratory Sciences, Gunma University Graduate School of
Health Sciences, Maebashi, Japan
| | - Kiminori Kimura
- Department of Hepatology, Tokyo Metropolitan
Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| | - Akira Watanabe
- Department of Hepatobiliary and Pancreatic
Surgery, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Hayato Ikota
- Clinical Department of Pathology, Gunma
University Hospital, Maebashi, Japan
| | - Ken Shirabe
- Department of Hepatobiliary and Pancreatic
Surgery, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Hideaki Yokoo
- Department of Human Pathology, Gunma University
Graduate School of Medicine, Maebashi, Japan
| | - Masanao Saio
- Laboratory of Histopathology and Cytopathology,
Department of Laboratory Sciences, Gunma University Graduate School of
Health Sciences, Maebashi, Japan
| |
Collapse
|
45
|
Song D, Oberai AA, Janmey PA. Hyperelastic continuum models for isotropic athermal fibrous networks. Interface Focus 2022; 12:20220043. [PMID: 36330327 PMCID: PMC9560787 DOI: 10.1098/rsfs.2022.0043] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 09/05/2022] [Indexed: 10/16/2023] Open
Abstract
Many biological materials contain fibrous protein networks as their main structural components. Understanding the mechanical properties of such networks is important for creating biomimicking materials for cell and tissue engineering, and for developing novel tools for detecting and diagnosing disease. In this work, we develop continuum models for isotropic, athermal fibrous networks by combining a single-fibre model that describes the axial response of individual fibres, with network models that assemble individual fibre properties into overall network behaviour. In particular, we consider four different network models, including the affine, three-chain, eight-chain, and micro-sphere models, which employ different assumptions about network structure and kinematics. We systematically investigate the ability of these models to describe the mechanical response of athermal collagen and fibrin networks by comparing model predictions with experimental data. We test how each model captures network behaviour under three different loading conditions: uniaxial tension, simple shear, and combined tension and shear. We find that the affine and three-chain models can accurately describe both the axial and shear behaviour, whereas the eight-chain and micro-sphere models fail to capture the shear response, leading to unphysical zero shear moduli at infinitesimal strains. Our study is the first to systematically investigate the applicability of popular network models for describing the macroscopic behaviour of athermal fibrous networks, offering insights for selecting efficient models that can be used for large-scale, finite-element simulations of athermal networks.
Collapse
Affiliation(s)
- Dawei Song
- Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA, USA
- Department of Physiology, and Department of Physics and Astronomy, University of Pennsylvania, Philadelphia, PA, USA
| | - Assad A. Oberai
- Department of Aerospace and Mechanical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Paul A. Janmey
- Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA, USA
- Department of Physiology, and Department of Physics and Astronomy, University of Pennsylvania, Philadelphia, PA, USA
- Department of Physics and Astronomy, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
46
|
Abstract
AIM Fibrosis is a common pathological feature of most types of chronic liver injuries. There is no specific treatment for liver fibrosis at present. The liver microenvironment, which fosters the survival and activity of liver cells, plays an important role in maintaining the normal structure and physiological function of the liver. The aim of this review is to deeply understand the role of the liver microenvironment in the dynamic and complicated development of liver fibrosis. METHODS After searching in Elsevier ScienceDirect, PubMed and Web of Science databases using 'liver fibrosis' and 'microenvironment' as keywords, studies related to microenvironment in liver fibrosis was compiled and examined. RESULTS The homeostasis of the liver microenvironment is disrupted during the development of liver fibrosis, affecting liver cell function, causing various types of cell reactions, and changing the cell-cell and cell-matrix interactions, eventually affecting fibrosis formation. CONCLUSION Liver microenvironment may be important for identifying potential therapeutic targets, and restoring microenvironment homeostasis may be an important strategy for promoting the reversal of liver fibrosis.KEY MESSAGESThe homeostasis of the liver microenvironment is disrupted in liver fibrosis;A pro-fibrotic microenvironment is formed during the development of liver fibrosis;Restoring microenvironment homeostasis may be an important strategy for promoting the reversal of liver fibrosis.
Collapse
Affiliation(s)
- Ying Meng
- Department of General Medicine, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Tong Zhao
- Department of Orthopedics, Lanzhou University First Hospital, Lanzhou, Gansu, China
| | - Zhengyi Zhang
- Department of General Medicine, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Dekui Zhang
- Department of Gastroenterology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| |
Collapse
|
47
|
Brougham-Cook A, Kimmel HRC, Monckton CP, Owen D, Khetani SR, Underhill GH. Engineered matrix microenvironments reveal the heterogeneity of liver sinusoidal endothelial cell phenotypic responses. APL Bioeng 2022; 6:046102. [PMID: 36345318 PMCID: PMC9637025 DOI: 10.1063/5.0097602] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 09/29/2022] [Indexed: 11/06/2022] Open
Abstract
Fibrosis is one of the hallmarks of chronic liver disease and is associated with aberrant wound healing. Changes in the composition of the liver microenvironment during fibrosis result in a complex crosstalk of extracellular cues that promote altered behaviors in the cell types that comprise the liver sinusoid, particularly liver sinusoidal endothelial cells (LSECs). Recently, it has been observed that LSECs may sustain injury before other fibrogenesis-associated cells of the sinusoid, implicating LSECs as key actors in the fibrotic cascade. A high-throughput cellular microarray platform was used to deconstruct the collective influences of defined combinations of extracellular matrix (ECM) proteins, substrate stiffness, and soluble factors on primary human LSEC phenotype in vitro. We observed remarkable heterogeneity in LSEC phenotype as a function of stiffness, ECM, and soluble factor context. LYVE-1 and CD-31 expressions were highest on 1 kPa substrates, and the VE-cadherin junction localization was highest on 25 kPa substrates. Also, LSECs formed distinct spatial patterns of LYVE-1 expression, with LYVE-1+ cells observed in the center of multicellular domains, and pattern size regulated by microenvironmental context. ECM composition also influenced a substantial dynamic range of expression levels for all markers, and the collagen type IV was observed to promote elevated expressions of LYVE-1, VE-cadherin, and CD-31. These studies highlight key microenvironmental regulators of LSEC phenotype and reveal unique spatial patterning of the sinusoidal marker LYVE-1. Furthermore, these data provide insight into understanding more precisely how LSECs respond to fibrotic microenvironments, which will aid drug development and identification of targets to treat liver fibrosis.
Collapse
Affiliation(s)
- Aidan Brougham-Cook
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Hannah R. C. Kimmel
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Chase P. Monckton
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, Illinois 60607, USA
| | - Daniel Owen
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Salman R. Khetani
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, Illinois 60607, USA
| | - Gregory H. Underhill
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA,Author to whom correspondence should be addressed:. Tel.: 217–244-2169
| |
Collapse
|
48
|
Kletukhina S, Mutallapova G, Titova A, Gomzikova M. Role of Mesenchymal Stem Cells and Extracellular Vesicles in Idiopathic Pulmonary Fibrosis. Int J Mol Sci 2022; 23:ijms231911212. [PMID: 36232511 PMCID: PMC9569825 DOI: 10.3390/ijms231911212] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 09/20/2022] [Accepted: 09/20/2022] [Indexed: 11/16/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive interstitial fibrotic disease that leads to disability and death within 5 years of diagnosis. Pulmonary fibrosis is a disease with a multifactorial etiology. The concept of aberrant regeneration of the pulmonary epithelium reveals the pathogenesis of IPF, according to which repeated damage and death of alveolar epithelial cells is the main mechanism leading to the development of progressive IPF. Cell death provokes the migration, proliferation and activation of fibroblasts, which overproduce extracellular matrix, resulting in fibrotic deformity of the lung tissue. Mesenchymal stem cells (MSCs) and extracellular vesicles (EVs) are promising therapies for pulmonary fibrosis. MSCs, and EVs derived from MSCs, modulate the activity of immune cells, inhibit the expression of profibrotic genes, reduce collagen deposition and promote the repair of damaged lung tissue. This review considers the molecular mechanisms of the development of IPF and the multifaceted role of MSCs in the therapy of IPF. Currently, EVs-MSCs are regarded as a promising cell-free therapy tool, so in this review we discuss the results available to date of the use of EVs-MSCs for lung tissue repair.
Collapse
Affiliation(s)
- Sevindzh Kletukhina
- Laboratory of Intercellular Communication, Kazan Federal University, 420008 Kazan, Russia
| | - Guzel Mutallapova
- Laboratory of Intercellular Communication, Kazan Federal University, 420008 Kazan, Russia
| | - Angelina Titova
- Morphology and General Pathology Department, Kazan Federal University, 420008 Kazan, Russia
| | - Marina Gomzikova
- Laboratory of Intercellular Communication, Kazan Federal University, 420008 Kazan, Russia
- Correspondence: ; Tel.: +7-917-8572269
| |
Collapse
|
49
|
Mowla A, Belford R, Köhn-Gaone J, Main N, Tirnitz-Parker JEE, Yeoh GC, Kennedy BF. Biomechanical assessment of chronic liver injury using quantitative micro-elastography. BIOMEDICAL OPTICS EXPRESS 2022; 13:5050-5066. [PMID: 36187256 PMCID: PMC9484444 DOI: 10.1364/boe.467684] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/17/2022] [Accepted: 08/19/2022] [Indexed: 06/16/2023]
Abstract
Hepatocellular carcinoma is one of the most lethal cancers worldwide, causing almost 700,000 deaths annually. It mainly arises from cirrhosis, which, in turn, results from chronic injury to liver cells and corresponding fibrotic changes. Although it is known that chronic liver injury increases the elasticity of liver tissue, the role of increased elasticity of the microenvironment as a possible hepatocarcinogen is yet to be investigated. One reason for this is the paucity of imaging techniques capable of mapping the micro-scale elasticity variation in liver and correlating that with cancerous mechanisms on the cellular scale. The clinical techniques of ultrasound elastography and magnetic resonance elastography typically do not provide micro-scale resolution, while atomic force microscopy can only assess the elasticity of a limited number of cells. We propose quantitative micro-elastography (QME) for mapping the micro-scale elasticity of liver tissue into images known as micro-elastograms, and therefore, as a technique capable of correlating the micro-environment elasticity of tissue with cellular scale cancerous mechanisms in liver. We performed QME on 13 freshly excised healthy and diseased mouse livers and present micro-elastograms, together with co-registered histology, in four representative cases. Our results indicate a significant increase in the mean (×6.3) and standard deviation (×6.0) of elasticity caused by chronic liver injury and demonstrate that the onset and progression of pathological features such as fibrosis, hepatocyte damage, and immune cell infiltration correlate with localized variations in micro-elastograms.
Collapse
Affiliation(s)
- Alireza Mowla
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, and Centre for Medical Research, The University of Western Australia, Perth, WA 6009, Australia
- Department of Electrical, Electronic & Computer Engineering, School of Engineering, The University of Western Australia, Perth, WA 6009, Australia
| | - Rose Belford
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, and Centre for Medical Research, The University of Western Australia, Perth, WA 6009, Australia
- Department of Electrical, Electronic & Computer Engineering, School of Engineering, The University of Western Australia, Perth, WA 6009, Australia
| | - Julia Köhn-Gaone
- Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, Australia
| | - Nathan Main
- Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, Australia
| | - Janina E. E. Tirnitz-Parker
- Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, Australia
- Centre for Medical Research, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, WA 6009, Australia
| | - George C. Yeoh
- Centre for Medical Research, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, WA 6009, Australia
- School of Molecular Sciences, The University of Western Australia, Perth, WA 6009, Australia
- Centre for Cell Therapy and Regenerative Medicine, School of Biomedical Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Brendan F. Kennedy
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, and Centre for Medical Research, The University of Western Australia, Perth, WA 6009, Australia
- Department of Electrical, Electronic & Computer Engineering, School of Engineering, The University of Western Australia, Perth, WA 6009, Australia
- Australian Research Council Centre for Personalised Therapeutics Technologies, Australia
| |
Collapse
|
50
|
Parihar K, Nukpezah J, Iwamoto DV, Janmey PA, Radhakrishnan R. Data driven and biophysical insights into the regulation of trafficking vesicles by extracellular matrix stiffness. iScience 2022; 25:104721. [PMID: 35865140 PMCID: PMC9293776 DOI: 10.1016/j.isci.2022.104721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/30/2022] [Accepted: 06/28/2022] [Indexed: 11/19/2022] Open
Abstract
Biomechanical signals from remodeled extracellular matrix (ECM) promote tumor progression. Here, we show that cell-matrix and cell-cell communication may be inherently linked and tuned through mechanisms of mechanosensitive biogenesis of trafficking vesicles. Pan-cancer analysis of cancer cells' mechanical properties (focusing primarily on cell stiffness) on substrates of varied stiffness and composition elucidated a heterogeneous cellular response to mechanical stimuli. Through machine learning, we identified a fingerprint of cytoskeleton-related proteins that accurately characterize cell stiffness in different ECM conditions. Expression of their respective genes correlates with patient prognosis across different tumor types. The levels of selected cytoskeleton proteins indicated that cortical tension mirrors the increase (or decrease) in cell stiffness with a change in ECM stiffness. A mechanistic biophysical model shows that the tendency for curvature generation by curvature-inducing proteins has an ultrasensitive dependence on cortical tension. This study thus highlights the effect of ECM stiffness, mediated by cortical tension, in modulating vesicle biogenesis.
Collapse
Affiliation(s)
- Kshitiz Parihar
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jonathan Nukpezah
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daniel V. Iwamoto
- Department of Physiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Paul A. Janmey
- Department of Physiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ravi Radhakrishnan
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|