1
|
Liao R, Dewey MJ, Rong J, Johnson SA, D’Angelo WA, Hussey GS, Badylak SF. Matrix-bound nanovesicles alleviate particulate-induced periprosthetic osteolysis. SCIENCE ADVANCES 2024; 10:eadn1852. [PMID: 39423278 PMCID: PMC11488533 DOI: 10.1126/sciadv.adn1852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 09/16/2024] [Indexed: 10/21/2024]
Abstract
Aseptic loosening of orthopedic implants is an inflammatory disease characterized by immune cell activation, chronic inflammation, and destruction of periprosthetic bone, and is one of the leading reasons for prosthetic failure, affecting 12% of total joint arthroplasty patients. Matrix-bound nanovesicles (MBVs) are a subclass of extracellular vesicle recently shown to mitigate inflammation in preclinical models of rheumatoid arthritis and influenza-mediated "cytokine storm." The molecular mechanism of these anti-inflammatory properties is only partially understood. The objective of the present study was to investigate the effects of MBV on RANKL-induced osteoclast formation in vitro and particulate-induced osteolysis in vivo. Results showed that MBV attenuated osteoclast differentiation and activity by suppressing the NF-κB signaling pathway and downstream NFATc1, DC-STAMP, c-Src, and cathepsin K expression. In vivo, local administration of MBV attenuated ultrahigh molecular weight polyethylene particle-induced osteolysis, bone reconstruction, and periosteal inflammation. The results suggest that MBV may be a therapeutic option for preventing periprosthetic loosening.
Collapse
Affiliation(s)
- Runzhi Liao
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Marley J. Dewey
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Jiayang Rong
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Scott A. Johnson
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - William A. D’Angelo
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - George S. Hussey
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Stephen F. Badylak
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15219, USA
| |
Collapse
|
2
|
Capella-Monsonís H, Crum RJ, Hussey GS, Badylak SF. Advances, challenges, and future directions in the clinical translation of ECM biomaterials for regenerative medicine applications. Adv Drug Deliv Rev 2024; 211:115347. [PMID: 38844005 DOI: 10.1016/j.addr.2024.115347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/29/2024] [Accepted: 06/03/2024] [Indexed: 06/11/2024]
Abstract
Extracellular Matrix (ECM) scaffolds and biomaterials have been widely used for decades across a variety of diverse clinical applications and have been implanted in millions of patients worldwide. ECM-based biomaterials have been especially successful in soft tissue repair applications but their utility in other clinical applications such as for regeneration of bone or neural tissue is less well understood. The beneficial healing outcome with the use of ECM biomaterials is the result of their biocompatibility, their biophysical properties and their ability to modify cell behavior after injury. As a consequence of successful clinical outcomes, there has been motivation for the development of next-generation formulations of ECM materials ranging from hydrogels, bioinks, powders, to whole organ or tissue scaffolds. The continued development of novel ECM formulations as well as active research interest in these materials ensures a wealth of possibilities for future clinical translation and innovation in regenerative medicine. The clinical translation of next generation formulations ECM scaffolds faces predictable challenges such as manufacturing, manageable regulatory pathways, surgical implantation, and the cost required to address these challenges. The current status of ECM-based biomaterials, including clinical translation, novel formulations and therapies currently under development, and the challenges that limit clinical translation of ECM biomaterials are reviewed herein.
Collapse
Affiliation(s)
- Héctor Capella-Monsonís
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA 15219, USA; Department of Surgery, School of Medicine, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA; Viscus Biologics LLC, 2603 Miles Road, Cleveland, OH 44128, USA
| | - Raphael J Crum
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA 15219, USA; Department of Surgery, School of Medicine, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - George S Hussey
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA 15219, USA; Department of Pathology, School of Medicine, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Stephen F Badylak
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA 15219, USA; Department of Surgery, School of Medicine, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA; Department of Bioengineering, University of Pittsburgh, 3700 O'Hara Street, Pittsburgh, PA 15261, USA.
| |
Collapse
|
3
|
Škarková A, Bizzarri M, Janoštiak R, Mašek J, Rosel D, Brábek J. Educate, not kill: treating cancer without triggering its defenses. Trends Mol Med 2024; 30:673-685. [PMID: 38658206 DOI: 10.1016/j.molmed.2024.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 04/02/2024] [Accepted: 04/03/2024] [Indexed: 04/26/2024]
Abstract
Traditionally, anticancer therapies focus on restraining uncontrolled proliferation. However, these cytotoxic therapies expose cancer cells to direct killing, instigating the process of natural selection favoring survival of resistant cells that become the foundation for tumor progression and therapy failure. Recognizing this phenomenon has prompted the development of alternative therapeutic strategies. Here we propose strategies targeting cancer hallmarks beyond proliferation, aiming at re-educating cancer cells towards a less malignant phenotype. These strategies include controlling cell dormancy, transdifferentiation therapy, normalizing the cancer microenvironment, and using migrastatic therapy. Adaptive resistance to these educative strategies does not confer a direct proliferative advantage to resistant cells, as non-resistant cells are not subject to eradication, thereby delaying or preventing the development of therapy-resistant tumors.
Collapse
Affiliation(s)
- Aneta Škarková
- Department of Cell Biology, BIOCEV, Faculty of Science, Charles University, Vestec, Czech Republic
| | - Mariano Bizzarri
- System Biology Group Laboratory, Sapienza University, Rome, Italy
| | - Radoslav Janoštiak
- First Faculty of Medicine, BIOCEV, Charles University, Vestec, Czech Republic
| | - Jan Mašek
- Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Daniel Rosel
- Department of Cell Biology, BIOCEV, Faculty of Science, Charles University, Vestec, Czech Republic.
| | - Jan Brábek
- Department of Cell Biology, BIOCEV, Faculty of Science, Charles University, Vestec, Czech Republic.
| |
Collapse
|
4
|
Martinez-Uribe O, Becker TC, Garman KS. Promises and Limitations of Current Models for Understanding Barrett's Esophagus and Esophageal Adenocarcinoma. Cell Mol Gastroenterol Hepatol 2024; 17:1025-1038. [PMID: 38325549 PMCID: PMC11041847 DOI: 10.1016/j.jcmgh.2024.01.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/16/2024] [Accepted: 01/18/2024] [Indexed: 02/09/2024]
Abstract
BACKGROUND & AIMS This review was developed to provide a thorough and effective update on models relevant to esophageal metaplasia, dysplasia, and carcinogenesis, focusing on the advantages and limitations of different models of Barrett's esophagus (BE) and esophageal adenocarcinoma (EAC). METHODS This expert review was written on the basis of a thorough review of the literature combined with expert interpretation of the state of the field. We emphasized advances over the years 2012-2023 and provided detailed information related to the characterization of established human esophageal cell lines. RESULTS New insights have been gained into the pathogenesis of BE and EAC using patient-derived samples and single-cell approaches. Relevant animal models include genetic as well as surgical mouse models and emphasize the development of lesions at the squamocolumnar junction in the mouse stomach. Rat models are generated using surgical approaches that directly connect the small intestine and esophagus. Large animal models have the advantage of including features in human esophagus such as esophageal submucosal glands. Alternatively, cell culture approaches remain important in the field and allow for personalized approaches, and scientific rigor can be ensured by authentication of cell lines. CONCLUSIONS Research in BE and EAC remains highly relevant given the morbidity and mortality associated with cancers of the tubular esophagus and gastroesophageal junction. Careful selection of models and inclusion of human samples whenever possible will ensure relevance to human health and disease.
Collapse
Affiliation(s)
- Omar Martinez-Uribe
- Division of Gastroenterology, Department of Medicine, Duke University, Durham, North Carolina
| | - Thomas C Becker
- Division of Endocrinology, Department of Medicine, Duke University, Durham, North Carolina
| | - Katherine S Garman
- Division of Gastroenterology, Department of Medicine, Duke University, Durham, North Carolina.
| |
Collapse
|
5
|
Gujjar S, Tyagi A, Sainger S, Bharti P, Nain V, Sood P, Jayabal P, Sharma JC, Sharma P, Rajput S, Pandey AK, Pandey AK, Abnave P, Mathapati S. Biocompatible Human Placental Extracellular Matrix Derived Hydrogels. Adv Biol (Weinh) 2024; 8:e2300349. [PMID: 37786307 DOI: 10.1002/adbi.202300349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/29/2023] [Indexed: 10/04/2023]
Abstract
Solubilizing extracellular matrix (ECM) materials and transforming them into hydrogels has expanded their potential applications both in vitro and in vivo. In this study, hydrogels are prepared by decellularization of human placental tissue using detergent and enzymes and by the subsequent creation of a homogenized acellular placental tissue powder (P-ECM). A perfusion-based decellularization approach is employed using detergent and enzymes. The P-ECM with and without gamma irradiation is then utilized to prepare P-ECM hydrogels. Physical and biological evaluations are conducted to assess the suitability of the P-ECM hydrogels for biocompatibility. The decellularized tissue has significantly reduced cellular content and retains the major ECM proteins. Increasing the concentration of P-ECM leads to improved mechanical properties of the P-ECM hydrogels. The biocompatibility of the P-ECM hydrogel is demonstrated through cell proliferation and viability assays. Notably, gamma-sterilized P-ECM does not support the formation of a stable hydrogel. Nonetheless, the use of HCl during the digestion process effectively decreases spore growth and bacterial bioburden. The study demonstrates that P-ECM hydrogels exhibit physical and biological attributes conducive to soft tissue reconstruction. These hydrogels establish a favorable microenvironment for cell growth and the need for investigating innovative sterilization methods.
Collapse
Affiliation(s)
- Sunil Gujjar
- Biomaterials Laboratory, Translational Health Science and Technology Institute, Faridabad, Haryana, 121001, India
| | - Anurag Tyagi
- Biomaterials Laboratory, Translational Health Science and Technology Institute, Faridabad, Haryana, 121001, India
| | - Saloni Sainger
- Biomaterials Laboratory, Translational Health Science and Technology Institute, Faridabad, Haryana, 121001, India
| | - Puja Bharti
- National Centre for Cell Science, Pune, Maharashtra, 411007, India
| | - Vaibhav Nain
- Mycobacterial Pathogenesis Laboratory, Translational Health Science and Technology Institute, Faridabad, Haryana, 121001, India
| | - Pratibha Sood
- Biomaterials Laboratory, Translational Health Science and Technology Institute, Faridabad, Haryana, 121001, India
| | - Prakash Jayabal
- Biomaterials Laboratory, Translational Health Science and Technology Institute, Faridabad, Haryana, 121001, India
| | - Jagadish Chandra Sharma
- Employees State Insurance Corporation Medical College and Hospital, Faridabad, Haryana, 121012, India
| | - Priyanka Sharma
- Employees State Insurance Corporation Medical College and Hospital, Faridabad, Haryana, 121012, India
| | - Sanjay Rajput
- Shriram Institute for Industrial Research, Delhi, 110007, India
| | - Anil Kumar Pandey
- Employees State Insurance Corporation Medical College and Hospital, Faridabad, Haryana, 121012, India
| | - Amit Kumar Pandey
- Mycobacterial Pathogenesis Laboratory, Translational Health Science and Technology Institute, Faridabad, Haryana, 121001, India
| | - Prasad Abnave
- National Centre for Cell Science, Pune, Maharashtra, 411007, India
| | - Santosh Mathapati
- Biomaterials Laboratory, Translational Health Science and Technology Institute, Faridabad, Haryana, 121001, India
| |
Collapse
|
6
|
Cruz-Acuña R, Kariuki SW, Sugiura K, Karaiskos S, Plaster EM, Loebel C, Efe G, Karakasheva T, Gabre JT, Hu J, Burdick JA, Rustgi AK. Engineered hydrogel reveals contribution of matrix mechanics to esophageal adenocarcinoma and identifies matrix-activated therapeutic targets. J Clin Invest 2023; 133:e168146. [PMID: 37788109 PMCID: PMC10688988 DOI: 10.1172/jci168146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 09/28/2023] [Indexed: 10/05/2023] Open
Abstract
Increased extracellular matrix (ECM) stiffness has been implicated in esophageal adenocarcinoma (EAC) progression, metastasis, and resistance to therapy. However, the underlying protumorigenic pathways are yet to be defined. Additional work is needed to develop physiologically relevant in vitro 3D culture models that better recapitulate the human tumor microenvironment and can be used to dissect the contributions of matrix stiffness to EAC pathogenesis. Here, we describe a modular, tumor ECM-mimetic hydrogel platform with tunable mechanical properties, defined presentation of cell-adhesive ligands, and protease-dependent degradation that supports robust in vitro growth and expansion of patient-derived EAC 3D organoids (EAC PDOs). Hydrogel mechanical properties control EAC PDO formation, growth, proliferation, and activation of tumor-associated pathways that elicit stem-like properties in the cancer cells, as highlighted through in vitro and in vivo environments. We also demonstrate that the engineered hydrogel serves as a platform for identifying potential therapeutic targets to disrupt the contribution of protumorigenic matrix mechanics in EAC. Together, these studies show that an engineered PDO culture platform can be used to elucidate underlying matrix-mediated mechanisms of EAC and inform the development of therapeutics that target ECM stiffness in EAC.
Collapse
Affiliation(s)
- Ricardo Cruz-Acuña
- Herbert Irving Comprehensive Cancer Center, Division of Digestive and Liver Diseases, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York, USA
| | - Secunda W. Kariuki
- Herbert Irving Comprehensive Cancer Center, Division of Digestive and Liver Diseases, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York, USA
| | - Kensuke Sugiura
- Herbert Irving Comprehensive Cancer Center, Division of Digestive and Liver Diseases, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York, USA
| | - Spyros Karaiskos
- Herbert Irving Comprehensive Cancer Center, Division of Digestive and Liver Diseases, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York, USA
| | | | - Claudia Loebel
- Department of Materials Science and Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Gizem Efe
- Herbert Irving Comprehensive Cancer Center, Division of Digestive and Liver Diseases, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York, USA
| | - Tatiana Karakasheva
- Division of Gastroenterology, Hepatology, and Nutrition, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Joel T. Gabre
- Herbert Irving Comprehensive Cancer Center, Division of Digestive and Liver Diseases, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York, USA
| | - Jianhua Hu
- Herbert Irving Comprehensive Cancer Center, Division of Digestive and Liver Diseases, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York, USA
| | - Jason A. Burdick
- BioFrontiers Institute and Department of Chemical and Biological Engineering, University of Colorado, Boulder, Colorado, USA
| | - Anil K. Rustgi
- Herbert Irving Comprehensive Cancer Center, Division of Digestive and Liver Diseases, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York, USA
| |
Collapse
|
7
|
Liu H, Wang X. Esophageal organoids: applications and future prospects. J Mol Med (Berl) 2023; 101:931-945. [PMID: 37380866 DOI: 10.1007/s00109-023-02340-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 05/26/2023] [Accepted: 06/14/2023] [Indexed: 06/30/2023]
Abstract
Organoids have been developed in the last decade as a new research tool to simulate organ cell biology and disease. Compared to traditional 2D cell lines and animal models, experimental data based on esophageal organoids are more reliable. In recent years, esophageal organoids derived from multiple cell sources have been established, and relatively mature culture protocols have been developed. Esophageal inflammation and cancer are two directions of esophageal organoid modeling, and organoid models of esophageal adenocarcinoma, esophageal squamous cell carcinoma, and eosinophilic esophagitis have been established. The properties of esophageal organoids, which mimic the real esophagus, contribute to research in drug screening and regenerative medicine. The combination of organoids with other technologies, such as organ chips and xenografts, can complement the deficiencies of organoids and create entirely new research models that are more advantageous for cancer research. In this review, we will summarize the development of tumor and non-tumor esophageal organoids, the current application of esophageal organoids in disease modeling, regenerative medicine, and drug screening. We will also discuss the future prospects of esophageal organoids.
Collapse
Affiliation(s)
- Hongyuan Liu
- Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xianli Wang
- Shanghai Jiao Tong University, School of Public Health, Shanghai, 200025, China.
| |
Collapse
|
8
|
Yeruva T, Yang S, Doski S, Duncan GA. Hydrogels for Mucosal Drug Delivery. ACS APPLIED BIO MATERIALS 2023; 6:1684-1700. [PMID: 37126538 PMCID: PMC11966650 DOI: 10.1021/acsabm.3c00050] [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] [Indexed: 05/02/2023]
Abstract
Mucosal tissues are often a desirable site of drug action to treat disease and engage the immune system. However, systemically administered drugs suffer from limited bioavailability in mucosal tissues where technologies to enable direct, local delivery to these sites would prove useful. In this Spotlight on Applications article, we discuss hydrogels as an attractive means for local delivery of therapeutics to address a range of conditions affecting the eye, nose, oral cavity, gastrointestinal, urinary bladder, and vaginal tracts. Considering the barriers to effective mucosal delivery, we provide an overview of the key parameters in the use of hydrogels for these applications. Finally, we highlight recent work demonstrating their use for inflammatory and infectious diseases affecting these tissues.
Collapse
Affiliation(s)
- Taj Yeruva
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| | - Sydney Yang
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| | - Shadin Doski
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| | - Gregg A. Duncan
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| |
Collapse
|
9
|
Marzaro M, Pozzato G, Tedesco S, Algeri M, Pozzato A, Tomao L, Montano I, Torroni F, Balassone V, Contini ACI, Guerra L, D’Angelo T, Federici di Abriola G, Lupoi L, Caristo ME, Boškoski I, Costamagna G, Francalanci P, Astori G, Bozza A, Bagno A, Todesco M, Trovalusci E, Oglio LD, Locatelli F, Caldaro T. Decellularized esophageal tubular scaffold microperforated by quantum molecular resonance technology and seeded with mesenchymal stromal cells for tissue engineering esophageal regeneration. Front Bioeng Biotechnol 2022; 10:912617. [PMID: 36267444 PMCID: PMC9576845 DOI: 10.3389/fbioe.2022.912617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 08/17/2022] [Indexed: 11/25/2022] Open
Abstract
Current surgical options for patients requiring esophageal replacement suffer from several limitations and do not assure a satisfactory quality of life. Tissue engineering techniques for the creation of customized “self-developing” esophageal substitutes, which are obtained by seeding autologous cells on artificial or natural scaffolds, allow simplifying surgical procedures and achieving good clinical outcomes. In this context, an appealing approach is based on the exploitation of decellularized tissues as biological matrices to be colonized by the appropriate cell types to regenerate the desired organs. With specific regard to the esophagus, the presence of a thick connective texture in the decellularized scaffold hampers an adequate penetration and spatial distribution of cells. In the present work, the Quantum Molecular Resonance® (QMR) technology was used to create a regular microchannel structure inside the connective tissue of full-thickness decellularized tubular porcine esophagi to facilitate a diffuse and uniform spreading of seeded mesenchymal stromal cells within the scaffold. Esophageal samples were thoroughly characterized before and after decellularization and microperforation in terms of residual DNA content, matrix composition, structure and biomechanical features. The scaffold was seeded with mesenchymal stromal cells under dynamic conditions, to assess the ability to be repopulated before its implantation in a large animal model. At the end of the procedure, they resemble the original esophagus, preserving the characteristic multilayer composition and maintaining biomechanical properties adequate for surgery. After the sacrifice we had histological and immunohistochemical evidence of the full-thickness regeneration of the esophageal wall, resembling the native organ. These results suggest the QMR microperforated decellularized esophageal scaffold as a promising device for esophagus regeneration in patients needing esophageal substitution.
Collapse
Affiliation(s)
| | | | | | - Mattia Algeri
- Department of Pediatric Onco-Hematology and Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | | | - Luigi Tomao
- Department of Pediatric Onco-Hematology and Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Ilaria Montano
- Department of Pediatric Onco-Hematology and Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Filippo Torroni
- Digestive Endoscopy and Surgical Unit, Bambino Gesù Children’s Hospital, Rome, Italy
| | - Valerio Balassone
- Digestive Endoscopy and Surgical Unit, Bambino Gesù Children’s Hospital, Rome, Italy
| | | | - Luciano Guerra
- Digestive Endoscopy and Surgical Unit, Bambino Gesù Children’s Hospital, Rome, Italy
| | - Tommaso D’Angelo
- Digestive Endoscopy and Surgical Unit, Bambino Gesù Children’s Hospital, Rome, Italy
| | | | - Lorenzo Lupoi
- Cen.Ri.S. Policlinico Gemelli UNICATT Rome, Rome, Italy
| | | | - Ivo Boškoski
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Digestive Endoscopy Unit, Rome, Italy
- Università Cattolica del Sacro Cuore, Centre For Endoscopic Research Therapeutics and Training (CERTT), Rome, Italy—CERTT Gemelli, Rome, Italy
- *Correspondence: Ivo Boškoski,
| | - Guido Costamagna
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Digestive Endoscopy Unit, Rome, Italy
- Università Cattolica del Sacro Cuore, Centre For Endoscopic Research Therapeutics and Training (CERTT), Rome, Italy—CERTT Gemelli, Rome, Italy
| | | | - Giuseppe Astori
- Advanced Cellular Therapy Laboratory, Haematology Unit, San Bortolo Hospital, Vicenza, Italy
| | - Angela Bozza
- Advanced Cellular Therapy Laboratory, Haematology Unit, San Bortolo Hospital, Vicenza, Italy
- Consorzio Per la Ricerca Sanitaria (CORIS) of the Veneto Region, Padova, Italy
| | - Andrea Bagno
- Department of Industrial Engineering, University of Padova, Padova, Italy
| | - Martina Todesco
- Department of Industrial Engineering, University of Padova, Padova, Italy
| | - Emanuele Trovalusci
- Pediatric Surgery Department AULSS2 Treviso, University of Padova, Padova, Italy
| | - Luigi Dall’ Oglio
- Digestive Endoscopy and Surgical Unit, Bambino Gesù Children’s Hospital, Rome, Italy
| | - Franco Locatelli
- Department of Pediatric Onco-Hematology and Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
- Department of Pediatrics, Sapienza University of Rome, Roma, Italy
| | - Tamara Caldaro
- Digestive Endoscopy and Surgical Unit, Bambino Gesù Children’s Hospital, Rome, Italy
| |
Collapse
|
10
|
Barbon S, Biccari A, Stocco E, Capovilla G, D’Angelo E, Todesco M, Sandrin D, Bagno A, Romanato F, Macchi V, De Caro R, Agostini M, Merigliano S, Valmasoni M, Porzionato A. Bio-Engineered Scaffolds Derived from Decellularized Human Esophagus for Functional Organ Reconstruction. Cells 2022; 11:cells11192945. [PMID: 36230907 PMCID: PMC9563623 DOI: 10.3390/cells11192945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/14/2022] [Accepted: 09/15/2022] [Indexed: 11/16/2022] Open
Abstract
Esophageal reconstruction through bio-engineered allografts that highly resemble the peculiar properties of the tissue extracellular matrix (ECM) is a prospective strategy to overcome the limitations of current surgical approaches. In this work, human esophagus was decellularized for the first time in the literature by comparing three detergent-enzymatic protocols. After decellularization, residual DNA quantification and histological analyses showed that all protocols efficiently removed cells, DNA (<50 ng/mg of tissue) and muscle fibers, preserving collagen/elastin components. The glycosaminoglycan fraction was maintained (70–98%) in the decellularized versus native tissues, while immunohistochemistry showed unchanged expression of specific ECM markers (collagen IV, laminin). The proteomic signature of acellular esophagi corroborated the retention of structural collagens, basement membrane and matrix–cell interaction proteins. Conversely, decellularization led to the loss of HLA-DR expression, producing non-immunogenic allografts. According to hydroxyproline quantification, matrix collagen was preserved (2–6 µg/mg of tissue) after decellularization, while Second-Harmonic Generation imaging highlighted a decrease in collagen intensity. Based on uniaxial tensile tests, decellularization affected tissue stiffness, but sample integrity/manipulability was still maintained. Finally, the cytotoxicity test revealed that no harmful remnants/contaminants were present on acellular esophageal matrices, suggesting allograft biosafety. Despite the different outcomes showed by the three decellularization methods (regarding, for example, tissue manipulability, DNA removal, and glycosaminoglycans/hydroxyproline contents) the ultimate validation should be provided by future repopulation tests and in vivo orthotopic implant of esophageal scaffolds.
Collapse
Affiliation(s)
- Silvia Barbon
- Section of Human Anatomy, Department of Neuroscience, University of Padova, 35121 Padova, Italy
- L.i.f.e.L.a.b. Program, Consorzio per la Ricerca Sanitaria, 35128 Padova, Italy
- Foundation for Biology and Regenerative Medicine, Tissue Engineering and Signaling—TES, Onlus, 35136 Padova, Italy
| | - Andrea Biccari
- L.i.f.e.L.a.b. Program, Consorzio per la Ricerca Sanitaria, 35128 Padova, Italy
- Department of Surgical Oncological and Gastroenterological Sciences, University of Padova, 35128 Padova, Italy
| | - Elena Stocco
- Section of Human Anatomy, Department of Neuroscience, University of Padova, 35121 Padova, Italy
- L.i.f.e.L.a.b. Program, Consorzio per la Ricerca Sanitaria, 35128 Padova, Italy
- Foundation for Biology and Regenerative Medicine, Tissue Engineering and Signaling—TES, Onlus, 35136 Padova, Italy
| | - Giovanni Capovilla
- Department of Surgical Oncological and Gastroenterological Sciences, University of Padova, 35128 Padova, Italy
| | - Edoardo D’Angelo
- Department of Surgical Oncological and Gastroenterological Sciences, University of Padova, 35128 Padova, Italy
| | - Martina Todesco
- L.i.f.e.L.a.b. Program, Consorzio per la Ricerca Sanitaria, 35128 Padova, Italy
- Department of Industrial Engineering, University of Padova, 35131 Padova, Italy
| | - Deborah Sandrin
- L.i.f.e.L.a.b. Program, Consorzio per la Ricerca Sanitaria, 35128 Padova, Italy
- Department of Physics and Astronomy “G. Galilei”, University of Padova, 35131 Padova, Italy
| | - Andrea Bagno
- L.i.f.e.L.a.b. Program, Consorzio per la Ricerca Sanitaria, 35128 Padova, Italy
- Department of Industrial Engineering, University of Padova, 35131 Padova, Italy
| | - Filippo Romanato
- L.i.f.e.L.a.b. Program, Consorzio per la Ricerca Sanitaria, 35128 Padova, Italy
- Department of Physics and Astronomy “G. Galilei”, University of Padova, 35131 Padova, Italy
| | - Veronica Macchi
- Section of Human Anatomy, Department of Neuroscience, University of Padova, 35121 Padova, Italy
- L.i.f.e.L.a.b. Program, Consorzio per la Ricerca Sanitaria, 35128 Padova, Italy
| | - Raffaele De Caro
- Section of Human Anatomy, Department of Neuroscience, University of Padova, 35121 Padova, Italy
- L.i.f.e.L.a.b. Program, Consorzio per la Ricerca Sanitaria, 35128 Padova, Italy
- Foundation for Biology and Regenerative Medicine, Tissue Engineering and Signaling—TES, Onlus, 35136 Padova, Italy
| | - Marco Agostini
- L.i.f.e.L.a.b. Program, Consorzio per la Ricerca Sanitaria, 35128 Padova, Italy
- Department of Surgical Oncological and Gastroenterological Sciences, University of Padova, 35128 Padova, Italy
- Correspondence: ; Tel.: +39-049-96-40-160
| | - Stefano Merigliano
- L.i.f.e.L.a.b. Program, Consorzio per la Ricerca Sanitaria, 35128 Padova, Italy
- Department of Surgical Oncological and Gastroenterological Sciences, University of Padova, 35128 Padova, Italy
| | - Michele Valmasoni
- L.i.f.e.L.a.b. Program, Consorzio per la Ricerca Sanitaria, 35128 Padova, Italy
- Department of Surgical Oncological and Gastroenterological Sciences, University of Padova, 35128 Padova, Italy
| | - Andrea Porzionato
- Section of Human Anatomy, Department of Neuroscience, University of Padova, 35121 Padova, Italy
- L.i.f.e.L.a.b. Program, Consorzio per la Ricerca Sanitaria, 35128 Padova, Italy
- Foundation for Biology and Regenerative Medicine, Tissue Engineering and Signaling—TES, Onlus, 35136 Padova, Italy
| |
Collapse
|
11
|
Brown M, Li J, Moraes C, Tabrizian M, Li-Jessen NY. Decellularized extracellular matrix: New promising and challenging biomaterials for regenerative medicine. Biomaterials 2022; 289:121786. [DOI: 10.1016/j.biomaterials.2022.121786] [Citation(s) in RCA: 117] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 08/22/2022] [Accepted: 08/29/2022] [Indexed: 11/28/2022]
|
12
|
Ullal TV, Marks SL, Belafsky PC, Conklin JL, Pandolfino JE. A Comparative Assessment of the Diagnosis of Swallowing Impairment and Gastroesophageal Reflux in Canines and Humans. Front Vet Sci 2022; 9:889331. [PMID: 35754550 PMCID: PMC9228035 DOI: 10.3389/fvets.2022.889331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 05/05/2022] [Indexed: 11/24/2022] Open
Abstract
Swallowing impairment is a highly prevalent and clinically significant problem affecting people and dogs. There are myriad causes of swallowing impairment of which gastroesophageal reflux is the most common in both species. Similarities in anatomy and physiology between humans and canines results in analogous swallowing disorders including cricopharyngeus muscle achalasia, esophageal achalasia, hiatal herniation, and gastroesophageal reflux with secondary esophagitis and esophageal dysmotility. Accordingly, the diagnostic approach to human and canine patients with swallowing impairment is similar. Diagnostic procedures such as swallowing fluoroscopy, high-resolution manometry, pH/impedance monitoring, and endolumenal functional luminal imaging probe can be performed in both species; however, nasofacial conformation, increased esophageal length, and the difficulty of completing several of these procedures in awake dogs are inherent challenges that need to be considered. Human patients can convey their symptoms and respond to verbal cues, whereas veterinarians must rely on clinical histories narrated by pet owners followed by comprehensive physical examination and observation of the animal eating different food consistencies and drinking water. Dogs may also be unwilling to drink or eat in the hospital setting and may be resistant to physical restraint during diagnostic procedures. Despite the species differences and diagnostic challenges, dogs are a natural animal model for many oropharyngeal and esophageal disorders affecting people, which presents a tremendous opportunity for shared learnings. This manuscript reviews the comparative aspects of esophageal anatomy and physiology between humans and canines, summarizes the diagnostic assessment of swallowing impairment in both species, and discusses future considerations for collaborative medicine and translational research.
Collapse
Affiliation(s)
- Tarini V Ullal
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Stanley L Marks
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Peter C Belafsky
- Department of Otolaryngology, Center for Voice and Swallowing, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Jeffrey L Conklin
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, UCLA Robert G. Kardashian Center for Esophageal Health, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - John E Pandolfino
- Division of Gastroenterology and Hepatology, Department of Medicine, Northwestern Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
13
|
Mai Y, Ouyang Y, Qin Y, Jia C, McCoubrey LE, Basit AW, Nie Y, Jia Y, Yu L, Dou L, Deng W, Deng Y, Liu Y. Poly(lactic acid)-hyperbranched polyglycerol nanoparticles enhance bioadhesive treatment of esophageal disease and reduce systemic drug exposure. NANOSCALE 2022; 14:8418-8428. [PMID: 35639565 DOI: 10.1039/d2nr01846b] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The effective treatment of esophageal disease represents a significant unmet clinical need, as existing treatments often lead to unnecessary systemic drug exposure and suboptimal concentrations at the disease site. Here, surface-modified bioadhesive poly(lactic acid)-hyperbranched polyglycerol nanoparticles (BNPs), with an average 100-200 nm diameter, were developed for local and sustained esophageal drug delivery. BNPs showed significantly higher adhesion and permeation into ex vivo human and rat esophageal tissue than non-adhesive nanoparticles (NNPs) and had longer residence times within the rat esophagus in vivo. Incubation with human esophagus (Het-1A) cells confirmed BNPs' biocompatibility at clinically relevant concentrations. In a rat model of achalasia, nifedipine-loaded BNPs significantly enhanced esophageal drug exposure, increased therapeutic efficacy, and reduced systemic drug exposure compared to NNPs and free drug. The safety of BNPs was demonstrated by an absence of intestinal, hepatic, and splenic toxicity following administration. This study is the first to demonstrate the efficacy of BNPs for esophageal drug delivery and highlight their potential for improving the lives of patients suffering with esophageal conditions.
Collapse
Affiliation(s)
- Yang Mai
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, 510275, China.
| | - Yaqi Ouyang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, 510275, China.
| | - Yujia Qin
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, 510275, China.
| | - Changchang Jia
- Cell-Gene Therapy Translational Medicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510000, China
| | - Laura E McCoubrey
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, UK
| | - Abdul W Basit
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, UK
| | - Yichu Nie
- Clinical Research Institute, The First People's Hospital of Foshan & Sun Yat-sen University Foshan Hospital, Foshan, 528000, China
| | - Yizhen Jia
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, 510275, China.
| | - Liu Yu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, 510275, China.
| | - Liu Dou
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, 510275, China.
| | - Wenbin Deng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, 510275, China.
| | - Yang Deng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, 510275, China.
| | - Yang Liu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, 510275, China.
| |
Collapse
|
14
|
Orozco-Vega A, Montes-Rodríguez MI, Luévano-Colmenero GH, Barros-Gómez J, Muñoz-González PU, Flores-Moreno M, Delgadillo-Holtfort I, Vega-González A, Rojo FJ, Guinea GV, Mendoza-Novelo B. Decellularization of porcine esophageal tissue at three diameters and the bioscaffold modification with EETs-ECM gel. J Biomed Mater Res A 2022; 110:1669-1680. [PMID: 35703732 DOI: 10.1002/jbm.a.37416] [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: 01/04/2022] [Revised: 05/30/2022] [Accepted: 06/02/2022] [Indexed: 11/07/2022]
Abstract
Damaged complex modular organs repair is a current clinical challenge in which one of the primary goals is to keep their biological response. An interesting case of study it is the porcine esophagus since it is a tubular muscular tissue selected as raw material for tissue engineering. The design of esophageal constructs can draw on properties of the processed homologous extracellular matrix (ECM). In this work, we report the decellularization of multilayered esophagus tissue from 1-, 21- and 45-days old piglets through the combination of reversible alkaline swelling and detergent perfusion. The bioscaffolds were characterized in terms of their residual composition and tensile mechanical properties. The biological response to esophageal submucosal derived bioscaffolds modified with ECM gel containing epoxyeicosatrienoic acids (EETs) was then evaluated. Results suggest that the composition (laminin, fibronectin, and sulphated glycosaminoglycans/sGAG) depends on the donor age: a better efficiency of the decellularization process combined with a higher retention of sGAG and fibronectin is observed in piglet esophageal scaffolds. The heterogeneity of this esophageal ECM is maintained, which implied the preservation of anisotropic tensile properties. Coating of bioscaffolds with ECM gel is suitable for carrying esophageal epithelial cells and EETs. Bioactivity of EETs-ECM gel modified esophageal submucosal bioscaffolds is observed to promote neovascularization and antiinflammatory after rabbit full-thickness esophageal defect replacement.
Collapse
Affiliation(s)
- Adriana Orozco-Vega
- División de Ciencias e Ingenierías, Universidad de Guanajuato, León, Gto, Mexico
| | - Metzeri I Montes-Rodríguez
- División de Ciencias e Ingenierías, Universidad de Guanajuato, León, Gto, Mexico.,Hospital Gineco-Pediatra No 48, Centro Médico Nacional del Bajío, UMAE, Instituto Mexicano del Seguro Social, León, Gto, Mexico
| | - Guadalupe H Luévano-Colmenero
- División de Ciencias e Ingenierías, Universidad de Guanajuato, León, Gto, Mexico.,Unidad Profesional Interdisciplinaria de Ingeniería, Campus Guanajuato, Instituto Politécnico Nacional, Silao de la Victoria, Gto, Mexico
| | - Jimena Barros-Gómez
- División de Ciencias e Ingenierías, Universidad de Guanajuato, León, Gto, Mexico
| | | | | | | | - Arturo Vega-González
- División de Ciencias e Ingenierías, Universidad de Guanajuato, León, Gto, Mexico
| | - Francisco J Rojo
- Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Pozuelo de Alarcón, Spain.,Departamento de Ciencia de Materiales, ETSI de Caminos, Canales y Puertos, Universidad Politécnica de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| | - Gustavo V Guinea
- Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Pozuelo de Alarcón, Spain.,Departamento de Ciencia de Materiales, ETSI de Caminos, Canales y Puertos, Universidad Politécnica de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain.,Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | | |
Collapse
|
15
|
Tang W, Qi J, Wang Q, Qu Y, Fu S, Luan J. Investigating the Adipogenic Effects of Different Tissue-Derived Decellularized Matrices. Front Bioeng Biotechnol 2022; 10:872897. [PMID: 35497363 PMCID: PMC9046558 DOI: 10.3389/fbioe.2022.872897] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 03/29/2022] [Indexed: 11/13/2022] Open
Abstract
Objective: Decellularized adipose-derived matrix (DAM) can promote adipogenic differentiation and adipose tissue remodeling, but the biological impact of tissue origin on DAM remains unknown. The present study aimed to investigate the effects of tissue origins on the adipogenic capacity of the decellularized matrix by comparing the cellular and tissue responses of DAM versus acellular dermal matrix (ADM). Methods: The in vitro response of adipose-derived stem/stromal cells (ADSCs) to DAM and ADM was characterized by proliferation and differentiation. The in vivo remodeling response was evaluated in the subcutaneous injection model of immunocompromised mice, using histology, protein expression, and transcriptome analysis. Results: Both DAM and ADM exhibited excellent decellularization effects and cytocompatibility. In the absence of exogenous stimuli, DAM could induce adipogenic differentiation of ADSCs compared with ADM. In the animal model, the levels of PDGF, VEGF, and ACRP30 were higher in the DAM groups than in the ADM group, and more neovascularization and extensive adipose tissue remodeling were observed. The mRNA-seq analysis indicated that the DAM implant regulated tissue remodeling by modulating Lat1/2 expression along with Hippo Signaling pathway in the early stage. Conclusion: Tissue origin can influence the biological response of the decellularized matrix. DAM can retain favorable tissue-specific characteristics after the decellularization process and have unique adipogenic effects in vitro and vivo, which can be fully utilized for soft tissue repair and regeneration.
Collapse
Affiliation(s)
| | | | | | | | - Su Fu
- *Correspondence: Su Fu, ; Jie Luan,
| | - Jie Luan
- *Correspondence: Su Fu, ; Jie Luan,
| |
Collapse
|
16
|
Murdock MH, Hussey GS, Chang JT, Hill RC, Nascari DG, Rao AV, Hansen KC, Foley LM, Hitchens TK, Amankulor NM, Badylak SF. A liquid fraction of extracellular matrix inhibits glioma cell viability in vitro and in vivo. Oncotarget 2022; 13:426-438. [PMID: 35198102 PMCID: PMC8860176 DOI: 10.18632/oncotarget.28203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 02/07/2022] [Indexed: 12/04/2022] Open
Abstract
Suppressive effects of extracellular matrix (ECM) upon various cancers have been reported. Glioblastoma multiforme has poor prognosis and new therapies are desired. This work investigated the effects of a saline-soluble fraction of urinary bladder ECM (ECM-SF) upon glioma cells. Viability at 24 hours in 1, 5, or 10 mg/mL ECM-SF-spiked media was evaluated in primary glioma cells (0319, 1015, 1119), glioma cell lines (A172, T98G, U87MG, C6), and brain cell lines (HCN-2, HMC3). Viability universally decreased at 5 and 10 mg/mL with U87MG, HCN-2, and HCM3 being least sensitive. Apoptosis in 0319 and 1119 cells was confirmed via NucView 488. Bi-weekly intravenous injection of ECM-SF (120 mg/kg) for 10 weeks in Sprague-Dawley rats did not affect weight, temperature, complete blood count, or multi-organ histology (N = 5). Intratumoral injection of ECM-SF (10 uL of 30 mg/mL) at weeks 2-4 post C6 inoculation in Wistar rats increased median survival from 24.5 to 51 days (hazard ratio for death 0.22) and decreased average tumor volume at time of death from 349 mm3 to 90 mm3 over 10 weeks (N = 6). Mass spectrometry identified 2,562 protein species in ECM-SF, parent ECM, and originating tissue. These results demonstrate the suppressive effects of ECM on glioma and warrant further study.
Collapse
Affiliation(s)
- Mark H. Murdock
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - George S. Hussey
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jordan T. Chang
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ryan C. Hill
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO, USA
| | - David G. Nascari
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Aparna V. Rao
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kirk C. Hansen
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO, USA
| | - Lesley M. Foley
- Animal Imaging Center, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - T. Kevin Hitchens
- Animal Imaging Center, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nduka M. Amankulor
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Stephen F. Badylak
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
17
|
The Immune Underpinnings of Barrett's-Associated Adenocarcinogenesis: a Retrial of Nefarious Immunologic Co-Conspirators. Cell Mol Gastroenterol Hepatol 2022; 13:1297-1315. [PMID: 35123116 PMCID: PMC8933845 DOI: 10.1016/j.jcmgh.2022.01.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 01/25/2022] [Accepted: 01/26/2022] [Indexed: 12/10/2022]
Abstract
There is no doubt that chronic gastroesophageal reflux disease increases the risk of esophageal adenocarcinoma (EAC) by several fold (odds ratio, 6.4; 95% CI, 4.6-9.1), and some relationships between reflux disease-mediated inflammation and oncogenic processes have been explored; however, the precise interconnections between the immune response and genomic instabilities underlying these pathologic processes only now are emerging. Furthermore, the precise cell of origin of the precancerous stages associated with EAC development, Barrett's esophagus, be it cardia resident or embryonic remnant, may shape our interpretation of the likely immune drivers. This review integrates the current collective knowledge of the immunology underlying EAC development and outlines a framework connecting proinflammatory pathways, such as those mediated by interleukin 1β, tumor necrosis factor α, leukemia inhibitory factor, interleukin 6, signal transduction and activator of transcription 3, nuclear factor-κB, cyclooxygenase-2, and transforming growth factor β, with oncogenic pathways in the gastroesophageal reflux disease-Barrett's esophagus-EAC cancer sequence. Further defining these immune and molecular railroads may show a map of the routes taken by gastroesophageal cells on their journey toward EAC tumor phylogeny. The selective pressures applied by this immune-induced journey likely impact the phenotype and genotype of the resulting oncogenic destination and further exploration of lesser-defined immune drivers may be useful in future individualized therapies or enhanced selective application of recent immune-driven therapeutics.
Collapse
|
18
|
Crum RJ, Hall K, Molina CP, Hussey GS, Graham E, Li H, Badylak SF. Immunomodulatory matrix-bound nanovesicles mitigate acute and chronic pristane-induced rheumatoid arthritis. NPJ Regen Med 2022; 7:13. [PMID: 35110573 PMCID: PMC8810774 DOI: 10.1038/s41536-022-00208-9] [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: 03/19/2021] [Accepted: 12/20/2021] [Indexed: 12/13/2022] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease characterized by chronic inflammation and destruction of synovial joints affecting ~7.5 million people worldwide. Disease pathology is driven by an imbalance in the ratio of pro-inflammatory vs. anti-inflammatory immune cells, especially macrophages. Modulation of macrophage phenotype, specifically an M1 to M2, pro- to anti-inflammatory transition, can be induced by biologic scaffold materials composed of extracellular matrix (ECM). The ECM-based immunomodulatory effect is thought to be mediated in part through recently identified matrix-bound nanovesicles (MBV) embedded within ECM. Isolated MBV was delivered via intravenous (i.v.) or peri-articular (p.a.) injection to rats with pristane-induced arthritis (PIA). The results of MBV administration were compared to intraperitoneal (i.p.) administration of methotrexate (MTX), the clinical standard of care. Relative to the diseased animals, i.p. MTX, i.v. MBV, and p.a. MBV reduced arthritis scores in both acute and chronic pristane-induced arthritis, decreased synovial inflammation, decreased adverse joint remodeling, and reduced the ratio of synovial and splenic M1 to M2 macrophages (p < 0.05). Both p.a. and i.v. MBV reduced the serum concentration of RA and PIA biomarkers CXCL10 and MCP-3 in the acute and chronic phases of disease (p < 0.05). Flow-cytometry revealed the presence of a systemic CD43hi/His48lo/CD206+, immunoregulatory monocyte population unique to p.a. and i.v. MBV treatment associated with disease resolution. The results show that the therapeutic efficacy of MBV is equal to that of MTX for the management of acute and chronic pristane-induced arthritis and, further, this effect is associated with modulation of local synovial macrophages and systemic myeloid populations.
Collapse
Affiliation(s)
- Raphael J Crum
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Suite 300, Pittsburgh, PA, 15219, USA
| | - Kelsey Hall
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Suite 300, Pittsburgh, PA, 15219, USA
| | - Catalina Pineda Molina
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Suite 300, Pittsburgh, PA, 15219, USA.,Department of Surgery, School of Medicine, University of Pittsburgh, University of Pittsburgh Medical Center Presbyterian Hospital, 200 Lothrop Street, Pittsburgh, PA, 15213, USA
| | - George S Hussey
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Suite 300, Pittsburgh, PA, 15219, USA.,Department of Surgery, School of Medicine, University of Pittsburgh, University of Pittsburgh Medical Center Presbyterian Hospital, 200 Lothrop Street, Pittsburgh, PA, 15213, USA.,ECM Therapeutics, Inc., 118 Marshall Dr., Warrendale, PA, 15086, USA
| | - Emma Graham
- Musculoskeletal Growth and Regeneration Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, 450 Technology Drive, Suite 206, Pittsburgh, PA, 15219, USA
| | - Hongshuai Li
- Department of Orthopedics and Rehabilitation, University of Iowa, 25 Grand Ave, Iowa City, IA, 52246, USA
| | - Stephen F Badylak
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Suite 300, Pittsburgh, PA, 15219, USA. .,Department of Surgery, School of Medicine, University of Pittsburgh, University of Pittsburgh Medical Center Presbyterian Hospital, 200 Lothrop Street, Pittsburgh, PA, 15213, USA. .,ECM Therapeutics, Inc., 118 Marshall Dr., Warrendale, PA, 15086, USA. .,Department of Bioengineering, University of Pittsburgh, 3700 O'Hara Street, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
19
|
Personalization of medical treatments in oncology: time for rethinking the disease concept to improve individual outcomes. EPMA J 2021; 12:545-558. [PMID: 34642594 PMCID: PMC8495186 DOI: 10.1007/s13167-021-00254-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 09/01/2021] [Indexed: 12/12/2022]
Abstract
The agenda of pharmacology discovery in the field of personalized oncology was dictated by the search of molecular targets assumed to deterministically drive tumor development. In this perspective, genes play a fundamental "causal" role while cells simply act as causal proxies, i.e., an intermediate between the molecular input and the organismal output. However, the ceaseless genomic change occurring across time within the same primary and metastatic tumor has broken the hope of a personalized treatment based only upon genomic fingerprint. Indeed, current models are unable in capturing the unfathomable complexity behind the outbreak of a disease, as they discard the contribution of non-genetic factors, environment constraints, and the interplay among different tiers of organization. Herein, we posit that a comprehensive personalized model should view at the disease as a "historical" process, in which different spatially and timely distributed factors interact with each other across multiple levels of organization, which collectively interact with a dynamic gene-expression pattern. Given that a disease is a dynamic, non-linear process - and not a static-stable condition - treatments should be tailored according to the "timing-frame" of each condition. This approach can help in detecting those critical transitions through which the system can access different attractors leading ultimately to diverse outcomes - from a pre-disease state to an overt illness or, alternatively, to recovery. Identification of such tipping points can substantiate the predictive and the preventive ambition of the Predictive, Preventive and Personalized Medicine (PPPM/3PM). However, an unusual effort is required to conjugate multi-omics approaches, data collection, and network analysis reconstruction (eventually involving innovative Artificial Intelligent tools) to recognize the critical phases and the relevant targets, which could help in patient stratification and therapy personalization.
Collapse
|
20
|
Zhang W, Du A, Liu S, Lv M, Chen S. Research progress in decellularized extracellular matrix-derived hydrogels. Regen Ther 2021; 18:88-96. [PMID: 34095366 PMCID: PMC8142036 DOI: 10.1016/j.reth.2021.04.002] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/21/2021] [Accepted: 04/27/2021] [Indexed: 12/17/2022] Open
Abstract
Decellularized extracellular matrix (dECM) is widely used in regenerative medicine as a scaffold material due to its unique biological activity and good biocompatibility. Hydrogel is a three-dimensional network structure polymer with high water content and high swelling that can simulate the water environment of human tissues, has good biocompatibility, and can exchange nutrients, oxygen, and waste with cells. At present, hydrogel is the ideal biological material for tissue engineering. In recent years, rapid development of the hydrogel theory and technology and progress in the use of dECM to form hydrogels have attracted considerable attention to dECM hydrogels as an innovative method for tissue engineering and regenerative medicine. This article introduces the classification of hydrogels, and focuses on the history and formation of dECM hydrogels, the source of dECM, the application of dECM hydrogels in tissue engineering and the commercial application of dECM materials.
Collapse
Affiliation(s)
- Wenhui Zhang
- Institute of Applied Anatomy and Reproductive Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Aoling Du
- Medical College, Hubei University of Arts and Science, Xiangyang, Hubei 441053, China
| | - Shun Liu
- Institute of Applied Anatomy and Reproductive Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Mingyue Lv
- Anesthesia Class 1 of Chuanshan College, South China University, Hengyang, Hunan 421001, China
| | - Shenghua Chen
- Institute of Applied Anatomy and Reproductive Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| |
Collapse
|
21
|
Baumeister T, Ingermann J, Marcazzan S, Fang HY, Oellinger R, Rad R, Engleitner T, Kleigrewe K, Anand A, Strangmann J, Schmid RM, Wang TC, Quante M. Anti-inflammatory chemoprevention attenuates the phenotype in a mouse model of esophageal adenocarcinoma. Carcinogenesis 2021; 42:1068-1078. [PMID: 33878160 DOI: 10.1093/carcin/bgab032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 01/29/2021] [Accepted: 04/16/2021] [Indexed: 01/20/2023] Open
Abstract
Barrett´s Esophagus (BE) is the main known precursor condition of Esophageal Adenocarcinoma (EAC). BE is defined by the presence of metaplasia above the normal squamous columnar junction and has mainly been attributed to gastroesophageal reflux disease (GERD) and chronic reflux esophagitis. Thus, the rising incidence of EAC in the Western world is likely mediated by chronic esophageal inflammation, secondary to GERD in combination with environmental risk factors such as a Western diet and obesity. However, (at present) risk prediction tools and endoscopic surveillance have shown limited effectiveness. Chemoprevention as an adjunctive approach remains an attractive option to reduce the incidence of neoplastic disease. Here, we investigate the feasibility of chemopreventive approaches in BE and EAC via inhibition of inflammatory signaling in a transgenic mouse model of BE and EAC (L2-IL1B mice), with accelerated tumor formation on a high fat diet (HFD). L2-IL1B mice were treated with the IL-1 receptor antagonist Anakinra and the nonsteroidal anti-inflammatory drugs (NSAIDs) aspirin or Sulindac. Interleukin-1b antagonism reduced tumor progression in L2-IL1B mice with or without a HFD, while both NSAIDs were effective chemoprevention agents in the accelerated HFD fed L2-IL1B mouse model. Sulindac treatment also resulted in a marked change in the immune profile of L2-IL-1B mice. In summary, anti-inflammatory treatment of HFD-treated L2-IL1B mice acted protectively on disease progression. These results from a mouse model of BE support results from clinical trials that suggest that anti-inflammatory medication may be effective in the chemoprevention of EAC.
Collapse
Affiliation(s)
- Theresa Baumeister
- Department of Medicine II, Klinikum rechts der Isar, Technical University Munich (TUM), Ismaninger Str, München, Germany
| | - Jonas Ingermann
- Department of Medicine II, Klinikum rechts der Isar, Technical University Munich (TUM), Ismaninger Str, München, Germany
| | - Sabrina Marcazzan
- Department of Medicine II, Klinikum rechts der Isar, Technical University Munich (TUM), Ismaninger Str, München, Germany
| | - Hsin-Yu Fang
- Department of Medicine II, Klinikum rechts der Isar, Technical University Munich (TUM), Ismaninger Str, München, Germany
| | - Rupert Oellinger
- Institute of Molecular Oncology and Functional Genomics, Klinikum rechts der Isar, Technical University Munich (TUM), Ismaninger Str, München, Germany
| | - Roland Rad
- Department of Medicine II, Klinikum rechts der Isar, Technical University Munich (TUM), Ismaninger Str, München, Germany.,Institute of Molecular Oncology and Functional Genomics, Klinikum rechts der Isar, Technical University Munich (TUM), Ismaninger Str, München, Germany
| | - Thomas Engleitner
- Institute of Molecular Oncology and Functional Genomics, Klinikum rechts der Isar, Technical University Munich (TUM), Ismaninger Str, München, Germany
| | - Karin Kleigrewe
- Leibniz-Institute for Food Systems Biology at the Technical University of Munich
| | - Akanksha Anand
- Department of Medicine II, Klinikum rechts der Isar, Technical University Munich (TUM), Ismaninger Str, München, Germany
| | - Julia Strangmann
- Department of Medicine II, Klinikum rechts der Isar, Technical University Munich (TUM), Ismaninger Str, München, Germany
| | - Roland M Schmid
- Department of Medicine II, Klinikum rechts der Isar, Technical University Munich (TUM), Ismaninger Str, München, Germany
| | - Timothy C Wang
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Michael Quante
- Department of Medicine II, Klinikum rechts der Isar, Technical University Munich (TUM), Ismaninger Str, München, Germany.,Innere Medizin II, Universitätskliniken Freiburg, Universität Freiburg, Germany
| |
Collapse
|
22
|
Sachdeva UM, Shimonosono M, Flashner S, Cruz-Acuña R, Gabre JT, Nakagawa H. Understanding the cellular origin and progression of esophageal cancer using esophageal organoids. Cancer Lett 2021; 509:39-52. [PMID: 33838281 DOI: 10.1016/j.canlet.2021.03.031] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/25/2021] [Accepted: 03/29/2021] [Indexed: 02/06/2023]
Abstract
Three-dimensional (3D) organoids are a novel tool to model epithelial cell biology and human diseases of the esophagus. 3D organoid culture systems have been utilized to investigate the pathobiology of esophageal cancer, including both squamous cell carcinoma and adenocarcinoma. Additional organoid-based approaches for study of esophageal development and benign esophageal diseases have provided key insights into esophageal keratinocyte differentiation and mucosal regeneration. These investigations have implications for the identification of esophageal cancer stem cells, as well as the potential to halt malignant progression through induction of differentiation pathways. Patient-derived organoids (PDOs) from human tissue samples allow for unique and faithful in vitro modeling of esophageal cancers, and provide an exciting platform for investigation into personalized medicine and targeted treatment approaches, as well as new models for understanding therapy resistance and recurrent disease. Future directions include high-throughput genomic screening using PDOs, and study of tumor-microenvironmental interactions through co-culture with immune and stromal cells and novel extracellular matrix complexes.
Collapse
Affiliation(s)
- Uma M Sachdeva
- Divison of Thoracic Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Masataka Shimonosono
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| | - Samuel Flashner
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| | - Ricardo Cruz-Acuña
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| | - Joel T Gabre
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, NY, USA; Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| | - Hiroshi Nakagawa
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, NY, USA; Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA.
| |
Collapse
|
23
|
The effect of normal, metaplastic, and neoplastic esophageal extracellular matrix upon macrophage activation. ACTA ACUST UNITED AC 2020; 13. [PMID: 34027260 DOI: 10.1016/j.regen.2020.100037] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Introduction Macrophages are capable of extreme plasticity and their activation state has been strongly associated with solid tumor growth progression and regression. Although the macrophage response to extracellular matrix (ECM) isolated from normal tissue is reasonably well understood, there is a relative dearth of information regarding their response to ECM isolated from chronically inflamed tissues, pre-neoplastic tissues, and neoplastic tissues. Esophageal adenocarcinoma (EAC) is a type of neoplasia driven by chronic inflammation in the distal esophagus, and the length of the esophagus provides the opportunity to investigate macrophage behavior in the presence of ECM isolated from a range of disease states within the same organ. Methods Normal, metaplastic, and neoplastic ECM hydrogels were prepared from decellularized EAC tissue. The hydrogels were evaluated for their nanofibrous structure (SEM), biochemical profile (targeted and global proteomics), and direct effect upon macrophage (THP-1 cell) activation state (qPCR, ELISA, immunolabeling) and indirect effect upon epithelial cell (Het-1A) migration (Boyden chamber). Results Nanofibrous ECM hydrogels from the three tissue types could be formed, and normal and neoplastic ECM showed distinctive protein profiles by targeted and global mass spectroscopy. ECM proteins functionally related to cancer and tumorigenesis were identified in the neoplastic esophageal ECM including collagen alpha-1(VIII) chain (COL8A1), lumican, and elastin. Metaplastic and neoplastic esophageal ECM induce distinctive effects upon THP-1 macrophage signaling compared to normal esophageal ECM. These effects include activation of pro-inflammatory IFNγ and TNFα gene expression and anti-inflammatory IL1RN gene expression. Most notably, neoplastic ECM robustly increased macrophage TNFα protein expression. The secretome of macrophages pre-treated with metaplastic and neoplastic ECM increases the migration of normal esophageal epithelial cells, similar behavior to that shown by tumor cells. Metaplastic ECM shows similar but less pronounced effects than neoplastic ECM suggesting the abnormal signals also exist within the pre-cancerous state. Conclusion A progressively diseased ECM, as exists within the esophagus exposed to chronic gastric reflux, can provide insights into novel biomarkers of early disease and identify potential therapeutic targets.
Collapse
|
24
|
Ha DH, Chae S, Lee JY, Kim JY, Yoon J, Sen T, Lee SW, Kim HJ, Cho JH, Cho DW. Therapeutic effect of decellularized extracellular matrix-based hydrogel for radiation esophagitis by 3D printed esophageal stent. Biomaterials 2020; 266:120477. [PMID: 33120198 DOI: 10.1016/j.biomaterials.2020.120477] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 09/26/2020] [Accepted: 10/18/2020] [Indexed: 02/07/2023]
Abstract
Radiation esophagitis, the most common acute adverse effect of radiation therapy, leads to unwanted consequences including discomfort, pain, an even death. However, no direct cure exists for patients suffering from this condition, with the harmful effect of ingestion and acid reflux on the damaged esophageal mucosa remaining an unresolved problem. Through the delivery of the hydrogel with stent platform, we aimed to evaluate the regenerative capacity of a tissue-specific decellularized extracellular matrix (dECM) hydrogel on damaged tissues. For this, an esophagus-derived dECM (EdECM) was developed and shown to have superior biofunctionality and rheological properties, as well as physical stability, potentially providing a better microenvironment for tissue development. An EdECM hydrogel-loaded stent was sequentially fabricated using a rotating rod combined 3D printing system that showed structural stability and protected a loaded hydrogel during delivery. Finally, following stent implantation, the therapeutic effect of EdECM was examined in a radiation esophagitis rat model. Our findings demonstrate that EdECM hydrogel delivery via a stent platform can rapidly resolve an inflammatory response, thus promoting a pro-regenerative microenvironment. The results suggest a promising therapeutic strategy for the treatment of radiation esophagitis.
Collapse
Affiliation(s)
- Dong-Heon Ha
- Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang, South Korea; EDmicBio, Inc., South Korea
| | - Suhun Chae
- Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang, South Korea
| | - Jae Yeon Lee
- Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang, South Korea; Department of Companion Animal Health, Daegu Haany University, Gyeongsan, South Korea
| | - Jae Yun Kim
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang, South Korea
| | - Jungbin Yoon
- Center for Rapid Prototyping based 3D Tissue/Organ Printing, Pohang University of Science and Technology, Pohang, South Korea
| | - Tugce Sen
- Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang, South Korea
| | - Sung-Woo Lee
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, South Korea; Combinatorial Tumor Immunotherapy Medical Research Center, Chonnam National University Medical School, Hwasun, South Korea
| | - Hak Jae Kim
- Department of Radiation Oncology, Seoul National University College of Medicine, Seoul, South Korea
| | - Jae Ho Cho
- Combinatorial Tumor Immunotherapy Medical Research Center, Chonnam National University Medical School, Hwasun, South Korea; CNU Biomed Center, Chonnam National University Hwasun Hospital, Hwason, South Korea
| | - Dong-Woo Cho
- Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang, South Korea; School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang, South Korea; Center for Rapid Prototyping based 3D Tissue/Organ Printing, Pohang University of Science and Technology, Pohang, South Korea; Postech-Catholic Biomedical Engineering Institute, Pohang University of Science and Technology, Pohang, South Korea; Institute of Convergence Science, Yonsei University, Seoul, South Korea.
| |
Collapse
|