1
|
White MJV, Ozkan M, Medellin JEG, Solanki A, Hubbell JA. Inhibition of Talin2 dedifferentiates myofibroblasts and reverses lung and kidney fibrosis. Sci Rep 2025; 15:18010. [PMID: 40410300 DOI: 10.1038/s41598-025-00939-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 05/02/2025] [Indexed: 05/25/2025] Open
Abstract
Fibrosis is involved in 45% of deaths in the United States, and no treatment exists to reverse progression of the disease. To find novel targets for fibrosis therapeutics, we developed a model for the differentiation of monocytes to myofibroblasts that allowed us to screen for proteins involved in myofibroblast differentiation. Inhibition of a novel protein target generated by our model, talin2, reduces myofibroblast-specific morphology, α-smooth muscle actin content, and collagen I content and lowers the pro-fibrotic secretome of myofibroblasts. We find that knockdown of talin2 de-differentiates myofibroblasts and reverses bleomycin-induced lung fibrosis in mice, and further that Tln2-/- mice are resistant to bleomycin-induced lung fibrosis and resistant to unilateral ureteral obstruction-induced kidney fibrosis. Talin2 inhibition is thus a potential treatment for reversing lung and kidney fibroses.
Collapse
Affiliation(s)
- Michael J V White
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, 60637, USA
| | - Melis Ozkan
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, 60637, USA
| | | | - Ani Solanki
- Animal Resources Center, University of Chicago, Chicago, IL, 60637, USA
| | - Jeffrey A Hubbell
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, 60637, USA.
- Committee on Immunology, University of Chicago, Chicago, IL, 60637, USA.
- Committee on Cancer Biology, University of Chicago, Chicago, IL, 60637, USA.
- Department of Chemical and Biomolecular Engineering, Tandon School of Engineering, New York University, New York, 11201, New York, United States.
- Departments of Biology and Chemistry, Faculty of Arts and Sciences, New York University, New York, 10012, New York, United States.
- Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, 10016, New York, United States.
| |
Collapse
|
2
|
Haaker MW, Chang JC, Chung BK, Pieper TS, Noé F, Wang T, Geijsen N, Houweling M, Wolfrum C, Vaandrager AB, Melum E, Spee B, Helms JB. Cellular Crosstalk Promotes Hepatic Progenitor Cell Proliferation and Stellate Cell Activation in 3D Co-culture. Cell Mol Gastroenterol Hepatol 2025; 19:101472. [PMID: 39892785 PMCID: PMC11968293 DOI: 10.1016/j.jcmgh.2025.101472] [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: 04/25/2024] [Revised: 01/23/2025] [Accepted: 01/23/2025] [Indexed: 02/04/2025]
Abstract
BACKGROUND & AIMS Following liver damage, ductular reaction often coincides with liver fibrosis. Proliferation of hepatic progenitor cells is observed in ductular reaction, whereas activated hepatic stellate cells (HSCs) are the main drivers of liver fibrosis. These observations may suggest a functional interaction between these 2 cell types. Here, we report on an in vitro co-culture system to examine these interactions and validate their co-expression in human liver explants. METHODS In a 3D organoid co-culture system, we combined freshly isolated quiescent mouse HSCs and fluorescently labeled progenitor cells (undifferentiated intrahepatic cholangiocyte organoids), permitting real-time observation of cell morphology and behavior. After 7 days, cells were sorted based on the fluorescent label and analyzed for changes in gene expression. RESULTS In the 3D co-culture system, the proliferation of progenitor cells is enhanced, and HSCs are activated, recapitulating the cellular events observed in the patient liver. Both effects in 3D co-culture require close contact between the 2 different cell types. HSC activation during 3D co-culture differs from quiescent (3D mono-cultured) HSCs and activated HSCs on plastic (2D mono-culture). Upregulation of a cluster of genes containing Aldh1a2, Cthrc1, and several genes related to frizzled binding/Wnt signaling were exclusively observed in 3D co-cultured HSCs. The localized co-expression of specific genes was confirmed by spatial transcriptomics in human liver explants. CONCLUSION An in vitro 3D co-culture system provides evidence for direct interactions between HSCs and progenitor cells, which are sufficient to drive responses that are similar to those seen during ductular reaction and fibrosis. This model paves the way for further research into the cellular basis of liver pathology.
Collapse
Affiliation(s)
- Maya W Haaker
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, The Netherlands
| | - Jung-Chin Chang
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, The Netherlands
| | - Brian K Chung
- Norwegian PSC Research Center, Department of Transplantation Medicine, Division of Surgery and Specialized Medicine, eDivision of Surgery and Specialized Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Tobias S Pieper
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, The Netherlands
| | - Falko Noé
- Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland
| | - Tongtong Wang
- Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland
| | - Niels Geijsen
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Martin Houweling
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, The Netherlands
| | - Christian Wolfrum
- Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland
| | - Arie B Vaandrager
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, The Netherlands
| | - Espen Melum
- Norwegian PSC Research Center, Department of Transplantation Medicine, Division of Surgery and Specialized Medicine, eDivision of Surgery and Specialized Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway; Section of Gastroenterology, Department of Transplantation Medicine, Division of Surgery and Specialized Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Hybrid Technology Hub-Centre of Excellence, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Bart Spee
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - J Bernd Helms
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, The Netherlands.
| |
Collapse
|
3
|
Zeng X, Liao Y, Cheng W. Transient receptor potential channel 6 knockout ameliorates hepatic fibrosis by inhibiting the activation and proliferation of hepatic stellate cells. J Gastroenterol Hepatol 2025; 40:294-303. [PMID: 39511967 DOI: 10.1111/jgh.16802] [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: 08/15/2024] [Revised: 09/25/2024] [Accepted: 10/24/2024] [Indexed: 11/15/2024]
Abstract
BACKGROUND AND AIM Hepatic fibrosis is a common outcome of chronic liver injury and can eventually lead to cirrhosis, which is a major public health concern. Hepatic stellate cells (HSCs) are the major producers of extracellular matrix (ECM) and regulate the synthesis and decomposition of ECM, but the specific mechanism of them remains unclear. Transient receptor potential channel 6 (TRPC6), a non-selective cation channel, plays an important role in organic fibrosis. However, the role of TRPC6 in liver fibrosis is rarely studied. METHODS Here, we investigated the function of TRPC6 in the activation of the human hepatic stellate cell line LX-2 in vitro and bile duct ligation (BDL)-induced hepatic fibrosis in vivo by western blot, Ca2+ imaging, and immunohistochemistry. RESULTS We first found that TRPC6 was upregulated in fibrotic liver tissues and TRPC6 knockout inhibited BDL-induced hepatic fibrosis. Transforming growth factor-β1 (TGF-β1) treatment increased TRPC6 expression and thapsigargin (Tg)-mediated SOCE in LX-2 cells, which was decreased by the TRPC6 specific inhibitor SAR7334. Blockage of TRPC6 by SAR7334 or TRPC6-shRNA transfection attenuated TGF-β1-induced LX-2 cell activation and proliferation via the PI3K/AKT/p70S6K signaling pathway. CONCLUSIONS These observations suggested that TRPC6 contribute to LX-2 cell activation and hepatic fibrosis, and downregulation of TRPC6 may become a therapeutic strategy for the treatment of hepatic fibrosis in the future.
Collapse
Affiliation(s)
- Xixi Zeng
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yanhong Liao
- Department of Anatomy, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Weiyi Cheng
- Department of Pain, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
4
|
Haaker MW, Goossens V, Hoogland NAN, van Doorne H, Wang Z, Jansen JWA, Kaloyanova DV, van de Lest CHA, Houweling M, Vaandrager AB, Helms JB. Early activation of hepatic stellate cells induces rapid initiation of retinyl ester breakdown while maintaining lecithin:retinol acyltransferase (LRAT) activity. Biochim Biophys Acta Mol Cell Biol Lipids 2024; 1869:159540. [PMID: 39068984 DOI: 10.1016/j.bbalip.2024.159540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 06/30/2024] [Accepted: 07/25/2024] [Indexed: 07/30/2024]
Abstract
Lecithin:retinol acyltransferase (LRAT) is the main enzyme producing retinyl esters (REs) in quiescent hepatic stellate cells (HSCs). When cultured on stiff plastic culture plates, quiescent HSCs activate and lose their RE stores in a process similar to that in the liver following tissue damage, leading to fibrosis. Here we validated HSC cultures in soft gels to study RE metabolism in stable quiescent HSCs and investigated RE synthesis and breakdown in activating HSCs. HSCs cultured in a soft gel maintained characteristics of quiescent HSCs, including the size, amount and composition of their characteristic large lipid droplets. Quiescent gel-cultured HSCs maintained high expression levels of Lrat and a RE storing phenotype with low levels of RE breakdown. Newly formed REs are highly enriched in retinyl palmitate (RP), similar to freshly isolated quiescent HSCs, which is associated with high LRAT activity. Comparison of these quiescent gel-cultured HSCs with activated plastic-cultured HSCs showed that although during early activation the total RE levels and RP-enrichment are reduced, levels of RE formation are maintained and mediated by LRAT. Loss of REs was caused by enhanced RE breakdown in activating HSCs. Upon prolonged culturing, activated HSCs have lost their LRAT activity and produce small amounts of REs by DGAT1. This study reveals unexpected dynamics in RE metabolism during early HSC activation, which might be important in liver disease as early stages are reversible. Soft gel cultures provide a promising model to study RE metabolism in quiescent HSCs, allowing detailed molecular investigations on the mechanisms for storage and release.
Collapse
Affiliation(s)
- Maya W Haaker
- Department of Biomolecular Health Sciences, Division of Cell Biology, Metabolism & Cancer, Faculty of Veterinary Medicine and Institute of Biomembranes, Utrecht University, 3584 CM Utrecht, the Netherlands
| | - Vera Goossens
- Department of Biomolecular Health Sciences, Division of Cell Biology, Metabolism & Cancer, Faculty of Veterinary Medicine and Institute of Biomembranes, Utrecht University, 3584 CM Utrecht, the Netherlands
| | - Nina A N Hoogland
- Department of Biomolecular Health Sciences, Division of Cell Biology, Metabolism & Cancer, Faculty of Veterinary Medicine and Institute of Biomembranes, Utrecht University, 3584 CM Utrecht, the Netherlands
| | - Hidde van Doorne
- Department of Biomolecular Health Sciences, Division of Cell Biology, Metabolism & Cancer, Faculty of Veterinary Medicine and Institute of Biomembranes, Utrecht University, 3584 CM Utrecht, the Netherlands
| | - Ziqiong Wang
- Department of Biomolecular Health Sciences, Division of Cell Biology, Metabolism & Cancer, Faculty of Veterinary Medicine and Institute of Biomembranes, Utrecht University, 3584 CM Utrecht, the Netherlands
| | - Jeroen W A Jansen
- Department of Biomolecular Health Sciences, Division of Cell Biology, Metabolism & Cancer, Faculty of Veterinary Medicine and Institute of Biomembranes, Utrecht University, 3584 CM Utrecht, the Netherlands
| | - Dora V Kaloyanova
- Department of Biomolecular Health Sciences, Division of Cell Biology, Metabolism & Cancer, Faculty of Veterinary Medicine and Institute of Biomembranes, Utrecht University, 3584 CM Utrecht, the Netherlands
| | - Chris H A van de Lest
- Department of Biomolecular Health Sciences, Division of Cell Biology, Metabolism & Cancer, Faculty of Veterinary Medicine and Institute of Biomembranes, Utrecht University, 3584 CM Utrecht, the Netherlands
| | - Martin Houweling
- Department of Biomolecular Health Sciences, Division of Cell Biology, Metabolism & Cancer, Faculty of Veterinary Medicine and Institute of Biomembranes, Utrecht University, 3584 CM Utrecht, the Netherlands
| | - A Bas Vaandrager
- Department of Biomolecular Health Sciences, Division of Cell Biology, Metabolism & Cancer, Faculty of Veterinary Medicine and Institute of Biomembranes, Utrecht University, 3584 CM Utrecht, the Netherlands
| | - J Bernd Helms
- Department of Biomolecular Health Sciences, Division of Cell Biology, Metabolism & Cancer, Faculty of Veterinary Medicine and Institute of Biomembranes, Utrecht University, 3584 CM Utrecht, the Netherlands.
| |
Collapse
|
5
|
Kalaitzidou C, Grekas G, Zilian A, Makridakis C, Rosakis P. Compressive instabilities enable cell-induced extreme densification patterns in the fibrous extracellular matrix: Discrete model predictions. PLoS Comput Biol 2024; 20:e1012238. [PMID: 38950077 PMCID: PMC11244807 DOI: 10.1371/journal.pcbi.1012238] [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: 07/24/2023] [Revised: 07/12/2024] [Accepted: 06/08/2024] [Indexed: 07/03/2024] Open
Abstract
We present a new model and extensive computations that explain the dramatic remodelling undergone by a fibrous collagen extracellular matrix (ECM), when subjected to contractile mechanical forces from embedded cells or cell clusters. This remodelling creates complex patterns, comprising multiple narrow localised bands of severe densification and fiber alignment, extending far into the ECM, often joining distant cells or cell clusters (such as tumours). Most previous models cannot capture this behaviour, as they assume stable mechanical fiber response with stress an increasing function of fiber stretch, and a restriction to small displacements. Our fully nonlinear network model distinguishes between two types of single-fiber nonlinearity: fibers that undergo stable (supercritical) buckling (as in previous work) versus fibers that suffer unstable (subcritical) buckling collapse. The model allows unrestricted, arbitrarily large displacements (geometric nonlinearity). Our assumptions on single-fiber instability are supported by recent simulations and experiments on buckling of individual beams with a hierarchical microstructure, such as collagen fibers. We use simple scenarios to illustrate, for the first time, two distinct compressive-instability mechanisms at work in our model: unstable buckling collapse of single fibers, and snap-through of multiple-fiber groups. The latter is possible even when single fibers are stable. Through simulations of large fiber networks, we show how these instabilities lead to spatially extended patterns of densification, fiber alignment and ECM remodelling induced by cell contraction. Our model is simple, but describes a very complex, multi-stable energy landscape, using sophisticated numerical optimisation methods that overcome the difficulties caused by instabilities in large systems. Our work opens up new ways of understanding the unique biomechanics of fibrous-network ECM, by fully accounting for nonlinearity and associated loss of stability in fiber networks. Our results provide new insights on tumour invasion and metastasis.
Collapse
Affiliation(s)
- Chrysovalantou Kalaitzidou
- Department of Engineering, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch sur Alzette, Luxembourg
| | - Georgios Grekas
- Aerospace Engineering and Mechanics, University of Minnesota, Minneapolis, Minnesota, United States of America
- Institute of Applied and Computational Mathematics, Foundation for Research and Technology-Hellas, Heraklion, Greece
| | - Andreas Zilian
- Department of Engineering, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch sur Alzette, Luxembourg
| | - Charalambos Makridakis
- Institute of Applied and Computational Mathematics, Foundation for Research and Technology-Hellas, Heraklion, Greece
- Department of Mathematics and Applied Mathematics, University of Crete, Heraklion, Greece
- Department of Mathematics, MPS, University of Sussex, Brighton, United Kingdom
| | - Phoebus Rosakis
- Institute of Applied and Computational Mathematics, Foundation for Research and Technology-Hellas, Heraklion, Greece
- Department of Mathematics and Applied Mathematics, University of Crete, Heraklion, Greece
| |
Collapse
|
6
|
Min K, Karuppannan SK, Tae G. The impact of matrix stiffness on hepatic cell function, liver fibrosis, and hepatocellular carcinoma-Based on quantitative data. BIOPHYSICS REVIEWS 2024; 5:021306. [PMID: 38846007 PMCID: PMC11151446 DOI: 10.1063/5.0197875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 05/14/2024] [Indexed: 06/09/2024]
Abstract
Over the past few decades, extensive research has explored the development of supportive scaffold materials for in vitro hepatic cell culture, to effectively mimic in vivo microenvironments. It is crucial for hepatic disease modeling, drug screening, and therapeutic evaluations, considering the ethical concerns and practical challenges associated with in vivo experiments. This review offers a comprehensive perspective on hepatic cell culture using bioscaffolds by encompassing all stages of hepatic diseases-from a healthy liver to fibrosis and hepatocellular carcinoma (HCC)-with a specific focus on matrix stiffness. This review begins by providing physiological and functional overviews of the liver. Subsequently, it explores hepatic cellular behaviors dependent on matrix stiffness from previous reports. For hepatic cell activities, softer matrices showed significant advantages over stiffer ones in terms of cell proliferation, migration, and hepatic functions. Conversely, stiffer matrices induced myofibroblastic activation of hepatic stellate cells, contributing to the further progression of fibrosis. Elevated matrix stiffness also correlates with HCC by increasing proliferation, epithelial-mesenchymal transition, metastasis, and drug resistance of HCC cells. In addition, we provide quantitative information on available data to offer valuable perspectives for refining the preparation and development of matrices for hepatic tissue engineering. We also suggest directions for further research on this topic.
Collapse
Affiliation(s)
- Kiyoon Min
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Sathish Kumar Karuppannan
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Giyoong Tae
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| |
Collapse
|
7
|
Asadollahi N, Hajari MA, Alipour Choshali M, Ajoudanian M, Ziai SA, Vosough M, Piryaei A. Bioengineering scalable and drug-responsive in vitro human multicellular non-alcoholic fatty liver disease microtissues encapsulated in the liver extracellular matrix-derived hydrogel. EXCLI JOURNAL 2024; 23:421-440. [PMID: 38741724 PMCID: PMC11089098 DOI: 10.17179/excli2023-6878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 03/06/2024] [Indexed: 05/16/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a high-prevalence and progressive disorder. Due to lack of reliable in vitro models to recapitulate the consecutive phases, the exact pathogenesis mechanism of this disease and approved therapeutic medications have not been revealed yet. It has been proven that the interplay between multiple hepatic cell types and liver extracellular matrix (ECM) are critical in NAFLD initiation and progression. Herein, a liver microtissue (LMT) consisting of Huh-7, THP-1, and LX-2 cell lines and human umbilical vein endothelial cells (HUVEC), which could be substituted for the main hepatic cells (hepatocyte, Kupffer, stellate, and sinusoidal endothelium, respectively), encapsulated in liver derived ECM-Alginate composite, was bioengineered. When the microtissues were treated with free fatty acids (FFAs) including Oleic acid (6.6×10-4M) and Palmitic acid (3.3×10-4M), they displayed the key features of NAFLD, including similar pattern of transcripts for genes involved in lipid metabolism, inflammation, insulin-resistance, and fibrosis, as well as pro-inflammatory and pro-fibrotic cytokines' secretions and intracellular lipid accumulation. Continuing FFAs supplementation, we demonstrated that the NAFLD phenomenon was established on day 3 and progressed to the initial fibrosis stage by day 8. Furthermore, this model was stable until day 12 post FFAs withdrawal on day 3. Moreover, administration of an anti-steatotic drug candidate, Liraglutide (15 μM), on the NAFLD microtissues significantly ameliorated the NAFLD phenomenon. Overall, we bioengineered a drug-responsive, cost-benefit liver microtissues which can simulate the initiation and progression of NAFLD. It is expected that this platform could potentially be used for studying molecular pathogenesis of NAFLD and high-throughput drug screening. See also the graphical abstract(Fig. 1).
Collapse
Affiliation(s)
- Negar Asadollahi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| | - Mohammad Amin Hajari
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mahmoud Alipour Choshali
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mohammad Ajoudanian
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Ali Ziai
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Huddinge, Sweden
| | - Abbas Piryaei
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
8
|
Zhang H, Xia T, Xia Z, Zhou H, Li Z, Wang W, Zhai X, Jin B. KIF18A inactivates hepatic stellate cells and alleviates liver fibrosis through the TTC3/Akt/mTOR pathway. Cell Mol Life Sci 2024; 81:96. [PMID: 38372748 PMCID: PMC10876760 DOI: 10.1007/s00018-024-05114-5] [Citation(s) in RCA: 62] [Impact Index Per Article: 62.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 12/03/2023] [Accepted: 01/04/2024] [Indexed: 02/20/2024]
Abstract
Activation of hepatic stellate cells (HSCs) has been demonstrated to play a pivotal role in the process of liver fibrogenesis. In this study, we observed a decrease in the expression of KIF18A in fibrotic liver tissues compared to healthy liver tissues, which exhibited a negative correlation with the activation of HSCs. To elucidate the molecular mechanisms underlying the involvement of KIF18A, we performed in vitro proliferation experiments and established a CCl4-induced liver fibrosis model. Our results revealed that KIF18A knockdown enhanced HSCs proliferation and reduced HSCs apoptosis in vitro. Mouse liver fibrosis grade was evaluated with Masson's trichrome and alpha-smooth muscle actin (α-SMA) staining. In addition, the expression of fibrosis markers Col1A1, Stat1, and Timp1 were detected. Animal experiments demonstrated that knockdown of KIF18A could promote liver fibrosis, whereas overexpression of KIF18A alleviated liver fibrosis in a CCl4-induced mouse model. Mechanistically, we found that KIF18A suppressed the AKT/mTOR pathway and exhibited direct binding to TTC3. Moreover, TTC3 was found to interact with p-AKT and could promote its ubiquitination and degradation. Our findings provide compelling evidence that KIF18A enhances the protein binding between TTC3 and p-AKT, promoting TTC3-mediated ubiquitination and degradation of p-AKT. These results refine the current understanding of the mechanisms underlying the pathogenesis of liver fibrosis and may offer new targets for treating this patient population.
Collapse
Affiliation(s)
- Hao Zhang
- Organ Transplant Department, Qilu Hospital of Shandong University, Jinan, China
- Department of Hepatobiliary Surgery, The Second Hospital of Shandong University, Jinan, China
| | - Tong Xia
- Organ Transplant Department, Qilu Hospital of Shandong University, Jinan, China
| | - Zhijia Xia
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Huaxin Zhou
- Department of Hepatobiliary Surgery, The Second Hospital of Shandong University, Jinan, China
| | - Zhipeng Li
- Department of Hepatobiliary Surgery, The Second Hospital of Shandong University, Jinan, China
| | - Wei Wang
- Medical Integration and Practice Center, Shandong University, Jinan, China.
| | - Xiangyu Zhai
- Organ Transplant Department, Qilu Hospital of Shandong University, Jinan, China.
- Department of Hepatobiliary Surgery, The Second Hospital of Shandong University, Jinan, China.
| | - Bin Jin
- Organ Transplant Department, Qilu Hospital of Shandong University, Jinan, China.
- Department of Hepatobiliary Surgery, The Second Hospital of Shandong University, Jinan, China.
| |
Collapse
|
9
|
Crawford JM, Bioulac-Sage P, Hytiroglou P. Structure, Function and Responses to Injury. MACSWEEN'S PATHOLOGY OF THE LIVER 2024:1-95. [DOI: 10.1016/b978-0-7020-8228-3.00001-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
10
|
LaGuardia JS, Shariati K, Bedar M, Ren X, Moghadam S, Huang KX, Chen W, Kang Y, Yamaguchi DT, Lee JC. Convergence of Calcium Channel Regulation and Mechanotransduction in Skeletal Regenerative Biomaterial Design. Adv Healthc Mater 2023; 12:e2301081. [PMID: 37380172 PMCID: PMC10615747 DOI: 10.1002/adhm.202301081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 06/20/2023] [Indexed: 06/30/2023]
Abstract
Cells are known to perceive their microenvironment through extracellular and intracellular mechanical signals. Upon sensing mechanical stimuli, cells can initiate various downstream signaling pathways that are vital to regulating proliferation, growth, and homeostasis. One such physiologic activity modulated by mechanical stimuli is osteogenic differentiation. The process of osteogenic mechanotransduction is regulated by numerous calcium ion channels-including channels coupled to cilia, mechanosensitive and voltage-sensitive channels, and channels associated with the endoplasmic reticulum. Evidence suggests these channels are implicated in osteogenic pathways such as the YAP/TAZ and canonical Wnt pathways. This review aims to describe the involvement of calcium channels in regulating osteogenic differentiation in response to mechanical loading and characterize the fashion in which those channels directly or indirectly mediate this process. The mechanotransduction pathway is a promising target for the development of regenerative materials for clinical applications due to its independence from exogenous growth factor supplementation. As such, also described are examples of osteogenic biomaterial strategies that involve the discussed calcium ion channels, calcium-dependent cellular structures, or calcium ion-regulating cellular features. Understanding the distinct ways calcium channels and signaling regulate these processes may uncover potential targets for advancing biomaterials with regenerative osteogenic capabilities.
Collapse
Affiliation(s)
- Jonnby S. LaGuardia
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Kaavian Shariati
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Meiwand Bedar
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Xiaoyan Ren
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, CA, 91343, USA
| | - Shahrzad Moghadam
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Kelly X. Huang
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Wei Chen
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Youngnam Kang
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Dean T. Yamaguchi
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, CA, 91343, USA
| | - Justine C. Lee
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, CA, 91343, USA
- Department of Orthopaedic Surgery, Los Angeles, CA, 90095, USA
- UCLA Molecular Biology Institute, Los Angeles, CA, 90095, USA
| |
Collapse
|
11
|
McDuffie D, Barr D, Helm M, Baumert T, Agarwal A, Thomas E. Physiomimetic In Vitro Human Models for Viral Infection in the Liver. Semin Liver Dis 2023; 43:31-49. [PMID: 36402129 PMCID: PMC10005888 DOI: 10.1055/a-1981-5944] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Viral hepatitis is a leading cause of liver morbidity and mortality globally. The mechanisms underlying acute infection and clearance, versus the development of chronic infection, are poorly understood. In vitro models of viral hepatitis circumvent the high costs and ethical considerations of animal models, which also translate poorly to studying the human-specific hepatitis viruses. However, significant challenges are associated with modeling long-term infection in vitro. Differentiated hepatocytes are best able to sustain chronic viral hepatitis infection, but standard two-dimensional models are limited because they fail to mimic the architecture and cellular microenvironment of the liver, and cannot maintain a differentiated hepatocyte phenotype over extended periods. Alternatively, physiomimetic models facilitate important interactions between hepatocytes and their microenvironment by incorporating liver-specific environmental factors such as three-dimensional ECM interactions and co-culture with non-parenchymal cells. These physiologically relevant interactions help maintain a functional hepatocyte phenotype that is critical for sustaining viral hepatitis infection. In this review, we provide an overview of distinct, novel, and innovative in vitro liver models and discuss their functionality and relevance in modeling viral hepatitis. These platforms may provide novel insight into mechanisms that regulate viral clearance versus progression to chronic infections that can drive subsequent liver disease.
Collapse
Affiliation(s)
- Dennis McDuffie
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida
| | - David Barr
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, Florida
| | - Madeline Helm
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida
| | - Thomas Baumert
- Inserm Research Institute for Viral and Liver Diseases, University of Strasbourg, Strasbourg, France
| | - Ashutosh Agarwal
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida
- Desai Sethi Urology Institute, Miller School of Medicine, University of Miami, Miami, Florida
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
| | - Emmanuel Thomas
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, Florida
- Schiff Center for Liver Diseases, University of Miami Miller School of Medicine, Miami, Florida
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
| |
Collapse
|
12
|
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: 8] [Impact Index Per Article: 4.0] [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
| |
Collapse
|
13
|
Chen S, Zhu J, Xue J, Wang X, Jing P, Zhou L, Cui Y, Wang T, Gong X, Lü S, Long M. Numerical simulation of flow characteristics in a permeable liver sinusoid with leukocytes. Biophys J 2022; 121:4666-4678. [PMID: 36271623 PMCID: PMC9748252 DOI: 10.1016/j.bpj.2022.10.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 08/31/2022] [Accepted: 10/17/2022] [Indexed: 02/07/2023] Open
Abstract
Double-layered channels of sinusoid lumen and Disse space separated by fenestrated liver sinusoidal endothelial cells (LSECs) endow the unique mechanical environment of the liver sinusoid network, which further guarantees its biological function. It is also known that this mechanical environment changes dramatically under liver fibrosis and cirrhosis, including the reduced plasma penetration and metabolite exchange between the two flow channels and the reduced Disse space deformability. The squeezing of leukocytes through narrow sinusoid lumen also affects the mechanical environment of liver sinusoid. To date, the detailed flow-field profile of liver sinusoid is still far from clear due to experimental limitations. It also remains elusive whether and how the varied physical properties of the pathological liver sinusoid regulate the fluid flow characteristics. Here a numerical model based on the immersed boundary method was established, and the effects of Disse space and leukocyte elasticities, endothelium permeability, and sinusoidal stenosis degree on fluid flow as well as leukocyte trafficking were specified upon a mimic liver sinusoid structure. Results showed that endothelium permeability dominantly controlled the plasma penetration velocity across the endothelium, whereas leukocyte squeezing promoted local penetration and significantly regulated wall shear stress on hepatocytes, which was strongly related to the Disse space and leukocyte deformability. Permeability and elasticity cooperatively regulated the process of leukocytes trafficking through the liver sinusoid, especially for stiffer leukocytes. This study will offer new insights into deeper understanding of the elaborate mechanical features of liver sinusoid and corresponding biological function.
Collapse
Affiliation(s)
- Shenbao Chen
- Center of Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), Beijing Key Laboratory of Engineered Construction and Mechanobiology, and CAS Center for Excellence in Complex System Mechanics, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China; School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Jingchen Zhu
- Center of Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), Beijing Key Laboratory of Engineered Construction and Mechanobiology, and CAS Center for Excellence in Complex System Mechanics, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China; School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Jian Xue
- School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China; State Key Laboratory of Nonlinear Mechanics (LNM), Institute of Mechanics, Chinese Academy of Sciences, Beijing, China
| | - Xiaolong Wang
- Key Laboratory of Hydrodynamics (Ministry of Education), Department of Engineering Mechanics, School of Naval Architecture, Ocean and Civil Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Peng Jing
- Key Laboratory of Hydrodynamics (Ministry of Education), Department of Engineering Mechanics, School of Naval Architecture, Ocean and Civil Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Lüwen Zhou
- Center of Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), Beijing Key Laboratory of Engineered Construction and Mechanobiology, and CAS Center for Excellence in Complex System Mechanics, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China; School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Yuhong Cui
- Department of Mechanics, Tianjin University, Tianjin, China
| | - Tianhao Wang
- Department of Mechanics, Tianjin University, Tianjin, China
| | - Xiaobo Gong
- Key Laboratory of Hydrodynamics (Ministry of Education), Department of Engineering Mechanics, School of Naval Architecture, Ocean and Civil Engineering, Shanghai Jiao Tong University, Shanghai, China.
| | - Shouqin Lü
- Center of Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), Beijing Key Laboratory of Engineered Construction and Mechanobiology, and CAS Center for Excellence in Complex System Mechanics, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China; School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China.
| | - Mian Long
- Center of Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), Beijing Key Laboratory of Engineered Construction and Mechanobiology, and CAS Center for Excellence in Complex System Mechanics, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China; School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
14
|
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: 1.0] [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
|
15
|
Prakash V, Christian Y, Redkar AS, Roy A, Anandalakshmi R, Ramakrishnan V. Antibacterial hydrogels of aromatic tripeptides. SOFT MATTER 2022; 18:6360-6371. [PMID: 35971808 DOI: 10.1039/d2sm00606e] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Self-assembled peptide hydrogels have emerged as alternatives to the conventional approaches employed in controlled drug release, wound-healing, and drug delivery, and as anti-infective agents. However, peptide hydrogels possessing antibacterial properties are less explored. In this work, we have designed three ultrashort antibacterial peptide hydrogels: Fmoc-FFH-CONH2, Fmoc-FHF-CONH2, and Fmoc-HFF-CONH2. The rheological study showed the higher storage modulus of Fmoc-FFH-CONH2 (30.43 kPa) compared to Fmoc-FHF-CONH2 and Fmoc-HFF-CONH2, which may be attributed to the enhanced aromatic interaction in Fmoc-FFH-CONH2 compared to the other two variants, resulting in more mechanical rigidity. Further, the prepared hydrogels were evaluated for their inherent antibacterial potency against Gram-positive (Staphylococcus aureus, strain MTCC 96) and Gram-negative (Pseudomonas aeruginosa, strain PA01) bacteria. Antibacterial experiments demonstrated the potency of the hydrogels in the order of Fmoc-FFH-CONH2 > Fmoc-FHF-CONH2 > Fmoc-HFF-CONH2. The antibacterial effect of the hydrogels was predominantly due to the osmotic stress and membrane disruption, which was verified by reactive oxygen species (ROS) generation and outer membrane permeabilization assays. Our findings point to the scope of using the synthesized peptide hydrogels as agents for topical applications.
Collapse
Affiliation(s)
- Vivek Prakash
- Molecular Informatics and Design Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam, 781039, India.
| | - Yvonne Christian
- Molecular Informatics and Design Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam, 781039, India.
| | - Amay Sanjay Redkar
- Molecular Informatics and Design Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam, 781039, India.
| | - Abhishek Roy
- Advanced Energy & Materials Systems Laboratory, Department of Chemical Engineering, Indian Institute of Technology Guwahati, Assam, 781039, India
| | - R Anandalakshmi
- Advanced Energy & Materials Systems Laboratory, Department of Chemical Engineering, Indian Institute of Technology Guwahati, Assam, 781039, India
| | - Vibin Ramakrishnan
- Molecular Informatics and Design Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam, 781039, India.
| |
Collapse
|
16
|
Lachowski D, Matellan C, Gopal S, Cortes E, Robinson BK, Saiani A, Miller AF, Stevens MM, del Río Hernández AE. Substrate Stiffness-Driven Membrane Tension Modulates Vesicular Trafficking via Caveolin-1. ACS NANO 2022; 16:4322-4337. [PMID: 35255206 PMCID: PMC9007531 DOI: 10.1021/acsnano.1c10534] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Liver fibrosis, a condition characterized by extensive deposition and cross-linking of extracellular matrix (ECM) proteins, is idiosyncratic in cases of chronic liver injury. The dysregulation of ECM remodeling by hepatic stellate cells (HSCs), the main mediators of fibrosis, results in an elevated ECM stiffness that drives the development of chronic liver disease such as cirrhosis and hepatocellular carcinoma. Tissue inhibitor of matrix metalloproteinase-1 (TIMP-1) is a key element in the regulation of ECM remodeling, which modulates the degradation and turnover of ECM components. We have previously reported that a rigid, fibrotic-like substrate can impact TIMP-1 expression at the protein level in HSCs without altering its mRNA expression. While HSCs are known to be highly susceptible to mechanical stimuli, the mechanisms through which mechanical cues regulate TIMP-1 at the post-translational level remain unclear. Here, we show a mechanism of regulation of plasma membrane tension by matrix stiffness. We found that this effect is orchestrated by the β1 integrin/RhoA axis and results in elevated exocytosis and secretion of TIMP-1 in a caveolin-1- and dynamin-2-dependent manner. We then show that TIMP-1 and caveolin-1 expression increases in cirrhosis and hepatocellular carcinoma. These conditions are associated with fibrosis, and this effect can be recapitulated in 3D fibrosis models consisting of hepatic stellate cells encapsulated in a self-assembling polypeptide hydrogel. This work positions stiffness-dependent membrane tension as a key regulator of enzyme secretion and function and a potential target for therapeutic strategies that aim at modulating ECM remodeling in chronic liver disease.
Collapse
Affiliation(s)
- Dariusz Lachowski
- Cellular
and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom
- Manchester
BIOGEL, Mereside, Alderley Park, Alderley Edge, Cheshire SK10 4TG, United Kingdom
| | - Carlos Matellan
- Cellular
and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - Sahana Gopal
- Department
of Materials, Department of Bioengineering and Institute of Biomedical
Engineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - Ernesto Cortes
- Cellular
and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - Benjamin K. Robinson
- Cellular
and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - Alberto Saiani
- Department
of Materials and Manchester Institute of Biotechnology, Faculty of
Science and Engineering, The University
of Manchester, Oxford Road, Manchester M13 9PL, United Kingdom
- Manchester
BIOGEL, Mereside, Alderley Park, Alderley Edge, Cheshire SK10 4TG, United Kingdom
| | - Aline F. Miller
- Department
of Chemical Engineering and Manchester Institute of Biotechnology,
Faculty of Science and Engineering, The
University of Manchester, Oxford Road, Manchester M13 9PL, United Kingdom
- Manchester
BIOGEL, Mereside, Alderley Park, Alderley Edge, Cheshire SK10 4TG, United Kingdom
| | - Molly M. Stevens
- Department
of Materials, Department of Bioengineering and Institute of Biomedical
Engineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - Armando E. del Río Hernández
- Cellular
and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom
| |
Collapse
|
17
|
Liu K, Wiendels M, Yuan H, Ruan C, Kouwer PH. Cell-matrix reciprocity in 3D culture models with nonlinear elasticity. Bioact Mater 2022; 9:316-331. [PMID: 34820573 PMCID: PMC8586441 DOI: 10.1016/j.bioactmat.2021.08.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 06/24/2021] [Accepted: 08/03/2021] [Indexed: 01/17/2023] Open
Abstract
Three-dimensional (3D) matrix models using hydrogels are powerful tools to understand and predict cell behavior. The interactions between the cell and its matrix, however is highly complex: the matrix has a profound effect on basic cell functions but simultaneously, cells are able to actively manipulate the matrix properties. This (mechano)reciprocity between cells and the extracellular matrix (ECM) is central in regulating tissue functions and it is fundamentally important to broadly consider the biomechanical properties of the in vivo ECM when designing in vitro matrix models. This manuscript discusses two commonly used biopolymer networks, i.e. collagen and fibrin gels, and one synthetic polymer network, polyisocyanide gel (PIC), which all possess the characteristic nonlinear mechanics in the biological stress regime. We start from the structure of the materials, then address the uses, advantages, and limitations of each material, to provide a guideline for tissue engineers and biophysicists in utilizing current materials and also designing new materials for 3D cell culture purposes.
Collapse
Affiliation(s)
- Kaizheng Liu
- Research Center for Human Tissue and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
- Radboud University, Institute for Molecules and Materials, Heyendaalseweg 135, 6525 AJ, Nijmegen, the Netherlands
| | - Maury Wiendels
- Radboud University, Institute for Molecules and Materials, Heyendaalseweg 135, 6525 AJ, Nijmegen, the Netherlands
| | - Hongbo Yuan
- Institute of Biophysics, Hebei University of Technology, Tianjin, 300401, PR China
- Molecular Imaging and Photonics, Chemistry Department, KU Leuven, Celestijnenlaan 200F, 3001, Heverlee, Belgium
| | - Changshun Ruan
- Research Center for Human Tissue and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
| | - Paul H.J. Kouwer
- Radboud University, Institute for Molecules and Materials, Heyendaalseweg 135, 6525 AJ, Nijmegen, the Netherlands
| |
Collapse
|
18
|
Ramos MJ, Bandiera L, Menolascina F, Fallowfield JA. In vitro models for non-alcoholic fatty liver disease: Emerging platforms and their applications. iScience 2022; 25:103549. [PMID: 34977507 PMCID: PMC8689151 DOI: 10.1016/j.isci.2021.103549] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) represents a global healthcare challenge, affecting 1 in 4 adults, and death rates are predicted to rise inexorably. The progressive form of NAFLD, non-alcoholic steatohepatitis (NASH), can lead to fibrosis, cirrhosis, and hepatocellular carcinoma. However, no medical treatments are licensed for NAFLD-NASH. Identifying efficacious therapies has been hindered by the complexity of disease pathogenesis, a paucity of predictive preclinical models and inadequate validation of pharmacological targets in humans. The development of clinically relevant in vitro models of the disease will pave the way to overcome these challenges. Currently, the combined application of emerging technologies (e.g., organ-on-a-chip/microphysiological systems) and control engineering approaches promises to unravel NAFLD biology and deliver tractable treatment candidates. In this review, we will describe advances in preclinical models for NAFLD-NASH, the recent introduction of novel technologies in this space, and their importance for drug discovery endeavors in the future.
Collapse
Affiliation(s)
- Maria Jimenez Ramos
- Centre for Inflammation Research, The University of Edinburgh, The Queen's Medical Research Institute, Edinburgh EH16 4TJ, UK
| | - Lucia Bandiera
- Institute for Bioengineering, The University of Edinburgh, Edinburgh EH9 3BF, UK.,Synthsys - Centre for Synthetic and Systems Biology, The University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Filippo Menolascina
- Institute for Bioengineering, The University of Edinburgh, Edinburgh EH9 3BF, UK.,Synthsys - Centre for Synthetic and Systems Biology, The University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Jonathan Andrew Fallowfield
- Centre for Inflammation Research, The University of Edinburgh, The Queen's Medical Research Institute, Edinburgh EH16 4TJ, UK
| |
Collapse
|
19
|
Ramos MJ, Bandiera L, Menolascina F, Fallowfield JA. In vitro models for non-alcoholic fatty liver disease: Emerging platforms and their applications. iScience 2022; 25:103549. [PMID: 34977507 DOI: 10.1016/j.isci] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) represents a global healthcare challenge, affecting 1 in 4 adults, and death rates are predicted to rise inexorably. The progressive form of NAFLD, non-alcoholic steatohepatitis (NASH), can lead to fibrosis, cirrhosis, and hepatocellular carcinoma. However, no medical treatments are licensed for NAFLD-NASH. Identifying efficacious therapies has been hindered by the complexity of disease pathogenesis, a paucity of predictive preclinical models and inadequate validation of pharmacological targets in humans. The development of clinically relevant in vitro models of the disease will pave the way to overcome these challenges. Currently, the combined application of emerging technologies (e.g., organ-on-a-chip/microphysiological systems) and control engineering approaches promises to unravel NAFLD biology and deliver tractable treatment candidates. In this review, we will describe advances in preclinical models for NAFLD-NASH, the recent introduction of novel technologies in this space, and their importance for drug discovery endeavors in the future.
Collapse
Affiliation(s)
- Maria Jimenez Ramos
- Centre for Inflammation Research, The University of Edinburgh, The Queen's Medical Research Institute, Edinburgh EH16 4TJ, UK
| | - Lucia Bandiera
- Institute for Bioengineering, The University of Edinburgh, Edinburgh EH9 3BF, UK
- Synthsys - Centre for Synthetic and Systems Biology, The University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Filippo Menolascina
- Institute for Bioengineering, The University of Edinburgh, Edinburgh EH9 3BF, UK
- Synthsys - Centre for Synthetic and Systems Biology, The University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Jonathan Andrew Fallowfield
- Centre for Inflammation Research, The University of Edinburgh, The Queen's Medical Research Institute, Edinburgh EH16 4TJ, UK
| |
Collapse
|
20
|
Wang AJ, Allen A, Sofman M, Sphabmixay P, Yildiz E, Griffith LG. Engineering Modular 3D Liver Culture Microenvironments In Vitro to Parse the Interplay between Biophysical and Biochemical Microenvironment Cues on Hepatic Phenotypes. ADVANCED NANOBIOMED RESEARCH 2022; 2:2100049. [PMID: 35872804 PMCID: PMC9307216 DOI: 10.1002/anbr.202100049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
In vitro models of human liver functions are used across a diverse range of applications in preclinical drug development and disease modeling, with particular increasing interest in models that capture facets of liver inflammatory status. This study investigates how the interplay between biophysical and biochemical microenvironment cues influence phenotypic responses, including inflammation signatures, of primary human hepatocytes (PHH) cultured in a commercially available perfused bioreactor. A 3D printing-based alginate microwell system was designed to form thousands of hepatic spheroids in a scalable manner as a comparator 3D culture modality to the bioreactor. Soft, synthetic extracellular matrix (ECM) hydrogel scaffolds with biophysical properties mimicking features of liver were engineered to replace polystyrene scaffolds, and the biochemical microenvironment was modulated with a defined set of growth factors and signaling modulators. The supplemented media significantly increased tissue density, albumin secretion, and CYP3A4 activity but also upregulated inflammatory markers. Basal inflammatory markers were lower for cells maintained in ECM hydrogel scaffolds or spheroid formats than polystyrene scaffolds, while hydrogel scaffolds exhibited the most sensitive response to inflammation as assessed by multiplexed cytokine and RNA-seq analyses. Together, these engineered 3D liver microenvironments provide insights for probing human liver functions and inflammatory response in vitro.
Collapse
Affiliation(s)
- Alex J Wang
- Biological Engineering Department, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Allysa Allen
- Biological Engineering Department, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Marianna Sofman
- Biological Engineering Department, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Pierre Sphabmixay
- Mechanical Engineering Department, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA; Whitehead Institute for Biomedical Research, 455 Main Street, Cambridge, MA, 02142, USA
| | - Ece Yildiz
- Biological Engineering Department, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA; Institute of Bioengineering, School of Life Science, École Polytechnique Fédérale de Lausanne, Route Cantonale, 1015 Lausanne, Switzerland
| | - Linda G. Griffith
- Biological Engineering Department, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA; Center for Gynepathology Research, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| |
Collapse
|
21
|
Nah EH, Cho S, Park H, Noh D, Kwon E, Cho HI. Subclinical steatohepatitis and advanced liver fibrosis in health examinees with nonalcoholic fatty liver disease (NAFLD) in 10 South Korean cities: A retrospective cross-sectional study. PLoS One 2021; 16:e0260477. [PMID: 34818372 PMCID: PMC8612540 DOI: 10.1371/journal.pone.0260477] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 11/10/2021] [Indexed: 01/17/2023] Open
Abstract
Background Nonalcoholic steatohepatitis (NASH) has a risk of progressing to cirrhosis. The prevalence of NASH and its associated risk factors in community populations are relatively unknown. This study aimed to determine the prevalence of NASH and advanced liver fibrosis using magnetic resonance elastography (MRE), and determine those risk factors in health examinees with asymptomatic fatty liver. Methods This study consecutively selected subjects who underwent health checkups at 13 health-promotion centers in 10 Korean cities between 2018 and 2020. Hepatic steatosis and stiffness were assessed using ultrasonography and MRE, respectively. Stages of liver stiffness were estimated using MRE with cutoff values for NASH and advanced liver fibrosis of 2.91 and 3.60 kPa, respectively. Results The overall prevalence of NASH and advanced liver fibrosis in the subjects with fatty liver were 8.35% and 2.04%, respectively. Multivariate logistic regression analysis indicated that central obesity (OR = 5.12, 95% CI = 2.70–9.71), increased triglyceride (OR = 3.29, 95% CI = 1.72–6.29), abnormal liver function test (OR = 3.09, 95% CI = 1.66–5.76) (all P<0.001), and decreased high-density lipoprotein cholesterol (OR = 5.18, 95% CI = 1.78–15.05) (P = 0.003) were associated with NASH. The main risk factor for advanced liver fibrosis was diabetes (OR = 4.46, 95% CI = 1.14–17.48) (P = 0.032). Conclusion NASH or advanced liver fibrosis is found in one-tenth of health examinees with asymptomatic fatty liver. This suggests that early detection of NASH should be considered to allow early interventions such as lifestyle changes to prevent the adverse effects of NASH and its progression in health examinees with asymptomatic fatty liver.
Collapse
Affiliation(s)
- Eun-Hee Nah
- Health Promotion Research Institute, Korea Association of Health Promotion, Seoul, Korea
| | - Seon Cho
- Health Promotion Research Institute, Korea Association of Health Promotion, Seoul, Korea
| | - Hyeran Park
- Health Promotion Research Institute, Korea Association of Health Promotion, Seoul, Korea
| | - Dongwon Noh
- Health Promotion Research Institute, Korea Association of Health Promotion, Seoul, Korea
| | - Eunjoo Kwon
- Health Promotion Research Institute, Korea Association of Health Promotion, Seoul, Korea
| | - Han-Ik Cho
- MEDIcheck LAB, Korea Association of Health Promotion, Seoul, Korea
| |
Collapse
|
22
|
Christ B, Collatz M, Dahmen U, Herrmann KH, Höpfl S, König M, Lambers L, Marz M, Meyer D, Radde N, Reichenbach JR, Ricken T, Tautenhahn HM. Hepatectomy-Induced Alterations in Hepatic Perfusion and Function - Toward Multi-Scale Computational Modeling for a Better Prediction of Post-hepatectomy Liver Function. Front Physiol 2021; 12:733868. [PMID: 34867441 PMCID: PMC8637208 DOI: 10.3389/fphys.2021.733868] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 10/26/2021] [Indexed: 01/17/2023] Open
Abstract
Liver resection causes marked perfusion alterations in the liver remnant both on the organ scale (vascular anatomy) and on the microscale (sinusoidal blood flow on tissue level). These changes in perfusion affect hepatic functions via direct alterations in blood supply and drainage, followed by indirect changes of biomechanical tissue properties and cellular function. Changes in blood flow impose compression, tension and shear forces on the liver tissue. These forces are perceived by mechanosensors on parenchymal and non-parenchymal cells of the liver and regulate cell-cell and cell-matrix interactions as well as cellular signaling and metabolism. These interactions are key players in tissue growth and remodeling, a prerequisite to restore tissue function after PHx. Their dysregulation is associated with metabolic impairment of the liver eventually leading to liver failure, a serious post-hepatectomy complication with high morbidity and mortality. Though certain links are known, the overall functional change after liver surgery is not understood due to complex feedback loops, non-linearities, spatial heterogeneities and different time-scales of events. Computational modeling is a unique approach to gain a better understanding of complex biomedical systems. This approach allows (i) integration of heterogeneous data and knowledge on multiple scales into a consistent view of how perfusion is related to hepatic function; (ii) testing and generating hypotheses based on predictive models, which must be validated experimentally and clinically. In the long term, computational modeling will (iii) support surgical planning by predicting surgery-induced perfusion perturbations and their functional (metabolic) consequences; and thereby (iv) allow minimizing surgical risks for the individual patient. Here, we review the alterations of hepatic perfusion, biomechanical properties and function associated with hepatectomy. Specifically, we provide an overview over the clinical problem, preoperative diagnostics, functional imaging approaches, experimental approaches in animal models, mechanoperception in the liver and impact on cellular metabolism, omics approaches with a focus on transcriptomics, data integration and uncertainty analysis, and computational modeling on multiple scales. Finally, we provide a perspective on how multi-scale computational models, which couple perfusion changes to hepatic function, could become part of clinical workflows to predict and optimize patient outcome after complex liver surgery.
Collapse
Affiliation(s)
- Bruno Christ
- Cell Transplantation/Molecular Hepatology Lab, Department of Visceral, Transplant, Thoracic and Vascular Surgery, University of Leipzig Medical Center, Leipzig, Germany
| | - Maximilian Collatz
- RNA Bioinformatics and High-Throughput Analysis, Faculty of Mathematics and Computer Science, Friedrich Schiller University Jena, Jena, Germany
- Optisch-Molekulare Diagnostik und Systemtechnologié, Leibniz Institute of Photonic Technology (IPHT), Jena, Germany
- InfectoGnostics Research Campus Jena, Jena, Germany
| | - Uta Dahmen
- Experimental Transplantation Surgery, Department of General, Visceral and Vascular Surgery, Jena University Hospital, Jena, Germany
| | - Karl-Heinz Herrmann
- Medical Physics Group, Institute of Diagnostic and Interventional Radiology, Jena University Hospital, Jena, Germany
| | - Sebastian Höpfl
- Faculty of Engineering Design, Production Engineering and Automotive Engineering, Institute for Systems Theory and Automatic Control, University of Stuttgart, Stuttgart, Germany
| | - Matthias König
- Systems Medicine of the Liver Lab, Institute for Theoretical Biology, Humboldt-University Berlin, Berlin, Germany
| | - Lena Lambers
- Faculty of Aerospace Engineering and Geodesy, Institute of Mechanics, Structural Analysis and Dynamics, University of Stuttgart, Stuttgart, Germany
| | - Manja Marz
- RNA Bioinformatics and High-Throughput Analysis, Faculty of Mathematics and Computer Science, Friedrich Schiller University Jena, Jena, Germany
| | - Daria Meyer
- RNA Bioinformatics and High-Throughput Analysis, Faculty of Mathematics and Computer Science, Friedrich Schiller University Jena, Jena, Germany
| | - Nicole Radde
- Faculty of Engineering Design, Production Engineering and Automotive Engineering, Institute for Systems Theory and Automatic Control, University of Stuttgart, Stuttgart, Germany
| | - Jürgen R. Reichenbach
- Medical Physics Group, Institute of Diagnostic and Interventional Radiology, Jena University Hospital, Jena, Germany
| | - Tim Ricken
- Faculty of Aerospace Engineering and Geodesy, Institute of Mechanics, Structural Analysis and Dynamics, University of Stuttgart, Stuttgart, Germany
| | - Hans-Michael Tautenhahn
- Department of General, Visceral and Vascular Surgery, Jena University Hospital, Jena, Germany
| |
Collapse
|
23
|
Cacopardo L, Ahluwalia A. Engineering and Monitoring 3D Cell Constructs with Time-Evolving Viscoelasticity for the Study of Liver Fibrosis In Vitro. Bioengineering (Basel) 2021; 8:106. [PMID: 34436109 PMCID: PMC8389340 DOI: 10.3390/bioengineering8080106] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/28/2021] [Accepted: 07/20/2021] [Indexed: 01/17/2023] Open
Abstract
Liver fibrosis is generally associated with an over-production and crosslinking of extracellular matrix proteins, causing a progressive increase in both the elastic and viscous properties of the hepatic tissue. We describe a strategy for mimicking and monitoring the mechano-dynamics of the 3D microenvironment associated with liver fibrosis. Cell-laden gelatin hydrogels were crosslinked with microbial transglutaminase using a purpose-designed cytocompatible two-step protocol, which allows for the exposure of cells to a mechanically changing environment during culturing. A bioreactor was re-engineered to monitor the mechanical properties of cell constructs over time. The results showed a shift towards a more elastic (i.e., solid-like) behaviour, which is likely related to an increase in cell stress. The method effectively mimics the time-evolving mechanical microenvironment associated with liver fibrosis and could provide novel insights into pathophysiological processes in which both elastic and viscous properties of tissues change over time.
Collapse
Affiliation(s)
| | - Arti Ahluwalia
- Research Center ‘E. Piaggio’, University of Pisa, 56122 Pisa, Italy;
- Department of Information Engineering, University of Pisa, 56122 Pisa, Italy
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research (Centro 3R), Italy
| |
Collapse
|
24
|
Yang J, Xu C, Wu M, Wu Y, Jia X, Zhou C, Zhang X, Ge S, Li Z, Zhang L. MicroRNA-124 inhibits hepatic stellate cells inflammatory cytokines secretion by targeting IQGAP1 through NF-κB pathway. Int Immunopharmacol 2021; 95:107520. [PMID: 33743313 DOI: 10.1016/j.intimp.2021.107520] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 02/17/2021] [Accepted: 02/17/2021] [Indexed: 01/01/2023]
Abstract
Liver fibrosis is a health concern that leads to organ failure mediated via production of inflammatory cytokines and fibrotic biomarkers. To date, there was no direct approved antifibrotic therapy, and current treatment was mainly the removal of the causative factor. Recent studies demonstrated that aberrant expression of miR-124 was involved in the progression of various liver diseases including hepatocellular carcinoma (HCC). However, whether miR-124 could function as a transcriptional regulator in the inflammatory cytokines secretion of liver fibrosis remains unclear. In this study, we demonstrated that the expression of miR-124 was downregulated in liver fibrosis tissues and TNF-α-induced LX-2 cells, concomitant with the upregulated expression of IQGAP1, suggesting that miR-124 and IQGAP1 might be associated with the development of inflammation in liver fibrosis. Therefore, we demonstrated that the overexpression of miR-124 and knockdown of IQGAP1 could lead to the downregulation of TNF-α, IL-1β and IL-6. While knockdown of miR-124 or overexpression of IQGAP1 showed reversed results. Moreover, dual luciferase reporter assays demonstrated that miR-124 specifically targeted the 3'-UTR of IQGAP1, and thus inhibited the expression of IQGAP1. Mechanistically, we found that the expression changes of miR-124 and IQGAP1 could be involved in inhibition or activation of NF-κB signaling pathway in response to TNF-α. In conclusion, these results indicated that miR-124 plays a crucial role in TNF-α-induced LX-2 cells via regulating NF-κB signaling pathway.
Collapse
Affiliation(s)
- Junfa Yang
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China; School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Changqing Xu
- The Third People's Hospital of Hefei (Hefei Third Clinical College of Anhui Medical University), Hefei, Anhui Province, China
| | - Maomao Wu
- Department of Pharmacy, Anhui Chest Hospital, Hefei, Anhui Province, China
| | - Ying Wu
- The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Xiaodi Jia
- Fujian Normal University, Fuzhou 350007, China
| | - Chang Zhou
- School of Basic Medical Science, Anhui Medical University, Hefei 230032, China
| | - Xianzheng Zhang
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Shenglin Ge
- The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China.
| | - Zeng Li
- School of Pharmacy, Anhui Medical University, Hefei 230032, China.
| | - Lingling Zhang
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China.
| |
Collapse
|
25
|
Kisseleva T, Brenner D. Molecular and cellular mechanisms of liver fibrosis and its regression. Nat Rev Gastroenterol Hepatol 2021; 18:151-166. [PMID: 33128017 DOI: 10.1038/s41575-020-00372-7] [Citation(s) in RCA: 1080] [Impact Index Per Article: 270.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/23/2020] [Indexed: 01/18/2023]
Abstract
Chronic liver injury leads to liver inflammation and fibrosis, through which activated myofibroblasts in the liver secrete extracellular matrix proteins that generate the fibrous scar. The primary source of these myofibroblasts are the resident hepatic stellate cells. Clinical and experimental liver fibrosis regresses when the causative agent is removed, which is associated with the elimination of these activated myofibroblasts and resorption of the fibrous scar. Understanding the mechanisms of liver fibrosis regression could identify new therapeutic targets to treat liver fibrosis. This Review summarizes studies of the molecular mechanisms underlying the reversibility of liver fibrosis, including apoptosis and the inactivation of hepatic stellate cells, the crosstalk between the liver and the systems that orchestrate the recruitment of bone marrow-derived macrophages (and other inflammatory cells) driving fibrosis resolution, and the interactions between various cell types that lead to the intracellular signalling that induces fibrosis or its regression. We also discuss strategies to target hepatic myofibroblasts (for example, via apoptosis or inactivation) and the myeloid cells that degrade the matrix (for example, via their recruitment to fibrotic liver) to facilitate fibrosis resolution and liver regeneration.
Collapse
Affiliation(s)
- Tatiana Kisseleva
- Department of Surgery, University of California, San Diego, La Jolla, CA, USA.
| | - David Brenner
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
26
|
Zhang X, Zhang X, Huang W, Ge X. The role of heat shock proteins in the regulation of fibrotic diseases. Biomed Pharmacother 2020; 135:111067. [PMID: 33383375 DOI: 10.1016/j.biopha.2020.111067] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 11/08/2020] [Accepted: 11/20/2020] [Indexed: 12/29/2022] Open
Abstract
Heat shock proteins (HSPs) are key players to restore cell homeostasis and act as chaperones by assisting the folding and assembly of newly synthesized proteins and preventing protein aggregation. Recently, evidence has been accumulating that HSPs have been proven to have other functions except for the classical molecular chaperoning in that they play an important role in a wider range of fibrotic diseases via modulating cytokine induction and inflammation response, including lung fibrosis, liver fibrosis, and idiopathic pulmonary fibrosis. The recruitment of inflammatory cells, a large number of secretion of pro-fibrotic cytokines such as transforming growth factor-β1 (TGF-β1) and increased apoptosis, oxidative stress, and proteasomal system degradation are all events occurring during fibrogenesis, which might be associated with HSPs. However, their role on fibrotic process is not yet fully understood. In this review, we discuss new discoveries regarding the involvement of HSPs in the regulation of organ and tissue fibrosis, and note recent findings suggesting that HSPs may be a promising therapeutic target for improving the current frustrating outcome of fibrotic disorders.
Collapse
Affiliation(s)
- Xiaoling Zhang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, PR China; School of Pharmacy, Nantong University, 19 Qixiu Road, Nantong, 226019, PR China.
| | - Xiaoyan Zhang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, PR China
| | - Wenmin Huang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, PR China
| | - Xiaoqun Ge
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, PR China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, 225001, PR China; Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, PR China.
| |
Collapse
|
27
|
Human Liver-Derived Extracellular Matrix for the Culture of Distinct Human Primary Liver Cells. Cells 2020; 9:cells9061357. [PMID: 32486126 PMCID: PMC7349413 DOI: 10.3390/cells9061357] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/17/2020] [Accepted: 05/22/2020] [Indexed: 01/17/2023] Open
Abstract
The lack of robust methods to preserve, purify and in vitro maintain the phenotype of the human liver’s highly specialized parenchymal and non-parenchymal cell types importantly hampers their exploitation for the development of research and clinical applications. There is in this regard a growing interest in the use of tissue-specific extracellular matrix (ECM) to provide cells with an in vitro environment that more closely resembles that of the native tissue. In the present study, we have developed a method that allows for the isolation and downstream application of the human liver’s main cell types from cryopreserved material. We also isolated and solubilized human liver ECM (HL-ECM), analyzed its peptidomic and proteomic composition by mass spectrometry and evaluated its interest for the culture of distinct primary human liver cells. Our analysis of the HL-ECM revealed proteomic diversity, type 1 collagen abundance and partial loss of integrity following solubilization. Solubilized HL-ECM was evaluated either as a coating or as a medium supplement for the culture of human primary hepatocytes, hepatic stellate cells and liver sinusoidal endothelial cells. Whereas the solubilized HL-ECM was suitable for cell culture, its impact on the phenotype and/or functionality of the human liver cells was limited. Our study provides a first detailed characterization of solubilized HL-ECM and a first report of its influence on the culture of distinct human primary liver cells.
Collapse
|
28
|
Cai L, Nauman EA, Pedersen CBW, Neu CP. Finite deformation elastography of articular cartilage and biomaterials based on imaging and topology optimization. Sci Rep 2020; 10:7980. [PMID: 32409711 PMCID: PMC7224212 DOI: 10.1038/s41598-020-64723-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Accepted: 04/17/2020] [Indexed: 01/17/2023] Open
Abstract
Tissues and engineered biomaterials exhibit exquisite local variation in stiffness that defines their function. Conventional elastography quantifies stiffness in soft (e.g. brain, liver) tissue, but robust quantification in stiff (e.g. musculoskeletal) tissues is challenging due to dissipation of high frequency shear waves. We describe new development of finite deformation elastography that utilizes magnetic resonance imaging of low frequency, physiological-level (large magnitude) displacements, coupled to an iterative topology optimization routine to investigate stiffness heterogeneity, including spatial gradients and inclusions. We reconstruct 2D and 3D stiffness distributions in bilayer agarose hydrogels and silicon materials that exhibit heterogeneous displacement/strain responses. We map stiffness in porcine and sheep articular cartilage deep within the bony articular joint space in situ for the first time. Elevated cartilage stiffness localized to the superficial zone is further related to collagen fiber compaction and loss of water content during cyclic loading, as assessed by independent T2 measurements. We additionally describe technical challenges needed to achieve in vivo elastography measurements. Our results introduce new functional imaging biomarkers, which can be assessed nondestructively, with clinical potential to diagnose and track progression of disease in early stages, including osteoarthritis or tissue degeneration.
Collapse
Affiliation(s)
- Luyao Cai
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, US
| | - Eric A Nauman
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, US
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, 47907, US
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, 47907, US
| | | | - Corey P Neu
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, US.
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO, 80309, US.
| |
Collapse
|
29
|
Lafoz E, Ruart M, Anton A, Oncins A, Hernández-Gea V. The Endothelium as a Driver of Liver Fibrosis and Regeneration. Cells 2020; 9:E929. [PMID: 32290100 PMCID: PMC7226820 DOI: 10.3390/cells9040929] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 04/05/2020] [Accepted: 04/06/2020] [Indexed: 02/07/2023] Open
Abstract
Liver fibrosis is a common feature of sustained liver injury and represents a major public health problem worldwide. Fibrosis is an active research field and discoveries in the last years have contributed to the development of new antifibrotic drugs, although none of them have been approved yet. Liver sinusoidal endothelial cells (LSEC) are highly specialized endothelial cells localized at the interface between the blood and other liver cell types. They lack a basement membrane and display open channels (fenestrae), making them exceptionally permeable. LSEC are the first cells affected by any kind of liver injury orchestrating the liver response to damage. LSEC govern the regenerative process initiation, but aberrant LSEC activation in chronic liver injury induces fibrosis. LSEC are also main players in fibrosis resolution. They maintain liver homeostasis and keep hepatic stellate cell and Kupffer cell quiescence. After sustained hepatic injury, they lose their phenotype and protective properties, promoting angiogenesis and vasoconstriction and contributing to inflammation and fibrosis. Therefore, improving LSEC phenotype is a promising strategy to prevent liver injury progression and complications. This review focuses on changes occurring in LSEC after liver injury and their consequences on fibrosis progression, liver regeneration, and resolution. Finally, a synopsis of the available strategies for LSEC-specific targeting is provided.
Collapse
Affiliation(s)
- Erica Lafoz
- Unidad de Hemodinámica Hepática, Servicio de Hepatología, Hospital Clínic, Universidad de Barcelona, Instituto de Investigaciones Biomédicas Augusto Pi Suñer (IDIBAPS), 08036 Barcelona, Spain; (E.L.); (M.R.); (A.A.); (A.O.)
| | - Maria Ruart
- Unidad de Hemodinámica Hepática, Servicio de Hepatología, Hospital Clínic, Universidad de Barcelona, Instituto de Investigaciones Biomédicas Augusto Pi Suñer (IDIBAPS), 08036 Barcelona, Spain; (E.L.); (M.R.); (A.A.); (A.O.)
| | - Aina Anton
- Unidad de Hemodinámica Hepática, Servicio de Hepatología, Hospital Clínic, Universidad de Barcelona, Instituto de Investigaciones Biomédicas Augusto Pi Suñer (IDIBAPS), 08036 Barcelona, Spain; (E.L.); (M.R.); (A.A.); (A.O.)
| | - Anna Oncins
- Unidad de Hemodinámica Hepática, Servicio de Hepatología, Hospital Clínic, Universidad de Barcelona, Instituto de Investigaciones Biomédicas Augusto Pi Suñer (IDIBAPS), 08036 Barcelona, Spain; (E.L.); (M.R.); (A.A.); (A.O.)
| | - Virginia Hernández-Gea
- Unidad de Hemodinámica Hepática, Servicio de Hepatología, Hospital Clínic, Universidad de Barcelona, Instituto de Investigaciones Biomédicas Augusto Pi Suñer (IDIBAPS), 08036 Barcelona, Spain; (E.L.); (M.R.); (A.A.); (A.O.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
30
|
Holmgren G, Ulfenborg B, Asplund A, Toet K, Andersson CX, Hammarstedt A, Hanemaaijer R, Küppers-Munther B, Synnergren J. Characterization of Human Induced Pluripotent Stem Cell-Derived Hepatocytes with Mature Features and Potential for Modeling Metabolic Diseases. Int J Mol Sci 2020; 21:ijms21020469. [PMID: 31940797 PMCID: PMC7014160 DOI: 10.3390/ijms21020469] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 01/06/2020] [Accepted: 01/07/2020] [Indexed: 01/17/2023] Open
Abstract
There is a strong anticipated future for human induced pluripotent stem cell-derived hepatocytes (hiPS-HEP), but so far, their use has been limited due to insufficient functionality. We investigated the potential of hiPS-HEP as an in vitro model for metabolic diseases by combining transcriptomics with multiple functional assays. The transcriptomics analysis revealed that 86% of the genes were expressed at similar levels in hiPS-HEP as in human primary hepatocytes (hphep). Adult characteristics of the hiPS-HEP were confirmed by the presence of important hepatocyte features, e.g., Albumin secretion and expression of major drug metabolizing genes. Normal energy metabolism is crucial for modeling metabolic diseases, and both transcriptomics data and functional assays showed that hiPS-HEP were similar to hphep regarding uptake of glucose, low-density lipoproteins (LDL), and fatty acids. Importantly, the inflammatory state of the hiPS-HEP was low under standard conditions, but in response to lipid accumulation and ER stress the inflammation marker tumor necrosis factor α (TNFα) was upregulated. Furthermore, hiPS-HEP could be co-cultured with primary hepatic stellate cells both in 2D and in 3D spheroids, paving the way for using these co-cultures for modeling non-alcoholic steatohepatitis (NASH). Taken together, hiPS-HEP have the potential to serve as an in vitro model for metabolic diseases. Furthermore, differently expressed genes identified in this study can serve as targets for future improvements of the hiPS-HEP.
Collapse
Affiliation(s)
- Gustav Holmgren
- Systems biology research center, School of Bioscience, University of Skövde, 54128 Skövde, Sweden; (G.H.); (J.S.)
| | - Benjamin Ulfenborg
- Systems biology research center, School of Bioscience, University of Skövde, 54128 Skövde, Sweden; (G.H.); (J.S.)
- Correspondence: (B.U.); (B.K.-M.)
| | - Annika Asplund
- R&D, Hepatocyte Product Development, Takara Bio Europe AB, 41346 Gothenburg, Sweden; (A.A.)
| | - Karin Toet
- Department of Metabolic Health Research, TNO, 2333 Leiden, The Netherlands; (K.T.); (R.H.)
| | - Christian X Andersson
- R&D, Hepatocyte Product Development, Takara Bio Europe AB, 41346 Gothenburg, Sweden; (A.A.)
| | - Ann Hammarstedt
- The Lundberg Laboratory for Diabetes Research, Departments of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 41345 Gothenburg, Sweden;
| | - Roeland Hanemaaijer
- Department of Metabolic Health Research, TNO, 2333 Leiden, The Netherlands; (K.T.); (R.H.)
| | - Barbara Küppers-Munther
- R&D, Hepatocyte Product Development, Takara Bio Europe AB, 41346 Gothenburg, Sweden; (A.A.)
- Correspondence: (B.U.); (B.K.-M.)
| | - Jane Synnergren
- Systems biology research center, School of Bioscience, University of Skövde, 54128 Skövde, Sweden; (G.H.); (J.S.)
| |
Collapse
|
31
|
Soydemir S, Comella O, Abdelmottaleb D, Pritchett J. Does Mechanocrine Signaling by Liver Sinusoidal Endothelial Cells Offer New Opportunities for the Development of Anti-fibrotics? Front Med (Lausanne) 2020; 6:312. [PMID: 31998732 PMCID: PMC6962108 DOI: 10.3389/fmed.2019.00312] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 12/11/2019] [Indexed: 01/17/2023] Open
Affiliation(s)
- Sumeyye Soydemir
- Department of Life Sciences, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, United Kingdom
| | - Olivia Comella
- Department of Life Sciences, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, United Kingdom
| | - Dina Abdelmottaleb
- Department of Life Sciences, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, United Kingdom.,Centre for Bioscience, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, United Kingdom
| | - James Pritchett
- Department of Life Sciences, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, United Kingdom.,Centre for Bioscience, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, United Kingdom
| |
Collapse
|
32
|
Lefebvre T, Petitclerc L, Hébert M, Bilodeau L, Sebastiani G, Olivié D, Gao ZH, Sylvestre MP, Cloutier G, Nguyen BN, Gilbert G, Tang A. MRI cine-tagging of cardiac-induced motion for noninvasive staging of liver fibrosis. J Magn Reson Imaging 2019; 51:1570-1580. [PMID: 31605412 DOI: 10.1002/jmri.26935] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Accepted: 09/05/2019] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND MR elastography is a noninvasive technique that provides high diagnostic accuracy for the staging of liver fibrosis; however, it requires external hardware and mainly assesses the right lobe. PURPOSE To evaluate the diagnostic performance of MRI cine-tagging for staging fibrosis in the left liver lobe, using biopsy as the reference standard. STUDY TYPE Institutional Review Board (IRB)-approved two-center prospective study. POPULATION Seventy-six patients with chronic liver disease who underwent an MRI cine-tagging examination and a liver biopsy within a 6-week interval. FIELD STRENGTH/SEQUENCE 2D-GRE multislice sequence at 3.0T with spatial modulation of the magnetization preparation sequence and peripheral pulse-wave triggering on two coronal slices chosen underneath the heart apex to capture maximal deformation with consecutive breath-holds adapted to patient cardiac frequency. ASSESSMENT A region of interest was selected in the liver close to the heart apex. Maximal strain was evaluated with the harmonic phase (HARP) technique. STATISTICAL TESTS Spearman's correlation, Kruskal-Wallis test, Mann-Whitney U-test, and receiver operating characteristic (ROC) analysis were performed. RESULTS Liver strain measured on tagged images decreased with higher histological fibrosis stage (ρ = -0.68, P < 0.0001). Strain values were significantly different between all fibrosis stages (P < 0.0001), and between groups of fibrosis stages ≤F3 vs. F4 (P < 0.05). Areas under the ROC curves were 0.95 (95% confidence interval: 0.89-1.00) to distinguish fibrosis stages F0 vs. F4, 0.81 (0.70-0.92) for stages F0 vs. ≥F1, 0.84 (0.76-0.93) for stages ≤F1 vs. ≥F2, 0.86 (0.78-0.94) for stages ≤F2 vs. ≥F3, and 0.87 (0.77-0.96) for stages ≤F3 vs. F4. DATA CONCLUSION MRI cine-tagging is a promising technique for measuring liver strain without additional elastography hardware. It could be used to assess the left liver lobe as a complement to current techniques assessing the right lobe. LEVEL OF EVIDENCE 1 Technical Efficacy: 3 J. Magn. Reson. Imaging 2020;51:1570-1580.
Collapse
Affiliation(s)
- Thierry Lefebvre
- Department of Radiology, Radio-Oncology and Nuclear Medicine, Université de Montréal, Montréal, Québec, Canada.,Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada.,Medical Physics Unit, McGill University, Montréal, Québec, Canada
| | - Léonie Petitclerc
- Department of Radiology, Radio-Oncology and Nuclear Medicine, Université de Montréal, Montréal, Québec, Canada.,Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada.,C.J. Gorter Center for High Field MRI, Department of Radiology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Mélanie Hébert
- Department of Radiology, Radio-Oncology and Nuclear Medicine, Université de Montréal, Montréal, Québec, Canada.,Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Laurent Bilodeau
- Department of Radiology, Radio-Oncology and Nuclear Medicine, Université de Montréal, Montréal, Québec, Canada.,Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Giada Sebastiani
- Department of Medicine, Division of Gastroenterology and Hepatology, McGill University Health Centre (MUHC), Montréal, Québec, Canada
| | - Damien Olivié
- Department of Radiology, Radio-Oncology and Nuclear Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Zu-Hua Gao
- Department of Pathology, McGill University, Montréal, Québec, Canada
| | - Marie-Pierre Sylvestre
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada.,Department of Social and Preventive Medicine, École de santé publique de l'Université de Montréal (ESPUM), Montréal, Québec, Canada
| | - Guy Cloutier
- Department of Radiology, Radio-Oncology and Nuclear Medicine, Université de Montréal, Montréal, Québec, Canada.,Institute of Biomedical Engineering, Université de Montréal, Montréal, Québec, Canada.,Laboratory of Biorheology and Medical Ultrasonics (LBUM), Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Bich N Nguyen
- Service of Pathology, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, Québec, Canada
| | - Guillaume Gilbert
- Department of Radiology, Radio-Oncology and Nuclear Medicine, Université de Montréal, Montréal, Québec, Canada.,MR Clinical Science, Philips Healthcare Canada, Montréal, Québec, Canada
| | - An Tang
- Department of Radiology, Radio-Oncology and Nuclear Medicine, Université de Montréal, Montréal, Québec, Canada.,Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada.,Institute of Biomedical Engineering, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
33
|
Reinhardt JW, Gooch KJ. An Agent-Based Discrete Collagen Fiber Network Model of Dynamic Traction Force-Induced Remodeling. J Biomech Eng 2019; 140:2654976. [PMID: 28975252 DOI: 10.1115/1.4037947] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Indexed: 01/17/2023]
Abstract
Microstructural properties of extracellular matrix (ECM) promote cell and tissue homeostasis as well as contribute to the formation and progression of disease. In order to understand how microstructural properties influence the mechanical properties and traction force-induced remodeling of ECM, we developed an agent-based model that incorporates repetitively applied traction force within a discrete fiber network. An important difference between our model and similar finite element models is that by implementing more biologically realistic dynamic traction, we can explore a greater range of matrix remodeling. Here, we validated our model by reproducing qualitative trends observed in three sets of experimental data reported by others: tensile and shear testing of cell-free collagen gels, collagen remodeling around a single isolated cell, and collagen remodeling between pairs of cells. In response to tensile and shear strain, simulated acellular networks with straight fibrils exhibited biphasic stress-strain curves indicative of strain-stiffening. When fibril curvature was introduced, stress-strain curves shifted to the right, delaying the onset of strain-stiffening. Our data support the notion that strain-stiffening might occur as individual fibrils successively align along the axis of strain and become engaged in tension. In simulations with a single, contractile cell, peak collagen displacement occurred closest to the cell and decreased with increasing distance. In simulations with two cells, compaction of collagen between cells appeared inversely related to the initial distance between cells. These results for cell-populated collagen networks match in vitro findings. A demonstrable benefit of modeling is that it allows for further analysis not feasible with experimentation. Within two-cell simulations, strain energy within the collagen network measured from the final state was relatively uniform around the outer surface of cells separated by 250 μm, but became increasingly nonuniform as the distance between cells decreased. For cells separated by 75 and 100 μm, strain energy peaked in the direction toward the other cell in the region in which fibrils become highly aligned and reached a minimum adjacent to this region, not on the opposite side of the cell as might be expected. This pattern of strain energy was partly attributable to the pattern of collagen compaction, but was still present when mapping strain energy divided by collagen density. Findings like these are of interest because fibril alignment, density, and strain energy may each contribute to contact guidance during tissue morphogenesis.
Collapse
Affiliation(s)
- James W Reinhardt
- Department of Biomedical Engineering, The Ohio State University, 270 Bevis Hall, 1080 Carmack Road, Columbus, OH 43210 e-mail:
| | - Keith J Gooch
- Department of Biomedical Engineering, The Ohio State University, 270 Bevis Hall, 1080 Carmack Road, Columbus, OH 43210.,Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University, 473 W. 12th Avenue, Columbus, OH 43210 e-mail:
| |
Collapse
|
34
|
Tossas-Milligan K, Shalabi S, Jones V, Keely PJ, Conklin MW, Elicerie KW, Winn R, Sistrunk C, Geradts J, Miranda-Carboni G, Dietze EC, Yee LD, Seewaldt VL. Mammographic density: intersection of advocacy, science, and clinical practice. CURRENT BREAST CANCER REPORTS 2019; 11:100-110. [PMID: 33312342 PMCID: PMC7728377 DOI: 10.1007/s12609-019-00316-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Purpose Here we aim to review the association between mammographic density, collagen structure and breast cancer risk. Findings While mammographic density is a strong predictor of breast cancer risk in populations, studies by Boyd show that mammographic density does not predict breast cancer risk in individuals. Mammographic density is affected by age, parity, menopausal status, race/ethnicity, and body mass index (BMI).New studies normalize mammographic density to BMI may provide a more accurate way to compare mammographic density in women of diverse race and ethnicity. Preclinical and tissue-based studies have investigated the role collagen composition and structure in predicting breast cancer risk. There is emerging evidence that collagen structure may activate signaling pathways associated with aggressive breast cancer biology. Summary Measurement of film mammographic density does not adequately capture the complex signaling that occurs in women with at-risk collagen. New ways to measure at-risk collagen potentially can provide a more accurate view of risk.
Collapse
Affiliation(s)
| | - Sundus Shalabi
- City of Hope Comprehensive Cancer Center, Duarte, CA
- Al Quds University, Jerusalem, West Bank
| | | | | | | | | | - Robert Winn
- University of Illinois, Chicago Cancer Center, Chicago, IL
| | | | | | | | | | - Lisa D. Yee
- City of Hope Comprehensive Cancer Center, Duarte, CA
| | | |
Collapse
|
35
|
Kunz HE, Dasari S, Lanza IR. EPA and DHA elicit distinct transcriptional responses to high-fat feeding in skeletal muscle and liver. Am J Physiol Endocrinol Metab 2019; 317:E460-E472. [PMID: 31265326 PMCID: PMC6766610 DOI: 10.1152/ajpendo.00083.2019] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Omega-3 polyunsaturated fatty acids (n-3 PUFAs) exert numerous beneficial biological effects and attenuate diet-induced insulin resistance in rodent models. In the present study, the independent, tissue-specific effects of two nutritionally relevant n-3 PUFAs, eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), were characterized in the context of a high-fat diet (HFD). EPA and DHA supplementation (3.2% of total fat) in 6-mo-old male C57BL/6 mice fed an HFD (60% fat) partially mitigated reductions in insulin sensitivity. At 5 wk, the area above the curve below baseline glucose following an intraperitoneal insulin tolerance test was 54.5% lower in HFD than control, whereas HFD + EPA and HFD + DHA showed 27.6% and 17.1% reductions, respectively. At 10 wk, HFD increased mitochondrial oxidative capacity supported by lipid and carbohydrate-based substrates in both liver and skeletal muscle (P < 0.05), with little effect of EPA or DHA supplementation. Whole genome transcriptomic analyses revealed HFD-induced transcriptional changes indicative of inflammation and fibrosis in both liver and muscle. Gene set enrichment analyses indicated a downregulation of transcripts associated with extracellular matrix in muscle (family-wise error rate P < 0.01) and liver (P = 0.04) and in transcripts associated with inflammation in muscle (P = 0.03) in HFD + DHA compared with HFD alone. In contrast, EPA appeared to potentiate some proinflammatory effects of the HFD. In the skeletal muscle, DHA increased the expression of stress-responsive genes, whereas EPA upregulated the expression of transcripts related to cell cycle. Therefore, although both EPA and DHA supplementation during HFD partially preserve insulin signaling, they modulate distinct processes, highlighting their unique biological effects in the context of obesity.
Collapse
Affiliation(s)
- Hawley E Kunz
- Division of Endocrinology and Metabolism, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Surendra Dasari
- Division of Biostatistics, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Ian R Lanza
- Division of Endocrinology and Metabolism, Mayo Clinic College of Medicine, Rochester, Minnesota
| |
Collapse
|
36
|
Nguyen TN, Podkowa AS, Tam AY, Arnold EC, Miller RJ, Park TH, Do MN, Oelze ML. Characterizing Fatty Liver in vivo in Rabbits, Using Quantitative Ultrasound. ULTRASOUND IN MEDICINE & BIOLOGY 2019; 45:2049-2062. [PMID: 31076231 PMCID: PMC6591075 DOI: 10.1016/j.ultrasmedbio.2019.03.021] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 03/06/2019] [Accepted: 03/28/2019] [Indexed: 05/09/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common cause of chronic liver disease and can often lead to fibrosis, cirrhosis, cancer and complete liver failure. Liver biopsy is the current standard of care to quantify hepatic steatosis, but it comes with increased patient risk and only samples a small portion of the liver. Imaging approaches to assess NAFLD include proton density fat fraction estimated via magnetic resonance imaging (MRI) and shear wave elastography. However, MRI is expensive and shear wave elastography is not proven to be sensitive to fat content of the liver (Kramer et al. 2016). On the other hand, ultrasonic attenuation and the backscatter coefficient (BSC) have been observed to be sensitive to levels of fat in the liver (Lin et al. 2015; Paige et al. 2017). In this study, we assessed the use of attenuation and the BSC to quantify hepatic steatosis in vivo in a rabbit model of fatty liver. Rabbits were maintained on a high-fat diet for 0, 1, 2, 3 or 6 wk, with 3 rabbits per diet group (total N = 15). An array transducer (L9-4) with a center frequency of 4.5 MHz connected to a SonixOne scanner was used to gather radio frequency (RF) backscattered data in vivo from rabbits. The RF signals were used to estimate an average attenuation and BSC for each rabbit. Two approaches were used to parameterize the BSC (i.e., the effective scatterer diameter and effective acoustic concentration using a spherical Gaussian model and a model-free approach using a principal component analysis [PCA]). The 2 major components of the PCA from the BSCs, which captured 96% of the variance of the transformed data, were used to generate input features to a support vector machine for classification. Rabbits were separated into two liver fat-level classes, such that approximately half of the rabbits were in the low-lipid class (≤9% lipid liver level) and half of the rabbits in the high-lipid class (>9% lipid liver level). The slope and the midband fit of the attenuation coefficient provided statistically significant differences (p value = 0.00014 and p value = 0.007, using a two-sample t test) between low and high-lipid fat classes. The proposed model-free and model-based parameterization of the BSC and attenuation coefficient parameters yielded classification accuracies of 84.11 %, 82.93 % and 78.91 % for differentiating low-lipid versus high-lipid classes, respectively. The results suggest that attenuation and BSC analysis can differentiate low-fat versus high-fat livers in a rabbit model of fatty liver disease.
Collapse
Affiliation(s)
- Trong N Nguyen
- Beckman Institute for Advanced Science and Technology, Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Anthony S Podkowa
- Beckman Institute for Advanced Science and Technology, Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Alex Y Tam
- Beckman Institute for Advanced Science and Technology, Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Eben C Arnold
- Beckman Institute for Advanced Science and Technology, Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Rita J Miller
- Beckman Institute for Advanced Science and Technology, Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Trevor H Park
- Beckman Institute for Advanced Science and Technology, Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Minh N Do
- Beckman Institute for Advanced Science and Technology, Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Michael L Oelze
- Beckman Institute for Advanced Science and Technology, Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
37
|
Retinoids in Stellate Cells: Development, Repair, and Regeneration. J Dev Biol 2019; 7:jdb7020010. [PMID: 31137700 PMCID: PMC6630434 DOI: 10.3390/jdb7020010] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 05/15/2019] [Accepted: 05/17/2019] [Indexed: 01/17/2023] Open
Abstract
Stellate cells, either hepatic (HSCs) or pancreatic (PSCs), are a type of interstitial cells characterized by their ability to store retinoids in lipid vesicles. In pathological conditions both HSCs and PSCs lose their retinoid content and transform into fibroblast-like cells, contributing to the fibrogenic response. HSCs also participate in other functions including vasoregulation, drug detoxification, immunotolerance, and maintenance of the hepatocyte population. PSCs maintain pancreatic tissue architecture and regulate pancreatic exocrine function. Recently, PSCs have attracted the attention of researchers due to their interactions with pancreatic ductal adenocarcinoma cells. PSCs promote tumour growth and angiogenesis, and their fibrotic activity increases the resistance of pancreatic cancer to chemotherapy and radiation. We are reviewing the current literature concerning the role played by retinoids in the physiology and pathophysiology of the stellate cells, paying attention to their developmental aspects as well as the function of stellate cells in tissue repair and organ regeneration.
Collapse
|
38
|
Thomas J, Sharma A, Panwar V, Chopra V, Ghosh D. Polysaccharide-Based Hybrid Self-Healing Hydrogel Supports the Paracrine Response of Mesenchymal Stem Cells. ACS APPLIED BIO MATERIALS 2019; 2:2013-2027. [DOI: 10.1021/acsabm.9b00074] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Jijo Thomas
- Institute of Nano Science and Technology, Habitat Centre, Phase 10, Mohali, Punjab 160062, India
| | - Anjana Sharma
- Institute of Nano Science and Technology, Habitat Centre, Phase 10, Mohali, Punjab 160062, India
| | - Vineeta Panwar
- Institute of Nano Science and Technology, Habitat Centre, Phase 10, Mohali, Punjab 160062, India
| | - Vianni Chopra
- Institute of Nano Science and Technology, Habitat Centre, Phase 10, Mohali, Punjab 160062, India
| | - Deepa Ghosh
- Institute of Nano Science and Technology, Habitat Centre, Phase 10, Mohali, Punjab 160062, India
| |
Collapse
|
39
|
Han Y, Pan L, Ran S, Song Y, Sun FF, Wang YZ, Hong Y. Rhizoma Paridis saponins ameliorates hepatic fibrosis in rats by downregulating expression of angiogenesis‑associated growth factors. Mol Med Rep 2019; 19:3548-3554. [PMID: 30864692 PMCID: PMC6471138 DOI: 10.3892/mmr.2019.10006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 12/18/2018] [Indexed: 02/07/2023] Open
Abstract
Previously, we demonstrated that Rhizoma Paridis saponins (RPS), the major active component of Rhizoma Paridis, may exhibit hepatoprotective effects. The present study aimed to identify the potential mechanism of RPS on hepatic injury and improvement in hepatic fibrosis (HF). A HF model was created in Sprague-Dawley rats by administration of carbon tetrachloride. RPS was administered for treatment following creation of the HF model. The protein and mRNA expression of vascular endothelial growth factor (VEGF), platelet-derived growth factor (PDGF), extracellular signal-regulated kinase (ERK)1/2 and α-smooth muscle actin (SMA) was detected by reverse transcription quantitative polymerase chain reaction and western blot analysis. RPS was demonstrated to improve hepatic inflammation and decrease HF severity according to hematoxylin and eosin and Masson trichrome staining. Following RPS treatment, the level of alanine aminotransferase, aspartate aminotransferase and malondialdehyde, and expression levels of the mRNA and protein of VEGF, ERK1/2, PDGF and α-SMA in the model group was decreased. By contrast, the content of glutathione-PX and superoxide dismutase was increased. These data suggest that RPS may treat HF primarily through downregulation of the expression levels of the mRNA and phosphorylated VEGF, ERK1/2, PDGF and α-SMA proteins.
Collapse
Affiliation(s)
- Yanquan Han
- Grade 3 Laboratory of Traditional Chinese Medicine Preparation, The First Affiliated Hospital, Anhui University of Chinese Medicine, State Administration of Anhui University of Chinese Medicine, Hefei, Anhui 230031, P.R. China
| | - Lingyu Pan
- Grade 3 Laboratory of Traditional Chinese Medicine Preparation, The First Affiliated Hospital, Anhui University of Chinese Medicine, State Administration of Anhui University of Chinese Medicine, Hefei, Anhui 230031, P.R. China
| | - Shan Ran
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230031, P.R. China
| | - Yan Song
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230031, P.R. China
| | - Fang-Fang Sun
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230031, P.R. China
| | - Yong-Zhong Wang
- Grade 3 Laboratory of Traditional Chinese Medicine Preparation, The First Affiliated Hospital, Anhui University of Chinese Medicine, State Administration of Anhui University of Chinese Medicine, Hefei, Anhui 230031, P.R. China
| | - Yan Hong
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230031, P.R. China
| |
Collapse
|
40
|
Cortes E, Lachowski D, Rice A, Chronopoulos A, Robinson B, Thorpe S, Lee DA, Possamai LA, Wang H, Pinato DJ, Del Río Hernández AE. Retinoic Acid Receptor-β Is Downregulated in Hepatocellular Carcinoma and Cirrhosis and Its Expression Inhibits Myosin-Driven Activation and Durotaxis in Hepatic Stellate Cells. Hepatology 2019; 69:785-802. [PMID: 30055117 DOI: 10.1002/hep.30193] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 07/25/2018] [Indexed: 01/17/2023]
Abstract
Hepatic stellate cells (HSCs) are essential perisinusoidal cells in both healthy and diseased liver. HSCs modulate extracellular matrix (ECM) homeostasis when quiescent, but in liver fibrosis, HSCs become activated and promote excess deposition of ECM molecules and tissue stiffening via force generation and mechanosensing. In hepatocellular carcinoma (HCC), activated HSCs infiltrate the stroma and migrate to the tumor core to facilitate paracrine signaling with cancer cells. Because the function of HSCs is known to be modulated by retinoids, we investigated the expression profile of retinoic acid receptor beta (RAR-β) in patients with cirrhosis and HCC, as well as the effects of RAR-β activation in HSCs. We found that RAR-β expression is significantly reduced in cirrhotic and HCC tissues. Using a comprehensive set of biophysical methods combined with cellular and molecular biology, we have elucidated the biomechanical mechanism by which all trans-retinoic acid promotes HSC deactivation via RAR-β-dependent transcriptional downregulation of myosin light chain 2 expression. Furthermore, this also abrogated mechanically driven migration toward stiffer substrates. Conclusion: Targeting mechanotransduction in HSCs at the transcriptional level may offer therapeutic options for a range of liver diseases.
Collapse
Affiliation(s)
- Ernesto Cortes
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Dariusz Lachowski
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Alistair Rice
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Antonios Chronopoulos
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Benjamin Robinson
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Stephen Thorpe
- School of Engineering and Materials Science, Queen Mary University of London, London, United Kingdom
| | - David A Lee
- School of Engineering and Materials Science, Queen Mary University of London, London, United Kingdom
| | - Lucia A Possamai
- Division of Integrative Systems Medicine and Digestive Disease, Imperial College London, London, United Kingdom
| | - Haiyun Wang
- School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - David J Pinato
- Department of Surgery and Cancer, Hammersmith Hospital Campus, Imperial College London, London, United Kingdom
| | - Armando E Del Río Hernández
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, United Kingdom
| |
Collapse
|
41
|
Stefanovic B, Manojlovic Z, Vied C, Badger CD, Stefanovic L. Discovery and evaluation of inhibitor of LARP6 as specific antifibrotic compound. Sci Rep 2019; 9:326. [PMID: 30674965 PMCID: PMC6344531 DOI: 10.1038/s41598-018-36841-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 11/27/2018] [Indexed: 01/17/2023] Open
Abstract
Fibrosis is characterized by excessive production of type I collagen. Biosynthesis of type I collagen in fibrosis is augmented by binding of protein LARP6 to the 5' stem-loop structure (5'SL), which is found exclusively in type I collagen mRNAs. A high throughput screen was performed to discover inhibitors of LARP6 binding to 5'SL, as potential antifibrotic drugs. The screen yielded one compound (C9) which was able to dissociate LARP6 from 5' SL RNA in vitro and to inactivate the binding of endogenous LARP6 in cells. Treatment of hepatic stellate cells (liver cells responsible for fibrosis) with nM concentrations of C9 reduced secretion of type I collagen. In precision cut liver slices, as an ex vivo model of hepatic fibrosis, C9 attenuated the profibrotic response at 1 μM. In prophylactic and therapeutic animal models of hepatic fibrosis C9 prevented development of fibrosis or hindered the progression of ongoing fibrosis when administered at 1 mg/kg. Toxicogenetics analysis revealed that only 42 liver genes changed expression after administration of C9 for 4 weeks, suggesting minimal off target effects. Based on these results, C9 represents the first LARP6 inhibitor with significant antifibrotic activity.
Collapse
Affiliation(s)
- Branko Stefanovic
- Department of Biomedical Sciences, College of Medicine, Florida State University, 1115 West Call Street, Tallahassee, FL, 32306, USA.
| | - Zarko Manojlovic
- Keck School of Medicine of University of Southern California, 1450 Biggy Street, NRT 4510, Los Angeles, CA, 90033, USA
| | - Cynthia Vied
- Translational Science Laboratory, College of Medicine, Florida State University, 1115 West Call Street, Tallahassee, FL, 32306, USA
| | - Crystal-Dawn Badger
- Translational Science Laboratory, College of Medicine, Florida State University, 1115 West Call Street, Tallahassee, FL, 32306, USA
- Proteomics and Metabolomics Facility, Colorado State University, 401 West Pitkin Street, Fort Collins, CO, 80521, USA
| | - Lela Stefanovic
- Department of Biomedical Sciences, College of Medicine, Florida State University, 1115 West Call Street, Tallahassee, FL, 32306, USA
| |
Collapse
|
42
|
Pan LX, Li LY, Zhou H, Cheng SQ, Liu YM, Lian PP, Li L, Wang LL, Rong SJ, Shen CP, Li J, Xu T. TMEM100 mediates inflammatory cytokines secretion in hepatic stellate cells and its mechanism research. Toxicol Lett 2019; 317:82-91. [PMID: 30639579 DOI: 10.1016/j.toxlet.2018.12.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 10/31/2018] [Accepted: 12/21/2018] [Indexed: 12/12/2022]
Abstract
Recent studies have shown that Transmembrane protein 100 (TMEM100) is a gene at locus 17q32 encoding a 134-amino acid protein with two hypothetical transmembrane domainsa, and first identified as a transcript from the mouse genome. As a downstream target gene of bone morphogenetic protein (BMP)-activin receptor-like kinase 1 (ALK1) signaling, it was activated to participate in inducing arterial endothelium differentiation, maintaining vascular integrity, promoting cell apoptosis, inhibiting metastasis and proliferation of cancer cells. However, evidence for the function of TMEM100 in inflammation is still limited. In this study, we explore the role of TMEM100 in inflammatory cytokine secretion and the role of MAPK signaling pathways in tumor necrosis factor-alpha (TNF-α)-induced TMEM100 expression in LX-2 cells. We found that the expression of TMEM100 was decreased markedly in human liver fibrosis tissues, and its expression was also inhibited in LX-2 cells induced by TNF-α, suggesting that it might be associated with the development of inflammation. Therefore, we demonstrated that overexpression of TMEM100 by transfecting pEGFP-C2-TMEM100 could lead to the down-regulation of IL-1β and IL-6 secretion. Moreover, we found that expression changes of TMEM100 could be involved in inhibition or activation of MAPK signaling pathways accompanied with regulating phosphorylation levels of ERK and JNK protein in response to TNF-α. These results suggested that TMEM100 might play an important role in the secretion of inflammatory cytokines (IL-1β and IL-6) of LX-2 cells induced by TNF-α, and MAPK (ERK and JNK) signaling pathways might participate in its induction of expression.
Collapse
Affiliation(s)
- Lin-Xin Pan
- School of Life Sciences, Anhui Medical University, Hefei, 230032, China
| | - Liang-Yun Li
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, 230032, China; Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Hong Zhou
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, 230032, China; Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, 230032, China; Anhui Provincial Cancer Hospital, West Branch of The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230031, China
| | - Shu-Qi Cheng
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, 230032, China; Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Yu-Min Liu
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, 230032, China; Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Pan-Pan Lian
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, 230032, China; Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Li Li
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, 230032, China; Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, 230032, China; Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Le-le Wang
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, 230032, China; Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Shan-Jie Rong
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, 230032, China; Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Chuan-Pu Shen
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, 230032, China; Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Jun Li
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, 230032, China; Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, 230032, China.
| | - Tao Xu
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, 230032, China; Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
43
|
Tamoxifen mechanically deactivates hepatic stellate cells via the G protein-coupled estrogen receptor. Oncogene 2018; 38:2910-2922. [PMID: 30575816 PMCID: PMC6755965 DOI: 10.1038/s41388-018-0631-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Revised: 11/12/2018] [Accepted: 11/28/2018] [Indexed: 02/07/2023]
Abstract
Tamoxifen has been used for many years to target estrogen receptor signalling in breast cancer cells. Tamoxifen is also an agonist of the G protein-coupled estrogen receptor (GPER), a GPCR ubiquitously expressed in tissues that mediates the acute response to estrogens. Here we report that tamoxifen promotes mechanical quiescence in hepatic stellate cells (HSCs), stromal fibroblast-like cells whose activation triggers and perpetuates liver fibrosis in hepatocellular carcinomas. This mechanical deactivation is mediated by the GPER/RhoA/myosin axis and induces YAP deactivation. We report that tamoxifen decreases the levels of hypoxia-inducible factor-1 alpha (HIF-1α) and the synthesis of extracellular matrix proteins through a mechanical mechanism that involves actomyosin-dependent contractility and mechanosensing of tissue stiffness. Our results implicate GPER-mediated estrogen signalling in the mechanosensory-driven activation of HSCs and put forward estrogenic signalling as an option for mechanical reprogramming of myofibroblast-like cells in the tumour microenvironment. Tamoxifen, with half a century of safe clinical use, might lead this strategy of drug repositioning.
Collapse
|
44
|
Thi Thanh Hai N, Thuy LTT, Shiota A, Kadono C, Daikoku A, Hoang DV, Dat NQ, Sato-Matsubara M, Yoshizato K, Kawada N. Selective overexpression of cytoglobin in stellate cells attenuates thioacetamide-induced liver fibrosis in mice. Sci Rep 2018; 8:17860. [PMID: 30552362 PMCID: PMC6294752 DOI: 10.1038/s41598-018-36215-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 11/15/2018] [Indexed: 02/07/2023] Open
Abstract
Cytoglobin (CYGB), discovered in hepatic stellate cells (HSCs), is known to possess a radical scavenger function, but its pathophysiological roles remain unclear. Here, for the first time, we generated a new transgenic (TG) mouse line in which both Cygb and mCherry reporter gene expression were under the control of the native Cygb gene promoter. We demonstrated that the expression of Cygb-mCherry was related to endogenous Cygb in adult tissues by tracing mCherry fluorescence together with DNA, mRNA, and protein analyses. Administration of a single dose (50 mg/kg) of thioacetamide (TAA) in Cygb-TG mice resulted in lower levels of alanine transaminase and oxidative stress than those in WT mice. After 10 weeks of TAA administration, Cygb-TG livers exhibited reduced neutrophil accumulation, cytokine expression and fibrosis but high levels of quiescent HSCs. Primary HSCs isolated from Cygb-TG mice (HSCCygb-TG) exhibited significantly decreased mRNA levels of α-smooth muscle actin (αSMA), collagen 1α1, and transforming growth factor β-3 after 4 days in culture relative to WT cells. HSCsCygb-TG were resistant to H2O2-induced αSMA expression. Thus, cell-specific overexpression of Cygb attenuates HSC activation and protects mice against TAA-induced liver fibrosis presumably by maintaining HSC quiescence. Cygb is a potential new target for antifibrotic approaches.
Collapse
Affiliation(s)
- Nguyen Thi Thanh Hai
- Departments of Hepatology, Graduate School of Medicine, Osaka City University, Osaka, Japan
- Department of Biochemistry, Hanoi Medical University, Hanoi, Vietnam
| | - Le Thi Thanh Thuy
- Departments of Hepatology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | | | - Chiho Kadono
- Departments of Hepatology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Atsuko Daikoku
- Departments of Hepatology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Dinh Viet Hoang
- Departments of Hepatology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Ninh Quoc Dat
- Departments of Hepatology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Misako Sato-Matsubara
- Departments of Hepatology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Katsutoshi Yoshizato
- Departments of Hepatology, Graduate School of Medicine, Osaka City University, Osaka, Japan
- PhoenixBio Co. Ltd., Hiroshima, Japan
- Endowed Laboratory of Synthetic Biology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Norifumi Kawada
- Departments of Hepatology, Graduate School of Medicine, Osaka City University, Osaka, Japan.
| |
Collapse
|
45
|
Ahmad A, Ahmad R. Proteomic approach to identify molecular signatures during experimental hepatic fibrosis and resveratrol supplementation. Int J Biol Macromol 2018; 119:1218-1227. [DOI: 10.1016/j.ijbiomac.2018.08.062] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 08/03/2018] [Accepted: 08/12/2018] [Indexed: 12/15/2022]
|
46
|
Wei W, Jiang F, Liu XC, Su Q. TMEM9 mediates IL-6 and IL-1β secretion and is modulated by the Wnt pathway. Int Immunopharmacol 2018; 63:253-260. [PMID: 30119033 DOI: 10.1016/j.intimp.2018.07.036] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Revised: 07/27/2018] [Accepted: 07/28/2018] [Indexed: 02/07/2023]
Abstract
Novel studies have shown that the Transmembrane protein 9 (TMEM9) gene is localized at 1q41 and encodes a protein consisting of 183 amino acids with an N-terminus containing many important domains. As a novel human transmembrane protein, TMEM9 is highly conserved in species from Caenorhabditis elegans to humans and is widely expressed in many tissues and cells. Moreover, TMEM9 may play an important role in intracellular transport and the growth of hepatoma cells. However, evidence for the function of TMEM9 in inflammation is still limited. We studied the expression of TMEM9 and its effect on cytokine secretion in tumor necrosis factor-alpha (TNF-α)-induced LX-2 cells. We proved that overexpression of TMEM9 by transfection with pEGFP-C2-TMEM9 may increase the expression of IL-6 and IL-1β in LX-2 cells. At the same time, knockdown of TMEM9 expression by transfection with a TMEM9-siRNA decreased IL-6 and IL-1β secretion in LX-2 cells. Additionally, our results proved that overexpression of TMEM9 enhanced the protein expression levels of the canonical Wnt/β-catenin accompanied by an upregulation of wnt2b, wnt3a and β-catenin protein levels in LX-2 cells treated with TNF-α. These results indicate that TMEM9 plays a significant role in TNF-α-enhanced cytokines (IL-6 and IL-1β) secretion in LX-2 cells and that the canonical Wnt/β-catenin signaling pathway is involved in the induction of these cytokine expressions.
Collapse
Affiliation(s)
- Wei Wei
- Department of Pathology, The First People's Hospital of Huainan, Huainan 232007, China
| | - Fei Jiang
- Hefei institute for food and drug control, Hefei 230001, China
| | - Xiao-Chang Liu
- Department of Gastroenterology, The First Affiliate Hospital of Anhui Medical University, Jixi Road No 218, Hefei, Anhui Province, 230032, China.
| | - Qian Su
- Department of infectious diseases, The First Affiliate Hospital of Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
47
|
Rohn F, Kordes C, Castoldi M, Götze S, Poschmann G, Stühler K, Herebian D, Benk AS, Geiger F, Zhang T, Spatz JP, Häussinger D. Laminin-521 promotes quiescence in isolated stellate cells from rat liver. Biomaterials 2018; 180:36-51. [PMID: 30014965 DOI: 10.1016/j.biomaterials.2018.07.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 07/06/2018] [Indexed: 12/15/2022]
Abstract
The laminin α5 protein chain is an element of basement membranes and important to maintain stem cells. Hepatic stellate cells (HSC) are liver-resident mesenchymal stem cells, which reside in a quiescent state on a basement membrane-like structure in the space of Dissé. In the present study, laminin α5 chain was detected in the space of Dissé of normal rat liver. Since HSC are critical for liver regeneration and can contribute to fibrosis in chronic liver diseases, the effect of laminins on HSC maintenance was investigated. Therefore, isolated rat HSC were seeded on uncoated polystyrene (PS) or PS coated with either laminin-521 (PS/LN-521) or laminin-211 (PS/LN-211). PS/LN-521 improved HSC adhesion and better preserved their retinoid stores as well as quiescence- and stem cell-associated phenotype, whereas HSC on PS/LN-211 or PS developed into myofibroblasts-like cells. To improve the homogeneity as well as the presentation of laminin molecules on the culture surface to HSC, laminin-functionalized, gold-nanostructured glass surfaces were generated. This approach further enhanced the expression of quiescence-associated genes in HSC. In conclusion, the results indicate that LN-521 supports the quiescent state of HSC and laminin α5 can be regarded as an important element of their niche in the space of Dissé.
Collapse
Affiliation(s)
- Friederike Rohn
- Clinic of Gastroenterology, Hepatology and Infectious Diseases, Heinrich Heine University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany
| | - Claus Kordes
- Clinic of Gastroenterology, Hepatology and Infectious Diseases, Heinrich Heine University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany
| | - Mirco Castoldi
- Clinic of Gastroenterology, Hepatology and Infectious Diseases, Heinrich Heine University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany
| | - Silke Götze
- Clinic of Gastroenterology, Hepatology and Infectious Diseases, Heinrich Heine University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany
| | - Gereon Poschmann
- Molecular Proteomics Laboratory, Biologisch-Medizinisches Forschungszentrum, Heinrich Heine University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany
| | - Kai Stühler
- Molecular Proteomics Laboratory, Biologisch-Medizinisches Forschungszentrum, Heinrich Heine University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany; Institute of Molecular Medicine, University Hospital Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany
| | - Diran Herebian
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Amelie S Benk
- Max-Planck-Institute for Medical Research, Department of Cellular Biophysics, Jahnstraße 29, 69120 Heidelberg, Germany; Department of Biophysical Chemistry, University of Heidelberg, Im Neuenheimer Feld 253, 69120 Heidelberg, Germany
| | - Fania Geiger
- Max-Planck-Institute for Medical Research, Department of Cellular Biophysics, Jahnstraße 29, 69120 Heidelberg, Germany; Department of Biophysical Chemistry, University of Heidelberg, Im Neuenheimer Feld 253, 69120 Heidelberg, Germany
| | - Tingyu Zhang
- Max-Planck-Institute for Medical Research, Department of Cellular Biophysics, Jahnstraße 29, 69120 Heidelberg, Germany; Department of Biophysical Chemistry, University of Heidelberg, Im Neuenheimer Feld 253, 69120 Heidelberg, Germany
| | - Joachim P Spatz
- Max-Planck-Institute for Medical Research, Department of Cellular Biophysics, Jahnstraße 29, 69120 Heidelberg, Germany; Department of Biophysical Chemistry, University of Heidelberg, Im Neuenheimer Feld 253, 69120 Heidelberg, Germany
| | - Dieter Häussinger
- Clinic of Gastroenterology, Hepatology and Infectious Diseases, Heinrich Heine University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany.
| |
Collapse
|
48
|
Hong Y, Li S, Wang J, Li Y. In vitro inhibition of hepatic stellate cell activation by the autophagy-related lipid droplet protein ATG2A. Sci Rep 2018; 8:9232. [PMID: 29915313 PMCID: PMC6006255 DOI: 10.1038/s41598-018-27686-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 05/18/2018] [Indexed: 02/07/2023] Open
Abstract
Clinical studies have found that moderate intake of retinol or oleic acid can enlarge the lipid droplets of hepatic stellate cells and suppress their activation. However, the link between lipid droplets and cell activation is unknown. This study compared the dynamics of lipid droplet-associated protein expression between activated and reverted stellate cells. Reversion of the activated human stellate cell line LX-2 and inhibition of primary mouse stellate cell activation were induced by retinol or oleic acid, which resulted in larger lipid droplets and the downregulation of cell activation markers. Quantitative proteomics and immunoblotting were performed to compare lipid-droplet protein profiles between activated and reverted LX-2 cells. Compared to expression in activated cells, 50 lipid-droplet proteins were upregulated, whereas 28 were downregulated upon reversion. ATG2A was significantly enriched in lipid droplets of retinol/oleic acid-treated LX-2 cells and quiescent primary stellate cells. Reduced expression of α-SMA, increased expression of perilipin-3, enlarged lipid droplets, and suppression of autophagic flux were observed in ATG2A-deficient LX2 cells. Lipid-droplet protein profile changes during the reversion of activated stellate cells might provide new insights into the molecular mechanisms linking lipid droplets to liver fibrosis. ATG2A could represent a potential new drug target for hepatic fibrosis.
Collapse
Affiliation(s)
- Yun Hong
- First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China. .,Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.
| | - Sirui Li
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Jifeng Wang
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Youming Li
- First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
49
|
Karimi F, O'Connor AJ, Qiao GG, Heath DE. Integrin Clustering Matters: A Review of Biomaterials Functionalized with Multivalent Integrin-Binding Ligands to Improve Cell Adhesion, Migration, Differentiation, Angiogenesis, and Biomedical Device Integration. Adv Healthc Mater 2018; 7:e1701324. [PMID: 29577678 DOI: 10.1002/adhm.201701324] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 01/24/2018] [Indexed: 01/17/2023]
Abstract
Material systems that exhibit tailored interactions with cells are a cornerstone of biomaterial and tissue engineering technologies. One method of achieving these tailored interactions is to biofunctionalize materials with peptide ligands that bind integrin receptors present on the cell surface. However, cell biology research has illustrated that both integrin binding and integrin clustering are required to achieve a full adhesion response. This biophysical knowledge has motivated researchers to develop material systems biofunctionalized with nanoscale clusters of ligands that promote both integrin occupancy and clustering of the receptors. These materials have improved a wide variety of biological interactions in vitro including cell adhesion, proliferation, migration speed, gene expression, and stem cell differentiation; and improved in vivo outcomes including increased angiogenesis, tissue healing, and biomedical device integration. This review first introduces the techniques that enable the fabrication of these nanopatterned materials, describes the improved biological effects that have been achieved, and lastly discusses the current limitations of the technology and where future advances may occur. Although this technology is still in its nascency, it will undoubtedly play an important role in the future development of biomaterials and tissue engineering scaffolds for both in vitro and in vivo applications.
Collapse
Affiliation(s)
- Fatemeh Karimi
- School of Chemical and Biomedical Engineering; Particulate Fluids Processing Centre; University of Melbourne; Parkville VIC 3010 Australia
- Polymer Science Group; Department of Chemical Engineering; Particulate Fluid Processing Centre; University of Melbourne; Parkville VIC 3010 Australia
| | - Andrea J. O'Connor
- School of Chemical and Biomedical Engineering; Particulate Fluids Processing Centre; University of Melbourne; Parkville VIC 3010 Australia
| | - Greg G. Qiao
- Polymer Science Group; Department of Chemical Engineering; Particulate Fluid Processing Centre; University of Melbourne; Parkville VIC 3010 Australia
| | - Daniel E. Heath
- School of Chemical and Biomedical Engineering; Particulate Fluids Processing Centre; University of Melbourne; Parkville VIC 3010 Australia
| |
Collapse
|
50
|
EphB2 receptor tyrosine kinase promotes hepatic fibrogenesis in mice via activation of hepatic stellate cells. Sci Rep 2018; 8:2532. [PMID: 29416088 PMCID: PMC5803231 DOI: 10.1038/s41598-018-20926-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 01/26/2018] [Indexed: 02/06/2023] Open
Abstract
Hepatic fibrosis is the result of an excessive wound-healing response subsequent to chronic liver injury. A feature of liver fibrogenesis is the secretion and deposition of extracellular matrix proteins by activated hepatic stellate cells (HSCs). Here we report that upregulation of EphB2 is a prominent feature of two mouse models of hepatic fibrosis and also observed in humans with liver cirrhosis. EphB2 is upregulated and activated in mouse HSCs following chronic carbon tetrachloride (CCl4) exposure. Moreover, we show that EphB2 deficiency attenuates liver fibrosis and inflammation and this is correlated with an overall reduction in pro-fibrotic markers, inflammatory chemokines and cytokines. In an in vitro system of HSCs activation we observed an impaired proliferation and sub-optimal differentiation into fibrogenic myofibroblasts of HSCs isolated from EphB2-/- mice compared to HSCs isolated from wild type mice. This supports the hypothesis that EphB2 promotes liver fibrosis partly via activation of HSCs. Cellular apoptosis which is generally observed during the regression of liver fibrogenesis was increased in liver specimens of CCl4-treated EphB2-/- mice compared to littermate controls. This data is suggestive of an active repair/regeneration system in the absence of EphB2. Altogether, our data validate this novel pro-fibrotic function of EphB2 receptor tyrosine kinase.
Collapse
|