1
|
Johnston EK, Fang Z, Soto-Gutierrez A, Taner CB, Cook KE, Yang L, Abbott RD. Engineering a three-dimensional liver steatosis model. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167888. [PMID: 40328412 DOI: 10.1016/j.bbadis.2025.167888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 04/28/2025] [Accepted: 05/02/2025] [Indexed: 05/08/2025]
Abstract
Liver transplantation is the key treatment for liver failure, yet organ scarcity, exacerbated by high discard rates of steatotic livers, leads to high waitlist mortality. Preclinical models of steatosis are necessary to understand the pathophysiology of the disease and to develop pharmacological interventions to decrease disease burden and liver discard rate. In this paper, we develop an expedited 3D steatotic organoid model containing primary human hepatocytes and non-parenchymal cells. We present our iterative approach as we transition from 2D to 3D models and from immortalized to primary cells to optimize conditions for the development of a 3D human steatosis model. Both primary cell aggregation and steatosis induction time were reduced from the standard, 5-7 days, to 2 days. Our 3D model incorporates human primary hepatocytes from discarded liver tissues, which have not been used in organoids previously due to their rapid loss of phenotype in culture. After optimizing our steatosis induction media there was a mix of macro- and micro-steatosis in these primary hepatocytes which is consistent with the human pathology. Our approach achieves a model reflective of the liver pathology, preserving cellular phenotypes and viability while exhibiting markers of oxidative stress, a key factor contributing to complications in the transplantation of steatotic livers.
Collapse
Affiliation(s)
- Elizabeth K Johnston
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Zhou Fang
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | | | - C Burcin Taner
- Department of Transplantation, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Keith E Cook
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Liu Yang
- Department of Transplantation, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Rosalyn D Abbott
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA.
| |
Collapse
|
2
|
Ros-Tarraga P, Villanueva-Badenas E, Sanchez-Gonzalez E, Gallego-Ferrer G, Donato MT, Tolosa L. Challenges of in vitro modelling of liver fibrosis. Front Cell Dev Biol 2025; 13:1567916. [PMID: 40371390 PMCID: PMC12075197 DOI: 10.3389/fcell.2025.1567916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 04/10/2025] [Indexed: 05/16/2025] Open
Abstract
Liver fibrosis has been proposed as the most important predictive indicator affecting prognosis of patients with chronic liver disease. It is defined by an abnormal accumulation of extracellular matrix components that results from necrotic and inflammatory processes and eventually impairs organ function. With no approved therapy, comprehensive cellular models directly derived from patient's cells are necessary to understand the mechanisms behind fibrosis and the response to anti-fibrotic therapies. Primary human cells, human hepatic cell lines and human stem cells-derived hepatic stellate-like cells have been widely used for studying fibrosis pathogenesis. In this paper, we depict the cellular crosstalk and the role of extracellular matrix during fibrosis pathogenesis and summarize different in vitro models from simple monolayers to multicellular 3D cultures used to gain deeper mechanistic understanding of the disease and the therapeutic response, discussing their major advantages and disadvantages for liver fibrosis modelling.
Collapse
Affiliation(s)
- Patricia Ros-Tarraga
- Experimental Hepatology Unit, Health Research Institute La Fe (IISLAFE), Valencia, Spain
| | - Estela Villanueva-Badenas
- Experimental Hepatology Unit, Health Research Institute La Fe (IISLAFE), Valencia, Spain
- Faculty of Medicine and Dentistry, Department of Biochemistry and Molecular Biology, University of Valencia, Valencia, Spain
| | - Estela Sanchez-Gonzalez
- Center for Biomaterials and Tissue Engineering (CBIT), Universitat Politècnica de València, Valencia, Spain
- Biomedical Research Networking Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Carlos III Health Institute, Valencia, Spain
| | - Gloria Gallego-Ferrer
- Center for Biomaterials and Tissue Engineering (CBIT), Universitat Politècnica de València, Valencia, Spain
- Biomedical Research Networking Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Carlos III Health Institute, Valencia, Spain
| | - M. Teresa Donato
- Experimental Hepatology Unit, Health Research Institute La Fe (IISLAFE), Valencia, Spain
- Faculty of Medicine and Dentistry, Department of Biochemistry and Molecular Biology, University of Valencia, Valencia, Spain
- Biomedical Research Networking Center in Hepatic and Digestive Diseases (CIBERehd), Carlos III Health Institute, Madrid, Spain
| | - Laia Tolosa
- Experimental Hepatology Unit, Health Research Institute La Fe (IISLAFE), Valencia, Spain
- Biomedical Research Networking Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Carlos III Health Institute, Valencia, Spain
| |
Collapse
|
3
|
Myrka AM, Frost R, Di Stefano D, Plotnikov SV, Buck LT. Cultured primary turtle hepatocytes: a cellular model for the study of temperature and anoxia. Am J Physiol Cell Physiol 2025; 328:C179-C198. [PMID: 39555638 DOI: 10.1152/ajpcell.00510.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 11/10/2024] [Accepted: 11/11/2024] [Indexed: 11/19/2024]
Abstract
Turtle hepatocytes are a nonexcitable model for metabolic depression during low-temperature and/or anoxic overwintering conditions. Cytoskeletal structure and mitochondrial distribution are continuously modified in cells, and we hypothesized that metabolic depression would inhibit such processes as cell attachment and spreading and promote withdrawal of cell protrusions and peripheral mitochondria. After developing a methodology for culturing painted turtle hepatocytes, two-dimensional (2-D) area and maintenance of cell attachment after a media change were used as indicators of structural rearrangement and spreading/volume. These were measured after incubating cells at varying temperatures and with or without the inclusion of cyanide (chemical proxy for anoxia). Experiments were performed using cells from 22°C- or 5°C-acclimated turtles. Live-cell imaging was used to monitor the effect of cyanide exposure on the distribution of mitochondria. We also acclimated cultured cells from 22°C-acclimated turtles to 4°C in vitro and scored withdrawal of protrusions. Only cells isolated from 5°C-acclimated turtles and incubated at 4°C had reduced attachment to fibronectin substrate, but cyanide exposure had no effect. These cells also had a 30% smaller 2-D area than those from 22°C-acclimated turtles. There was no change in mitochondrial distribution during cyanide perfusion. Finally, 4°C acclimation in vitro resulted in the withdrawal of protrusions over 14 days. Taken together with the results from cells acclimated to low temperature in vivo, this suggests inhibition of structural rearrangement and protrusion stability by low temperature acclimation, but not cyanide exposure. Our cultured primary hepatocyte system will facilitate further study of the role of structural dynamics in reversible metabolic depression.NEW & NOTEWORTHY We have optimized a methodology for two-dimensional (2-D) culturing of primary western painted turtle hepatocytes and used this model to study the effects of cyanide and temperature on structural rearrangement, and the effect of cyanide on mitochondrial distribution. Our results suggest that low temperature acclimation, either in vivo or in vitro, inhibits cell protrusions and structural rearrangement. Acute cyanide exposure did not inhibit structural rearrangement or alter mitochondrial distribution.
Collapse
Affiliation(s)
- Alexander M Myrka
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Ryan Frost
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Domenic Di Stefano
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Sergey V Plotnikov
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Leslie T Buck
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
- Department of Ecology and Evolutionary Biology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
4
|
Obmann VC, Ardoino M, Klaus J, Catucci D, Berzigotti A, Montani M, Peters A, Todorski I, Wagner B, Zbinden L, Gräni C, Ebner L, Heverhagen JT, Christe A, Huber AT. MRI Extracellular Volume Fraction in Liver Fibrosis-A Comparison of Different Time Points and Blood Pool Measurements. J Magn Reson Imaging 2024; 60:1678-1688. [PMID: 38553860 DOI: 10.1002/jmri.29259] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/12/2024] [Accepted: 01/14/2024] [Indexed: 11/15/2024] Open
Abstract
BACKGROUND Extracellular volume (ECV) correlates with the degree of liver fibrosis. PURPOSE To analyze the performance of liver MRI-based ECV evaluations with different blood pool measurements at different time points. STUDY TYPE Prospective. SAMPLE 73 consecutive patients (n = 31 females, mean age 56 years) with histopathology-proven liver fibrosis. FIELD STRENGTH/SEQUENCE 3T acquisition within 90 days of biopsy, including shortened modified look-locker inversion recovery T1 mapping. ASSESSMENT Polygonal regions of interest were manually drawn in the liver, aorta, vena cava, and in the main, left and right portal vein on four slices before and after Gd-DOTA administration at 5/10/15 minutes. ECV was calculated 1) on one single slice on portal bifurcation level, and 2) averaged over all four slices. STATISTICAL TESTS Parameters were compared between patients with fibrosis grades F0-2 and F3-F4 with the Mann-Whitney U and fishers exact test. ROC analysis was used to assess the performance of the parameters to predict F3-4 fibrosis. A P-value <0.05 was considered statistically significant. RESULTS ECV was significantly higher in F3-4 fibrosis (35.4% [33.1%-37.6%], 36.1% [34.2%-37.5%], and 37.0% [34.8%-39.2%] at 5/10/15 minutes) than in patients with F0-2 fibrosis (33.3% [30.8%-34.8%], 33.7% [31.6%-34.7%] and 34.9% [32.2%-36.0%]; AUC = 0.72-0.75). Blood pool T1 relaxation times in the aorta and vena cava were longer on the upper vs. lower slices at 5 minutes, but not at 10/15 minutes. AUC values were similar when measured on a single slice (AUC = 0.69-0.72) or based on blood pool measurements in the cava or portal vein (AUC = 0.63-0.67 and AUC = 0.65-0.70). DATA CONCLUSION Liver ECV is significantly higher in F3-4 fibrosis compared to F0-2 fibrosis with blood pool measurements performed in the aorta, inferior vena cava, and portal vein at 5, 10, and 15 minutes. However, a smaller variability was observed for blood pool measurements between slices at 15 minutes. LEVEL OF EVIDENCE 1 TECHNICAL EFFICACY: Stage 3.
Collapse
Affiliation(s)
- Verena Carola Obmann
- Department of Diagnostic, Interventional and Pediatric Radiology, Inselspital Bern, University Hospital, University of Bern, Bern, Switzerland
- Liver Elastography Center, Translational Imaging Center (TIC), Swiss Institute for Translational and Entrepreneurial Medicine, Bern, Switzerland
| | - Marie Ardoino
- Department of Diagnostic, Interventional and Pediatric Radiology, Inselspital Bern, University Hospital, University of Bern, Bern, Switzerland
| | - Jeremias Klaus
- Department of Diagnostic, Interventional and Pediatric Radiology, Inselspital Bern, University Hospital, University of Bern, Bern, Switzerland
- Institute of Forensic Medicine, University of Bern, Bern, Switzerland
| | - Damiano Catucci
- Department of Diagnostic, Interventional and Pediatric Radiology, Inselspital Bern, University Hospital, University of Bern, Bern, Switzerland
- Graduate School for Health Sciences, University of Bern, Bern, Switzerland
| | - Annalisa Berzigotti
- Department of Visceral Surgery and Medicine, Inselspital Bern University Hospital, University of Bern, Bern, Switzerland
| | - Matteo Montani
- Department of Pathology, University of Bern, Bern, Switzerland
| | - Alan Peters
- Department of Diagnostic, Interventional and Pediatric Radiology, Inselspital Bern, University Hospital, University of Bern, Bern, Switzerland
| | - Inga Todorski
- Department of Diagnostic, Interventional and Pediatric Radiology, Inselspital Bern, University Hospital, University of Bern, Bern, Switzerland
| | - Benedikt Wagner
- Department of Diagnostic, Interventional and Pediatric Radiology, Inselspital Bern, University Hospital, University of Bern, Bern, Switzerland
| | - Lukas Zbinden
- Department of Diagnostic, Interventional and Pediatric Radiology, Inselspital Bern, University Hospital, University of Bern, Bern, Switzerland
- ARTORG Center for Biomedical Engineering Research, University of Bern, Bern, Switzerland
| | - Christoph Gräni
- Department of Cardiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Lukas Ebner
- Department of Diagnostic, Interventional and Pediatric Radiology, Inselspital Bern, University Hospital, University of Bern, Bern, Switzerland
- Radiology and Nuclear Medicine (RUN), Luzerner Kantonsspital, University of Lucerne, Switzerland, Lucerne, Switzerland
| | - Johannes Thomas Heverhagen
- Department of Diagnostic, Interventional and Pediatric Radiology, Inselspital Bern, University Hospital, University of Bern, Bern, Switzerland
| | - Andreas Christe
- Department of Diagnostic, Interventional and Pediatric Radiology, Inselspital Bern, University Hospital, University of Bern, Bern, Switzerland
- Liver Elastography Center, Translational Imaging Center (TIC), Swiss Institute for Translational and Entrepreneurial Medicine, Bern, Switzerland
| | - Adrian Thomas Huber
- Department of Diagnostic, Interventional and Pediatric Radiology, Inselspital Bern, University Hospital, University of Bern, Bern, Switzerland
- Liver Elastography Center, Translational Imaging Center (TIC), Swiss Institute for Translational and Entrepreneurial Medicine, Bern, Switzerland
- Radiology and Nuclear Medicine (RUN), Luzerner Kantonsspital, University of Lucerne, Switzerland, Lucerne, Switzerland
| |
Collapse
|
5
|
Arteel GE. Hepatic Extracellular Matrix and Its Role in the Regulation of Liver Phenotype. Semin Liver Dis 2024; 44:343-355. [PMID: 39191427 PMCID: PMC12057067 DOI: 10.1055/a-2404-7973] [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] [Indexed: 08/29/2024]
Abstract
The hepatic extracellular matrix (ECM) is most accurately depicted as a dynamic compartment that comprises a diverse range of players that work bidirectionally with hepatic cells to regulate overall homeostasis. Although the classic meaning of the ECM referred to only proteins directly involved in generating the ECM structure, such as collagens, proteoglycans, and glycoproteins, the definition of the ECM is now broader and includes all components associated with this compartment. The ECM is critical in mediating phenotype at the cellular, organ, and even organismal levels. The purpose of this review is to summarize the prevailing mechanisms by which ECM mediates hepatic phenotype and discuss the potential or established role of this compartment in the response to hepatic injury in the context of steatotic liver disease.
Collapse
Affiliation(s)
- Gavin E. Arteel
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
6
|
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]
|
7
|
Wang Q, Liu J, Yin W, Wang A, Zheng J, Wang Y, Dong J. Microscale tissue engineering of liver lobule models: advancements and applications. Front Bioeng Biotechnol 2023; 11:1303053. [PMID: 38144540 PMCID: PMC10749204 DOI: 10.3389/fbioe.2023.1303053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 11/28/2023] [Indexed: 12/26/2023] Open
Abstract
The liver, as the body's primary organ for maintaining internal balance, is composed of numerous hexagonal liver lobules, each sharing a uniform architectural framework. These liver lobules serve as the basic structural and functional units of the liver, comprised of central veins, hepatic plates, hepatic sinusoids, and minute bile ducts. Meanwhile, within liver lobules, distinct regions of hepatocytes carry out diverse functions. The in vitro construction of liver lobule models, faithfully replicating their structure and function, holds paramount significance for research in liver development and diseases. Presently, two primary technologies for constructing liver lobule models dominate the field: 3D bioprinting and microfluidic techniques. 3D bioprinting enables precise deposition of cells and biomaterials, while microfluidics facilitates targeted transport of cells or other culture materials to specified locations, effectively managing culture media input and output through micro-pump control, enabling dynamic simulations of liver lobules. In this comprehensive review, we provide an overview of the biomaterials, cells, and manufacturing methods employed by recent researchers in constructing liver lobule models. Our aim is to explore strategies and technologies that closely emulate the authentic structure and function of liver lobules, offering invaluable insights for research into liver diseases, drug screening, drug toxicity assessment, and cell replacement therapy.
Collapse
Affiliation(s)
- Qi Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Juan Liu
- Research Unit of Precision Hepatobiliary Surgery Paradigm, Chinese Academy of Medical Sciences, Beijing, China
- Hepato-Pancreato-Biliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
- Institute for Organ Transplant and Bionic Medicine, Tsinghua University, Beijing, China
- Key Laboratory of Digital Intelligence Hepatology, Ministry of Education, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Wenzhen Yin
- Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - Anqi Wang
- Hepato-Pancreato-Biliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Jingjing Zheng
- Hepato-Pancreato-Biliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Yunfang Wang
- Research Unit of Precision Hepatobiliary Surgery Paradigm, Chinese Academy of Medical Sciences, Beijing, China
- Hepato-Pancreato-Biliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
- Institute for Organ Transplant and Bionic Medicine, Tsinghua University, Beijing, China
- Key Laboratory of Digital Intelligence Hepatology, Ministry of Education, School of Clinical Medicine, Tsinghua University, Beijing, China
- Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - Jiahong Dong
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Jilin University, Changchun, China
- Research Unit of Precision Hepatobiliary Surgery Paradigm, Chinese Academy of Medical Sciences, Beijing, China
- Hepato-Pancreato-Biliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
- Institute for Organ Transplant and Bionic Medicine, Tsinghua University, Beijing, China
- Key Laboratory of Digital Intelligence Hepatology, Ministry of Education, School of Clinical Medicine, Tsinghua University, Beijing, China
| |
Collapse
|
8
|
Ferrari E, Monti E, Cerutti C, Visone R, Occhetta P, Griffith LG, Rasponi M. A method to generate perfusable physiologic-like vascular channels within a liver-on-chip model. BIOMICROFLUIDICS 2023; 17:064103. [PMID: 38058462 PMCID: PMC10697721 DOI: 10.1063/5.0170606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 11/13/2023] [Indexed: 12/08/2023]
Abstract
The human vasculature is essential in organs and tissues for the transport of nutrients, metabolic waste products, and the maintenance of homeostasis. The integration of vessels in in vitro organs-on-chip may, therefore, improve the similarity to the native organ microenvironment, ensuring proper physiological functions and reducing the gap between experimental research and clinical outcomes. This gap is particularly evident in drug testing and the use of vascularized models may provide more realistic insights into human responses to drugs in the pre-clinical phases of the drug development pipeline. In this context, different vascularized liver models have been developed to recapitulate the architecture of the hepatic sinusoid, exploiting either porous membranes or bioprinting techniques. In this work, we developed a method to generate perfusable vascular channels with a circular cross section within organs-on-chip without any interposing material between the parenchyma and the surrounding environment. Through this technique, vascularized liver sinusoid-on-chip systems with and without the inclusion of the space of Disse were designed and developed. The recapitulation of the Disse layer, therefore, a gap between hepatocytes and endothelial cells physiologically present in the native liver milieu, seems to enhance hepatic functionality (e.g., albumin production) compared to when hepatocytes are in close contact with endothelial cells. These findings pave the way to numerous further uses of microfluidic technologies coupled with vascularized tissue models (e.g., immune system perfusion) as well as the integration within multiorgan-on-chip settings.
Collapse
Affiliation(s)
| | - E. Monti
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, via Camillo Golgi 39, 20134 Milano (MI), Italy
| | - C. Cerutti
- Istituto Europeo di Oncologia, via Adamello 16, 20139 Milano (MI), Italy
| | - R. Visone
- BiomimX Srl, viale Decumano 41, 20157 Milano (MI), Italy
| | | | - L. G. Griffith
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, Massachusetts 02139, USA
| | - M. Rasponi
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, via Camillo Golgi 39, 20134 Milano (MI), Italy
| |
Collapse
|
9
|
Vasudevan A, Majumder N, Sharma I, Kaur I, Sundarrajan S, Venugopal JR, Vijayaraghavan P, Singh N, Ramakrishna S, Ghosh S, M Tripathi D, Kaur S. Liver Extracellular Matrix-Based Nanofiber Scaffolds for the Culture of Primary Hepatocytes and Drug Screening. ACS Biomater Sci Eng 2023; 9:6357-6368. [PMID: 37847169 DOI: 10.1021/acsbiomaterials.3c01216] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2023]
Abstract
Immortalized liver cell lines and primary hepatocytes are currently used as in vitro models for hepatotoxic drug screening. However, a decline in the viability and functionality of hepatocytes with time is an important limitation of these culture models. Advancements in tissue engineering techniques have allowed us to overcome this challenge by designing suitable scaffolds for maintaining viable and functional primary hepatocytes for a longer period of time in culture. In the current study, we fabricated liver-specific nanofiber scaffolds with polylactic acid (PLA) along with a decellularized liver extracellular matrix (LEM) by the electrospinning technique. The fabricated hybrid PLA-LEM scaffolds were more hydrophilic and had better swelling properties than the PLA scaffolds. The hybrid scaffolds had a pore size of 38 ± 8 μm and supported primary rat hepatocyte cultures for 10 days. Increased viability (2-fold increase in the number of live cells) and functionality (5-fold increase in albumin secretion) were observed in primary hepatocytes cultured on the PLA-LEM scaffolds as compared to those on conventional collagen-coated plates on day 10 of culture. A significant increase in CYP1A2 enzyme activity was observed in hepatocytes cultured on PLA-LEM hybrid scaffolds in comparison to those on collagen upon induction with phenobarbital. Drugs like acetaminophen and rifampicin showed the highest toxicity in hepatocytes cultured on hybrid scaffolds. Also, the lethal dose of these drugs in rodents was accurately predicted as 1.6 g/kg and 594 mg/kg, respectively, from the corresponding IC50 values obtained from drug-treated hepatocytes on hybrid scaffolds. Thus, the fabricated liver-specific electrospun scaffolds maintained primary hepatocyte viability and functionality for an extended period in culture and served as an effective ex vivo drug screening platform to predict an accurate in vivo drug-induced hepatotoxicity.
Collapse
Affiliation(s)
- Ashwini Vasudevan
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi 110070, India
- Amity Institute of Biotechnology, Sector-125, Amity University Uttar Pradesh, Noida 201301, India
| | - Nilotpal Majumder
- Department of Textile and Fibre Engineering, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Indu Sharma
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi 110070, India
| | - Impreet Kaur
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi 110070, India
| | - Subramanian Sundarrajan
- Department of Mechanical Engineering, National University of Singapore, Singapore 117581, Singapore
- Department of Prosthodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai 600077, India
| | - Jayarama Reddy Venugopal
- Faculty of Industrial Sciences and Technology, Universiti Malaysia Pahang, Kuantan 26600, Malaysia
| | - Pooja Vijayaraghavan
- Amity Institute of Biotechnology, Sector-125, Amity University Uttar Pradesh, Noida 201301, India
| | - Neetu Singh
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - Seeram Ramakrishna
- Department of Mechanical Engineering, National University of Singapore, Singapore 117581, Singapore
| | - Sourabh Ghosh
- Department of Textile and Fibre Engineering, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Dinesh M Tripathi
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi 110070, India
| | - Savneet Kaur
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi 110070, India
| |
Collapse
|
10
|
Allu I, Sahi AK, Koppadi M, Gundu S, Sionkowska A. Decellularization Techniques for Tissue Engineering: Towards Replicating Native Extracellular Matrix Architecture in Liver Regeneration. J Funct Biomater 2023; 14:518. [PMID: 37888183 PMCID: PMC10607724 DOI: 10.3390/jfb14100518] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/09/2023] [Accepted: 10/10/2023] [Indexed: 10/28/2023] Open
Abstract
The process of tissue regeneration requires the utilization of a scaffold, which serves as a structural framework facilitating cellular adhesion, proliferation, and migration within a physical environment. The primary aim of scaffolds in tissue engineering is to mimic the structural and functional properties of the extracellular matrix (ECM) in the target tissue. The construction of scaffolds that accurately mimic the architecture of the extracellular matrix (ECM) is a challenging task, primarily due to the intricate structural nature and complex composition of the ECM. The technique of decellularization has gained significant attention in the field of tissue regeneration because of its ability to produce natural scaffolds by removing cellular and genetic components from the extracellular matrix (ECM) while preserving its structural integrity. The present study aims to investigate the various decellularization techniques employed for the purpose of isolating the extracellular matrix (ECM) from its native tissue. Additionally, a comprehensive comparison of these methods will be presented, highlighting their respective advantages and disadvantages. The primary objective of this study is to gain a comprehensive understanding of the anatomical and functional features of the native liver, as well as the prevalence and impact of liver diseases. Additionally, this study aims to identify the limitations and difficulties associated with existing therapeutic methods for liver diseases. Furthermore, the study explores the potential of tissue engineering techniques in addressing these challenges and enhancing liver performance. By investigating these aspects, this research field aims to contribute to the advancement of liver disease treatment and management.
Collapse
Affiliation(s)
- Ishita Allu
- Department of Biomedical Engineering, University College of Engineering (UCE), Osmania University, Hyderabad 500007, India; (I.A.); (M.K.)
| | - Ajay Kumar Sahi
- School of Medicine, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA;
| | - Meghana Koppadi
- Department of Biomedical Engineering, University College of Engineering (UCE), Osmania University, Hyderabad 500007, India; (I.A.); (M.K.)
| | - Shravanya Gundu
- Department of Biomedical Engineering, University College of Engineering (UCE), Osmania University, Hyderabad 500007, India; (I.A.); (M.K.)
| | - Alina Sionkowska
- Faculty of Chemistry, Nicolaus Copernicus University in Torun, Jurija Gagarina 11, 87-100 Torun, Poland
- Faculty of Health Sciences, Calisia University, Nowy Świat 4, 62-800 Kalisz, Poland
| |
Collapse
|
11
|
Mir TA, Alzhrani A, Nakamura M, Iwanaga S, Wani SI, Altuhami A, Kazmi S, Arai K, Shamma T, Obeid DA, Assiri AM, Broering DC. Whole Liver Derived Acellular Extracellular Matrix for Bioengineering of Liver Constructs: An Updated Review. Bioengineering (Basel) 2023; 10:1126. [PMID: 37892856 PMCID: PMC10604736 DOI: 10.3390/bioengineering10101126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 09/13/2023] [Accepted: 09/15/2023] [Indexed: 10/29/2023] Open
Abstract
Biomaterial templates play a critical role in establishing and bioinstructing three-dimensional cellular growth, proliferation and spatial morphogenetic processes that culminate in the development of physiologically relevant in vitro liver models. Various natural and synthetic polymeric biomaterials are currently available to construct biomimetic cell culture environments to investigate hepatic cell-matrix interactions, drug response assessment, toxicity, and disease mechanisms. One specific class of natural biomaterials consists of the decellularized liver extracellular matrix (dECM) derived from xenogeneic or allogeneic sources, which is rich in bioconstituents essential for the ultrastructural stability, function, repair, and regeneration of tissues/organs. Considering the significance of the key design blueprints of organ-specific acellular substrates for physiologically active graft reconstruction, herein we showcased the latest updates in the field of liver decellularization-recellularization technologies. Overall, this review highlights the potential of acellular matrix as a promising biomaterial in light of recent advances in the preparation of liver-specific whole organ scaffolds. The review concludes with a discussion of the challenges and future prospects of liver-specific decellularized materials in the direction of translational research.
Collapse
Affiliation(s)
- Tanveer Ahmed Mir
- Laboratory of Tissue/Organ Bioengineering & BioMEMS, Organ Transplant Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia (T.S.)
| | - Alaa Alzhrani
- Laboratory of Tissue/Organ Bioengineering & BioMEMS, Organ Transplant Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia (T.S.)
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21423, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh 11211, Saudi Arabia
| | - Makoto Nakamura
- Division of Biomedical System Engineering, Graduate School of Science and Engineering for Education, University of Toyama, 3190 Gofuku, Toyama 930-8555, Japan; (M.N.); (S.I.)
| | - Shintaroh Iwanaga
- Division of Biomedical System Engineering, Graduate School of Science and Engineering for Education, University of Toyama, 3190 Gofuku, Toyama 930-8555, Japan; (M.N.); (S.I.)
| | - Shadil Ibrahim Wani
- Division of Biomedical System Engineering, Graduate School of Science and Engineering for Education, University of Toyama, 3190 Gofuku, Toyama 930-8555, Japan; (M.N.); (S.I.)
| | - Abdullah Altuhami
- Laboratory of Tissue/Organ Bioengineering & BioMEMS, Organ Transplant Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia (T.S.)
| | - Shadab Kazmi
- Laboratory of Tissue/Organ Bioengineering & BioMEMS, Organ Transplant Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia (T.S.)
- Department of Child Health, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Kenchi Arai
- Department of Clinical Biomaterial Applied Science, Faculty of Medicine, University of Toyama, Toyama 930-0194, Japan
| | - Talal Shamma
- Laboratory of Tissue/Organ Bioengineering & BioMEMS, Organ Transplant Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia (T.S.)
| | - Dalia A. Obeid
- Laboratory of Tissue/Organ Bioengineering & BioMEMS, Organ Transplant Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia (T.S.)
| | - Abdullah M. Assiri
- Laboratory of Tissue/Organ Bioengineering & BioMEMS, Organ Transplant Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia (T.S.)
- College of Medicine, Alfaisal University, Riyadh 11211, Saudi Arabia
| | - Dieter C. Broering
- Laboratory of Tissue/Organ Bioengineering & BioMEMS, Organ Transplant Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia (T.S.)
- College of Medicine, Alfaisal University, Riyadh 11211, Saudi Arabia
| |
Collapse
|
12
|
Patel N, Davis Z, Hofmann C, Vlasak J, Loughney JW, DePhillips P, Mukherjee M. Development and Characterization of an In Vitro Cell-Based Assay to Predict Potency of mRNA-LNP-Based Vaccines. Vaccines (Basel) 2023; 11:1224. [PMID: 37515040 PMCID: PMC10383996 DOI: 10.3390/vaccines11071224] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/03/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
Messenger RNA (mRNA) vaccines have emerged as a flexible platform for vaccine development. The evolution of lipid nanoparticles as effective delivery vehicles for modified mRNA encoding vaccine antigens was demonstrated by the response to the COVID-19 pandemic. The ability to rapidly develop effective SARS-CoV-2 vaccines from the spike protein genome, and to then manufacture multibillions of doses per year was an extraordinary achievement and a vaccine milestone. Further development and application of this platform for additional pathogens is clearly of interest. This comes with the associated need for new analytical tools that can accurately predict the performance of these mRNA vaccine candidates and tie them to an immune response expected in humans. Described here is the development and characterization of an imaging based in vitro assay able to quantitate transgene protein expression efficiency, with utility to measure lipid nanoparticles (LNP)-encapsulated mRNA vaccine potency, efficacy, and stability. Multiple biologically relevant adherent cell lines were screened to identify a suitable cell substrate capable of providing a wide dose-response curve and dynamic range. Biologically relevant assay attributes were examined and optimized, including cell monolayer morphology, antigen expression kinetics, and assay sensitivity to LNP properties, such as polyethylene glycol-lipid (or PEG-lipid) composition, mRNA mass, and LNP size. Collectively, this study presents a strategy to quickly optimize and develop a robust cell-based potency assay for the development of future mRNA-based vaccines.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Malini Mukherjee
- Analytical Research & Development, Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA 19486, USA; (N.P.); (Z.D.); (C.H.); (J.V.); (J.W.L.); (P.D.)
| |
Collapse
|
13
|
Frtús A, Smolková B, Uzhytchak M, Lunova M, Jirsa M, Petrenko Y, Dejneka A, Lunov O. Mechanical Regulation of Mitochondrial Dynamics and Function in a 3D-Engineered Liver Tumor Microenvironment. ACS Biomater Sci Eng 2023; 9:2408-2425. [PMID: 37001010 PMCID: PMC10170482 DOI: 10.1021/acsbiomaterials.2c01518] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
Abstract
It has become evident that physical stimuli of the cellular microenvironment transmit mechanical cues regulating key cellular functions, such as proliferation, migration, and malignant transformation. Accumulating evidence suggests that tumor cells face variable mechanical stimuli that may induce metabolic rewiring of tumor cells. However, the knowledge of how tumor cells adapt metabolism to external mechanical cues is still limited. We therefore designed soft 3D collagen scaffolds mimicking a pathological mechanical environment to decipher how liver tumor cells would adapt their metabolic activity to physical stimuli of the cellular microenvironment. Here, we report that the soft 3D microenvironment upregulates the glycolysis of HepG2 and Alexander cells. Both cell lines adapt their mitochondrial activity and function under growth in the soft 3D microenvironment. Cells grown in the soft 3D microenvironment exhibit marked mitochondrial depolarization, downregulation of mitochondrially encoded cytochrome c oxidase I, and slow proliferation rate in comparison with stiff monolayer cultures. Our data reveal the coupling of liver tumor glycolysis to mechanical cues. It is proposed here that soft 3D collagen scaffolds can serve as a useful model for future studies of mechanically regulated cellular functions of various liver (potentially other tissues as well) tumor cells.
Collapse
Affiliation(s)
- Adam Frtús
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, Prague 18221, Czech Republic
| | - Barbora Smolková
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, Prague 18221, Czech Republic
| | - Mariia Uzhytchak
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, Prague 18221, Czech Republic
| | - Mariia Lunova
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, Prague 18221, Czech Republic
- Institute for Clinical & Experimental Medicine (IKEM), Prague 14021, Czech Republic
| | - Milan Jirsa
- Institute for Clinical & Experimental Medicine (IKEM), Prague 14021, Czech Republic
| | - Yuriy Petrenko
- Department of Neuroregeneration, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague 14220, Czech Republic
| | - Alexandr Dejneka
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, Prague 18221, Czech Republic
| | - Oleg Lunov
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, Prague 18221, Czech Republic
| |
Collapse
|
14
|
Seidemann L, Prinz S, Scherbel JC, Götz C, Seehofer D, Damm G. Optimization of extracellular matrix for primary human hepatocyte cultures using mixed collagen-Matrigel matrices. EXCLI JOURNAL 2023; 22:12-34. [PMID: 36660192 PMCID: PMC9837384 DOI: 10.17179/excli2022-5459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 11/17/2022] [Indexed: 01/21/2023]
Abstract
Loss of differentiation of primary human hepatocytes (PHHs) ex vivo is a known problem of in vitro liver models. Culture optimizations using collagen type I and Matrigel reduce the dedifferentiation process but are not able to prevent it. While neither of these extracellular matrices (ECMs) on their own correspond to the authentic hepatic ECM, a combination of them could more closely resemble the in vivo situation. Our study aimed to systematically analyze the influence of mixed matrices composed of collagen type I and Matrigel on the maintenance and reestablishment of hepatic functions. Therefore, PHHs were cultured on mixed collagen-Matrigel matrices in monolayer and sandwich cultures and viability, metabolic capacity, differentiation markers, cellular arrangement and the cells' ability to repolarize and form functional bile canaliculi were assessed by reverse transcription quantitative real-time polymerase chain reaction (RT-qPCR), functional assays and immunofluorescence microscopy. Our results show that mixed matrices were superior to pure matrices in maintaining metabolic capacity and hepatic differentiation. In contrast, Matrigel supplementation can impair the development of a proper hepatocytic polarization. Our systematic study helps to compose an optimized ECM to maintain and reestablish hepatic differentiation on cellular and multicellular levels in human liver models.
Collapse
Affiliation(s)
- Lena Seidemann
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, Liebigstr. 20, 04103 Leipzig, Germany,Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, Philipp-Rosenthal-Str. 55, 04103 Leipzig, Germany
| | - Sarah Prinz
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, Liebigstr. 20, 04103 Leipzig, Germany,Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, Philipp-Rosenthal-Str. 55, 04103 Leipzig, Germany
| | - Jan-Constantin Scherbel
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, Liebigstr. 20, 04103 Leipzig, Germany,Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, Philipp-Rosenthal-Str. 55, 04103 Leipzig, Germany
| | - Christina Götz
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, Liebigstr. 20, 04103 Leipzig, Germany,Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, Philipp-Rosenthal-Str. 55, 04103 Leipzig, Germany
| | - Daniel Seehofer
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, Liebigstr. 20, 04103 Leipzig, Germany,Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, Philipp-Rosenthal-Str. 55, 04103 Leipzig, Germany
| | - Georg Damm
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, Liebigstr. 20, 04103 Leipzig, Germany,Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, Philipp-Rosenthal-Str. 55, 04103 Leipzig, Germany,*To whom correspondence should be addressed: Georg Damm, Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, Liebigstr. 20, 04103 Leipzig, Germany; Tel.: +49-341-9739656, E-mail:
| |
Collapse
|
15
|
Lv W, Zhou H, Aazmi A, Yu M, Xu X, Yang H, Huang YYS, Ma L. Constructing biomimetic liver models through biomaterials and vasculature engineering. Regen Biomater 2022; 9:rbac079. [PMID: 36338176 PMCID: PMC9629974 DOI: 10.1093/rb/rbac079] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 09/19/2022] [Accepted: 10/08/2022] [Indexed: 04/04/2024] Open
Abstract
The occurrence of various liver diseases can lead to organ failure of the liver, which is one of the leading causes of mortality worldwide. Liver tissue engineering see the potential for replacing liver transplantation and drug toxicity studies facing donor shortages. The basic elements in liver tissue engineering are cells and biomaterials. Both mature hepatocytes and differentiated stem cells can be used as the main source of cells to construct spheroids and organoids, achieving improved cell function. To mimic the extracellular matrix (ECM) environment, biomaterials need to be biocompatible and bioactive, which also help support cell proliferation and differentiation and allow ECM deposition and vascularized structures formation. In addition, advanced manufacturing approaches are required to construct the extracellular microenvironment, and it has been proved that the structured three-dimensional culture system can help to improve the activity of hepatocytes and the characterization of specific proteins. In summary, we review biomaterials for liver tissue engineering, including natural hydrogels and synthetic polymers, and advanced processing techniques for building vascularized microenvironments, including bioassembly, bioprinting and microfluidic methods. We then summarize the application fields including transplant and regeneration, disease models and drug cytotoxicity analysis. In the end, we put the challenges and prospects of vascularized liver tissue engineering.
Collapse
Affiliation(s)
- Weikang Lv
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou 310058, China
- School of Mechanical Engineering, Zhejiang University, Hangzhou 310058, China
| | - Hongzhao Zhou
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou 310058, China
- School of Mechanical Engineering, Zhejiang University, Hangzhou 310058, China
| | - Abdellah Aazmi
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou 310058, China
- School of Mechanical Engineering, Zhejiang University, Hangzhou 310058, China
| | - Mengfei Yu
- The Affiliated Stomatologic Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Xiaobin Xu
- School of Materials Science and Engineering, Tongji University, Shanghai 201804, China
| | - Huayong Yang
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou 310058, China
- School of Mechanical Engineering, Zhejiang University, Hangzhou 310058, China
| | | | - Liang Ma
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou 310058, China
- School of Mechanical Engineering, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
16
|
Liu M, Xiang Y, Yang Y, Long X, Xiao Z, Nan Y, Jiang Y, Qiu Y, Huang Q, Ai K. State-of-the-art advancements in Liver-on-a-chip (LOC): Integrated biosensors for LOC. Biosens Bioelectron 2022; 218:114758. [DOI: 10.1016/j.bios.2022.114758] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 09/24/2022] [Accepted: 09/24/2022] [Indexed: 12/12/2022]
|
17
|
Abstract
Liver regeneration is a well-orchestrated process that is typically studied in animal models. Although previous animal studies have offered many insights into liver regeneration, human biology is less well understood. To this end, we developed a three-dimensional (3D) platform called structurally vascularized hepatic ensembles for analyzing regeneration (SHEAR) to model multiple aspects of human liver regeneration. SHEAR enables control over hemodynamic alterations to mimic those that occur during liver injury and regeneration and supports the administration of biochemical inputs such as cytokines and paracrine interactions with endothelial cells. We found that exposing the endothelium-lined channel to fluid flow led to increased secretion of regeneration-associated factors. Stimulation with relevant cytokines not only amplified the secretory response, but also induced cell-cycle entry of primary human hepatocytes (PHHs) embedded within the device. Further, we identified endothelial-derived mediators that are sufficient to initiate proliferation of PHHs in this context. Collectively, the data presented here underscore the importance of multicellular models that can recapitulate high-level tissue functions and demonstrate that the SHEAR device can be used to discover and validate conditions that promote human liver regeneration.
Collapse
|
18
|
Setiawati A, Jeong S, Brillian AI, Lee SH, Shim JG, Jung KH, Shin K. Fabrication of a Tailored, Hybrid Extracellular Matrix Composite. Macromol Biosci 2022; 22:e2200106. [PMID: 35765216 DOI: 10.1002/mabi.202200106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 05/24/2022] [Indexed: 11/10/2022]
Abstract
The extracellular matrix (ECM) is a network of connective fibers that supports cells living in their surroundings. Native ECM, generated by the secretory products of each tissue's resident cells, has a unique architecture with different protein composition depending on the tissue. Therefore, it is very difficult to artificially design in vivo architecture in tissue engineering. In this study, we fabricated a hybrid ECM scaffold from the basic structure of fibroblast-derived cellular ECMs by adding major ECM components of fibronectin (FN) and collagen (COL I) externally. It was confirmed that while maintaining the basic structure of the native ECM, major protein components can be regulated. Then, decellularization was performed to prepare hybrid ECM scaffolds with various protein compositions and we demonstrated that a liver-mimicking fibronectin (FN)-rich hybrid ECM promoted successful settling of H4IIE rat hepatoma cells. We believe that our method holds promise for the fabrication of scaffolds that provide a tailored cellular microenvironment for specific organs and serve as novel pathways for the replacement or regeneration of specific organ tissues. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Agustina Setiawati
- Department of Chemistry and Institute of Biological Interfaces, 35-Baekbeom-ro, Sogang University, Mapo-gu, Seoul, 04107, Republic of Korea.,Department of Life Science, 35-Baekbeom-ro, Sogang University, Mapo-gu, Seoul, 04107, Republic of Korea.,Faculty of Pharmacy, Paingan, Maguwoharjo, Depok, Sanata Dharma University, Sleman, Yogyakarta, 55284, Indonesia
| | - Sungwoo Jeong
- Department of Chemistry and Institute of Biological Interfaces, 35-Baekbeom-ro, Sogang University, Mapo-gu, Seoul, 04107, Republic of Korea
| | - Albertus Ivan Brillian
- Department of Chemistry and Institute of Biological Interfaces, 35-Baekbeom-ro, Sogang University, Mapo-gu, Seoul, 04107, Republic of Korea
| | - Sang Ho Lee
- Department of Chemistry and Institute of Biological Interfaces, 35-Baekbeom-ro, Sogang University, Mapo-gu, Seoul, 04107, Republic of Korea
| | - Jin-Gon Shim
- Department of Life Science, 35-Baekbeom-ro, Sogang University, Mapo-gu, Seoul, 04107, Republic of Korea
| | - Kwang-Hwan Jung
- Department of Life Science, 35-Baekbeom-ro, Sogang University, Mapo-gu, Seoul, 04107, Republic of Korea
| | - Kwanwoo Shin
- Department of Chemistry and Institute of Biological Interfaces, 35-Baekbeom-ro, Sogang University, Mapo-gu, Seoul, 04107, Republic of Korea
| |
Collapse
|
19
|
Guo L, Zhu Z, Gao C, Chen K, Lu S, Yan H, Liu W, Wang M, Ding Y, Huang L, Wang X. Development of Biomimetic Hepatic Lobule-Like Constructs on Silk-Collagen Composite Scaffolds for Liver Tissue Engineering. Front Bioeng Biotechnol 2022; 10:940634. [PMID: 35814001 PMCID: PMC9260023 DOI: 10.3389/fbioe.2022.940634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 05/31/2022] [Indexed: 11/13/2022] Open
Abstract
Constructing an engineered hepatic lobule-mimetic model is challenging owing to complicated lobular architecture and crucial hepatic functionality. Our previous study has demonstrated the feasibility of using silk fibroin (SF) scaffolds as functional templates for engineering hepatic lobule-like constructs. But the unsatisfactory chemical and physical performances of the SF-only scaffold and the inherent defect in the functional activity of the carcinoma-derived seeding cells remain to be addressed to satisfy the downstream application demand. In this study, SF-collagen I (SFC) composite scaffolds with improved physical and chemical properties were fabricated, and their utilization for bioengineering a more hepatic lobule-like construct was explored using the immortalized human hepatocyte-derived liver progenitor-like cells (iHepLPCs) and endothelial cells incorporated in the dynamic culture system. The SFC scaffolds prepared through the directional lyophilization process showed radially aligned porous structures with increased swelling ratio and porosity, ameliorative mechanical stiffness that resembled the normal liver matrix more closely, and improved biocompatibility. The iHepLPCs displayed a hepatic plate-like distribution and differentiated into matured hepatocytes with improved hepatic function in vitro and in vivo. Moreover, hepatocyte–endothelial cell interphase arrangement was generated in the co-culture compartment with improved polarity, bile capillary formation, and enhanced liver functions compared with the monocultures. Thus, a more biomimetic hepatic lobule-like model was established and could provide a valuable and robust platform for various applications, including bioartificial liver and drug screening.
Collapse
Affiliation(s)
- Lina Guo
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Ziqing Zhu
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou, China
| | - Chuanzhou Gao
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Kaiwen Chen
- School of Bioengineering, State Key Laboratory of Fine Chemistry, Dalian University of Technology, Dalian, China
| | - Shenzhou Lu
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou, China
| | - Hexin Yan
- Department of Anesthesiology and Critical Care Medicine, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Engineering Research Center of Peri-operative Organ Support and Function Preservation, Shanghai, China
| | - Wenming Liu
- Department of Anesthesiology and Critical Care Medicine, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Engineering Research Center of Peri-operative Organ Support and Function Preservation, Shanghai, China
| | - Mingqi Wang
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Yanfang Ding
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Lin Huang
- College of Basic Medical Science, Dalian Medical University, Dalian, China
- *Correspondence: Lin Huang, ; Xiuli Wang,
| | - Xiuli Wang
- College of Basic Medical Science, Dalian Medical University, Dalian, China
- General Surgery Center, Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- *Correspondence: Lin Huang, ; Xiuli Wang,
| |
Collapse
|
20
|
Karlsson S, Nyström H. The extracellular matrix in colorectal cancer and its metastatic settling – alterations and biological implications. Crit Rev Oncol Hematol 2022; 175:103712. [DOI: 10.1016/j.critrevonc.2022.103712] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 05/05/2022] [Accepted: 05/11/2022] [Indexed: 12/12/2022] Open
|
21
|
Morales-Guerrero NA, Varela-Echavarría A, Lozano Flores C, Vázquez-Cuevas FG, Velázquez-Miranda E, Reyes-López JV, García-Solís P, Solís-S JC, Hernández-Montiel HL. A new strategy for the decellularization of whole organs by hydrostatic pressure. Biotechnol Prog 2022; 38:e3248. [PMID: 35201677 DOI: 10.1002/btpr.3248] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 02/21/2022] [Accepted: 02/23/2022] [Indexed: 11/06/2022]
Abstract
Tissue engineering has been able to develop novel decellularization-recellularization techniques, which facilitates the research for the generation of functional organs. This is based in the initial obtention of the organ's extracellular matrix (ECM). Therefore, any improvement in the decellularization process would have a positive impact in the results of the recellularization process. Nevertheless, commonly the methods and equipment employed for this process are expensive and thus limit the access of this technique to various research groups globally. AIM To develop a decellularization technique with the exclusive use of hydrostatic pressure of detergent solutions, to have an easily accessible and low-cost technique that meets the basic requirements of acellularity and functionality of the ECM. METHODS This experimental study was performed in 10 male Wistar rats, obtaining the liver to carry out serial washes, with 1, 2 and 3% Triton X-100 solutions and 0.1% SDS. The washes were performed by using a Gravity Perfusion System (GPS), which assured us a continuous hydrostatic pressure of 7.5 mmHg. The obtained ECM was processed using stains and immunostaining to determine the residual cell content and preservation of its components. RESULTS The staining showed a removal of cellular and nuclear components of approximately 97% of the acellular ECM, with an adequate three-dimensional pattern of collagen and proteoglycans. Furthermore, the acellular ECM allowed the viability of a primary hepatocyte culture. CONCLUSIONS The use of the GPS decellularization technique allowed us to obtain an acellular and functional ECM, drastically reducing experimentation costs. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Nelly A Morales-Guerrero
- Department of Biomedical Research, School of Medicine, Autonomous University of Queretaro, Qro., Mexico
| | | | - Carlos Lozano Flores
- Institute of Neurobiology, National Autonomous University of Mexico, Qro., Mexico
| | | | | | - Julián V Reyes-López
- Laboratory of Neurobiology and Cellular Bioengineering, Neurodiagnostic and Rehabilitation Unit "Dr. Moisés López González ", Faculty of Natural Sciences, Autonomous University of Querétaro
| | - Pablo García-Solís
- Department of Biomedical Research, School of Medicine, Autonomous University of Queretaro, Qro., Mexico
| | - Juan Carlos Solís-S
- Department of Biomedical Research, School of Medicine, Autonomous University of Queretaro, Qro., Mexico
| | - Hebert Luis Hernández-Montiel
- Laboratory of Neurobiology and Cellular Bioengineering, Neurodiagnostic and Rehabilitation Unit "Dr. Moisés López González ", Faculty of Natural Sciences, Autonomous University of Querétaro
| |
Collapse
|
22
|
Dias ML, Paranhos BA, Goldenberg RCDS. Liver scaffolds obtained by decellularization: A transplant perspective in liver bioengineering. J Tissue Eng 2022; 13:20417314221105305. [PMID: 35756167 PMCID: PMC9218891 DOI: 10.1177/20417314221105305] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 05/19/2022] [Indexed: 11/15/2022] Open
Abstract
Liver transplantation is the only definitive treatment for many diseases that affect this organ, however, its quantity and viability are reduced. The study of liver scaffolds based on an extracellular matrix is a tissue bioengineering strategy with great application in regenerative medicine. Collectively, recent studies suggest that liver scaffold transplantation may assist in reestablishing hepatic function in preclinical diseased animals, which represents a great potential for application as a treatment for patients with liver disease in the future. This review focuses on useful strategies to promote liver scaffold transplantation and the main open questions about this context. We outline the current knowledge about ex vivo bioengineered liver transplantation, including the surgical techniques, recipient survival time, scaffold preparation before transplantation, and liver disease models. We also highlight the current limitations and future directions regarding in vivo bioengineering techniques.
Collapse
Affiliation(s)
- Marlon Lemos Dias
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil.,Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa - INCT - REGENERA, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Bruno Andrade Paranhos
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil.,Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa - INCT - REGENERA, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Regina Coeli Dos Santos Goldenberg
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil.,Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa - INCT - REGENERA, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| |
Collapse
|
23
|
Patarashvili L, Gvidiani S, Azmaipharashvili E, Tsomaia K, Sareli M, Kordzaia D, Chanukvadze I. Porta-caval fibrous connections - the lesser-known structure of intrahepatic connective-tissue framework: A unified view of liver extracellular matrix. World J Hepatol 2021; 13:1484-1493. [PMID: 34904025 PMCID: PMC8637665 DOI: 10.4254/wjh.v13.i11.1484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 05/17/2021] [Accepted: 08/30/2021] [Indexed: 02/06/2023] Open
Abstract
Knowledge about the connective-tissue framework of the liver is not systematized, the terminology is inconsistent and some perspectives on the construction of the hepatic matrix components are contradictory. In addition, until the last two decades of the 20th century, the connective-tissue sheaths of the portal tracts and the hepatic veins were considered to be independent from each other in the liver and that they do not make contact with each other. The results of the research carried out by Professor Shalva Toidze and his colleagues started in the 1970s in the Department of Operative Surgery and Topographic Anatomy at the Tbilisi State Medical Institute have changed this perception. In particular, Chanukvadze I showed that in some regions where they intersect with each other, the connective tissue sheaths of the large portal complexes and hepatic veins fuse. The areas of such fusion are called porta-caval fibrous connections (PCFCs). This opinion review aims to promote a systematic understanding of the hepatic connective-tissue skeleton and to demonstrate the hitherto underappreciated PCFC as a genuine structure with high biological and clinical significance. The components of the liver connective-tissue framework - the capsules, plates, sheaths, covers - are described, and their intercommunication is discussed. The analysis of the essence of the PCFC and a description of its various forms are provided. It is also mentioned that analogs of different forms of PCFC are found in different mammals.
Collapse
Affiliation(s)
- Leila Patarashvili
- Department of Clinical Anatomy and Operative Surgery, Ivane Javakhishvili Tbilisi State University, Tbilisi 0159, Georgia
| | - Salome Gvidiani
- Faculty of Medicine, Ivane Javakhishvili Tbilisi State University, Tbilisi 0159, Georgia
| | - Elza Azmaipharashvili
- Faculty of Medicine, Ivane Javakhishvili Tbilisi State University, Tbilisi 0159, Georgia
| | - Keti Tsomaia
- Clinical Anatomy and Experimental Modeling, Institute of Morphology, Ivane Javakhishvili Tbilisi State University, Tbilisi 0159, Georgia
| | - Marom Sareli
- Department of Surgical Oncology (Surgery C), Chaim Sheba Medical Center at HaShomer, Ramat Gan, Tel Aviv 52621, Israel
| | - Dimitri Kordzaia
- Department of Clinical Anatomy and Operative Surgery, Ivane Javakhishvili Tbilisi State University, Tbilisi 0159, Georgia.
| | - Ilia Chanukvadze
- Faculty of Medicine, Tbilisi State Medical University, Tbilsi 0177, Georgia
| |
Collapse
|
24
|
Bobrova MM, Safonova LA, Efimov AE, Iljinsky IM, Agapova OI, Agapov II. Relation between micro- and nanostructure features and biological properties of the decellularized rat liver. Biomed Mater 2021; 16. [PMID: 34100773 DOI: 10.1088/1748-605x/ac058b] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 05/26/2021] [Indexed: 12/12/2022]
Abstract
Organ decellularization is one of the promising technologies of regenerative medicine, which allows obtaining cell-free extracellular matrix (ECM), which provide preservation of the composition, architecture, vascular network and biological activity of the ECM. The method of decellularization opens up wide prospects for its practical application not only in the field of creating full-scale bioengineered structures, but also in the manufacture of vessels, microcarriers, hydrogels, and coatings. The main goal of our work was the investigation of structure and biological properties of lyophilized decellularized Wistar rat liver fragments (LDLFs), as well as we assessed the regenerative potential of the obtained ECM. We obtained decellularized liver of a Wistar rat, the vascular network and the main components of the ECM of tissue were preserved. H&E staining of histological sections confirmed the removal of cells. DNA content of ECM is equal to 0.7% of native tissue DNA content. Utilizing scanning probe nanotomogrphy method, we showed sinuous, rough topography and highly nanoporous structure of ECM, which provide high level of mouse 3T3 fibroblast and Hep-G2cells biocompatibility. Obtained LDLF had a high regenerative potential, which we studied in an experimental model of a full-thickness rat skin wound healing: we observed the acceleration of wound healing by 2.2 times in comparison with the control.
Collapse
Affiliation(s)
- Maria M Bobrova
- Laboratory of Bionanotechnologies, Academician V.I. Shumakov National Medical Research Center of Transplantology and Artificial Organs, Ministry of Health of the Russian Federation, 123182 Moscow, Russia
| | - Liubov A Safonova
- Laboratory of Bionanotechnologies, Academician V.I. Shumakov National Medical Research Center of Transplantology and Artificial Organs, Ministry of Health of the Russian Federation, 123182 Moscow, Russia
| | - Anton E Efimov
- Laboratory of Bionanotechnologies, Academician V.I. Shumakov National Medical Research Center of Transplantology and Artificial Organs, Ministry of Health of the Russian Federation, 123182 Moscow, Russia.,SNOTRA LLC., 121205 Moscow, Russia
| | - Igor M Iljinsky
- Laboratory of Bionanotechnologies, Academician V.I. Shumakov National Medical Research Center of Transplantology and Artificial Organs, Ministry of Health of the Russian Federation, 123182 Moscow, Russia
| | - Olga I Agapova
- Laboratory of Bionanotechnologies, Academician V.I. Shumakov National Medical Research Center of Transplantology and Artificial Organs, Ministry of Health of the Russian Federation, 123182 Moscow, Russia
| | - Igor I Agapov
- Laboratory of Bionanotechnologies, Academician V.I. Shumakov National Medical Research Center of Transplantology and Artificial Organs, Ministry of Health of the Russian Federation, 123182 Moscow, Russia
| |
Collapse
|
25
|
Marino GE, Weeraratna AT. A glitch in the matrix: Age-dependent changes in the extracellular matrix facilitate common sites of metastasis. AGING AND CANCER 2020; 1:19-29. [PMID: 35694033 PMCID: PMC9187055 DOI: 10.1002/aac2.12013] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 09/07/2020] [Indexed: 12/15/2022]
Abstract
People over 55 years old represent the majority of cancer patients and suffer from increased metastatic burden compared to the younger patient population. As the aging population increases globally, it is prudent to understand how the intrinsic aging process contributes to cancer progression. As we age, we incur aberrant changes in the extracellular matrix (ECM) of our organs, which contribute to numerous pathologies, including cancer. Notably, the lung, liver, and bone represent the most common sites of distal metastasis for all cancer types. In this review, we describe how age-dependent changes in the ECM of these organs influence cancer progression. Further, we outline how these alterations prime the premetastatic niche and why these may help explain the disparity in outcome for older cancer patients.
Collapse
Affiliation(s)
- Gloria E. Marino
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
- Department of Oncology, Sidney Kimmel Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Ashani T. Weeraratna
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
- Department of Oncology, Sidney Kimmel Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| |
Collapse
|
26
|
Frtús A, Smolková B, Uzhytchak M, Lunova M, Jirsa M, Hof M, Jurkiewicz P, Lozinsky VI, Wolfová L, Petrenko Y, Kubinová Š, Dejneka A, Lunov O. Hepatic Tumor Cell Morphology Plasticity under Physical Constraints in 3D Cultures Driven by YAP-mTOR Axis. Pharmaceuticals (Basel) 2020; 13:ph13120430. [PMID: 33260691 PMCID: PMC7759829 DOI: 10.3390/ph13120430] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/23/2020] [Accepted: 11/26/2020] [Indexed: 02/06/2023] Open
Abstract
Recent studies undoubtedly show that the mammalian target of rapamycin (mTOR) and the Hippo–Yes-associated protein 1 (YAP) pathways are important mediators of mechanical cues. The crosstalk between these pathways as well as de-regulation of their signaling has been implicated in multiple tumor types, including liver tumors. Additionally, physical cues from 3D microenvironments have been identified to alter gene expression and differentiation of different cell lineages. However, it remains incompletely understood how physical constraints originated in 3D cultures affect cell plasticity and what the key mediators are of such process. In this work, we use collagen scaffolds as a model of a soft 3D microenvironment to alter cellular size and study the mechanotransduction that regulates that process. We show that the YAP-mTOR axis is a downstream effector of 3D cellular culture-driven mechanotransduction. Indeed, we found that cell mechanics, dictated by the physical constraints of 3D collagen scaffolds, profoundly affect cellular proliferation in a YAP–mTOR-mediated manner. Functionally, the YAP–mTOR connection is key to mediate cell plasticity in hepatic tumor cell lines. These findings expand the role of YAP–mTOR-driven mechanotransduction to the control hepatic tumor cellular responses under physical constraints in 3D cultures. We suggest a tentative mechanism, which coordinates signaling rewiring with cytoplasmic restructuring during cell growth in 3D microenvironments.
Collapse
Affiliation(s)
- Adam Frtús
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic; (A.F.); (B.S.); (M.U.); (M.L.); (Š.K.)
| | - Barbora Smolková
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic; (A.F.); (B.S.); (M.U.); (M.L.); (Š.K.)
| | - Mariia Uzhytchak
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic; (A.F.); (B.S.); (M.U.); (M.L.); (Š.K.)
| | - Mariia Lunova
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic; (A.F.); (B.S.); (M.U.); (M.L.); (Š.K.)
- Institute for Clinical & Experimental Medicine (IKEM), 14021 Prague, Czech Republic;
| | - Milan Jirsa
- Institute for Clinical & Experimental Medicine (IKEM), 14021 Prague, Czech Republic;
| | - Martin Hof
- J. Heyrovský Institute of Physical Chemistry of the Czech Academy of Sciences, 18223 Prague, Czech Republic; (M.H.); (P.J.)
| | - Piotr Jurkiewicz
- J. Heyrovský Institute of Physical Chemistry of the Czech Academy of Sciences, 18223 Prague, Czech Republic; (M.H.); (P.J.)
| | - Vladimir I. Lozinsky
- A.N. Nesmeyanov Institute of Organoelement Compounds, Russian Academy of Sciences, Vavilov Street, 28, 119991 Moscow, Russia;
| | - Lucie Wolfová
- Department of Biomaterials and Biophysical Methods, Institute of Experimental Medicine of the Czech Academy of Sciences, 14220 Prague, Czech Republic; (L.W.); (Y.P.)
- Department of Tissue Engineering, Contipro a.s., 56102 Dolni Dobrouc, Czech Republic
| | - Yuriy Petrenko
- Department of Biomaterials and Biophysical Methods, Institute of Experimental Medicine of the Czech Academy of Sciences, 14220 Prague, Czech Republic; (L.W.); (Y.P.)
| | - Šárka Kubinová
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic; (A.F.); (B.S.); (M.U.); (M.L.); (Š.K.)
- Department of Biomaterials and Biophysical Methods, Institute of Experimental Medicine of the Czech Academy of Sciences, 14220 Prague, Czech Republic; (L.W.); (Y.P.)
| | - Alexandr Dejneka
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic; (A.F.); (B.S.); (M.U.); (M.L.); (Š.K.)
- Correspondence: (A.D.); (O.L.); Tel.: +420-2660-52141 (A.D.); +420-2660-52131 (O.L.)
| | - Oleg Lunov
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic; (A.F.); (B.S.); (M.U.); (M.L.); (Š.K.)
- Correspondence: (A.D.); (O.L.); Tel.: +420-2660-52141 (A.D.); +420-2660-52131 (O.L.)
| |
Collapse
|
27
|
Meurer SK, Karsdal MA, Weiskirchen R. Advances in the clinical use of collagen as biomarker of liver fibrosis. Expert Rev Mol Diagn 2020; 20:947-969. [PMID: 32865433 DOI: 10.1080/14737159.2020.1814746] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Hepatic fibrosis is the excessive synthesis and deposition of extracellular matrix including collagen in the tissue. Chronic liver insult leads to progressive parenchymal damage, portal hypertension, and cirrhosis. Determination of hepatic collagen by invasive liver biopsy is the gold standard to estimate severity and stage of fibrosis. However, this procedure is associated with pain, carries the risk of infection and bleeding, and is afflicted with a high degree of sampling error. Therefore, there is urgent need for serological collagen-derived markers to assess collagen synthesis/turnover. AREAS COVERED Biochemical properties of collagens, cellular sources of hepatic collagen synthesis, and regulatory aspects in collagen expression. Markers are discussed suitable to estimate hepatic collagen synthesis and/or turnover. Discussed studies were identified through a PubMed search done in May 2020 and the authors' topic knowledge. EXPERT OPINION Hepatic fibrosis is mainly characterized by accumulation of collagen-rich scar tissue. Although traditionally performed liver biopsy is still standard in estimating hepatic fibrosis, there is evidence that noninvasive diagnostic scores and collagen-derived neo-epitopes provide clinical useful information. These noninvasive tests are less expensive than liver biopsy, better tolerated, safer, and more acceptable to patients. Therefore, these tests will lead to dramatic changes in diagnosis.
Collapse
Affiliation(s)
- Steffen K Meurer
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen , Aachen, Germany
| | - Morten A Karsdal
- Nordic Bioscience, Fibrosis Biomarkers and Research , Herlev, Denmark
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen , Aachen, Germany
| |
Collapse
|
28
|
Rada M, Lazaris A, Kapelanski-Lamoureux A, Mayer TZ, Metrakos P. Tumor microenvironment conditions that favor vessel co-option in colorectal cancer liver metastases: A theoretical model. Semin Cancer Biol 2020; 71:52-64. [PMID: 32920126 DOI: 10.1016/j.semcancer.2020.09.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/03/2020] [Accepted: 09/03/2020] [Indexed: 02/07/2023]
Abstract
Vessel co-option is an alternative strategy by which tumour cells vascularize and gain access to nutrients to support tumour growth, survival and metastasis. In vessel co-option, the cancer cells move towards the pre-existing vasculature and hijack them. Vessel co-option is adopted by a wide range of human tumours including colorectal cancer liver metastases (CRCLM) and is responsible for the effectiveness of treatment in CRCLM. Furthermore, vessel co-option is an intrinsic feature and an acquired mechanism of resistance to anti-angiogenic treatment. In this review, we describe the microenvironment, the molecular players, discovered thus far of co-opting CRCLM lesions and propose a theoretical model. We also highlight key unanswered questions that are critical to improving our understanding of CRCLM vessel co-option and for the development of effective approaches for the treatment of co-opting tumours.
Collapse
Affiliation(s)
- Miran Rada
- Cancer Research Program, McGill University Health Centre Research Institute, Montreal, Quebec, H4A3J1, Canada
| | - Anthoula Lazaris
- Cancer Research Program, McGill University Health Centre Research Institute, Montreal, Quebec, H4A3J1, Canada
| | - Audrey Kapelanski-Lamoureux
- Cancer Research Program, McGill University Health Centre Research Institute, Montreal, Quebec, H4A3J1, Canada
| | - Thomas Z Mayer
- Cancer Research Program, McGill University Health Centre Research Institute, Montreal, Quebec, H4A3J1, Canada
| | - Peter Metrakos
- Cancer Research Program, McGill University Health Centre Research Institute, Montreal, Quebec, H4A3J1, Canada.
| |
Collapse
|
29
|
Laskowski J, Renner B, Pickering MC, Serkova NJ, Smith-Jones PM, Clambey ET, Nemenoff RA, Thurman JM. Complement factor H-deficient mice develop spontaneous hepatic tumors. J Clin Invest 2020; 130:4039-4054. [PMID: 32369457 PMCID: PMC7410061 DOI: 10.1172/jci135105] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 04/22/2020] [Indexed: 12/13/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is difficult to detect, carries a poor prognosis, and is one of few cancers with an increasing yearly incidence. Molecular defects in complement factor H (CFH), a critical regulatory protein of the complement alternative pathway (AP), are typically associated with inflammatory diseases of the eye and kidney. Little is known regarding the role of CFH in controlling complement activation within the liver. While studying aging CFH-deficient (fH-/-) mice, we observed spontaneous hepatic tumor formation in more than 50% of aged fH-/- males. Examination of fH-/- livers (3-24 months) for evidence of complement-mediated inflammation revealed widespread deposition of complement-activation fragments throughout the sinusoids, elevated transaminase levels, increased hepatic CD8+ and F4/80+ cells, overexpression of hepatic mRNA associated with inflammatory signaling pathways, steatosis, and increased collagen deposition. Immunostaining of human HCC biopsies revealed extensive deposition of complement fragments within the tumors. Investigating the Cancer Genome Atlas also revealed that increased CFH mRNA expression is associated with improved survival in patients with HCC, whereas mutations are associated with worse survival. These results indicate that CFH is critical for controlling complement activation in the liver, and in its absence, AP activation leads to chronic inflammation and promotes hepatic carcinogenesis.
Collapse
Affiliation(s)
- Jennifer Laskowski
- Department of Medicine, Nephrology and Hypertension, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Brandon Renner
- Department of Medicine, Nephrology and Hypertension, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Matthew C. Pickering
- Centre for Inflammatory Disease, Division of Immunology and Inflammation, Department of Medicine, Imperial College of London, London, United Kingdom
| | - Natalie J. Serkova
- Department of Medicine, Radiology
- Department of Medicine, Radiation Oncology, and
- Department of Anesthesiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Peter M. Smith-Jones
- Department of Medicine, Radiology
- Department of Medicine, Radiation Oncology, and
- Department of Anesthesiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Eric T. Clambey
- Department of Anesthesiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Raphael A. Nemenoff
- Department of Medicine, Nephrology and Hypertension, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Joshua M. Thurman
- Department of Medicine, Nephrology and Hypertension, University of Colorado School of Medicine, Aurora, Colorado, USA
| |
Collapse
|
30
|
Arteel GE, Naba A. The liver matrisome - looking beyond collagens. JHEP Rep 2020; 2:100115. [PMID: 32637906 PMCID: PMC7330160 DOI: 10.1016/j.jhepr.2020.100115] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 03/17/2020] [Accepted: 03/22/2020] [Indexed: 02/07/2023] Open
Abstract
The extracellular matrix (ECM) is a diverse microenvironment that maintains bidirectional communication with surrounding cells to regulate cell and tissue homeostasis. The classical definition of the ECM has more recently been extended to include non-fibrillar proteins that either interact or are structurally affiliated with the ECM, termed the 'matrisome.' In addition to providing the structure and architectural support for cells and tissue, the matrisome serves as a reservoir for growth factors and cytokines, as well as a signaling hub via which cells can communicate with their environment and vice-versa. The matrisome is a master regulator of tissue homeostasis and organ function, which can dynamically and appropriately respond to any stress or injury. Failure to properly regulate these responses can lead to changes in the matrisome that are maladaptive. Hepatic fibrosis is a canonical example of ECM dyshomeostasis, leading to accumulation of predominantly collagenous ECM; indeed, hepatic fibrosis is considered almost synonymous with collagen accumulation. However, the qualitative and quantitative alterations of the hepatic matrisome during fibrosis are much more diverse than simple accumulation of collagens and occur long before fibrosis is histologically detected. A deeper understanding of the hepatic matrisome and its response to injury could yield new mechanistic insights into disease progression and regression, as well as potentially identify new biomarkers for both. In this review, we discuss the role of the ECM in liver diseases and look at new "omic" approaches to study this compartment.
Collapse
Key Words
- AUROC, area under the receiver operating characteristic curve
- CCl4, carbon tetrachloride
- ECM
- ECM, extracellular matrix
- Extracellular matrix
- Fibrosis
- HCC, hepatocellular carcinoma
- Liver disease
- MMP, matrix metalloproteinase
- NAFLD, non-alcoholic fatty liver disease
- NPV, negative predictive value
- POSTN, periostin
- PPV, positive predictive values
- Proteomics
- Regeneration
- TGFβ, transforming growth factor beta
Collapse
Affiliation(s)
- Gavin E. Arteel
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Pittsburgh Liver Research Center, Pittsburgh, PA, USA
| | - Alexandra Naba
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, USA
- University of Illinois Cancer Center, Chicago, IL, USA
| |
Collapse
|
31
|
Nyström H. Extracellular matrix proteins in metastases to the liver - Composition, function and potential applications. Semin Cancer Biol 2020; 71:134-142. [PMID: 32526353 DOI: 10.1016/j.semcancer.2020.06.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 06/04/2020] [Accepted: 06/04/2020] [Indexed: 02/08/2023]
Abstract
The rising evidence of the tumor microenvironment (TME) and its role in cancer have made this an area of increased research efforts. The focus is both on the primary tumor but also on the metastatic setting. The TME though, does not only consist of the non-malignant cells of a tumor, but also of the acellular compartment: The Extracellular Matrix (ECM). The liver is a common organ for metastasis of many cancers and for some of these cancers' liver surgery is a standard treatment with long-term cure, whereas for other cancers not considered meaningful. Blood supply and anatomical reasons plays one part for the establishment of liver metastasis. It is however a well-known fact that the "soil" of a metastatic organ is of utter importance in the process of metastasis. The "soil" consists of the TME where the ECM is a critical and active part. This review focuses what is known about the normal ECM of the human liver, what is known about ECM proteins in human liver metastasis, challenges of studying the ECM in liver metastases and lastly, potential applications of this field of knowledge.
Collapse
Affiliation(s)
- Hanna Nyström
- Department of Surgical and Perioperative Sciences, Surgery, Umeå University, Sweden; Associated Researcher Wallenberg Centre for Molecular Medicine, Umeå University, Sweden.
| |
Collapse
|
32
|
Das P, DiVito MD, Wertheim JA, Tan LP. Collagen-I and fibronectin modified three-dimensional electrospun PLGA scaffolds for long-term in vitro maintenance of functional hepatocytes. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 111:110723. [DOI: 10.1016/j.msec.2020.110723] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 12/26/2019] [Accepted: 02/03/2020] [Indexed: 12/20/2022]
|
33
|
Abstract
Chronic fatty liver disease is common worldwide. This disease is a spectrum of disease states, ranging from simple steatosis (fat accumulation) to inflammation, and eventually to fibrosis and cirrhosis if untreated. The fibrotic stage of chronic liver disease is primarily characterized by robust accumulation of extracellular matrix (ECM) proteins (collagens) that ultimately impairs the function of the organ. The role of the ECM in early stages of chronic liver disease is less well-understood, but recent research has demonstrated that several changes in the hepatic ECM in prefibrotic liver disease are not only present but may also contribute to disease progression. The purpose of this review is to summarize the established and proposed changes to the hepatic ECM that may contribute to inflammation during earlier stages of disease development, and to discuss potential mechanisms by which these changes may mediate the progression of the disease.
Collapse
Affiliation(s)
- Christine E. Dolin
- Department of Pharmacology and Toxicology, University of Louisville Health Sciences Center, Louisville, Kentucky
| | - Gavin E. Arteel
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania,Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
34
|
Hepatocyte Injury and Hepatic Stem Cell Niche in the Progression of Non-Alcoholic Steatohepatitis. Cells 2020; 9:cells9030590. [PMID: 32131439 PMCID: PMC7140508 DOI: 10.3390/cells9030590] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 02/21/2020] [Accepted: 02/27/2020] [Indexed: 02/07/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a chronic liver disease characterized by lipid accumulation in hepatocytes in the absence of excessive alcohol consumption. The global prevalence of NAFLD is constantly increasing. NAFLD is a disease spectrum comprising distinct stages with different prognoses. Non-alcoholic steatohepatitis (NASH) is a progressive condition, characterized by liver inflammation and hepatocyte ballooning, with or without fibrosis. The natural history of NAFLD is negatively influenced by NASH onset and by the progression towards advanced fibrosis. Pathogenetic mechanisms and cellular interactions leading to NASH and fibrosis involve hepatocytes, liver macrophages, myofibroblast cell subpopulations, and the resident progenitor cell niche. These cells are implied in the regenerative trajectories following liver injury, and impairment or perturbation of these mechanisms could lead to NASH and fibrosis. Recent evidence underlines the contribution of extra-hepatic organs/tissues (e.g., gut, adipose tissue) in influencing NASH development by interacting with hepatic cells through various molecular pathways. The present review aims to summarize the role of hepatic parenchymal and non-parenchymal cells, their mutual influence, and the possible interactions with extra-hepatic tissues and organs in the pathogenesis of NAFLD.
Collapse
|
35
|
Schaschkow A, Sigrist S, Mura C, Barthes J, Vrana NE, Czuba E, Lemaire F, Neidl R, Dissaux C, Lejay A, Lavalle P, Bruant-Rodier C, Bouzakri K, Pinget M, Maillard E. Glycaemic control in diabetic rats treated with islet transplantation using plasma combined with hydroxypropylmethyl cellulose hydrogel. Acta Biomater 2020; 102:259-272. [PMID: 31811957 DOI: 10.1016/j.actbio.2019.11.047] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 11/15/2019] [Accepted: 11/22/2019] [Indexed: 12/18/2022]
Abstract
Islet transplantation is one of the most efficient cell therapies used in clinics and could treat a large proportion of patients with diabetes. However, it is limited by the high requirement of pancreas necessary to provide the sufficient surviving islet mass in the hepatic tissue and restore normoglycaemia. Reduction in organ procurement requirements could be achieved by extrahepatic transplantation using a biomaterial that enhances islet survival and function. We report a plasma-supplemented hydroxypropyl methylcellulose (HPMC) hydrogel, engineered specifically using a newly developed technique for intra-omental islet infusion, known as hOMING (h-Omental Matrix Islet filliNG). The HPMC hydrogel delivered islets with better performance than that of the classical intrahepatic infusion. After the validation of the HPMC suitability for islets in vivo and in vitro, plasma supplementation modified the rheological properties of HPMC without affecting its applicability with hOMING. The biomaterial association was proven to be more efficient both in vitro and in vivo, with better islet viability and function than that of the current clinical intrahepatic delivery technique. Indeed, when the islet mass was decreased by 25% or 35%, glycaemia control was observed in the group of plasma-supplemented hydrogels, whereas no regulation was observed in the hepatic group. Plasma gelation, observed immediately post infusion, decreased anoïkis and promoted vascularisation. To conclude, the threshold mass for islet transplantation could be decreased using HPMC-Plasma combined with the hOMING technique. The simplicity of the hOMING technique and the already validated use of its components could facilitate its transfer to clinics. STATEMENT OF SIGNIFICANCE: One of the major limitations for the broad deployment of current cell therapy for brittle type 1 diabetes is the islets' destruction during the transplantation process. Retrieved from their natural environment, the islets are grafted into a foreign tissue, which triggers massive cell loss. It is mandatory to provide the islets with an 3D environment specifically designed for promoting isletimplantation to improve cell therapy outcomes. For this aim, we combined HPMC and plasma. HPMC provides suitable rheological properties to the plasma to be injectable and be maintained in the omentum. Afterwards, the plasma polymerises around the graft in vivo, thereby allowing their optimal integration into their transplantation site. As a result, the islet mass required to obtain glycaemic control was reduced by 35%.
Collapse
|
36
|
Velázquez-Miranda E, Díaz-Muñoz M, Vázquez-Cuevas FG. Purinergic signaling in hepatic disease. Purinergic Signal 2019; 15:477-489. [PMID: 31576486 DOI: 10.1007/s11302-019-09680-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Accepted: 09/02/2019] [Indexed: 12/11/2022] Open
Abstract
Extracellular purines (ATP and adenosine) are ubiquitous intercellular messengers. During tissular damage, they function as damage-associated molecular patterns (DAMPs). In this context, purines announce tissue alterations to initiate a reparative response that involve the formation of the inflammasome complex and the recruitment of specialized cells of the immune system. The present review focuses on the role of the purinergic system in liver damage, mainly during the onset and development of fibrosis. After hepatocellular injury, extracellular ATP promotes a signaling cascade that ameliorates tissue alterations to restore the hepatic function. However, if cellular damage becomes chronic, ATP orchestrates an aberrant reparative process that results in severe liver diseases such as fibrosis and cirrhosis. ATP and adenosine, their receptors, and extracellular ectonucleotidases are mediators of unique processes that will be reviewed in detail.
Collapse
Affiliation(s)
- E Velázquez-Miranda
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Boulevard Juriquilla 3001, C.P. 76230, Juriquilla, Querétaro, México
| | - M Díaz-Muñoz
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Boulevard Juriquilla 3001, C.P. 76230, Juriquilla, Querétaro, México
| | - F G Vázquez-Cuevas
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Boulevard Juriquilla 3001, C.P. 76230, Juriquilla, Querétaro, México.
| |
Collapse
|
37
|
Wu L, Ferracci G, Wang Y, Fan TF, Cho NJ, Chow PKH. Porcine hepatocytes culture on biofunctionalized 3D inverted colloidal crystal scaffolds as an in vitro model for predicting drug hepatotoxicity. RSC Adv 2019; 9:17995-18007. [PMID: 35520590 PMCID: PMC9064660 DOI: 10.1039/c9ra03225h] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 05/27/2019] [Indexed: 01/03/2023] Open
Abstract
As drug-induced hepatotoxicity represents one of the most common causes of drug failure, three-dimensional (3D) in vitro liver platforms represent a fantastic toolbox to predict drug toxicity and thus reduce in vivo animal studies and lessen drug attrition rates. The aim of this study is to establish a functional porcine hepatocyte culture using a biofunctionalized 3D inverted colloidal crystal (ICC) hydrogel platform. The performances of non-adhesive bare poly(ethylene glycol)diacrylate (PEGDA) ICCs and PEGDA ICCs coated with either collagen type I or fibronectin have been investigated. Porcine hepatocytes viability, morphology, hepatic-specific functions and patterns of gene expression have been evaluated over a period of two weeks in culture to test diclofenac, a well-known hepatotoxic drug. Interestingly, cells in the fibronectin-functionalized scaffold exhibit different aggregation patterns and maintain better liver-specific function than those in bare ICCs and in collagen functionalized scaffold. We concluded that the 3D cell culture environment and the presence of extracellular matrix (ECM) proteins, especially fibronectin, facilitate hepatocyte viability and maintenance of the liver-specific phenotype in vitro, and enable us to predict hepatotoxicity. As drug-induced hepatotoxicity represents one of the most common causes of drug failure, three-dimensional in vitro liver platforms represent a fantastic toolbox to predict drug toxicity and reduce in vivo studies and drug attrition rates.![]()
Collapse
Affiliation(s)
- Lingyan Wu
- Division of Surgical Oncology, National Cancer Centre Singapore 11 Hospital Drive 169610 Singapore
| | - Gaia Ferracci
- Interdisciplinary Graduate School, NTU Institute for Health Technologies, Nanyang Technological University 50 Nanyang Drive 637553 Singapore.,School of Materials Science and Engineering, Nanyang Technological University 50 Nanyang Avenue 639798 Singapore .,Centre for Biomimetic Sensor Science, Nanyang Technological University 50 Nanyang Drive 637553 Singapore
| | - Yan Wang
- School of Materials Science and Engineering, Nanyang Technological University 50 Nanyang Avenue 639798 Singapore .,Centre for Biomimetic Sensor Science, Nanyang Technological University 50 Nanyang Drive 637553 Singapore
| | - Teng Fei Fan
- School of Materials Science and Engineering, Nanyang Technological University 50 Nanyang Avenue 639798 Singapore .,Centre for Biomimetic Sensor Science, Nanyang Technological University 50 Nanyang Drive 637553 Singapore
| | - Nam-Joon Cho
- School of Materials Science and Engineering, Nanyang Technological University 50 Nanyang Avenue 639798 Singapore .,Centre for Biomimetic Sensor Science, Nanyang Technological University 50 Nanyang Drive 637553 Singapore.,School of Chemical and Biomedical Engineering, Nanyang Technological University 62 Nanyang Drive 637459 Singapore
| | - Pierce K H Chow
- Division of Surgical Oncology, National Cancer Centre Singapore 11 Hospital Drive 169610 Singapore .,Duke-NUS Medical School 8 College Road 169857 Singapore.,Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital Outram Road 169608 Singapore
| |
Collapse
|
38
|
Grant R, Hallett J, Forbes S, Hay D, Callanan A. Blended electrospinning with human liver extracellular matrix for engineering new hepatic microenvironments. Sci Rep 2019; 9:6293. [PMID: 31000735 PMCID: PMC6472345 DOI: 10.1038/s41598-019-42627-7] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 04/03/2019] [Indexed: 12/21/2022] Open
Abstract
Tissue engineering of a transplantable liver could provide an alternative to donor livers for transplant, solving the problem of escalating donor shortages. One of the challenges for tissue engineers is the extracellular matrix (ECM); a finely controlled in vivo niche which supports hepatocytes. Polymers and decellularized tissue scaffolds each provide some of the necessary biological cues for hepatocytes, however, neither alone has proved sufficient. Enhancing microenvironments using bioactive molecules allows researchers to create more appropriate niches for hepatocytes. We combined decellularized human liver tissue with electrospun polymers to produce a niche for hepatocytes and compared the human liver ECM to its individual components; Collagen I, Laminin-521 and Fibronectin. The resulting scaffolds were validated using THLE-3 hepatocytes. Immunohistochemistry confirmed retention of proteins in the scaffolds. Mechanical testing demonstrated significant increases in the Young's Modulus of the decellularized ECM scaffold; providing significantly stiffer environments for hepatocytes. Each scaffold maintained hepatocyte growth, albumin production and influenced expression of key hepatic genes, with the decellularized ECM scaffolds exerting an influence which is not recapitulated by individual ECM components. Blended protein:polymer scaffolds provide a viable, translatable niche for hepatocytes and offers a solution to current obstacles in disease modelling and liver tissue engineering.
Collapse
Affiliation(s)
- Rhiannon Grant
- Institute for Bioengineering, School of Engineering, University of Edinburgh, Scotland, UK
| | - John Hallett
- Scottish Centre for Regenerative Medicine, University of Edinburgh, Scotland, UK
| | - Stuart Forbes
- Scottish Centre for Regenerative Medicine, University of Edinburgh, Scotland, UK
| | - David Hay
- Scottish Centre for Regenerative Medicine, University of Edinburgh, Scotland, UK
| | - Anthony Callanan
- Institute for Bioengineering, School of Engineering, University of Edinburgh, Scotland, UK.
| |
Collapse
|
39
|
Kobayashi J, Okano T. Design of Temperature-Responsive Polymer-Grafted Surfaces for Cell Sheet Preparation and Manipulation. BULLETIN OF THE CHEMICAL SOCIETY OF JAPAN 2019. [DOI: 10.1246/bcsj.20180378] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Jun Kobayashi
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women’s Medical University, TWIns, 8-1 Kawadacho, Shinjuku-ku, Tokyo 162-8666, Japan
| | - Teruo Okano
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women’s Medical University, TWIns, 8-1 Kawadacho, Shinjuku-ku, Tokyo 162-8666, Japan
- Cell Sheet Tissue Engineering Center and Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, 30 South 2000 East, Salt Lake City, Utah 84112, USA
| |
Collapse
|
40
|
Blau BJ, Miki T. The role of cellular interactions in the induction of hepatocyte polarity and functional maturation in stem cell-derived hepatic cells. Differentiation 2019; 106:42-48. [PMID: 30878880 DOI: 10.1016/j.diff.2019.02.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 02/25/2019] [Accepted: 02/28/2019] [Indexed: 02/08/2023]
Abstract
The unique microenvironment found within the liver in vivo plays a key role in the induction of functional maturation in the developing hepatocyte. During organogenesis, hepatocytes acquire a polar phenotype that allows them to perform their functions of bile production and transport, protein synthesis, metabolism, and detoxification simultaneously, independently, and efficiently. It is thought that the induction of polarity and functional maturation in hepatocytes is dependent on the complex interplay of cell-cell and cell-extracellular matrix (ECM) interactions. While this process is highly efficient in the human liver, it has been shown that hepatocytes rapidly lose their functions when placed in cell culture. This poses a challenge for the development of a bioartificial liver (BAL) support system, which utilizes a live cellular source to perform hepatic functions in the event of acute liver failure or primary nonfunction. However, once the molecular mechanisms underlying the induction of hepatocyte polarity are fully identified, it will be possible to develop highly functional hepatic cells from human pluripotent stem cells (hPSCs). This new cell line would be an ideal cellular source for a BAL system, as it would have both the functionality and longevity to support a patient through the entire clinical course of treatment. In this review, we explore the literature that has examined the potential mechanisms that induce polarity in the developing hepatocyte and discuss the future implications of this knowledge in a clinical setting from a bioengineering perspective.
Collapse
Affiliation(s)
- Brandon J Blau
- Department of Surgery, Keck School of Medicine, University of Southern California, USA
| | - Toshio Miki
- Department of Surgery, Keck School of Medicine, University of Southern California, USA.
| |
Collapse
|
41
|
Gao S, Silasi-Mansat R, Behar M, Lupu F, Griffin CT. Excessive Plasmin Compromises Hepatic Sinusoidal Vascular Integrity After Acetaminophen Overdose. Hepatology 2018; 68:1991-2003. [PMID: 29729197 PMCID: PMC6204085 DOI: 10.1002/hep.30070] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 04/20/2018] [Accepted: 04/29/2018] [Indexed: 12/19/2022]
Abstract
The serine protease plasmin degrades extracellular matrix (ECM) components both directly and indirectly through activation of matrix metalloproteinases. Excessive plasmin activity and subsequent ECM degradation cause hepatic sinusoidal fragility and hemorrhage in developing embryos. We report here that excessive plasmin activity in a murine acetaminophen (APAP) overdose model likewise compromises hepatic sinusoidal vascular integrity in adult animals. We found that hepatic plasmin activity is up-regulated significantly at 6 hours after APAP overdose. This plasmin up-regulation precedes both degradation of the ECM component fibronectin around liver vasculature and bleeding from centrilobular sinusoids. Importantly, administration of the pharmacological plasmin inhibitor tranexamic acid or genetic reduction of plasminogen, the circulating zymogen of plasmin, ameliorates APAP-induced hepatic fibronectin degradation and sinusoidal bleeding. Conclusion: These studies demonstrate that reduction of plasmin stabilizes hepatic sinusoidal vascular integrity after APAP overdose. (Hepatology 2018; 00:1-13).
Collapse
Affiliation(s)
- Siqi Gao
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK,Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Robert Silasi-Mansat
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Mandi Behar
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Florea Lupu
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK,Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Courtney T. Griffin
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK,Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| |
Collapse
|
42
|
Grant R, Hay D, Callanan A. From scaffold to structure: the synthetic production of cell derived extracellular matrix for liver tissue engineering. Biomed Phys Eng Express 2018. [DOI: 10.1088/2057-1976/aacbe1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
43
|
Joshi P, Datar A, Yu KN, Kang SY, Lee MY. High-content imaging assays on a miniaturized 3D cell culture platform. Toxicol In Vitro 2018; 50:147-159. [PMID: 29501531 DOI: 10.1016/j.tiv.2018.02.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 02/19/2018] [Accepted: 02/20/2018] [Indexed: 12/17/2022]
Abstract
The majority of high-content imaging (HCI) assays have been performed on two-dimensional (2D) cell monolayers for its convenience and throughput. However, 2D-cultured cell models often do not represent the in vivo characteristics accurately and therefore reduce the predictability of drug toxicity/efficacy in vivo. Recently, three-dimensional (3D) cell-based HCI assays have been demonstrated to improve predictability, but its use is limited due to difficulty in maneuverability and low throughput in cell imaging. To alleviate these issues, we have developed miniaturized 3D cell culture on a micropillar/microwell chip and demonstrated high-throughput HCI assays for mechanistic toxicity. Briefly, Hep3B human hepatoma cell line was encapsulated in a mixture of alginate and fibrin gel on the micropillar chip, cultured in 3D, and exposed to six model compounds in the microwell chip for rapidly assessing mechanistic hepatotoxicity. Several toxicity parameters, including DNA damage, mitochondrial impairment, intracellular glutathione level, and cell membrane integrity were measured on the chip, and the IC50 values of the compounds at different readouts were determined to investigate the mechanism of toxicity. Overall, the Z' factors were between 0.6 and 0.8 for the HCI assays, and the coefficient of variation (CV) were below 20%. These results indicate high robustness and reproducibility of the HCI assays established on the miniaturized 3D cell culture chip. In addition, it was possible to determine the predominant mechanism of toxicity using the 3D HCI assays. Therefore, our miniaturized 3D cell culture coupled with HCI assays has great potential for high-throughput screening (HTS) of compounds and mechanistic toxicity profiling.
Collapse
Affiliation(s)
- Pranav Joshi
- Department of Chemical and Biomedical Engineering, Cleveland State University, 455 Fenn Hall, 1960 East 24th Street, Cleveland, OH 44115-2214, USA
| | - Akshata Datar
- Department of Chemical and Biomedical Engineering, Cleveland State University, 455 Fenn Hall, 1960 East 24th Street, Cleveland, OH 44115-2214, USA
| | - Kyeong-Nam Yu
- Department of Chemical and Biomedical Engineering, Cleveland State University, 455 Fenn Hall, 1960 East 24th Street, Cleveland, OH 44115-2214, USA
| | - Soo-Yeon Kang
- Department of Chemical and Biomedical Engineering, Cleveland State University, 455 Fenn Hall, 1960 East 24th Street, Cleveland, OH 44115-2214, USA
| | - Moo-Yeal Lee
- Department of Chemical and Biomedical Engineering, Cleveland State University, 455 Fenn Hall, 1960 East 24th Street, Cleveland, OH 44115-2214, USA.
| |
Collapse
|
44
|
Müsch A. From a common progenitor to distinct liver epithelial phenotypes. Curr Opin Cell Biol 2018; 54:18-23. [PMID: 29505983 DOI: 10.1016/j.ceb.2018.02.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 02/12/2018] [Accepted: 02/13/2018] [Indexed: 12/23/2022]
Abstract
The vertebrate liver presents a fascinating case study for how cell form is optimized for function. To execute its duties the liver assembles two distinct lumen-forming epithelial phenotypes: Firstly, cords with a branched, capillary-like luminal network formed between hepatocytes (bile canaliculi); and secondly, tubular ducts formed by biliary epithelial cells arranged around a central cavity and connected to the bile canaliculi. How these remarkably different epithelial polarity phenotypes are generated and joined into a contiguous luminal network are major unresolved questions. Recent studies have characterized the divergence of the two epithelial lineages from common progenitors, described the coordination of bile canaliculi formation with bile duct branching during biliary tree morphogenesis and implicated RhoA-dependent E-cadherin adhesion in the decision to polarize with hepatocytic or biliary phenotype.
Collapse
Affiliation(s)
- Anne Müsch
- Department of Developmental and Molecular Biology at Albert-Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY 10461, USA.
| |
Collapse
|
45
|
Lord MS, Tang F, Rnjak-Kovacina J, Smith JGW, Melrose J, Whitelock JM. The multifaceted roles of perlecan in fibrosis. Matrix Biol 2018; 68-69:150-166. [PMID: 29475023 DOI: 10.1016/j.matbio.2018.02.013] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Revised: 02/15/2018] [Accepted: 02/16/2018] [Indexed: 12/11/2022]
Abstract
Perlecan, or heparan sulfate proteoglycan 2 (HSPG2), is a ubiquitous heparan sulfate proteoglycan that has major roles in tissue and organ development and wound healing by orchestrating the binding and signaling of mitogens and morphogens to cells in a temporal and dynamic fashion. In this review, its roles in fibrosis are reviewed by drawing upon evidence from tissue and organ systems that undergo fibrosis as a result of an uncontrolled response to either inflammation or traumatic cellular injury leading to an over production of a collagen-rich extracellular matrix. This review focuses on examples of fibrosis that occurs in lung, liver, kidney, skin, kidney, neural tissues and blood vessels and its link to the expression of perlecan in that particular organ system.
Collapse
Affiliation(s)
- Megan S Lord
- Graduate School of Biomedical Engineering, UNSW Sydney, NSW 2052, Australia.
| | - Fengying Tang
- Graduate School of Biomedical Engineering, UNSW Sydney, NSW 2052, Australia
| | | | - James G W Smith
- University of Nottingham, Nottingham NG7 2RD, United Kingdom
| | - James Melrose
- Graduate School of Biomedical Engineering, UNSW Sydney, NSW 2052, Australia; Raymond Purves Bone and Joint Research Laboratory, Kolling Institute Northern Sydney Local Health District, St. Leonards, NSW 2065, Australia; Sydney Medical School, Northern, The University of Sydney, Royal North Shore Hospital, St. Leonards, NSW 2065, Australia
| | - John M Whitelock
- Graduate School of Biomedical Engineering, UNSW Sydney, NSW 2052, Australia
| |
Collapse
|
46
|
Jaramillo M, Yeh H, Yarmush ML, Uygun BE. Decellularized human liver extracellular matrix (hDLM)-mediated hepatic differentiation of human induced pluripotent stem cells (hIPSCs). J Tissue Eng Regen Med 2018; 12:e1962-e1973. [PMID: 29222839 DOI: 10.1002/term.2627] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 08/31/2017] [Accepted: 11/30/2017] [Indexed: 12/30/2022]
Abstract
Liver tissue engineering has emerged as a promising approach in organ transplantation but has been hampered by the lack of a reliable and readily available cell source. Human induced pluripotent stem cells hiPSCs have been highlighted as a desirable source, due to their differentiation potential, ability to self-renew, and the possibility of making patient-specific cells. We developed a decellularization protocol that efficiently removes cellular material while retaining extracellular matrix components. Subsequently, hiPSCs were differentiated on decellularized human liver extracellular matrix (hDLM) scaffolds using an established hepatic differentiation protocol. We demonstrate that using hDLM leads to upregulation markers of hepatic functions when compared with standard differentiation conditions. In addition, expression of a number of hepatic transcription and nuclear factors were found to be within levels comparable with those of primary human adult hepatocytes. Analysis of progression of differentiation on hDLM demonstrated that hepatic developmental marker expression was consistent with hepatic development. The hDLM-derived cells exhibited key hepatic characteristics that were comparable with those observed in primary neonatal human hepatocytes. We investigated the optimal timing of the introduction of hDLM into the differentiation protocol and found that the best results are obtained when cells are plated on hDLM since the earliest stages and accompanied by a progressive loss of sensitivity to substrate composition at later stages. The significance of this work is that it allows for the development of differentiation protocols that take into account signals from extracellular matrix, closely recapitulating of the in vivo micro-environment and resulting in cells that are phenotypically closer to mature hepatocytes.
Collapse
Affiliation(s)
- Maria Jaramillo
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School, Shriners Hospitals for Children, Boston, MA, USA.,Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Heidi Yeh
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Martin L Yarmush
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School, Shriners Hospitals for Children, Boston, MA, USA.,Department of Surgery, Massachusetts General Hospital, Boston, MA, USA.,Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, USA
| | - Basak E Uygun
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School, Shriners Hospitals for Children, Boston, MA, USA.,Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
47
|
Crawford JM, Bioulac-Sage P, Hytiroglou P. Structure, Function, and Responses to Injury. MACSWEEN'S PATHOLOGY OF THE LIVER 2018:1-87. [DOI: 10.1016/b978-0-7020-6697-9.00001-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
48
|
Karsdal MA, Nielsen SH, Leeming DJ, Langholm LL, Nielsen MJ, Manon-Jensen T, Siebuhr A, Gudmann NS, Rønnow S, Sand JM, Daniels SJ, Mortensen JH, Schuppan D. The good and the bad collagens of fibrosis - Their role in signaling and organ function. Adv Drug Deliv Rev 2017; 121:43-56. [PMID: 28736303 DOI: 10.1016/j.addr.2017.07.014] [Citation(s) in RCA: 351] [Impact Index Per Article: 43.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 07/14/2017] [Accepted: 07/17/2017] [Indexed: 12/11/2022]
Abstract
Usually the dense extracellular structure in fibrotic tissues is described as extracellular matrix (ECM) or simply as collagen. However, fibrosis is not just fibrosis, which is already exemplified by the variant morphological characteristics of fibrosis due to viral versus cholestatic, autoimmune or toxic liver injury, with reticular, chicken wire and bridging fibrosis. Importantly, the overall composition of the ECM, especially the relative amounts of the many types of collagens, which represent the most abundant ECM molecules and which centrally modulate cellular functions and physiological processes, changes dramatically during fibrosis progression. We hypothesize that there are good and bad collagens in fibrosis and that a change of location alone may change the function from good to bad. Whereas basement membrane collagen type IV anchors epithelial and other cells in a polarized manner, the interstitial fibroblast collagens type I and III do not provide directional information. In addition, feedback loops from biologically active degradation products of some collagens are examples of the importance of having the right collagen at the right place and at the right time controlling cell function, proliferation, matrix production and fate. Examples are the interstitial collagen type VI and basement membrane collagen type XVIII. Their carboxyterminal propeptides serve as an adipose tissue hormone, endotrophin, and as a regulator of angiogenesis, endostatin, respectively. We provide an overview of the 28 known collagen types and propose that the molecular composition of the ECM in fibrosis needs careful attention to assess its impact on organ function and its potential to progress or reverse. Consequently, to adequately assess fibrosis and to design optimal antifibrotic therapies, we need to dissect the molecular entity of fibrosis for the molecular composition and spatial distribution of collagens and the associated ECM.
Collapse
Affiliation(s)
- M A Karsdal
- Nordic Bioscience Biomarkers & Research A/S, Herlev, Denmark.
| | - S H Nielsen
- Nordic Bioscience Biomarkers & Research A/S, Herlev, Denmark
| | - D J Leeming
- Nordic Bioscience Biomarkers & Research A/S, Herlev, Denmark
| | - L L Langholm
- Nordic Bioscience Biomarkers & Research A/S, Herlev, Denmark
| | - M J Nielsen
- Nordic Bioscience Biomarkers & Research A/S, Herlev, Denmark
| | - T Manon-Jensen
- Nordic Bioscience Biomarkers & Research A/S, Herlev, Denmark
| | - A Siebuhr
- Nordic Bioscience Biomarkers & Research A/S, Herlev, Denmark
| | - N S Gudmann
- Nordic Bioscience Biomarkers & Research A/S, Herlev, Denmark
| | - S Rønnow
- Nordic Bioscience Biomarkers & Research A/S, Herlev, Denmark
| | - J M Sand
- Nordic Bioscience Biomarkers & Research A/S, Herlev, Denmark
| | - S J Daniels
- Nordic Bioscience Biomarkers & Research A/S, Herlev, Denmark
| | - J H Mortensen
- Nordic Bioscience Biomarkers & Research A/S, Herlev, Denmark
| | - D Schuppan
- Institute of Translational Immunology and Research Center for Immune Therapy, University Medical Center, Johannes Gutenberg University, Mainz, Germany; Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
49
|
Bircher K, Ehret AE, Mazza E. Microstructure based prediction of the deformation behavior of soft collagenous membranes. SOFT MATTER 2017; 13:5107-5116. [PMID: 28492654 DOI: 10.1039/c7sm00101k] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The response of human amnion (HA) and bovine Glisson's capsule (GC) to uniaxial and biaxial tensile loading is analyzed on tissue (∼mm) and collagen fiber (∼μm) length scales. The mechanical behavior of the membranes is rationalized based on a discrete fiber network model that relates model parameters with microstructural features of the tissues. Parameters were first determined for GC based on the quantity and organization of collagen fibers in the tissue. Next, parameters for HA were defined by comparing the microstructures of the two membranes, which differ in fiber organization in that collagen forms μm-thick fiber bundles in GC while 50 nm-thin fibrils constitute the network in HA. The flexural behavior of these structures is phenomenologically represented in the model, indicating that shear forces are transmitted through fibrils within GC bundles, but to a much lesser extent than in a corresponding solid cross section. The model provides excellent predictions of the uniaxial and biaxial mechanical response, as well as of the progressive reorientation of fibers associated with uniaxial loading. The results are particularly relevant since model parameters were not obtained through a fitting procedure of the tissue's tension-stretch curve. Furthermore, simulations of representative in vivo deformation states indicated that a large part of the fibers are expected to be un-crimped under physiological loading conditions. Thus, the crimped shape of collagen fibers in the initial test configuration, and typically observed in histological analyses, might be a consequence of the contraction occurring when membranes are extracted from their environment in the body.
Collapse
Affiliation(s)
- Kevin Bircher
- Institute for Mechanical Systems, ETH Zurich, 8092 Zurich, Switzerland.
| | | | | |
Collapse
|
50
|
Grant R, Hay DC, Callanan A. A Drug-Induced Hybrid Electrospun Poly-Capro-Lactone: Cell-Derived Extracellular Matrix Scaffold for Liver Tissue Engineering. Tissue Eng Part A 2017; 23:650-662. [PMID: 28437180 DOI: 10.1089/ten.tea.2016.0419] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Liver transplant is the only treatment option for patients with end-stage liver failure, however, there are too few donor livers available for transplant. Whole organ tissue engineering presents a potential solution to the problem of rapidly escalating donor liver shortages worldwide. A major challenge for liver tissue engineers is the creation of a hepatocyte microenvironment; a niche in which liver cells can survive and function optimally. While polymers and decellularized tissues pose an attractive option for scaffold manufacturing, neither alone has thus far proved sufficient. This study exploited cell's native extracellular matrix (ECM) producing capabilities using two different histone deacetylase inhibitors, and combined these with the customizability and reproducibility of electrospun polymer scaffolds to produce a "best of both worlds" niche microenvironment for hepatocytes. The resulting hybrid poly-capro-lactone (PCL)-ECM scaffolds were validated using HepG2 hepatocytes. The hybrid PCL-ECM scaffolds maintained hepatocyte growth and function, as evidenced by metabolic activity and DNA quantitation. Mechanical testing revealed little significant difference between scaffolds, indicating that cells were responding to a biochemical and topographical profile rather than mechanical changes. Immunohistochemistry showed that the biochemical profile of the drug-derived and nondrug-derived ECMs differed in ratio of Collagen I, Laminin, and Fibronectin. Furthermore, the hybrid PCL-ECM scaffolds influence the gene expression profile of the HepG2s drastically; with expression of Albumin, Cytochrome P450 Family 1 Subfamily A Polypeptide 1, Cytochrome P450 Family 1 Subfamily A Polypeptide 2, Cytochrome P450 Family 3 Subfamily A Polypeptide 4, Fibronectin, Collagen I, and Collagen IV undergoing significant changes. Our results demonstrate that drug-induced hybrid PCL-ECM scaffolds provide a viable, translatable platform for creating a niche microenvironment for hepatocytes, supporting in vivo phenotype and function. These scaffolds offer great potential for tissue engineering and regenerative medicine strategies for whole organ tissue engineering.
Collapse
Affiliation(s)
- Rhiannon Grant
- 1 Institute for Bioengineering, School of Engineering, University of Edinburgh , Edinburgh, United Kingdom
| | - David C Hay
- 2 MRC Scottish Centre for Regenerative Medicine, University of Edinburgh , Edinburgh, United Kingdom
| | - Anthony Callanan
- 1 Institute for Bioengineering, School of Engineering, University of Edinburgh , Edinburgh, United Kingdom
| |
Collapse
|