1
|
Lee HJ, Park SW, Lee JH, Chang SY, Oh SM, Mun S, Kang J, Park JE, Choi JK, Kim TI, Kim JY, Kim P. Differential cellular origins of the extracellular matrix of tumor and normal tissues according to colorectal cancer subtypes. Br J Cancer 2025; 132:770-782. [PMID: 40032993 PMCID: PMC12041468 DOI: 10.1038/s41416-025-02964-z] [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: 04/16/2024] [Revised: 01/07/2025] [Accepted: 02/13/2025] [Indexed: 03/05/2025] Open
Abstract
BACKGROUND Understanding the proteomic-level heterogeneity of the tumor microenvironment (TME) in colorectal cancer (CRC) is crucial due to its well-known heterogeneity. While heterogenous CRC has been extensively characterized at the molecular subtype level, research into the functional heterogeneity of fibroblasts, particularly their relationship with extracellular matrix (ECM) alterations, remains limited. Addressing this gap is essential for a comprehensive understanding of CRC progression and the development of targeted therapies. METHODS 24 tissue samples from 21 CRC patients, along with adjacent normal tissues (NAT), were collected and decellularized using a detergent-based method to enrich the ECM component. Proteomic analysis of ECM-enriched samples was performed using tandem mass tag (TMT) spectrometry, followed by statistical analysis including differential expression protein (DEP) analysis. Single-cell RNA sequencing (scRNA-Seq) data from public datasets were integrated and analyzed to delineate cell states within the TME. Bulk tissue RNA-Seq and bioinformatics analysis, including consensus molecular subtype (CMS) classification and single-cell level deconvolution of TCGA bulk RNA-seq data, were conducted to further explore gene expression patterns and TME composition. RESULTS Differential cellular origin of the NAT and tumorous ECM proteins were identified, revealing 110 ECM proteins enriched in NAT and 28 ECM proteins in tumor tissues. Desmoplastic and WNT5A+ inflammatory fibroblasts were indicated as the sources of tumor-enriched ECM proteins, while ADAMDEC1+ expressing fibroblasts and PI16+ expressing fibroblast were identified as the sources of NAT-enriched ECM proteins. Deconvolution of bulk RNA-seq of CRC tissues discriminated CMS-specific fibroblast state, reflecting the biological traits of each CMS subtype. Specially, seven ECM genes specific to mesenchymal subtype (CMS4), including PI16+ fibroblast-related 4 genes (SFRP2, PRELP, OGN, SRPX) and desmoplastic fibroblast-related 3 genes (THBS2, CTHRC1, BGN), showed a significant association with poorer survival in patient with CRC. CONCLUSION We conducted an extracellular matrix (ECM)-focused profiling of the TME by integrating quantitative proteomics with single-cell RNA sequencing (scRNA-seq) data from CRC patients. We identified the ECM proteins of NAT and tumor tissue, and established a cell-matrisome database. We defined mesenchymal subtype-specific molecules associated with specific fibroblast subtypes showing a significant association with poorer survival in patients with CRC. Our ECM-focused profiling of tumor stroma provides new insights as indicators for biological processes and clinical endpoints.
Collapse
Affiliation(s)
- Hyun Jin Lee
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, Republic of Korea
| | - Sang Woo Park
- Korea Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Ochang, 28119, Republic of Korea
| | - Jun Hyeong Lee
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, Republic of Korea
| | - Shin Young Chang
- Department of Internal Medicine, Institute of Gastroenterology, Brain Korea 21 Project for Medical Science, Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Sang Mi Oh
- Department of Internal Medicine, Institute of Gastroenterology, Brain Korea 21 Project for Medical Science, Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Siwon Mun
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, Republic of Korea
| | - Junho Kang
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
| | - Jong-Eun Park
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
- SCL-KAIST Institute of Translational Research, KAIST, Daejeon, Republic of Korea
| | - Jung Kyoon Choi
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, Republic of Korea
- SCL-KAIST Institute of Translational Research, KAIST, Daejeon, Republic of Korea
| | - Tae Il Kim
- Department of Internal Medicine, Institute of Gastroenterology, Brain Korea 21 Project for Medical Science, Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
| | - Jin Young Kim
- Korea Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Ochang, 28119, Republic of Korea.
| | - Pilnam Kim
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, Republic of Korea.
- SCL-KAIST Institute of Translational Research, KAIST, Daejeon, Republic of Korea.
| |
Collapse
|
2
|
Fukunaga I, Takebe T. In vitro liver models for toxicological research. Drug Metab Pharmacokinet 2025; 62:101478. [PMID: 40203632 DOI: 10.1016/j.dmpk.2025.101478] [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: 12/19/2024] [Revised: 02/25/2025] [Accepted: 03/04/2025] [Indexed: 04/11/2025]
Abstract
Drug-induced liver injury (DILI) presents a major challenge not only in new drug development but also in post-marketing withdrawals and the safety of food, cosmetics, and chemicals. Experimental model organisms such as the rodents have been widely used for preclinical toxicological testing. However, the tension exists associated with the ethical and sustainable use of animals in part because animals do not necessarily inform the human-specific ADME (adsorption, dynamics, metabolism and elimination) profiling. To establish alternative models in humans, in vitro hepatic tissue models have been proposed, ranging from primary hepatocytes, immortal hepatocytes, to the development of new cell resources such as stem cell-derived hepatocytes. Given the evolving number of novel alternative methods, understanding possible combinations of cell sources and culture methods will be crucial to develop the context-of-use assays. This review primarily focuses on 3D liver organoid models for conducting. We will review the relevant cell sources, bioengineering methods, selection of training compounds, and biomarkers towards the rationale design of in vitro toxicology testing.
Collapse
Affiliation(s)
- Ichiro Fukunaga
- Center for Genomic and Regenerative Medicine, Juntendo University Graduate School of Medicine, 2-1-1, Hongo, Bunkyo-ku, Tokyo, 113-8431, Japan.
| | - Takanori Takebe
- Human Biology Research Unit, Institute of Integrated Research, Institute of Science Tokyo, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan; Department of Genome Biology, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan; Divisions of Gastroenterology, Hepatology & Nutrition, Developmental Biology and Biomedical Informatics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229-3039, USA; Department of Pediatrics, University of Cincinnati College of Medicine, 3333 Burnet Avenue, Cincinnati, OH, 45229-3039, USA; Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229-3039, USA; Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), Osaka University, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
3
|
Qian Y, Ding J, Zhao R, Song Y, Yoo J, Moon H, Koo S, Kim JS, Shen J. Intrinsic immunomodulatory hydrogels for chronic inflammation. Chem Soc Rev 2025; 54:33-61. [PMID: 39499495 DOI: 10.1039/d4cs00450g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2024]
Abstract
The immune system plays a pivotal role in maintaining physiological homeostasis and influencing disease processes. Dysregulated immune responses drive chronic inflammation, which in turn results in a range of diseases that are among the leading causes of death globally. Traditional immune interventions, which aim to regulate either insufficient or excessive inflammation, frequently entail lifelong comorbidities and the risk of severe side effects. In this context, intrinsic immunomodulatory hydrogels, designed to precisely control the local immune microenvironment, have recently attracted increasing attention. In particular, these advanced hydrogels not only function as delivery mechanisms but also actively engage in immune modulation, optimizing interactions with the immune system for enhanced tissue repair, thereby providing a sophisticated strategy for managing chronic inflammation. In this tutorial review, we outline key elements of chronic inflammation and subsequently explore the strategic design principles of intrinsic immunomodulatory hydrogels based on these elements. Finally, we examine the challenges and prospects of such immunomodulatory hydrogels, which are expected to inspire further preclinical research and clinical translation in addressing chronic inflammation.
Collapse
Affiliation(s)
- Yuna Qian
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China.
| | - Jiayi Ding
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Institute of Imaging Diagnosis and Minimally Invasive Intervention, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Rui Zhao
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
| | - Yang Song
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610065, China
| | - Jiyoung Yoo
- Department of Chemistry, Korea University, Seoul 02841, Korea
| | - Huiyeon Moon
- Department of Chemistry, Korea University, Seoul 02841, Korea
| | - Seyoung Koo
- Department of Chemical and Molecular Engineering, Hanyang University ERICA, Ansan, Gyeonggi-do 15588, Korea.
| | - Jong Seung Kim
- Department of Chemical and Molecular Engineering, Hanyang University ERICA, Ansan, Gyeonggi-do 15588, Korea.
| | - Jianliang Shen
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China.
| |
Collapse
|
4
|
You P, Sun H, Chen H, Li C, Mao Y, Zhang T, Yang H, Dong H. Composite bioink incorporating cell-laden liver decellularized extracellular matrix for bioprinting of scaffolds for bone tissue engineering. BIOMATERIALS ADVANCES 2024; 165:214017. [PMID: 39236580 DOI: 10.1016/j.bioadv.2024.214017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 08/26/2024] [Accepted: 08/29/2024] [Indexed: 09/07/2024]
Abstract
The field of bone tissue engineering (BTE) has witnessed a revolutionary breakthrough with the advent of three-dimensional (3D) bioprinting technology, which is considered an ideal choice for constructing scaffolds for bone regeneration. The key to realizing scaffold biofunctions is the selection and design of an appropriate bioink, and existing bioinks have significant limitations. In this study, a composite bioink based on natural polymers (gelatin and alginate) and liver decellularized extracellular matrix (LdECM) was developed and used to fabricate scaffolds for BTE using 3D bioprinting. Through in vitro studies, the concentration of LdECM incorporated into the bioink was optimized to achieve printability and stability and to improve the proliferation and osteogenic differentiation of loaded rat bone mesenchymal stem cells (rBMSCs). Furthermore, in vivo experiments were conducted using a Sprague Dawley rat model of critical-sized calvarial defects. The proposed rBMSC-laden LdECM-gelatin-alginate scaffold, bioprinted layer-by-layer, was implanted in the rat calvarial defect and the development of new bone growth was studied for four weeks. The findings showed that the proposed bioactive scaffolds facilitated angiogenesis and osteogenesis at the defect site. The findings of this study suggest that the developed rBMSC-laden LdECM-gelatin-alginate bioink has great potential for clinical translation and application in solving bone regeneration problems.
Collapse
Affiliation(s)
- Pengyue You
- Department of Stomatology, Peking Union Medical College (PUMC) Hospital, Peking Union Medical College (PUMC) & Chinese Academy of Medical Sciences (CAMS), Beijing 100730, China
| | - Hang Sun
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, Peking Union Medical College (PUMC) & Chinese Academy of Medical Sciences (CAMS), Beijing 100730, China
| | - Haotian Chen
- Department of Stomatology, Peking Union Medical College (PUMC) Hospital, Peking Union Medical College (PUMC) & Chinese Academy of Medical Sciences (CAMS), Beijing 100730, China
| | - Changcan Li
- Department of General Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Yilei Mao
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, Peking Union Medical College (PUMC) & Chinese Academy of Medical Sciences (CAMS), Beijing 100730, China
| | - Tao Zhang
- Department of Stomatology, Peking Union Medical College (PUMC) Hospital, Peking Union Medical College (PUMC) & Chinese Academy of Medical Sciences (CAMS), Beijing 100730, China.
| | - Huayu Yang
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, Peking Union Medical College (PUMC) & Chinese Academy of Medical Sciences (CAMS), Beijing 100730, China.
| | - Haitao Dong
- Department of Stomatology, Peking Union Medical College (PUMC) Hospital, Peking Union Medical College (PUMC) & Chinese Academy of Medical Sciences (CAMS), Beijing 100730, China.
| |
Collapse
|
5
|
Li C, An N, Song Q, Hu Y, Yin W, Wang Q, Le Y, Pan W, Yan X, Wang Y, Liu J. Enhancing organoid culture: harnessing the potential of decellularized extracellular matrix hydrogels for mimicking microenvironments. J Biomed Sci 2024; 31:96. [PMID: 39334251 PMCID: PMC11429032 DOI: 10.1186/s12929-024-01086-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Over the past decade, organoids have emerged as a prevalent and promising research tool, mirroring the physiological architecture of the human body. However, as the field advances, the traditional use of animal or tumor-derived extracellular matrix (ECM) as scaffolds has become increasingly inadequate. This shift has led to a focus on developing synthetic scaffolds, particularly hydrogels, that more accurately mimic three-dimensional (3D) tissue structures and dynamics in vitro. The ECM-cell interaction is crucial for organoid growth, necessitating hydrogels that meet organoid-specific requirements through modifiable physical and compositional properties. Advanced composite hydrogels have been engineered to more effectively replicate in vivo conditions, offering a more accurate representation of human organs compared to traditional matrices. This review explores the evolution and current uses of decellularized ECM scaffolds, emphasizing the application of decellularized ECM hydrogels in organoid culture. It also explores the fabrication of composite hydrogels and the prospects for their future use in organoid systems.
Collapse
Affiliation(s)
- Chen Li
- Beijing International Science and Technology Cooperation Base for Antiviral Drugs, Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing, 100124, China
- School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Hepato-Pancreato-Biliary Center, Tsinghua University, Beijing, 102218, China
| | - Ni An
- School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Clinical Translational Science Center, Tsinghua University, Beijing, 102218, China
| | - Qingru Song
- School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Hepato-Pancreato-Biliary Center, Tsinghua University, Beijing, 102218, China
- School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Clinical Translational Science Center, Tsinghua University, Beijing, 102218, China
| | - Yuelei Hu
- School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Hepato-Pancreato-Biliary Center, Tsinghua University, Beijing, 102218, China
- Key Laboratory of Digital Intelligence Hepatology (Ministry of Education/Beijing), School of Clinical Medicine, Tsinghua University, Beijing, 100084, China
| | - Wenzhen Yin
- School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Clinical Translational Science Center, Tsinghua University, Beijing, 102218, China
| | - Qi Wang
- School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Hepato-Pancreato-Biliary Center, Tsinghua University, Beijing, 102218, China
- Key Laboratory of Digital Intelligence Hepatology (Ministry of Education/Beijing), School of Clinical Medicine, Tsinghua University, Beijing, 100084, China
| | - Yinpeng Le
- School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Hepato-Pancreato-Biliary Center, Tsinghua University, Beijing, 102218, China
- School of Materials Science and Engineering, Institute of Smart Biomedical Materials, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Wenting Pan
- Beijing International Science and Technology Cooperation Base for Antiviral Drugs, Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing, 100124, China
| | - Xinlong Yan
- Beijing International Science and Technology Cooperation Base for Antiviral Drugs, Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing, 100124, China.
| | - Yunfang Wang
- School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Hepato-Pancreato-Biliary Center, Tsinghua University, Beijing, 102218, China.
- School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Clinical Translational Science Center, Tsinghua University, Beijing, 102218, China.
- Key Laboratory of Digital Intelligence Hepatology (Ministry of Education/Beijing), School of Clinical Medicine, Tsinghua University, Beijing, 100084, China.
| | - Juan Liu
- School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Hepato-Pancreato-Biliary Center, Tsinghua University, Beijing, 102218, China.
- Key Laboratory of Digital Intelligence Hepatology (Ministry of Education/Beijing), School of Clinical Medicine, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
6
|
Wu TY, Hsieh YC, Yin WR, Cheng KY, Hou YT. Fabrication of a decellularized liver matrix-based hepatic patch for the repair of CCl4-induced liver injury. Biotechnol J 2024; 19:e2300570. [PMID: 38864387 DOI: 10.1002/biot.202300570] [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: 10/21/2023] [Revised: 04/23/2024] [Accepted: 05/16/2024] [Indexed: 06/13/2024]
Abstract
This article primarily introduces a new treatment for liver fibrosis/cirrhosis. We developed a hepatic patch by combining decellularized liver matrix (DLM) with the hepatocyte growth factor (HGF)/heparin-complex and evaluated its restorative efficacy. In vitro prophylactic results, the HGF/heparin-DLM patches effectively mitigated CCl4-induced hepatocyte toxicity and restored the cytotoxicity levels to the baseline levels by day 5. Furthermore, these patches restored albumin synthesis of injured hepatocytes to more than 70% of the normal levels within 5 days. In vitro therapeutic results, the urea synthesis of the injured hepatocytes reached 91% of the normal levels after 10 days of culture, indicating successful restoration of hepatic function by the HGF/heparin-DLM patches in both prophylactic and therapeutic models. In vivo results, HGF/heparin-DLM patches attached to the liver and gut exhibited a significant decrease in collagen content (4.44 times and 2.77 times, respectively) and an increase in glycogen content (1.19 times and 1.12 times, respectively) compared to the fibrosis group after 1 week, separately. In summary, liver function was restored and inflammation was inhibited through the combined effects of DLM and the HGF/heparin-complex in fibrotic liver. The newly designed hepatic patch holds promise for both in vitro and in vivo regeneration therapy and preventive health care for liver tissue engineering.
Collapse
Affiliation(s)
- Ting-Yi Wu
- Department of Biomechatronics Engineering, National Taiwan University, Taipei, Taiwan
| | - Yi-Cheng Hsieh
- Department of Biomechatronics Engineering, National Taiwan University, Taipei, Taiwan
| | - Wei-Rong Yin
- Department of Biomechatronics Engineering, National Taiwan University, Taipei, Taiwan
| | - Kai-Yi Cheng
- Department of Biomechatronics Engineering, National Taiwan University, Taipei, Taiwan
| | - Yung-Te Hou
- Department of Biomechatronics Engineering, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
7
|
Wang J, Jin X. Strategies for decellularization, re-cellularIzation and crosslinking in liver bioengineering. Int J Artif Organs 2024; 47:129-139. [PMID: 38253541 DOI: 10.1177/03913988231218566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Liver transplantation is the only definitive treatment for end-stage liver disease and its availability is restricted by organ donor shortages. The development of liver bioengineering provides the probability to create a functional alternative to reduce the gap in organ demand and supply. Decellularized liver scaffolds have been widely applied in bioengineering because they can mimic the native liver microenvironment and retain extracellular matrix (ECM) components. Multiple approaches including chemical, physical and biological methods have been developed for liver decellularization in current studies, but a full set of unified criteria has not yet been established. Each method has its advantages and drawbacks that influence the microstructure and ligand landscape of decellularized liver scaffolds. Optimizing a decellularization method to eliminate cell material while retaining as much of the ECM intact as possible is therefore important for biological scaffold applications. Furthermore, crosslinking strategies can improve the biological performance of scaffolds, including reinforcing biomechanics, delaying degradation in vivo and reducing immune rejection, which can better promote the integration of re-cellularized scaffolds with host tissue and influence the reconstruction process. In this review, we aim to present the different liver decellularization techniques, the crosslinking methods to improve scaffold characteristics with crosslinking and the preparation of soluble ECM.
Collapse
Affiliation(s)
- Jiajia Wang
- Department of Obstetrics and Gynecology, School of Clinical Medicine, Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Xiaojun Jin
- School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
8
|
Diedrich AM, Daneshgar A, Tang P, Klein O, Mohr A, Onwuegbuchulam OA, von Rueden S, Menck K, Bleckmann A, Juratli MA, Becker F, Sauer IM, Hillebrandt KH, Pascher A, Struecker B. Proteomic analysis of decellularized mice liver and kidney extracellular matrices. J Biol Eng 2024; 18:17. [PMID: 38389090 PMCID: PMC10885605 DOI: 10.1186/s13036-024-00413-8] [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: 10/01/2023] [Accepted: 02/07/2024] [Indexed: 02/24/2024] Open
Abstract
BACKGROUND The extracellular matrix (ECM) is a three-dimensional network of proteins that encases and supports cells within a tissue and promotes physiological and pathological cellular differentiation and functionality. Understanding the complex composition of the ECM is essential to decrypt physiological processes as well as pathogenesis. In this context, the method of decellularization is a useful technique to eliminate cellular components from tissues while preserving the majority of the structural and functional integrity of the ECM. RESULTS In this study, we employed a bottom-up proteomic approach to elucidate the intricate network of proteins in the decellularized extracellular matrices of murine liver and kidney tissues. This approach involved the use of a novel, perfusion-based decellularization protocol to generate acellular whole organ scaffolds. Proteomic analysis of decellularized mice liver and kidney ECM scaffolds revealed tissue-specific differences in matrisome composition, while we found a predominantly stable composition of the core matrisome, consisting of collagens, glycoproteins, and proteoglycans. Liver matrisome analysis revealed unique proteins such as collagen type VI alpha-6, fibrillin-2 or biglycan. In the kidney, specific ECM-regulators such as cathepsin z were detected. CONCLUSION The identification of distinct proteomic signatures provides insights into how different matrisome compositions might influence the biological properties of distinct tissues. This experimental workflow will help to further elucidate the proteomic landscape of decellularized extracellular matrix scaffolds of mice in order to decipher complex cell-matrix interactions and their contribution to a tissue-specific microenvironment.
Collapse
Affiliation(s)
- Anna-Maria Diedrich
- Department of General, Visceral, and Transplant Surgery, University Hospital Muenster, 48149, Muenster, Germany
| | - Assal Daneshgar
- Department of Surgery, Charité Mitte | Campus Virchow-Klinikum, Charité -Universitaetsmedizin Berlin, Campus, 13353, Berlin, Germany
- Berlin Institute of Health at Charité - Universitaetsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Clinician Scientist Program, Charitéplatz 1, 10117, Berlin, Germany
| | - Peter Tang
- Department of Surgery, Charité Mitte | Campus Virchow-Klinikum, Charité -Universitaetsmedizin Berlin, Campus, 13353, Berlin, Germany
| | - Oliver Klein
- Berlin Institute of Health at Charité - Universitaetsmedizin Berlin, Core Facility Imaging Mass Spectrometry, 13353, Berlin, Germany
| | - Annika Mohr
- Department of General, Visceral, and Transplant Surgery, University Hospital Muenster, 48149, Muenster, Germany
| | - Olachi A Onwuegbuchulam
- Department of General, Visceral, and Transplant Surgery, University Hospital Muenster, 48149, Muenster, Germany
| | - Sabine von Rueden
- Department of General, Visceral, and Transplant Surgery, University Hospital Muenster, 48149, Muenster, Germany
| | - Kerstin Menck
- Department of Medicine A for Hematology, Oncology, Hemostaseology and Pneumology, University Hospital Muenster, 48149, Muenster, Germany
- West German Cancer Center, University Hospital Muenster, 48149, Muenster, Germany
| | - Annalen Bleckmann
- Department of Medicine A for Hematology, Oncology, Hemostaseology and Pneumology, University Hospital Muenster, 48149, Muenster, Germany
- West German Cancer Center, University Hospital Muenster, 48149, Muenster, Germany
| | - Mazen A Juratli
- Department of General, Visceral, and Transplant Surgery, University Hospital Muenster, 48149, Muenster, Germany
- West German Cancer Center, University Hospital Muenster, 48149, Muenster, Germany
| | - Felix Becker
- Department of General, Visceral, and Transplant Surgery, University Hospital Muenster, 48149, Muenster, Germany
- West German Cancer Center, University Hospital Muenster, 48149, Muenster, Germany
| | - Igor M Sauer
- Department of Surgery, Charité Mitte | Campus Virchow-Klinikum, Charité -Universitaetsmedizin Berlin, Campus, 13353, Berlin, Germany
| | - Karl H Hillebrandt
- Department of Surgery, Charité Mitte | Campus Virchow-Klinikum, Charité -Universitaetsmedizin Berlin, Campus, 13353, Berlin, Germany
- Berlin Institute of Health at Charité - Universitaetsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Clinician Scientist Program, Charitéplatz 1, 10117, Berlin, Germany
| | - Andreas Pascher
- Department of General, Visceral, and Transplant Surgery, University Hospital Muenster, 48149, Muenster, Germany
- West German Cancer Center, University Hospital Muenster, 48149, Muenster, Germany
| | - Benjamin Struecker
- Department of General, Visceral, and Transplant Surgery, University Hospital Muenster, 48149, Muenster, Germany.
- West German Cancer Center, University Hospital Muenster, 48149, Muenster, Germany.
| |
Collapse
|
9
|
Cardinale V, Lanthier N, Baptista PM, Carpino G, Carnevale G, Orlando G, Angelico R, Manzia TM, Schuppan D, Pinzani M, Alvaro D, Ciccocioppo R, Uygun BE. Cell transplantation-based regenerative medicine in liver diseases. Stem Cell Reports 2023; 18:1555-1572. [PMID: 37557073 PMCID: PMC10444572 DOI: 10.1016/j.stemcr.2023.06.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 06/11/2023] [Accepted: 06/12/2023] [Indexed: 08/11/2023] Open
Abstract
This review aims to evaluate the current preclinical state of liver bioengineering, the clinical context for liver cell therapies, the cell sources, the delivery routes, and the results of clinical trials for end-stage liver disease. Different clinical settings, such as inborn errors of metabolism, acute liver failure, chronic liver disease, liver cirrhosis, and acute-on-chronic liver failure, as well as multiple cellular sources were analyzed; namely, hepatocytes, hepatic progenitor cells, biliary tree stem/progenitor cells, mesenchymal stromal cells, and macrophages. The highly heterogeneous clinical scenario of liver disease and the availability of multiple cellular sources endowed with different biological properties make this a multidisciplinary translational research challenge. Data on each individual liver disease and more accurate endpoints are urgently needed, together with a characterization of the regenerative pathways leading to potential therapeutic benefit. Here, we critically review these topics and identify related research needs and perspectives in preclinical and clinical settings.
Collapse
Affiliation(s)
- Vincenzo Cardinale
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy.
| | - Nicolas Lanthier
- Service d'Hépato-gastroentérologie, Cliniques Universitaires Saint-Luc, Laboratory of Hepatogastroenterology, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Pedro M Baptista
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), Zaragoza, Spain; Centro de Investigación Biomédica en Red en el Área Temática de Enfermedades Hepáticas (CIBERehd), Madrid, Spain; Fundación ARAID, Zaragoza, Spain; Department of Biomedical and Aerospace Engineering, Universidad Carlos III de Madrid, Madrid, Spain
| | - Guido Carpino
- Department of Anatomical, Histological, Forensic Medicine and Orthopedic Sciences, Sapienza University of Rome, Italy
| | - Gianluca Carnevale
- Department of Surgery, Medicine, Dentistry, and Morphological Sciences with Interest in Transplant, Oncology, and Regenerative Medicine, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Giuseppe Orlando
- Section of Transplantation, Department of Surgery, Wake Forest University School of Medicine, Winston Salem, NC, USA
| | - Roberta Angelico
- Hepatobiliary Surgery and Transplant Unit, Department of Surgical Sciences, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Tommaso Maria Manzia
- Hepatobiliary Surgery and Transplant Unit, Department of Surgical Sciences, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Detlef Schuppan
- Institute of Translational Immunology, Research Center for Immune Therapy, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany; Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Massimo Pinzani
- UCL Institute for Liver and Digestive Health, Division of Medicine, Royal Free Hospital, London, UK
| | - Domenico Alvaro
- Department of Translation and Precision Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Rachele Ciccocioppo
- Gastroenterology Unit, Department of Medicine, A.O.U.I. Policlinico G.B. Rossi & University of Verona, Verona, Italy.
| | - Basak E Uygun
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Harvard Medical School, Shriners Hospitals for Children, Boston, MA 02114, USA; Department of Surgery, Massachusetts General Hospital, Boston, MA 02114, USA.
| |
Collapse
|
10
|
Hoffman ET, Uriarte JJ, Uhl FE, Eckstrom K, Tanneberger AE, Becker C, Moulin C, Asarian L, Ikonomou L, Kotton DN, Weiss DJ. Human alveolar hydrogels promote morphological and transcriptional differentiation in iPSC-derived alveolar type 2 epithelial cells. Sci Rep 2023; 13:12057. [PMID: 37491483 PMCID: PMC10368739 DOI: 10.1038/s41598-023-37685-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 06/26/2023] [Indexed: 07/27/2023] Open
Abstract
Alveolar type 2 epithelial cells (AT2s) derived from human induced pluripotent stem cells (iAT2s) have rapidly contributed to our understanding of AT2 function and disease. However, while iAT2s are primarily cultured in three-dimensional (3D) Matrigel, a matrix derived from cancerous mouse tissue, it is unclear how a physiologically relevant matrix will impact iAT2s phenotype. As extracellular matrix (ECM) is recognized as a vital component in directing cellular function and differentiation, we sought to derive hydrogels from decellularized human lung alveolar-enriched ECM (aECM) to provide an ex vivo model to characterize the role of physiologically relevant ECM on iAT2 phenotype. We demonstrate aECM hydrogels retain critical in situ ECM components, including structural and basement membrane proteins. While aECM hydrogels facilitate iAT2 proliferation and alveolosphere formation, a subset of iAT2s rapidly change morphology to thin and elongated ring-like cells. This morphological change correlates with upregulation of recently described iAT2-derived transitional cell state genetic markers. As such, we demonstrate a potentially underappreciated role of physiologically relevant aECM in iAT2 differentiation.
Collapse
Affiliation(s)
- Evan T Hoffman
- Department of Medicine, Larner College of Medicine, University of Vermont, 149 Beaumont Avenue, Burlington, VT, 05405, USA
| | - Juan J Uriarte
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, 40506, USA
| | - Franziska E Uhl
- Department of Experimental Medical Science, Lund University, Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - Korin Eckstrom
- Department of Microbiology and Molecular Genetics, Larner College of Medicine, University of Vermont, Burlington, VT, 05405, USA
| | - Alicia E Tanneberger
- Department of Medicine, Larner College of Medicine, University of Vermont, 149 Beaumont Avenue, Burlington, VT, 05405, USA
| | - Chloe Becker
- Department of Medicine, Larner College of Medicine, University of Vermont, 149 Beaumont Avenue, Burlington, VT, 05405, USA
| | - Chloe Moulin
- Department of Medicine, Larner College of Medicine, University of Vermont, 149 Beaumont Avenue, Burlington, VT, 05405, USA
| | - Loredana Asarian
- Department of Medicine, Larner College of Medicine, University of Vermont, 149 Beaumont Avenue, Burlington, VT, 05405, USA
| | - Laertis Ikonomou
- Department of Oral Biology, University of Buffalo, The State University of New York, Buffalo, NY, 14260, USA
- Cell, Gene and Tissue Engineering Center, University at Buffalo, The State University of New York, Buffalo, NY, USA
| | - Darrell N Kotton
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA, 02118, USA
| | - Daniel J Weiss
- Department of Medicine, Larner College of Medicine, University of Vermont, 149 Beaumont Avenue, Burlington, VT, 05405, USA.
| |
Collapse
|
11
|
Zhang Q, Chiu Y, Chen Y, Wu Y, Dunne LW, Largo RD, Chang EI, Adelman DM, Schaverien MV, Butler CE. Harnessing the synergy of perfusable muscle flap matrix and adipose-derived stem cells for prevascularization and macrophage polarization to reconstruct volumetric muscle loss. Bioact Mater 2023; 22:588-614. [PMID: 36382023 PMCID: PMC9646752 DOI: 10.1016/j.bioactmat.2022.10.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 10/09/2022] [Accepted: 10/21/2022] [Indexed: 11/06/2022] Open
Abstract
Muscle flaps must have a strong vascular network to support a large tissue volume and ensure successful engraftment. We developed porcine stomach musculofascial flap matrix (PDSF) comprising extracellular matrix (ECM) and intact vasculature. PDSF had a dominant vascular pedicle, microcirculatory vessels, a nerve network, well-retained 3-dimensional (3D) nanofibrous ECM structures, and no allo- or xenoantigenicity. In-depth proteomic analysis demonstrated that PDSF was composed of core matrisome proteins (e.g., collagens, glycoproteins, proteoglycans, and ECM regulators) that, as shown by Gene Ontology term enrichment analysis, are functionally related to musculofascial biological processes. Moreover, PDSF-human adipose-derived stem cell (hASC) synergy not only induced monocytes towards IL-10-producing M2 macrophage polarization through the enhancement of hASCs' paracrine effect but also promoted the proliferation and interconnection of both human skeletal muscle myoblasts (HSMMs) and human umbilical vein endothelial cells (HUVECs) in static triculture conditions. Furthermore, PDSF was successfully prevascularized through a dynamic perfusion coculture of hASCs and HUVECs, which integrated with PDSF and induced the maturation of vascular networks in vitro. In a xenotransplantation model, PDSF demonstrated myoconductive and immunomodulatory properties associated with the predominance of M2 macrophages and regulatory T cells. In a volumetric muscle loss (VML) model, prevascularized PDSF augmented neovascularization and constructive remodeling, which was characterized by the predominant infiltration of M2 macrophages and significant musculofascial tissue formation. These results indicate that hASCs' integration with PDSF enhances the cells' dual function in immunomodulation and angiogenesis. Owing in part to this PDSF-hASC synergy, our platform shows promise for vascularized muscle flap engineering for VML reconstruction.
Collapse
Affiliation(s)
- Qixu Zhang
- Department of Plastic Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yulun Chiu
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Youbai Chen
- Department of Plastic Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Department of Plastic Surgery, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yewen Wu
- Department of Plastic Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Lina W. Dunne
- Department of Plastic Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Rene D. Largo
- Department of Plastic Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Edward I. Chang
- Department of Plastic Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - David M. Adelman
- Department of Plastic Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Mark V. Schaverien
- Department of Plastic Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Charles E. Butler
- Department of Plastic Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| |
Collapse
|
12
|
Hoffman ET, Uhl FE, Asarian L, Deng B, Becker C, Uriarte JJ, Downs I, Young B, Weiss DJ. Regional and disease specific human lung extracellular matrix composition. Biomaterials 2023; 293:121960. [PMID: 36580718 PMCID: PMC9868084 DOI: 10.1016/j.biomaterials.2022.121960] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 11/25/2022] [Accepted: 12/13/2022] [Indexed: 12/25/2022]
Abstract
Chronic lung diseases, such as chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF), are characterized by regional extracellular matrix (ECM) remodeling which contributes to disease progression. Previous proteomic studies on whole decellularized lungs have provided detailed characterization on the impact of COPD and IPF on total lung ECM composition. However, such studies are unable to determine the differences in ECM composition between individual anatomical regions of the lung. Here, we employ a post-decellularization dissection method to compare the ECM composition of whole decellularized lungs (wECM) and specific anatomical lung regions, including alveolar-enriched ECM (aECM), airway ECM (airECM), and vasculature ECM (vECM), between non-diseased (ND), COPD, and IPF human lungs. We demonstrate, using mass spectrometry, that individual regions possess a unique ECM signature characterized primarily by differences in collagen composition and basement-membrane associated proteins, including ECM glycoproteins. We further demonstrate that both COPD and IPF lead to alterations in lung ECM composition in a region-specific manner, including enrichment of type-III collagen and fibulin in IPF aECM. Taken together, this study provides methodology for future studies, including isolation of region-specific lung biomaterials, as well as a dataset that may be applied for the identification of novel ECM targets for therapeutics.
Collapse
Affiliation(s)
- Evan T. Hoffman
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, 05405, USA
| | - Franziska E. Uhl
- Department of Experimental Medical Science, Lund University, Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - Loredana Asarian
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, 05405, USA
| | - Bin Deng
- Department of Biology, University of Vermont, Burlington, VT, 05405, USA
| | - Chloe Becker
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, 05405, USA
| | - Juan J. Uriarte
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, 40506, USA
| | - Isaac Downs
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, 05405, USA
| | - Brad Young
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, 05405, USA
| | - Daniel J. Weiss
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, 05405, USA
| |
Collapse
|
13
|
Yuan Y. Clinical Translation of Engineered Pulmonary Vascular Models. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1413:273-288. [PMID: 37195536 DOI: 10.1007/978-3-031-26625-6_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Diseases in pulmonary vasculature remain a major cause of morbidity and mortality worldwide. Numerous pre-clinical animal models were developed to understand lung vasculature during diseases and development. However, these systems are typically limited in their ability to represent human pathophysiology for the study of disease and drug mechanisms. In recent years, a growing number of studies have focused on developing in vitro experimental platforms that mimic human tissues/organs. In this chapter, we discuss the key components involved in developing engineered pulmonary vascular modeling systems and provide perspectives on ways to improve the translational potential of existing models.
Collapse
Affiliation(s)
- Yifan Yuan
- Department of Medicine (Pulmonary), Department of Anesthesiology, Yale University, New Haven, CT, USA.
| |
Collapse
|
14
|
Wang H, Sun WQ, Wang J. Complete proteomic profiling of regenerative bio-scaffolds with a two-step trypsinization method. J Biomed Mater Res B Appl Biomater 2023; 111:62-72. [PMID: 35822935 DOI: 10.1002/jbm.b.35132] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 06/20/2022] [Accepted: 06/28/2022] [Indexed: 01/01/2023]
Abstract
Regenerative bio-scaffolds, widely used for clinical tissue reconstruction and tissue repairs, are functionally diversified and structurally complex decellularized tissue materials (e.g., extracellular matrix, ECM). ECM is naturally cross-linked and can be further selectively cross-linked upon processing. Identification, quantification and bioinformatics functional comparison of all ECM proteins are challenging for regenerative bio-scaffolds. In this study, we have applied proteomic profiling with a two-step sequential trypsinization method, and identified and quantified 300-400 constituent proteins in three commercially available regenerative bio-scaffolds (BioDesign Surgisis, ReGen tissue matrix, and ThormalGEN mesh). These proteins were classified into four categories and 14 subcategories based on their mainly biological function. The main components of regenerative bio-scaffolds were highly abundant ECM structural proteins, and the minor parts of bio-scaffolds were lowly abundant, less cross-linked, functionally more diversified proteins, especially extracellular fluid proteins that were easily solubilized by trypsin. The comparative analysis has revealed large differences in the number, type, abundance and function of identified proteins, as well as the extent of decellularization and cross-linking among regenerative bio-scaffolds. So, the proteomic profiling with a two-step sequential trypsinization method could not only provide the molecular basis to better understand the degradation process of regenerative bio-scaffolds in vivo and different clinical outcomes among various regenerative bio-scaffolds, facilitate the exploration of the response mechanisms in the host's early clinical stages of ECM-induced tissue regeneration that is still poorly understood, but also can be used for optimization of the decellularization and cross-linking process, product characterization and rational design of new ECM products.
Collapse
Affiliation(s)
- Huidan Wang
- Institute of Biothermal Science and Technology, School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Wendell Q Sun
- Institute of Biothermal Science and Technology, School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Jian Wang
- National Institutes for Food and Drug Control, Beijing, China
| |
Collapse
|
15
|
Pien N, Bray F, Gheysens T, Tytgat L, Rolando C, Mantovani D, Dubruel P, Vlierberghe SV. Proteomics as a tool to gain next level insights into photo-crosslinkable biopolymer modifications. Bioact Mater 2022; 17:204-220. [PMID: 35386456 PMCID: PMC8965084 DOI: 10.1016/j.bioactmat.2022.01.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 01/04/2022] [Accepted: 01/16/2022] [Indexed: 12/25/2022] Open
Abstract
The distribution of photo-crosslinkable moieties onto a protein backbone can affect a biomaterial's crosslinking behavior, and therefore also its mechanical and biological properties. A profound insight in this respect is essential for biomaterials exploited in tissue engineering and regenerative medicine. In the present work, photo-crosslinkable moieties have been introduced on the primary amine groups of: (i) a recombinant collagen peptide (RCPhC1) with a known amino acid (AA) sequence, and (ii) bovine skin collagen (COL BS) with an unknown AA sequence. The degree of substitution (DS) was quantified with two conventional techniques: an ortho-phthalic dialdehyde (OPA) assay and 1H NMR spectroscopy. However, neither of both provides information on the exact type and location of the modified AAs. Therefore, for the first time, proteomic analysis was evaluated herein as a tool to identify functionalized AAs as well as the exact position of photo-crosslinkable moieties along the AA sequence, thereby enabling an in-depth, unprecedented characterization of functionalized photo-crosslinkable biopolymers. Moreover, our strategy enabled to visualize the spatial distribution of the modifications within the overall structure of the protein. Proteomics has proven to provide unprecedented insight in the distribution of photo-crosslinkable moieties along the protein backbone, undoubtedly contributing to superior functional biomaterial design to serve regenerative medicine.
Collapse
Affiliation(s)
- Nele Pien
- Ghent University, Polymer Chemistry and Biomaterials Research Group, Centre of Macromolecular Chemistry (CMaC), Krijgslaan 281 S4bis, 9000 Gent, Belgium
- Laval University, Laboratory for Biomaterials and Bioengineering, CRC-I, Pavillon Pouliot, Québec G1V 0A6, Canada
| | - Fabrice Bray
- Univ. Lille, CNRS, USR 3290, MSAP, Miniaturisation pour la Synthèse l'Analyse et la Protéomique, F-59 000, Lille, France
| | - Tom Gheysens
- Ghent University, Polymer Chemistry and Biomaterials Research Group, Centre of Macromolecular Chemistry (CMaC), Krijgslaan 281 S4bis, 9000 Gent, Belgium
| | - Liesbeth Tytgat
- Ghent University, Polymer Chemistry and Biomaterials Research Group, Centre of Macromolecular Chemistry (CMaC), Krijgslaan 281 S4bis, 9000 Gent, Belgium
| | - Christian Rolando
- Univ. Lille, CNRS, USR 3290, MSAP, Miniaturisation pour la Synthèse l'Analyse et la Protéomique, F-59 000, Lille, France
| | - Diego Mantovani
- Laval University, Laboratory for Biomaterials and Bioengineering, CRC-I, Pavillon Pouliot, Québec G1V 0A6, Canada
| | - Peter Dubruel
- Ghent University, Polymer Chemistry and Biomaterials Research Group, Centre of Macromolecular Chemistry (CMaC), Krijgslaan 281 S4bis, 9000 Gent, Belgium
| | - Sandra Van Vlierberghe
- Ghent University, Polymer Chemistry and Biomaterials Research Group, Centre of Macromolecular Chemistry (CMaC), Krijgslaan 281 S4bis, 9000 Gent, Belgium
| |
Collapse
|
16
|
Dong Y, Zhang C, Zhang Q, Li Z, Wang Y, Yan J, Wu G, Qiu L, Zhu Z, Wang B, Gu H, Zhang Y. Identification of nanoparticle-mediated siRNA-ASPN as a key gene target in the treatment of keloids. Front Bioeng Biotechnol 2022; 10:1025546. [PMID: 36394011 PMCID: PMC9649824 DOI: 10.3389/fbioe.2022.1025546] [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: 08/23/2022] [Accepted: 10/17/2022] [Indexed: 12/03/2022] Open
Abstract
Background: Keloid, also known as connective tissue hyperplasia, is a benign proliferative disorder with a global distribution. The available therapeutic interventions are steroid injections, surgical removal of keloids, radiotherapy, compression therapy, the application of cryosurgery, and many other methods. Objectives: Existing treatments or approaches for keloids may lead to similar or even larger lesions at the site of keloid excision, leading to a high recurrence rate. Therefore, this study aims at identifying a new gene-based therapy for the treatment of keloids. Methods: An ASPN-siRNA/nanoparticle combination (si-ASPN) and a negative siRNA/nanoparticle complex (NC) was developed on the basis of bioinformatics studies and used in vitro and in vivo experiments. Results: The results showed a strong correlation between the development of keloids and high expression of ASPN protein. With the expression of ASPN protein greatly reduced in keloid fibroblasts and nude mice allografts after treatment with si-ASPN, the collagen and fibroblasts were also uniform, thinner, parallel and regular. Conclusion: All the above experimental results suggest that keloid and ASPN are closely related and both fibroblast growth and metabolism of keloid are inhibited after silencing ASPN. Therefore, ASPN-siRNA delivered via nanoparticles can serve as a novel intervention therapy for the treatment of keloids.
Collapse
Affiliation(s)
- Yipeng Dong
- Department of Burns and Plastic Surgery, Affiliated Hospital of Nantong University, Nantong, China
- Medical School of Nantong University, Nantong, China
| | - Chuwei Zhang
- Department of Burns and Plastic Surgery, Affiliated Hospital of Nantong University, Nantong, China
- Medical School of Nantong University, Nantong, China
| | - Qingrong Zhang
- Department of Burns and Plastic Surgery, Affiliated Hospital of Nantong University, Nantong, China
- Institute of Burn Research, Third Military Medical University (Army Medical University), Chongqing, China
| | - Zihan Li
- Department of Burns and Plastic Surgery, Affiliated Hospital of Nantong University, Nantong, China
- Medical School of Nantong University, Nantong, China
| | - Yixiao Wang
- Medical School of Nantong University, Nantong, China
| | - Jun Yan
- Department of Burns and Plastic Surgery, Affiliated Hospital of Nantong University, Nantong, China
- Medical School of Nantong University, Nantong, China
| | - Gujie Wu
- Medical School of Nantong University, Nantong, China
| | - Ling Qiu
- Department of Burns and Plastic Surgery, Affiliated Hospital of Nantong University, Nantong, China
- Medical School of Nantong University, Nantong, China
| | - Zhihan Zhu
- Department of Burns and Plastic Surgery, Affiliated Hospital of Nantong University, Nantong, China
- Medical School of Nantong University, Nantong, China
| | - Bolin Wang
- Department of Burns and Plastic Surgery, Affiliated Hospital of Nantong University, Nantong, China
- Medical School of Nantong University, Nantong, China
| | - Haiying Gu
- Institute of Analytical Chemistry for Life Science, Nantong University, Nantong, China
- School of Public Health, Nantong University, Nantong, China
- *Correspondence: Yi Zhang, ; Haiying Gu,
| | - Yi Zhang
- Department of Burns and Plastic Surgery, Affiliated Hospital of Nantong University, Nantong, China
- *Correspondence: Yi Zhang, ; Haiying Gu,
| |
Collapse
|
17
|
Talaei-Khozani T, Yaghoubi A. An overview of post transplantation events of decellularized scaffolds. Transpl Immunol 2022; 74:101640. [PMID: 35667545 DOI: 10.1016/j.trim.2022.101640] [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: 03/23/2022] [Revised: 05/29/2022] [Accepted: 05/31/2022] [Indexed: 12/19/2022]
Abstract
Regenerative medicine and tissue engineering are reasonable techniques for repairing failed tissues and could be a suitable alternative to organ transplantation. One of the most widely used methods for preparing bioscaffolds is the decellularization procedure. Although cell debris and DNA are removed from the decellularized tissues, important compositions of the extracellular matrix including proteins, proteoglycans, and glycoproteins are nearly preserved. Moreover, the obtained scaffolds have a 3-dimensional (3D) structure, appropriate naïve mechanical properties, and good biocompatibility. After transplantation, different types of host cells migrate to the decellularized tissues. Histological and immunohistochemical assessment of the different bioscaffolds after implantation reveals the migration of parenchymal cells, angiogenesis, as well as the invasion of inflammatory and giant foreign cells. In this review, the events after transplantation including angiogenesis, scaffold degradation, and the presence of immune and tissue-specific progenitor cells in the decellularized scaffolds in various hosts, are discussed.
Collapse
Affiliation(s)
- Tahereh Talaei-Khozani
- Histotomorphometry and stereology research center, Shiraz University of Medical Sciences, Shiraz, Iran; Tissue engineering lab, Anatomy Department, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Atefeh Yaghoubi
- Tissue engineering lab, Anatomy Department, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
18
|
Design by Nature: Emerging Applications of Native Liver Extracellular Matrix for Cholangiocyte Organoid-Based Regenerative Medicine. Bioengineering (Basel) 2022; 9:bioengineering9030110. [PMID: 35324799 PMCID: PMC8945468 DOI: 10.3390/bioengineering9030110] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/25/2022] [Accepted: 03/04/2022] [Indexed: 12/14/2022] Open
Abstract
Organoid technology holds great promise for regenerative medicine. Recent studies show feasibility for bile duct tissue repair in humans by successfully transplanting cholangiocyte organoids in liver grafts during perfusion. Large-scale expansion of cholangiocytes is essential for extending these regenerative medicine applications. Human cholangiocyte organoids have a high and stable proliferation capacity, making them an attractive source of cholangiocytes. Commercially available basement membrane extract (BME) is used to expand the organoids. BME allows the cells to self-organize into 3D structures and stimulates cell proliferation. However, the use of BME is limiting the clinical applications of the organoids. There is a need for alternative tissue-specific and clinically relevant culture substrates capable of supporting organoid proliferation. Hydrogels prepared from decellularized and solubilized native livers are an attractive alternative for BME. These hydrogels can be used for the culture and expansion of cholangiocyte organoids in a clinically relevant manner. Moreover, the liver-derived hydrogels retain tissue-specific aspects of the extracellular microenvironment. They are composed of a complex mixture of bioactive and biodegradable extracellular matrix (ECM) components and can support the growth of various hepatobiliary cells. In this review, we provide an overview of the clinical potential of native liver ECM-based hydrogels for applications with human cholangiocyte organoids. We discuss the current limitations of BME for the clinical applications of organoids and how native ECM hydrogels can potentially overcome these problems in an effort to unlock the full regenerative clinical potential of the organoids.
Collapse
|
19
|
Hsieh YC, Yin WR, Xu YY, Hou YT. HGF/heparin-immobilized decellularized liver matrices as novel hepatic patches for hepatocyte regeneration in an acute liver injury model. Biochem Eng J 2022. [DOI: 10.1016/j.bej.2022.108354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
20
|
Wan J, Wu T, Liu Y, Yang M, Fichna J, Guo Y, Yin L, Chen C. Mast Cells Tryptase Promotes Intestinal Fibrosis in Natural Decellularized Intestinal Scaffolds. Tissue Eng Regen Med 2022; 19:717-726. [PMID: 35218507 PMCID: PMC9294124 DOI: 10.1007/s13770-022-00433-9] [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: 09/13/2021] [Revised: 12/18/2021] [Accepted: 01/08/2022] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Standard two-dimensional (2D) culture has confirmed the mechanism of mast cells (MCs) in the pathogenesis of inflammatory bowel disease (IBD), but the regulation of signaling responses of MCs may well differ in three-dimensional (3D) microenvironments. The aim of the study was to develop a 3D culture model based on decellularized intestinal scaffolds (DIS) and verify how MCs influenced fibroblasts phenotype in the 3D model. METHODS DIS were achieved using the detergent technique and extracellular matrix (ECM) components were verified by histologic analysis, quantification and scanning electron microscope. After human colon fibroblasts recellularized into the scaffolds and activated by MCs tryptase and TGFβ1, the changes in genes and signaling pathways during fibroblasts activation in 3D were studied and compared with the changes in 2D cell culture on plastic plates. RESULTS Decellularization process effectively removed native cell debris while retaining natural ECM components and structure. The engrafted fibroblasts could penetrate into the scaffolds and maintain its phenotype. No matter whether fibroblasts were cultured in 2D or 3D, MCs tryptase and transforming growth factor β1 (TGF-β1) could promote the differentiation of fibroblasts into fibrotic-phenotype myofibroblasts through Akt and Smad2/3 signaling pathways. Furthermore, the pro-collagen1α1 and fibronectin synthesis of myofibroblasts in 3D was higher than in 2D culture. CONCLUSION Our results demonstrated that the DIS can be used as a bioactive microenvironment for the study of intestinal fibrosis, providing an innovative platform for future intestinal disease modeling and screening of genes and signaling pathways.
Collapse
Affiliation(s)
- Jian Wan
- Center for Difficult and Complicated Abdominal Surgery, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, 200072 China
| | - Tianqi Wu
- Center for Difficult and Complicated Abdominal Surgery, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, 200072 China
| | - Ying Liu
- Department of General Surgery, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, 200072 China
| | - Muqing Yang
- Center for Difficult and Complicated Abdominal Surgery, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, 200072 China
| | - Jakub Fichna
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 6/8, 92-215 Lodz, Poland
| | - Yibing Guo
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, 226000 China
| | - Lu Yin
- Center for Difficult and Complicated Abdominal Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China.
| | - Chunqiu Chen
- Center for Difficult and Complicated Abdominal Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China.
| |
Collapse
|
21
|
Xue W, Kong Y, Abu R, Roy P, Huh SH, Kuss M, Kumar V, Duan B. Regulation of Schwann Cell and DRG Neurite Behaviors within Decellularized Peripheral Nerve Matrix. ACS APPLIED MATERIALS & INTERFACES 2022; 14:8693-8704. [PMID: 35148064 DOI: 10.1021/acsami.1c20320] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Decellularized nerve hydrogels (dNHs) containing bioactive molecules are promising biomaterials for peripheral nerve injury (PNI) treatment and have been extensively applied in clinical and preclinical practice. However, most previous research projects studied their influences on nerve-related cellular behaviors in two dimensions (2D) without taking hydrogel biomechanics into consideration. The molecular mechanisms underlying the beneficial microenvironment provided by dNHs also remain unclear. In this study, dNHs from rat sciatic nerves were prepared, and their effects on Schwann cell (SC) and dorsal root ganglion (DRG) neurite behaviors were evaluated and compared to commercial rat tail type I collagen (Col) hydrogels in three-dimensional (3D) environments. We found that dNHs could promote SC proliferation and neurite outgrowth, and both the hydrogel mechanics and components contributed to the dNH functionalization. Through proteomics analysis, we found that laminin (LAM) and type V collagen (COLV) exclusively and abundantly existed in dNHs. By adding exogenous LAM and COLV into Col hydrogels, we demonstrated that they regulated SC gene expression and that LAM could promote SC spreading and neurite outgrowth, while COLV improved SC proliferation. Lastly, dNHs were fabricated into paper-like, aligned nerve scaffolds through unidirectional freezing to expand the dNH applications in PNI treatment.
Collapse
Affiliation(s)
- Wen Xue
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
- Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Yunfan Kong
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
- Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Rafay Abu
- Mass Spectrometry & Proteomics Core, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Pooja Roy
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Sung-Ho Huh
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Mitchell Kuss
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
- Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Vikas Kumar
- Mass Spectrometry & Proteomics Core, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Bin Duan
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
- Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
- Department of Mechanical and Materials Engineering, University of Nebraska─Lincoln, Lincoln, Nebraska 68588, United States
- Department of Surgery, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| |
Collapse
|
22
|
Zhu X, Wu Q, He Y, Gao M, Li Y, Peng W, Li S, Liu Y, Zhang R, Bao J. Fabrication of Size-Controllable and Arrangement-Orderly HepG2 Spheroids for Drug Screening via Decellularized Liver Matrix-Derived Micropattern Array Chips. ACS OMEGA 2022; 7:2364-2376. [PMID: 35071924 PMCID: PMC8772313 DOI: 10.1021/acsomega.1c06302] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 12/20/2021] [Indexed: 02/08/2023]
Abstract
![]()
Three-dimensional
(3D) culture via micropattern arrays to generate
cellular spheroids seems a promising in vitro biomimetic
system for liver tissue engineering applications, such as drug screening.
Recently, organ-derived decellularized extracellular matrix emerges
as arguably the most biomimetic bioink. Herein, decellularized liver
matrix (DLM)-derived micropattern array chips were developed to fabricate
size-controllable and arrangement-orderly HepG2 spheroids for drug
screening. The porcine DLM was obtained by the removal of cellular
components and then ground into powder, followed by enzymolysis. DLM
as a coating substrate was compared with collagen type I (Col I) and
Matrigel in terms of biological performance for enhancing cell adhesion,
proliferation, and functions. Subsequently, we used poly(dimethylsiloxane)
(PDMS) to adsorb DLM as the bioink to fabricate micropattern array
chips. The optimal shape and size of micropattern were determined
by evaluating the morphology, viability, and functions of HepG2 3D
cellular aggregates. In addition, drug-susceptibility testing (paclitaxel,
doxorubicin HCl, and disulfiram) was performed on this novel platform.
The DLM provided the tissue-specific microenvironment that provided
suitable supports for HepG2 cells, compared to Col I and Matrigel.
A circular micropattern with a diameter of 100 μm was the optimal
processing parameter to rapidly fabricate large-scale, size-controllable,
and arrangement-orderly HepG2 cellular aggregates with 3D spheroid’s
shape and high cell viability. Drug screening testing showed that
the effect of a drug could be directly demonstrated on-chip by confocal
microscopy measuring the viability of spheroids. We provide a novel
platform for the large-scale generation of HepG2 spheroids with uniform
size and arrangement, thus bringing convenience, reducing error, and
increasing reproducibility for a rapid drug discovery by fluorescence
quantitative analysis. This methodology may be possible to apply in
advancing personalized medicine and drug discovery.
Collapse
Affiliation(s)
- Xinglong Zhu
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Qiong Wu
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China.,Laboratory of Liver Transplantation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Yuting He
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Mengyu Gao
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China.,Department of Pathology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Yi Li
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China.,Precision Medicine Key Laboratory, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wanliu Peng
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Shengfu Li
- Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Yong Liu
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Rundong Zhang
- West China School of Medicine, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Ji Bao
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| |
Collapse
|
23
|
Yuan Y, Leiby KL, Greaney AM, Raredon MSB, Qian H, Schupp JC, Engler AJ, Baevova P, Adams TS, Kural MH, Wang J, Obata T, Yoder MC, Kaminski N, Niklason LE. A Pulmonary Vascular Model From Endothelialized Whole Organ Scaffolds. Front Bioeng Biotechnol 2021; 9:760309. [PMID: 34869270 PMCID: PMC8640093 DOI: 10.3389/fbioe.2021.760309] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 10/27/2021] [Indexed: 12/12/2022] Open
Abstract
The development of an in vitro system for the study of lung vascular disease is critical to understanding human pathologies. Conventional culture systems fail to fully recapitulate native microenvironmental conditions and are typically limited in their ability to represent human pathophysiology for the study of disease and drug mechanisms. Whole organ decellularization provides a means to developing a construct that recapitulates structural, mechanical, and biological features of a complete vascular structure. Here, we developed a culture protocol to improve endothelial cell coverage in whole lung scaffolds and used single-cell RNA-sequencing analysis to explore the impact of decellularized whole lung scaffolds on endothelial phenotypes and functions in a biomimetic bioreactor system. Intriguingly, we found that the phenotype and functional signals of primary pulmonary microvascular revert back—at least partially—toward native lung endothelium. Additionally, human induced pluripotent stem cell-derived endothelium cultured in decellularized lung systems start to gain various native human endothelial phenotypes. Vascular barrier function was partially restored, while small capillaries remained patent in endothelial cell-repopulated lungs. To evaluate the ability of the engineered endothelium to modulate permeability in response to exogenous stimuli, lipopolysaccharide (LPS) was introduced into repopulated lungs to simulate acute lung injury. After LPS treatment, proinflammatory signals were significantly increased and the vascular barrier was impaired. Taken together, these results demonstrate a novel platform that recapitulates some pulmonary microvascular functions and phenotypes at a whole organ level. This development may help pave the way for using the whole organ engineering approach to model vascular diseases.
Collapse
Affiliation(s)
- Yifan Yuan
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, United States.,Department of Anesthesiology, Yale University, New Haven, CT, United States
| | - Katherine L Leiby
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, United States.,Department of Biomedical Engineering, Yale University, New Haven, CT, United States.,Medical Scientist Training Program, Yale University, New Haven, CT, United States
| | - Allison M Greaney
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, United States.,Department of Biomedical Engineering, Yale University, New Haven, CT, United States
| | - Micha Sam Brickman Raredon
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, United States.,Department of Biomedical Engineering, Yale University, New Haven, CT, United States.,Medical Scientist Training Program, Yale University, New Haven, CT, United States
| | - Hong Qian
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, United States.,Department of Anesthesiology, Yale University, New Haven, CT, United States
| | - Jonas C Schupp
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, New Haven, CT, United States.,Department of Respiratory Medicine, Hannover Medical School and Biomedical Research in End-stage and Obstructive Lung Disease Hannover, German Lung Research Center (DZL), Hannover, Germany
| | - Alexander J Engler
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, United States.,Department of Biomedical Engineering, Yale University, New Haven, CT, United States
| | - Pavlina Baevova
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, United States.,Department of Anesthesiology, Yale University, New Haven, CT, United States
| | - Taylor S Adams
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, New Haven, CT, United States
| | - Mehmet H Kural
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, United States.,Department of Anesthesiology, Yale University, New Haven, CT, United States
| | - Juan Wang
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, United States.,Department of Anesthesiology, Yale University, New Haven, CT, United States
| | - Tomohiro Obata
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, United States.,Department of Anesthesiology, Yale University, New Haven, CT, United States
| | - Mervin C Yoder
- Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Naftali Kaminski
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, New Haven, CT, United States
| | - Laura E Niklason
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, United States.,Department of Anesthesiology, Yale University, New Haven, CT, United States.,Department of Biomedical Engineering, Yale University, New Haven, CT, United States
| |
Collapse
|
24
|
Translational considerations for adipose-derived biological scaffolds for soft tissue repair. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2021. [DOI: 10.1016/j.cobme.2021.100321] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
25
|
Abstract
INTRODUCTION The matrisome and adhesome comprise proteins that are found within or are associated with the extracellular matrix (ECM) and adhesion complexes, respectively. Interactions between cells and their microenvironment are mediated by key matrisome and adhesome proteins, which direct fundamental processes, including growth and development. Due to their underlying complexity, it has historically been challenging to undertake mass spectrometry (MS)-based profiling of these proteins. New developments in sample preparative workflows, informatics databases, and MS techniques have enabled in-depth proteomic characterization of the matrisome and adhesome, resulting in a comprehensive understanding of the interactomes, and cellular signaling that occur at the cell-ECM interface. AREA COVERED This review summarizes recent advances in proteomic characterization of the matrisome and adhesome. It focuses on the importance of curated databases and discusses key strengths and limitations of different workflows. EXPERT OPINION MS-based proteomics has shown promise in characterizing the matrisome and topology of adhesome networks in health and disease. Moving forward, it will be important to incorporate integrative analysis to define the bidirectional signaling between the matrisome and adhesome, and adopt new methods for post-translational modification and in vivo analyses to better dissect the critical roles that these proteins play in human pathophysiology.
Collapse
Affiliation(s)
- Lukas Krasny
- Division of Molecular Pathology, The Institute of Cancer Research, London, United Kingdom
| | - Paul H Huang
- Division of Molecular Pathology, The Institute of Cancer Research, London, United Kingdom
| |
Collapse
|
26
|
Li M, Yang S, Song J, Fu T, Liang P, Gao Z, Tang J, Guo L. Different grinding speeds affect induced regeneration capacity of human treated dentin matrix. J Biomed Mater Res B Appl Biomater 2021; 110:755-767. [PMID: 34637601 DOI: 10.1002/jbm.b.34954] [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: 10/26/2020] [Revised: 09/18/2021] [Accepted: 09/29/2021] [Indexed: 11/07/2022]
Abstract
Human-treated dentin matrix (hTDM) is a biomaterial scaffold, which can induce implant cells to differentiate into odontoblasts and then form neo-dentin. However, hTDM with long storage or prepared by high-speed handpiece would not to form neo-dentin. In this research, we developed two fresh hTDM with different grinding speeds, which were low-speed hTDM (LTDM) with maximum speed of 500 rpm and high-speed hTDM (HTDM) with a speed of 3,80,000 rpm. Here, we aim to understand whether there were induced regeneration capacity differences between LTDM and HTDM. Scanning electron microscope showed that DFCs grew well on both materials, but the morphology of DFCs and the extracellular matrix was different. Especially, the secreted extracellular matrixes on the inner surface of LTDM were regular morphology and ordered arrangement around the dentin tubules. The transcription-quantitative polymerase chain reaction (qRT-PCR), western blot and immunofluorescence assay showed that the dentin markers DSPP and DMP-1 were about 2× greater in DFCs induced by LTDM than by HTDM, and osteogenic marker BSP was about 2× greater in DFCs induced by HTDM than by LTDM. Histological examinations of the harvested grafts observed the formation of neo-tissue were different, and there were neo-dentin formed on the inner surface of LTDM and neo-cementum formed on the outer surface of HTDM. In summary, it found that the induction abilities of LTDM and HTDM are different, and the dentin matrix is directional. This study lays a necessary foundation for searching the key factors of dentin regeneration in future.
Collapse
Affiliation(s)
- Min Li
- Department of Stomatology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Department of Medical Cosmetology, Suining Central Hospital, Suining, China.,Department of Stomatology, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Sen Yang
- Stomatology Center, Suining Central Hospital, Suining, China
| | - Jinlin Song
- Chongqing Medical University Stomatology College, Chongqing, China.,Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Tiwei Fu
- Chongqing Medical University Stomatology College, Chongqing, China.,Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Panpan Liang
- Chongqing Medical University Stomatology College, Chongqing, China.,Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Zhi Gao
- Department of Stomatology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jing Tang
- Department of Stomatology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lijuan Guo
- Department of Medical Cosmetology, Suining Central Hospital, Suining, China
| |
Collapse
|
27
|
Assis A, Camargo S, Margalit R, Mitrani E. Creation of a vascular inducing device using mesenchymal stem cells to induce angiogenesis. J Biosci Bioeng 2021; 132:408-416. [PMID: 34326013 DOI: 10.1016/j.jbiosc.2021.06.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 06/14/2021] [Accepted: 06/29/2021] [Indexed: 12/13/2022]
Abstract
Conventional treatments of peripheral vascular disease and coronary artery disease have partial success but are still limited. Methods to deliver angiogenic factors into ischemic areas using gene, protein and cell therapies are faced with difficult issues such a delivery, effective concentration and duration of action. Tissue engineering offers the possibility of creating a functional self-contained three-dimensional (3D) unit that works as a coordinated biological pump that can secrete a whole range of angiogenic factors. We report a tissue engineering approach using decellularized micro-fragments and mesenchymal stem cells (MSCs) to create a vascular inducing device (VID). Proteomic analysis of the decellularized micro-fragments and of the VIDs reveals a large number of extracellular-matrix (ECM) proteins. Moreover, the VIDs were found to transcribe and secrete a whole repertoire of angiogenic factors in a sustained manner. Furthermore, preliminary results of implantation VIDs into non-obese diabetic/severe combined immunodeficiency (NOD/SCID) mice indicate formation of vascular network at the site within a week. We propose that those VIDs could serve as a safe, localized, simple and powerful method for the treatment of certain types of vascular diseases.
Collapse
Affiliation(s)
- Assaf Assis
- Department of Cell and Developmental Biology, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Edmond J. Safra Givat Ram Campus, Jerusalem 91904, Israel
| | - Sandra Camargo
- Department of Cell and Developmental Biology, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Edmond J. Safra Givat Ram Campus, Jerusalem 91904, Israel
| | | | - Eduardo Mitrani
- Department of Cell and Developmental Biology, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Edmond J. Safra Givat Ram Campus, Jerusalem 91904, Israel.
| |
Collapse
|
28
|
Decellularization of kidney tissue: comparison of sodium lauryl ether sulfate and sodium dodecyl sulfate for allotransplantation in rat. Cell Tissue Res 2021; 386:365-378. [PMID: 34424397 DOI: 10.1007/s00441-021-03517-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 08/04/2021] [Indexed: 10/20/2022]
Abstract
An automatic decellularization device was developed to perfuse and decellularize male rats' kidneys using both sodium lauryl ether sulfate (SLES) and sodium dodecyl sulfate (SDS) and to compare their efficacy in kidney decellularization and post-transplantation angiogenesis. Kidneys were perfused with either 1% SDS solution for 4 h or 1% SLES solution for 6 h. The decellularized scaffolds were stained with hematoxylin and eosin, periodic acid Schiff, Masson's trichrome, and Alcian blue to determine cell removal and glycogen, collagen, and glycosaminoglycan contents, respectively. Moreover, scanning electron microscopy was performed to evaluate the cell removal and preservation of microarchitecture of both SDS and SLES scaffolds. Additionally, DNA quantification assay was applied for all groups in order to measure residual DNA in the scaffolds and normal kidney. In order to demonstrate biocompatibility of the decellularized scaffolds, human umbilical cord mesenchymal stromal/stem cells (hUC-MSCs) were seeded on the scaffolds. In addition, the allotransplantation was performed in back muscle and angiogenesis was evaluated. Complete cell removal in both SLES and SDS groups was observed in scanning electron microscopy and DNA quantification assays. Moreover, the extracellular matrix (ECM) architecture of rat kidney in the SLES group was significantly preserved better than the SDS group. The hUC-MSCs were successfully migrated from the cell culture plate surface into the SDS and SLES decellularized scaffolds. The formation of blood vessels was observed in the kidney in both SLES and SDS decellularized kidneys. The better preservation of ECM than SDS introduces SLES as the solvent of choice for kidney decellularization.
Collapse
|
29
|
Sonpho E, Mann FG, Levy M, Ross EJ, Guerrero-Hernández C, Florens L, Saraf A, Doddihal V, Ounjai P, Sánchez Alvarado A. Decellularization Enables Characterization and Functional Analysis of Extracellular Matrix in Planarian Regeneration. Mol Cell Proteomics 2021; 20:100137. [PMID: 34416386 PMCID: PMC8503668 DOI: 10.1016/j.mcpro.2021.100137] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 07/22/2021] [Accepted: 08/10/2021] [Indexed: 10/25/2022] Open
Abstract
The extracellular matrix (ECM) is a three-dimensional network of macromolecules that provides a microenvironment capable of supporting and regulating cell functions. However, only a few research organisms are available for the systematic dissection of the composition and functions of the ECM, particularly during regeneration. We utilized the free-living flatworm Schmidtea mediterranea to develop an integrative approach consisting of decellularization, proteomics, and RNAi to characterize and investigate ECM functions during tissue homeostasis and regeneration. ECM-enriched samples were isolated from planarians, and their proteomes were characterized by LC-MS/MS. The functions of identified ECM components were interrogated using RNA interference. Using this approach, we found that heparan sulfate proteoglycan is essential for tissue regeneration. Our strategy provides an experimental approach for identifying both known and novel ECM components involved in regeneration.
Collapse
Affiliation(s)
- Ekasit Sonpho
- Stowers Institute for Medical Research, Kansas City, Missouri, USA; Department of Biology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Frederick G Mann
- Stowers Institute for Medical Research, Kansas City, Missouri, USA; Howard Hughes Medical Institute, Kansas City, Missouri, USA
| | - Michaella Levy
- Stowers Institute for Medical Research, Kansas City, Missouri, USA
| | - Eric J Ross
- Stowers Institute for Medical Research, Kansas City, Missouri, USA; Howard Hughes Medical Institute, Kansas City, Missouri, USA
| | | | - Laurence Florens
- Stowers Institute for Medical Research, Kansas City, Missouri, USA
| | - Anita Saraf
- Stowers Institute for Medical Research, Kansas City, Missouri, USA
| | - Viraj Doddihal
- Stowers Institute for Medical Research, Kansas City, Missouri, USA
| | - Puey Ounjai
- Department of Biology, Faculty of Science, Mahidol University, Bangkok, Thailand; Center of Excellence on Environmental Health and Toxicology (EHT), Office of Higher Education Commission, Ministry of Education, Bangkok, Thailand
| | - Alejandro Sánchez Alvarado
- Stowers Institute for Medical Research, Kansas City, Missouri, USA; Howard Hughes Medical Institute, Kansas City, Missouri, USA.
| |
Collapse
|
30
|
Evangelista-Leite D, Carreira ACO, Gilpin SE, Miglino MA. Protective Effects of Extracellular Matrix-Derived Hydrogels in Idiopathic Pulmonary Fibrosis. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:517-530. [PMID: 33899554 DOI: 10.1089/ten.teb.2020.0357] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic progressive disease with significant gas exchange impairment owing to exaggerated extracellular matrix (ECM) deposition and myofibroblast activation. IPF has no cure, and although nintedanib and pirfenidone are two approved medications for symptom management, the total treatment cost is exuberant and prohibitive to a global uninsured patient population. New therapeutic alternatives with moderate costs are needed to treat IPF. ECM hydrogels derived from decellularized lungs are cost-effective therapeutic candidates to treat pulmonary fibrosis because of their reported antioxidant properties. Oxidative stress contributes to IPF pathophysiology by damaging macromolecules, interfering with tissue remodeling, and contributing to myofibroblast activation. Thus, preventing oxidative stress has beneficial outcomes in IPF. For this purpose, this review describes ECM hydrogel's properties to regulate oxidative stress and tissue remodeling in IPF.
Collapse
Affiliation(s)
- Daniele Evangelista-Leite
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Ana Claudia O Carreira
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil.,NUCEL (Cell and Molecular Therapy Center), University of São Paulo, São Paulo, Brazil
| | - Sarah E Gilpin
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts, USA
| | - Maria Angélica Miglino
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
31
|
Goldman SM, Janakiram NB, Valerio MS, Dearth CL. Evaluation of licofelone as an adjunct anti-inflammatory therapy to biologic scaffolds in the treatment of volumetric muscle loss. Cell Tissue Res 2021; 385:149-159. [PMID: 33852076 DOI: 10.1007/s00441-021-03449-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 03/08/2021] [Indexed: 12/24/2022]
Abstract
Biologic scaffolds (BS) are the most widely studied therapeutics for the treatment of volumetric muscle loss (VML) owing to their purported effects on cell proliferation, chemotaxis, migration, and differentiation. Despite these claims, variability in reports on the nature of the immune response to their implantation suggests that BS-associated inflammation may be limiting their regenerative efficacy. To address this shortcoming, this study sought to evaluate licofelone (ML3000), a dual 5-LOX/COX inhibitor, as an anti-inflammatory adjunct therapy to a BS in the treatment of VML. Utilizing a well-established rat VML model, a micronized BS was used to treat the VML injury, with or without administration of licofelone. Functional, molecular, and histological outcomes were assessed at both 7- and 28-day post-injury time points. While the BS + licofelone group exhibited decreased transcription of pro-inflammatory markers (Tnf, Ccl5, Nos2) relative to the BS only control group, no differences in expression profile of a panel of inflammatory-related soluble factors were observed between groups. A modest reduction in type I collagen was observed in the licofelone-treated group, but no meaningful differences in histologic presentation of repaired tissue were observed between groups. Furthermore, no differences in end organ functional capacity were observed between groups. Moving forward, efforts related to modulating the wound healing environment of VML should focus on polypharmaceutical strategies that target multiple aspects of the early pathophysiology of VML so as to provide an environment that is sufficiently permissive for local regenerative therapies to promote restoration of myofiber number.
Collapse
Affiliation(s)
- Stephen M Goldman
- DoD-VA Extremity Trauma and Amputation Center of Excellence, Bethesda, MD, USA.,Department of Surgery, Uniformed Services University of the Health Sciences and Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Naveena Basa Janakiram
- DoD-VA Extremity Trauma and Amputation Center of Excellence, Bethesda, MD, USA.,Department of Surgery, Uniformed Services University of the Health Sciences and Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Michael S Valerio
- DoD-VA Extremity Trauma and Amputation Center of Excellence, Bethesda, MD, USA.,Department of Surgery, Uniformed Services University of the Health Sciences and Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Christopher L Dearth
- DoD-VA Extremity Trauma and Amputation Center of Excellence, Bethesda, MD, USA. .,Department of Surgery, Uniformed Services University of the Health Sciences and Walter Reed National Military Medical Center, Bethesda, MD, USA.
| |
Collapse
|
32
|
Joyce K, Fabra GT, Bozkurt Y, Pandit A. Bioactive potential of natural biomaterials: identification, retention and assessment of biological properties. Signal Transduct Target Ther 2021; 6:122. [PMID: 33737507 PMCID: PMC7973744 DOI: 10.1038/s41392-021-00512-8] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 12/29/2020] [Accepted: 01/19/2021] [Indexed: 02/07/2023] Open
Abstract
Biomaterials have had an increasingly important role in recent decades, in biomedical device design and the development of tissue engineering solutions for cell delivery, drug delivery, device integration, tissue replacement, and more. There is an increasing trend in tissue engineering to use natural substrates, such as macromolecules native to plants and animals to improve the biocompatibility and biodegradability of delivered materials. At the same time, these materials have favourable mechanical properties and often considered to be biologically inert. More importantly, these macromolecules possess innate functions and properties due to their unique chemical composition and structure, which increase their bioactivity and therapeutic potential in a wide range of applications. While much focus has been on integrating these materials into these devices via a spectrum of cross-linking mechanisms, little attention is drawn to residual bioactivity that is often hampered during isolation, purification, and production processes. Herein, we discuss methods of initial material characterisation to determine innate bioactivity, means of material processing including cross-linking, decellularisation, and purification techniques and finally, a biological assessment of retained bioactivity of a final product. This review aims to address considerations for biomaterials design from natural polymers, through the optimisation and preservation of bioactive components that maximise the inherent bioactive potency of the substrate to promote tissue regeneration.
Collapse
Affiliation(s)
- Kieran Joyce
- School of Medicine, National University of Ireland, Galway, Ireland
- CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland, Galway, Ireland
| | - Georgina Targa Fabra
- CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland, Galway, Ireland
| | - Yagmur Bozkurt
- CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland, Galway, Ireland
| | - Abhay Pandit
- CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland, Galway, Ireland.
| |
Collapse
|
33
|
Proteome-wide and matrisome-specific alterations during human pancreas development and maturation. Nat Commun 2021; 12:1020. [PMID: 33589611 PMCID: PMC7884717 DOI: 10.1038/s41467-021-21261-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 01/19/2021] [Indexed: 01/31/2023] Open
Abstract
The extracellular matrix (ECM) is unique to each tissue and capable of guiding cell differentiation, migration, morphology, and function. The ECM proteome of different developmental stages has not been systematically studied in the human pancreas. In this study, we apply mass spectrometry-based quantitative proteomics strategies using N,N-dimethyl leucine isobaric tags to delineate proteome-wide and ECM-specific alterations in four age groups: fetal (18-20 weeks gestation), juvenile (5-16 years old), young adults (21-29 years old) and older adults (50-61 years old). We identify 3,523 proteins including 185 ECM proteins and quantify 117 of them. We detect previously unknown proteome and matrisome features during pancreas development and maturation. We also visualize specific ECM proteins of interest using immunofluorescent staining and investigate changes in ECM localization within islet or acinar compartments. This comprehensive proteomics analysis contributes to an improved understanding of the critical roles that ECM plays throughout human pancreas development and maturation.
Collapse
|
34
|
Zhou Q, Fan L, Li J. Liver Regeneration and Tissue Engineering. ARTIFICIAL LIVER 2021:73-94. [DOI: 10.1007/978-981-15-5984-6_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
35
|
Asthana A, Tamburrini R, Chaimov D, Gazia C, Walker SJ, Van Dyke M, Tomei A, Lablanche S, Robertson J, Opara EC, Soker S, Orlando G. Comprehensive characterization of the human pancreatic proteome for bioengineering applications. Biomaterials 2020; 270:120613. [PMID: 33561625 DOI: 10.1016/j.biomaterials.2020.120613] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 11/27/2020] [Accepted: 12/14/2020] [Indexed: 02/07/2023]
Abstract
Interactions between the pancreatic extracellular matrix (ECM) and islet cells are known to regulate multiple aspects of islet physiology, including survival, proliferation, and glucose-stimulated insulin secretion. Recognizing the essential role of ECM in islet survival and function, various engineering approaches have been developed that aim to utilize ECM-based materials to recreate a native-like microenvironment. However, a major impediment to the success of these approaches has been the lack of a robust and comprehensive characterization of the human pancreatic proteome. Herein, by combining mass spectrometry (MS) and multiplex ELISA, we have provided an improved workflow for the in-depth profiling of the proteome, including minor constituents that are generally underrepresented. Moreover, we have further validated the effectiveness of our detergent-free decellularization protocol in the removal of cellular proteins and retention of the matrisome. It has also been established that the decellularized ECM and its derivatives can provide more tissue-specific cues than traditionally used biological scaffolds and are therefore more physiologically relevant for the development of hydrogels, bioinks and medium additives, in order to create a pancreatic niche. The data generated in this study would contribute significantly to the efforts of comprehensively defining the ECM atlas and also serve as a standard for the human pancreatic proteome to provide further guidance for design and engineering strategies for improved tissue engineering scaffolds.
Collapse
Affiliation(s)
- Amish Asthana
- Department of Surgery, Wake Forest Baptist Medical Center, Medical Center Boulevard, Winston Salem, USA; Wake Forest Institute for Regenerative Medicine, Winston Salem, USA; Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, USA
| | - Riccardo Tamburrini
- Department of Surgery, Wake Forest Baptist Medical Center, Medical Center Boulevard, Winston Salem, USA; Wake Forest Institute for Regenerative Medicine, Winston Salem, USA; Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, USA
| | - Deborah Chaimov
- Department of Surgery, Wake Forest Baptist Medical Center, Medical Center Boulevard, Winston Salem, USA; Wake Forest Institute for Regenerative Medicine, Winston Salem, USA; Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, USA
| | - Carlo Gazia
- Department of Surgery, Wake Forest Baptist Medical Center, Medical Center Boulevard, Winston Salem, USA; Wake Forest Institute for Regenerative Medicine, Winston Salem, USA; Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, USA
| | - Stephen J Walker
- Wake Forest Institute for Regenerative Medicine, Winston Salem, USA; Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, USA
| | - Mark Van Dyke
- Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Alice Tomei
- Diabetes Research Institute, University of Miami, Miami, USA
| | - Sandrine Lablanche
- Grenoble Alps University, Laboratory of Fundamental and Applied Bioenergetics (LBFA), And Environmental and System Biology (BEeSy), Grenoble, France; Inserm, U1055, Grenoble, France
| | - John Robertson
- Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, USA
| | - Emmanuel C Opara
- Wake Forest Institute for Regenerative Medicine, Winston Salem, USA; Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, USA
| | - Shay Soker
- Wake Forest Institute for Regenerative Medicine, Winston Salem, USA; Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, USA
| | - Giuseppe Orlando
- Department of Surgery, Wake Forest Baptist Medical Center, Medical Center Boulevard, Winston Salem, USA; Wake Forest Institute for Regenerative Medicine, Winston Salem, USA; Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, USA.
| |
Collapse
|
36
|
Li M, Zhang A, Li J, Zhou J, Zheng Y, Zhang C, Xia D, Mao H, Zhao J. Osteoblast/fibroblast coculture derived bioactive ECM with unique matrisome profile facilitates bone regeneration. Bioact Mater 2020; 5:938-948. [PMID: 32637756 PMCID: PMC7330453 DOI: 10.1016/j.bioactmat.2020.06.017] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 06/15/2020] [Accepted: 06/23/2020] [Indexed: 02/08/2023] Open
Abstract
Extracellular matrix (ECM) with mimetic tissue niches was attractive to facilitate tissue regeneration in situ via recruitment of endogenous cells and stimulation of self-healing process. However, how to engineer the complicate tissue specific ECM with unique matrisome in vitro was a challenge of ECM-based biomaterials in tissue engineering and regenerative medicine. Here, we introduced coculture system to engineer bone mimetic ECM niche guided by cell-cell communication. In the cocultures, fibroblasts promoted osteogenic differentiation of osteoblasts via extracellular vesicles. The generated ECM (MN-ECM) displayed a unique appearance of morphology and biological components. The advantages of MN-ECM were demonstrated with promotion of multiple cellular behaviors (proliferation, adhesion and osteogenic mineralization) in vitro and bone regeneration in vivo. Moreover, proteomic analysis was used to clarify the molecular mechanism of MN-ECM, which revealed a specific matrisome signature. The present study provides a novel strategy to generate ECM with tissue mimetic niches via cell-cell communication in a coculture system, which forwards the development of tissue-bioactive ECM engineering along with deepening the understanding of ECM niches regulated by cells for bone tissue engineering.
Collapse
Affiliation(s)
- Mei Li
- Zhejiang Key Laboratory of Pathophysiology, Medical School in Ningbo University, Ningbo, Zhejiang, PR China
- Ningbo Institute of Medical Sciences, Ningbo, Zhejiang, PR China
| | - Anqi Zhang
- Zhejiang Key Laboratory of Pathophysiology, Medical School in Ningbo University, Ningbo, Zhejiang, PR China
| | - Jiajing Li
- Zhejiang Key Laboratory of Pathophysiology, Medical School in Ningbo University, Ningbo, Zhejiang, PR China
| | - Jing Zhou
- Zhejiang Key Laboratory of Pathophysiology, Medical School in Ningbo University, Ningbo, Zhejiang, PR China
| | - Yanan Zheng
- Zhejiang Key Laboratory of Pathophysiology, Medical School in Ningbo University, Ningbo, Zhejiang, PR China
| | - Chi Zhang
- Orthopedic Department, Ningbo First Hospital, Ningbo, Zhejiang, PR China
| | - Dongdong Xia
- Orthopedic Department, Ningbo First Hospital, Ningbo, Zhejiang, PR China
| | - Haijiao Mao
- Department of Orthopaedic Surgery, The Affiliated Hospital of Medical School, Ningbo University, Ningbo, Zhejiang, PR China
| | - Jiyuan Zhao
- Zhejiang Key Laboratory of Pathophysiology, Medical School in Ningbo University, Ningbo, Zhejiang, PR China
| |
Collapse
|
37
|
Lei R, Kumar S. Getting the big picture of cell-matrix interactions: High-throughput biomaterial platforms and systems-level measurements. CURRENT OPINION IN SOLID STATE & MATERIALS SCIENCE 2020; 24:100871. [PMID: 33244294 PMCID: PMC7685248 DOI: 10.1016/j.cossms.2020.100871] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Living cells interact with the extracellular matrix (ECM) in a complex and reciprocal manner. Much has been learned over the past few decades about cell-ECM interactions from targeted studies in which a specific matrix parameter (e.g. stiffness, adhesivity) has been varied across a few discrete values, or in which the level or activity of a protein is controlled in an isolated fashion. As the field moves forward, there is growing interest in addressing cell-matrix interactions from a systems perspective, which has spurred a new generation of matrix platforms capable of interrogating multiple ECM inputs in a combinatorial and parallelized fashion. Efforts are also actively underway to integrate specialized, synthetic ECM platforms with global measures of cell behaviors, including at the transcriptomic, proteomic and epigenomic levels. Here we review recent advances in both areas. We describe how new combinatorial ECM technologies are revealing unexpected crosstalk and nonlinearity in the relationship between cell phenotype and matrix properties. Similarly, efforts to integrate "omics" measurements with synthetic ECM platforms are illuminating how ECM properties can control cell biology in surprising and functionally important ways. We expect that advances in both areas will deepen the field's understanding of cell-ECM interactions and offer valuable insight into the design of biomaterials for specific biomedical applications.
Collapse
Affiliation(s)
- Ruoxing Lei
- Department of Chemistry, University of California, Berkeley, CA, 94720
- Department of Bioengineering, University of California, Berkeley, CA, 94720
| | - Sanjay Kumar
- Department of Bioengineering, University of California, Berkeley, CA, 94720
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, 94720
| |
Collapse
|
38
|
Lee JS, Mitulović G, Panahipour L, Gruber R. Proteomic Analysis of Porcine-Derived Collagen Membrane and Matrix. MATERIALS 2020; 13:ma13225187. [PMID: 33212864 PMCID: PMC7698422 DOI: 10.3390/ma13225187] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 11/06/2020] [Accepted: 11/11/2020] [Indexed: 12/13/2022]
Abstract
Collagen membranes and matrices being widely used in guided bone regeneration and soft tissue augmentation have characteristic properties based on their composition. The respective proteomic signatures have not been identified. Here, we performed a high-resolution shotgun proteomic analysis on two porcine collagen-based biomaterials designed for guided bone regeneration and soft tissue augmentation. Three lots each of a porcine-derived collagen membrane and a matrix derived from peritoneum and/or skin were digested and separated by nano-reverse-phase high-performance liquid chromatography. The peptides were subjected to mass spectrometric detection and analysis. A total of 37 proteins identified by two peptides were present in all collagen membranes and matrices, with 11 and 16 proteins being exclusively present in the membrane and matrix, respectively. The common extracellular matrix proteins include fibrillar collagens (COL1A1, COL1A2, COL2A1, COL3A1, COL5A1, COL5A2, COL5A3, COL11A2), non-fibrillar collagens (COL4A2, COL6A1, COL6A2, COL6A3, COL7A1, COL16A1, COL22A1), and leucine-rich repeat proteoglycans (DCN, LUM, BGN, PRELP, OGN). The structural proteins vimentin, actin-based microfilaments (ACTB), annexins (ANXA1, ANXA5), tubulins (TUBA1B, TUBB), and histones (H2A, H2B, H4) were also identified. Examples of membrane-only proteins are COL12A1 and COL14A1, and, of matrix only proteins, elastin (ELN). The proteomic signature thus revealed the similarities between but also some individual proteins of collagen membrane and matrix.
Collapse
Affiliation(s)
- Jung-Seok Lee
- Department of Oral Biology, School of Dentistry, Medical University of Vienna, 1090 Vienna, Austria; (J.-S.L.); (L.P.)
- Department of Periodontology, Research Institute for Periodontal Regeneration, College of Dentistry, Yonsei University, Seoul 03722, Korea
| | - Goran Mitulović
- Proteomics Core Facility, Clinical Institute of Laboratory Medicine, Medical University of Vienna, 1090 Vienna, Austria;
| | - Layla Panahipour
- Department of Oral Biology, School of Dentistry, Medical University of Vienna, 1090 Vienna, Austria; (J.-S.L.); (L.P.)
| | - Reinhard Gruber
- Department of Oral Biology, School of Dentistry, Medical University of Vienna, 1090 Vienna, Austria; (J.-S.L.); (L.P.)
- Proteomics Core Facility, Clinical Institute of Laboratory Medicine, Medical University of Vienna, 1090 Vienna, Austria;
- Department of Periodontology, School of Dental Medicine, University of Bern, 3010 Bern, Switzerland
- Correspondence:
| |
Collapse
|
39
|
Engineering an endothelialized, endocrine Neo-Pancreas: Evaluation of islet functionality in an ex vivo model. Acta Biomater 2020; 117:213-225. [PMID: 32949822 DOI: 10.1016/j.actbio.2020.09.022] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 09/08/2020] [Accepted: 09/10/2020] [Indexed: 12/15/2022]
Abstract
Islet-based recellularization of decellularized, repurposed rat livers may form a transplantable Neo-Pancreas. The aim of this study is the establishment of the necessary protocols, the evaluation of the organ structure and the analysis of the islet functionality ex vivo. After perfusion-based decellularization of rat livers, matrices were repopulated with endothelial cells and mesenchymal stromal cells, incubated for 8 days in a perfusion chamber, and finally repopulated on day 9 with intact rodent islets. Integrity and quality of re-endothelialization was assessed by histology and FITC-dextran perfusion assay. Functionality of the islets of Langerhans was determined on day 10 and day 12 via glucose stimulated insulin secretion. Blood gas analysis variables confirmed the stability of the perfusion cultivation. Histological staining showed that cells formed a monolayer inside the intact vascular structure. These findings were confirmed by electron microscopy. Islets infused via the bile duct could histologically be found in the parenchymal space. Adequate insulin secretion after glucose stimulation after 1-day and 3-day cultivation verified islet viability and functionality after the repopulation process. We provide the first proof-of-concept for the functionality of islets of Langerhans engrafted in a decellularized rat liver. Furthermore, a re-endothelialization step was implemented to provide implantability. This technique can serve as a bioengineered platform to generate implantable and functional endocrine Neo-Pancreases.
Collapse
|
40
|
Zanardo TÉC, Amorim FG, Taufner GH, Pereira RHA, Baiense IM, Destefani AC, Iwai LK, Maranhão RC, Nogueira BV. Decellularized Splenic Matrix as a Scaffold for Spleen Bioengineering. Front Bioeng Biotechnol 2020; 8:573461. [PMID: 33123515 PMCID: PMC7567156 DOI: 10.3389/fbioe.2020.573461] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 09/08/2020] [Indexed: 01/15/2023] Open
Abstract
The spleen is considered a non-essential organ. However, its importance is increasingly clear, given the serious disorders caused by its absence or dysfunction, e.g., greater susceptibility to infections, thromboembolism and cancer. Surgical techniques to preserve the spleen and maintain splenic function have become increasingly common. However, the morbidity and mortality associated with its absence and dysfunction are still high. We used the decellularization technique to obtain a viable splenic scaffold for recellularization in vitro and propose the idea of bioengineered spleen transplantation to the host. We observed the maintenance of important structural components such as white pulp, marginal zone and red pulp, in addition to the network of vascular ducts. The decellularized scaffold presents minimal residual DNA and SDS, which are essential to prevent immunogenic responses and transplantation failure. Also, the main components of the splenic matrix were preserved after decellularization, with retention of approximately 72% in the matrisomal protein content. The scaffold we developed was partially recellularized with stromal cells from the spleen of neonatal rats, demonstrating adhesion, proliferation and viability of cells. Therefore, the splenic scaffold is very promising for use in studies on spleen reconstruction and transplantation, with the aim of complete recovery of splenic function.
Collapse
Affiliation(s)
- Tadeu Ériton Caliman Zanardo
- Biotechnology Graduate Program, Rede Nordeste de Biotecnologia (RENORBIO), Vitória, Brazil.,Tissue Engineering Core, Department of Morphology, Federal University of Espírito Santo, Vitória, Brazil
| | - Fernanda Gobbi Amorim
- Biotechnology Graduate Program, Rede Nordeste de Biotecnologia (RENORBIO), Vitória, Brazil.,Pharmaceutical Sciences Graduate Program, University of Vila Velha, Vila Velha, Brazil
| | - Gabriel Henrique Taufner
- Biotechnology Graduate Program, Rede Nordeste de Biotecnologia (RENORBIO), Vitória, Brazil.,Tissue Engineering Core, Department of Morphology, Federal University of Espírito Santo, Vitória, Brazil
| | - Rayssa Helena Arruda Pereira
- Biotechnology Graduate Program, Rede Nordeste de Biotecnologia (RENORBIO), Vitória, Brazil.,Tissue Engineering Core, Department of Morphology, Federal University of Espírito Santo, Vitória, Brazil
| | - Ian Manhoni Baiense
- Tissue Engineering Core, Department of Morphology, Federal University of Espírito Santo, Vitória, Brazil
| | - Afrânio Côgo Destefani
- Biotechnology Graduate Program, Rede Nordeste de Biotecnologia (RENORBIO), Vitória, Brazil.,Tissue Engineering Core, Department of Morphology, Federal University of Espírito Santo, Vitória, Brazil
| | - Leo Kei Iwai
- Laboratory of Proteomics and Mass Spectrometry-Special Laboratory of Applied Toxinology LETA/CETICS, Instituto Butantan, São Paulo, Brazil
| | | | - Breno Valentim Nogueira
- Biotechnology Graduate Program, Rede Nordeste de Biotecnologia (RENORBIO), Vitória, Brazil.,Tissue Engineering Core, Department of Morphology, Federal University of Espírito Santo, Vitória, Brazil
| |
Collapse
|
41
|
Guo B, Zhou Q, Jiang J, Chen J, Liang X, Jiaojiao X, Shi D, Ren K, Zhou X, Hassan HM, Li J. Functionalized Vascular Structure in Bioengineered Liver Identified with Proteomics. ACS Biomater Sci Eng 2020; 6:6394-6404. [PMID: 33449649 DOI: 10.1021/acsbiomaterials.0c01353] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Vascularization has been a major challenge in the development of a bioengineered liver. We aimed to develop a functionalized vascular structure in bioengineered liver and to identify the biological vascularization processes at different time points using proteomics. Decellularized rat liver scaffolds were vascularized with human umbilical vein endothelial cells (HUVECs) for 1, 3, 7, 14, and 21 days. HUVECs adhered to the internal surface and formed a functional barrier structure within 7 days. Vascularized liver scaffolds with biological activity were sustained for more than 21 days in vitro. Proteomics analysis indicated distinct characteristics after 14 days of culture compared with other time points. The biological processes of proteins expressed at days 1, 3, and 7 mainly involved cell adhesion, protein synthesis, and energy metabolism; however, different biological processes associated with muscle contraction and muscle filament sliding were identified at days 14 and 21. Coexpressed proteins at days 14 and 21 participated in 7 biological processes that could be classified as angiogenesis, myogenesis, or vascular function. Furthermore, the validation of related proteins revealed that basement membrane assembly, phenotype plasticity of HUVECs, and the regulation of adherence junctions contribute to the formation of a functionalized vascular structure. The biological vascularization processes at different time points identified with proteomics revealed development characteristics of vascular structure in a bioengineered liver, and at least 14 days of in vitro culture should be recommended for developing a functionalized vascular structure. This study may help to provide a better understanding of the mechanism of vascularization and facilitate the construction of a functional bioengineered liver for future clinical applications.
Collapse
Affiliation(s)
- Beibei Guo
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Qian Zhou
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jing Jiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.,Precision Medicine Center of Taizhou Central Hospital, Taizhou University Medical School, Taizhou 318000, China
| | - Jiaxian Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Xi Liang
- Precision Medicine Center of Taizhou Central Hospital, Taizhou University Medical School, Taizhou 318000, China
| | - Xin Jiaojiao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.,Precision Medicine Center of Taizhou Central Hospital, Taizhou University Medical School, Taizhou 318000, China
| | - Dongyan Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.,Precision Medicine Center of Taizhou Central Hospital, Taizhou University Medical School, Taizhou 318000, China
| | - Keke Ren
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Xingping Zhou
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Hozeifa M Hassan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jun Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.,Precision Medicine Center of Taizhou Central Hospital, Taizhou University Medical School, Taizhou 318000, China
| |
Collapse
|
42
|
Liu B, Zhang S, Wang W, Yun Z, Lv L, Chai M, Wu Z, Zhu Y, Ma J, Leng L. Matrisome Provides a Supportive Microenvironment for Skin Functions of Diverse Species. ACS Biomater Sci Eng 2020; 6:5720-5733. [PMID: 33320565 DOI: 10.1021/acsbiomaterials.0c00479] [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] [Indexed: 12/12/2022]
Abstract
A biomaterial scaffold is a promising tool employed to drive tissue regeneration. This technology has been successfully applied in human tissue rebuilding, particularly for the skin. Meanwhile, there is still room for further improvement, such as maintaining sufficient functionality of a biomaterial scaffold. Here, we developed a new decellularization method to generate a complete anatomical skin biomatrix scaffold with a preserved extracellular matrix (ECM) architecture. We performed proteomic and bioinformatic analyses of the skin scaffold maps of humans, pigs, and rats and systematically analyzed the interaction between ECM proteins and different cell types in the skin microenvironment. These interactions served to quantify the structure and function of the skin's Matrisome comprising core ECM components and ECM-associated soluble signaling molecules required for the regulation of epidermal development. We primarily found that the properties of the skin ECM were species-specific. For example, the composition and function of the ECM of the human skin were more similar to those of pigs compared with those of rats. However, the skin ECM of the pig was significantly deficient in its enzyme systems and immune regulatory factors compared with that of humans. These findings provide a new understanding of the role of the skin ECM niche as well as an attractive strategy that can apply tissue engineering principles to skin biomatrix scaffold materials, which promises to accelerate and enhance tissue regeneration.
Collapse
Affiliation(s)
- Binghui Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Life Omics, No. 38 Life Science Park Road, Beijing 102206, China
| | - Shikun Zhang
- Department of Stem Cell and Regenerative Medicine Laboratory, Institute of Health Service and Transfusion Medicine, No. 27 Taiping Road, Beijing 100850, China
| | - Wenjuan Wang
- Department of Dermatology, Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing 100853, China
| | - Zhimin Yun
- Department of Stem Cell and Regenerative Medicine Laboratory, Institute of Health Service and Transfusion Medicine, No. 27 Taiping Road, Beijing 100850, China
| | - Luye Lv
- Institute of NBC Defense, No. 1 Central North Street, Beijing 102205, China
| | - Mi Chai
- Department of Plastic and Reconstruction Surgery, Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing 100853, China
| | - Zhihong Wu
- Department of Medical Science Research Center, Translational Medicine Center, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Beijing 100730, China
| | - Yunping Zhu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Life Omics, No. 38 Life Science Park Road, Beijing 102206, China.,Basic Medical School, Anhui Medical University, No. 81 Meishan Road, Anhui 230032, China
| | - Jie Ma
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Life Omics, No. 38 Life Science Park Road, Beijing 102206, China
| | - Ling Leng
- Department of Medical Science Research Center, Translational Medicine Center, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Beijing 100730, China
| |
Collapse
|
43
|
The human liver matrisome - Proteomic analysis of native and fibrotic human liver extracellular matrices for organ engineering approaches. Biomaterials 2020; 257:120247. [PMID: 32739662 DOI: 10.1016/j.biomaterials.2020.120247] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 07/05/2020] [Accepted: 07/16/2020] [Indexed: 02/06/2023]
Abstract
The production of biomaterials that endow significant morphogenic and microenvironmental cues for the constitution of cell integration and regeneration remains a key challenge in the successful implementation of functional organ replacements. Despite the vast development in the production of biological and architecturally native matrices, the complex compositions and pivotal figures by which the human matrisome mediates many of its essential functions are yet to be defined. Here we present a thorough analysis of the native human liver proteomic landscape using decellularization and defatting protocols to create extracellular matrix scaffolds of natural origin that can further be used in both bottom-up and top-down approaches in tissue engineering based organ replacements. Furthermore, by analyzing human liver extracellular matrices in different stages of fibrosis and cirrhosis, we have identified distinct attributes of these tissues that could potentially be exploited therapeutically and thus require further investigation. The general experimental pipeline presented in this study is applicable to any type of tissue and can be widely used for different approaches in regenerative medicine and in the construction of novel biomaterials for organ engineering approaches.
Collapse
|
44
|
Herrera JA, Mallikarjun V, Rosini S, Montero MA, Lawless C, Warwood S, O’Cualain R, Knight D, Schwartz MA, Swift J. Laser capture microdissection coupled mass spectrometry (LCM-MS) for spatially resolved analysis of formalin-fixed and stained human lung tissues. Clin Proteomics 2020; 17:24. [PMID: 32565759 PMCID: PMC7302139 DOI: 10.1186/s12014-020-09287-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 06/11/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Haematoxylin and eosin (H&E)-which respectively stain nuclei blue and other cellular and stromal material pink-are routinely used for clinical diagnosis based on the identification of morphological features. A richer characterization can be achieved by laser capture microdissection coupled to mass spectrometry (LCM-MS), giving an unbiased assay of the proteins that make up the tissue. However, the process of fixing and H&E staining of tissues provides challenges with standard sample preparation methods for mass spectrometry, resulting in low protein yield. Here we describe a microproteomics technique to analyse H&E-stained, formalin-fixed paraffin-embedded (FFPE) tissues. METHODS Herein, we utilize heat extraction, physical disruption, and in column digestion for the analysis of H&E stained FFPE tissues. Micro-dissected morphologically normal human lung alveoli (0.082 mm3) and human lung blood vessels (0.094 mm3) from FFPE-fixed H&E-stained sections from Idiopathic Pulmonary Fibrosis (IPF) specimens (n = 3 IPF specimens) were then subject to a qualitative and then quantitative proteomics approach using BayesENproteomics. In addition, we tested the sensitivity of this method by processing and analysing a range of micro-dissected human lung blood vessel tissue volumes. RESULTS This approach yields 1252 uniquely expressed proteins (at a protein identification threshold of 3 unique peptides) with 892 differentially expressed proteins between these regions. In accord with prior knowledge, our methodology approach confirms that human lung blood vessels are enriched with smoothelin, CNN1, ITGA7, MYH11, TAGLN, and PTGIS; whereas morphologically normal human lung alveoli are enriched with cytokeratin-7, -8, -18, -19, 14, and -17. In addition, we identify a total of 137 extracellular matrix (ECM) proteins and immunohistologically validate that laminin subunit beta-1 localizes to morphologically normal human lung alveoli and tenascin localizes to human lung blood vessels. Lastly, we show that this micro-proteomics technique can be applied to tissue volumes as low as 0.0125 mm3. CONCLUSION Herein we show that our multistep sample preparation methodology of LCM-MS can identify distinct, characteristic proteomic compositions of anatomical features within complex fixed and stained tissues.
Collapse
Affiliation(s)
- Jeremy A. Herrera
- The Wellcome Centre for Cell-Matrix Research, University of Manchester, Manchester, M13 9PT UK
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PL UK
| | - Venkatesh Mallikarjun
- The Wellcome Centre for Cell-Matrix Research, University of Manchester, Manchester, M13 9PT UK
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PL UK
| | - Silvia Rosini
- The Wellcome Centre for Cell-Matrix Research, University of Manchester, Manchester, M13 9PT UK
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PL UK
| | - Maria Angeles Montero
- Histopathology Department, Manchester University NHS Foundation Trust, Southmoor Road, Wythenshawe, Manchester, M23 9LT UK
| | - Craig Lawless
- The Wellcome Centre for Cell-Matrix Research, University of Manchester, Manchester, M13 9PT UK
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PL UK
| | - Stacey Warwood
- The Wellcome Centre for Cell-Matrix Research, University of Manchester, Manchester, M13 9PT UK
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PL UK
| | - Ronan O’Cualain
- The Wellcome Centre for Cell-Matrix Research, University of Manchester, Manchester, M13 9PT UK
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PL UK
| | - David Knight
- The Wellcome Centre for Cell-Matrix Research, University of Manchester, Manchester, M13 9PT UK
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PL UK
| | - Martin A. Schwartz
- The Wellcome Centre for Cell-Matrix Research, University of Manchester, Manchester, M13 9PT UK
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PL UK
| | - Joe Swift
- The Wellcome Centre for Cell-Matrix Research, University of Manchester, Manchester, M13 9PT UK
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PL UK
| |
Collapse
|
45
|
Liao D, Li H. Dissecting the Niche for Alveolar Type II Cells With Alveolar Organoids. Front Cell Dev Biol 2020; 8:419. [PMID: 32582703 PMCID: PMC7287157 DOI: 10.3389/fcell.2020.00419] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 05/06/2020] [Indexed: 12/11/2022] Open
Affiliation(s)
- Danying Liao
- Department of Haematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huaibiao Li
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
46
|
Ferreira LP, Gaspar VM, Mano JF. Decellularized Extracellular Matrix for Bioengineering Physiomimetic 3D in Vitro Tumor Models. Trends Biotechnol 2020; 38:1397-1414. [PMID: 32416940 DOI: 10.1016/j.tibtech.2020.04.006] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/10/2020] [Accepted: 04/14/2020] [Indexed: 02/06/2023]
Abstract
Recent advances in the extraction and purification of decellularized extracellular matrix (dECM) obtained from healthy or malignant tissues open new avenues for engineering physiomimetic 3D in vitro tumor models, which closely recapitulate key biomolecular hallmarks and the dynamic cancer cell-ECM interactions in the tumor microenvironment. We review current and upcoming methodologies for chemical modification of dECM-based biomaterials and advanced bioprocessing into organotypic 3D solid tumor models. A comprehensive review of disruptive advances and shortcomings of exploring dECM-based biomaterials for recapitulating the native tumor-supporting matrix is also provided. We hope to drive the discussion on how 3D dECM testing platforms can be leveraged for generating microphysiological tumor surrogates that generate more robust and predictive data on therapeutic bioperformance.
Collapse
Affiliation(s)
- Luís P Ferreira
- Department of Chemistry, CICECO, Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - Vítor M Gaspar
- Department of Chemistry, CICECO, Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal.
| | - João F Mano
- Department of Chemistry, CICECO, Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal.
| |
Collapse
|
47
|
Rajab TK, O’Malley TJ, Tchantchaleishvili V. Decellularized scaffolds for tissue engineering: Current status and future perspective. Artif Organs 2020; 44:1031-1043. [DOI: 10.1111/aor.13701] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 03/10/2020] [Accepted: 04/02/2020] [Indexed: 12/11/2022]
Affiliation(s)
| | - Thomas J. O’Malley
- Division of Cardiac Surgery Thomas Jefferson University Philadelphia PA USA
| | | |
Collapse
|
48
|
Everwien H, Ariza de Schellenberger A, Haep N, Tzschätzsch H, Pratschke J, Sauer IM, Braun J, Hillebrandt KH, Sack I. Magnetic resonance elastography quantification of the solid-to-fluid transition of liver tissue due to decellularization. J Mech Behav Biomed Mater 2020; 104:103640. [DOI: 10.1016/j.jmbbm.2020.103640] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 01/13/2020] [Accepted: 01/13/2020] [Indexed: 12/12/2022]
|
49
|
Berger C, Bjørlykke Y, Hahn L, Mühlemann M, Kress S, Walles H, Luxenhofer R, Ræder H, Metzger M, Zdzieblo D. Matrix decoded - A pancreatic extracellular matrix with organ specific cues guiding human iPSC differentiation. Biomaterials 2020; 244:119766. [PMID: 32199284 DOI: 10.1016/j.biomaterials.2020.119766] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 11/29/2019] [Accepted: 01/04/2020] [Indexed: 12/19/2022]
Abstract
The extracellular matrix represents a dynamic microenvironment regulating essential cell functions in vivo. Tissue engineering approaches aim to recreate the native niche in vitro using biological scaffolds generated by organ decellularization. So far, the organ specific origin of such scaffolds was less considered and potential consequences for in vitro cell culture remain largely elusive. Here, we show that organ specific cues of biological scaffolds affect cellular behavior. In detail, we report on the generation of a well-preserved pancreatic bioscaffold and introduce a scoring system allowing standardized inter-study quality assessment. Using multiple analysis tools for in-depth-characterization of the biological scaffold, we reveal unique compositional, physico-structural, and biophysical properties. Finally, we prove the functional relevance of the biological origin by demonstrating a regulatory effect of the matrix on multi-lineage differentiation of human induced pluripotent stem cells emphasizing the significance of matrix specificity for cellular behavior in artificial microenvironments.
Collapse
Affiliation(s)
- Constantin Berger
- Chair Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Yngvild Bjørlykke
- Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
| | - Lukas Hahn
- Functional Polymer Materials, Department of Chemistry and Pharmacy and Bavarian Polymer Institute, Würzburg University, Würzburg, Germany
| | - Markus Mühlemann
- Chair Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Sebastian Kress
- Chair Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Heike Walles
- Translational Center Regenerative Therapies (TLC-RT), Fraunhofer Institute for Silicate Research ISC, Würzburg, Germany; Otto-von Guericke University, Core Facility Tissue Engineering, Magdeburg, Germany
| | - Robert Luxenhofer
- Functional Polymer Materials, Department of Chemistry and Pharmacy and Bavarian Polymer Institute, Würzburg University, Würzburg, Germany
| | - Helge Ræder
- Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
| | - Marco Metzger
- Chair Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Würzburg, Germany; Translational Center Regenerative Therapies (TLC-RT), Fraunhofer Institute for Silicate Research ISC, Würzburg, Germany
| | - Daniela Zdzieblo
- Chair Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Würzburg, Germany; Translational Center Regenerative Therapies (TLC-RT), Fraunhofer Institute for Silicate Research ISC, Würzburg, Germany.
| |
Collapse
|
50
|
Uhl FE, Zhang F, Pouliot RA, Uriarte JJ, Rolandsson Enes S, Han X, Ouyang Y, Xia K, Westergren-Thorsson G, Malmström A, Hallgren O, Linhardt RJ, Weiss DJ. Functional role of glycosaminoglycans in decellularized lung extracellular matrix. Acta Biomater 2020; 102:231-246. [PMID: 31751810 PMCID: PMC8713186 DOI: 10.1016/j.actbio.2019.11.029] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 11/09/2019] [Accepted: 11/12/2019] [Indexed: 01/17/2023]
Abstract
Despite progress in use of decellularized lung scaffolds in ex vivo lung bioengineering schemes, including use of gels and other materials derived from the scaffolds, the detailed composition and functional role of extracellular matrix (ECM) proteoglycans (PGs) and their glycosaminoglycan (GAG) chains remaining in decellularized lungs, is poorly understood. Using a commonly utilized detergent-based decellularization approach in human autopsy lungs resulted in disproportionate losses of GAGs with depletion of chondroitin sulfate/dermatan sulfate (CS/DS) > heparan sulfate (HS) > hyaluronic acid (HA). Specific changes in disaccharide composition of remaining GAGs were observed with disproportionate loss of NS and NS2S for HS groups and of 4S for CS/DS groups. No significant influence of smoking history, sex, time to autopsy, or age was observed in native vs. decellularized lungs. Notably, surface plasmon resonance demonstrated that GAGs remaining in decellularized lungs were unable to bind key matrix-associated growth factors FGF2, HGF, and TGFβ1. Growth of lung epithelial, pulmonary vascular, and stromal cells cultured on the surface of or embedded within gels derived from decellularized human lungs was differentially and combinatorially enhanced by replenishing specific GAGs and FGF2, HGF, and TGFβ1. In summary, lung decellularization results in loss and/or dysfunction of specific GAGs or side chains significantly affecting matrix-associated growth factor binding and lung cell metabolism. GAG and matrix-associated growth factor replenishment thus needs to be incorporated into schemes for investigations utilizing gels and other materials produced from decellularized human lungs. STATEMENT OF SIGNIFICANCE: Despite progress in use of decellularized lung scaffolds in ex vivo lung bioengineering schemes, including use of gels and other materials derived from the scaffolds, the detailed composition and functional role of extracellular matrix (ECM) proteoglycans (PGs) and their glycosaminoglycan (GAG) chains remaining in decellularized lungs, is poorly understood. In the current studies, we demonstrate that glycosaminoglycans (GAGs) are significantly depleted during decellularization and those that remain are dysfunctional and unable to bind matrix-associated growth factors critical for cell growth and differentiation. Systematically repleting GAGs and matrix-associated growth factors to gels derived from decellularized human lung significantly and differentially affects cell growth. These studies highlight the importance of considering GAGs in decellularized lungs and their derivatives.
Collapse
Affiliation(s)
- Franziska E Uhl
- University of Vermont, Larner College of Medicine, Burlington, VT, United States; Department of Experimental Medical Science, Faculty of Medicine, Lund University, Lund, Sweden
| | - Fuming Zhang
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, United States
| | - Robert A Pouliot
- University of Vermont, Larner College of Medicine, Burlington, VT, United States
| | - Juan J Uriarte
- University of Vermont, Larner College of Medicine, Burlington, VT, United States
| | - Sara Rolandsson Enes
- University of Vermont, Larner College of Medicine, Burlington, VT, United States; Department of Experimental Medical Science, Faculty of Medicine, Lund University, Lund, Sweden
| | - Xiaorui Han
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, United States
| | - Yilan Ouyang
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, United States
| | - Ke Xia
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, United States
| | | | - Anders Malmström
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, Lund, Sweden
| | - Oskar Hallgren
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, Lund, Sweden
| | - Robert J Linhardt
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, United States
| | - Daniel J Weiss
- University of Vermont, Larner College of Medicine, Burlington, VT, United States.
| |
Collapse
|