51
|
Shaha S, Rodrigues D, Mitragotri S. Locoregional drug delivery for cancer therapy: Preclinical progress and clinical translation. J Control Release 2024; 367:737-767. [PMID: 38325716 DOI: 10.1016/j.jconrel.2024.01.072] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/26/2024] [Accepted: 01/31/2024] [Indexed: 02/09/2024]
Abstract
Systemic drug delivery is the current clinically preferred route for cancer therapy. However, challenges associated with tumor localization and off-tumor toxic effects limit the clinical effectiveness of this route. Locoregional drug delivery is an emerging viable alternative to systemic therapies. With the improvement in real-time imaging technologies and tools for direct access to tumor lesions, the clinical applicability of locoregional drug delivery is becoming more prominent. Theoretically, locoregional treatments can bypass challenges faced by systemic drug delivery. Preclinically, locoregional delivery of drugs has demonstrated enhanced therapeutic efficacy with limited off-target effects while still yielding an abscopal effect. Clinically, an array of locoregional strategies is under investigation for the delivery of drugs ranging in target and size. Locoregional tumor treatment strategies can be classified into two main categories: 1) direct drug infusion via injection or implanted port and 2) extended drug elution via injected or implanted depot. The number of studies investigating locoregional drug delivery strategies for cancer treatment is rising exponentially, in both preclinical and clinical settings, with some approaches approved for clinical use. Here, we highlight key preclinical advances and the clinical relevance of such locoregional delivery strategies in the treatment of cancer. Furthermore, we critically analyze 949 clinical trials involving locoregional drug delivery and discuss emerging trends.
Collapse
Affiliation(s)
- Suyog Shaha
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Allston, MA 02134, USA; Wyss Institute for Biologically Inspired Engineering, Boston, MA 02115, USA
| | - Danika Rodrigues
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Allston, MA 02134, USA; Wyss Institute for Biologically Inspired Engineering, Boston, MA 02115, USA
| | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Allston, MA 02134, USA; Wyss Institute for Biologically Inspired Engineering, Boston, MA 02115, USA.
| |
Collapse
|
52
|
Bao H, Zhang Y, Xin H, Gao Y, Hou Y, Yue G, Wang N, Wang Y, Li C, Liu F, Zhao Y, Kong L. The Construction of Three-Layered Biomimetic Arterial Graft Balances Biomechanics and Biocompatibility for Dynamic Biological Reconstruction. ACS OMEGA 2024; 9:7609-7620. [PMID: 38405546 PMCID: PMC10882685 DOI: 10.1021/acsomega.3c06628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 12/24/2023] [Accepted: 01/15/2024] [Indexed: 02/27/2024]
Abstract
The process of reconstructing an arterial graft is a complex and dynamic process that is subject to the influence of various mechanical factors, including tissue regeneration and blood pressure. The attainment of favorable remodeling outcomes is contingent upon the biocompatibility and biomechanical properties of the arterial graft. A promising strategy involves the emulation of the three-layer structure of the native artery, wherein the inner layer is composed of polycaprolactone (PCL) fibers aligned with blood flow, exhibiting excellent biocompatibility that fosters endothelial cell growth and effectively prevents platelet adhesion. The middle layer, consisting of PCL and polyurethane (PU), offers mechanical support and stability by forming a contractile smooth muscle ring and antiexpansion PU network. The outer layer, composed of PCL fibers with an irregular arrangement, promotes the growth of nerves and pericytes for long-term vascular function. Prioritizing the reconstruction of the inner and outer layers establishes a stable environment for intermediate smooth muscle growth. Our three-layer arterial graft is designed to provide the blood vessel with mechanical support and stability through nondegradable PU, while the incorporation of degradable PCL generates potential spaces for tissue ingrowth, thereby transforming our graft into a living implant.
Collapse
Affiliation(s)
- Han Bao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, China
| | - Yanyuan Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, China
| | - He Xin
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, China
| | - Ye Gao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, China
| | - Yan Hou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, China
| | - Guichu Yue
- Key Laboratory of Bio-inspired Smart Interfacial Science and Technology of Ministry of Education, School of Chemistry, Beihang University, Beijing 100191, China
| | - Nü Wang
- Key Laboratory of Bio-inspired Smart Interfacial Science and Technology of Ministry of Education, School of Chemistry, Beihang University, Beijing 100191, China
| | - Yaqiong Wang
- Key Laboratory of Bio-inspired Smart Interfacial Science and Technology of Ministry of Education, School of Chemistry, Beihang University, Beijing 100191, China
| | - Chun Li
- Shandong Nafeibo Technology Development Co., Ltd, Yantai 264000, China
| | - Fuwei Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, China
| | - Yong Zhao
- Key Laboratory of Bio-inspired Smart Interfacial Science and Technology of Ministry of Education, School of Chemistry, Beihang University, Beijing 100191, China
| | - Liang Kong
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, China
| |
Collapse
|
53
|
Wang J, Zhang X, Chen H, Ren H, Zhou M, Zhao Y. Engineered stem cells by emerging biomedical stratagems. Sci Bull (Beijing) 2024; 69:248-279. [PMID: 38101962 DOI: 10.1016/j.scib.2023.12.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/24/2023] [Accepted: 11/09/2023] [Indexed: 12/17/2023]
Abstract
Stem cell therapy holds immense potential as a viable treatment for a widespread range of intractable disorders. As the safety of stem cell transplantation having been demonstrated in numerous clinical trials, various kinds of stem cells are currently utilized in medical applications. Despite the achievements, the therapeutic benefits of stem cells for diseases are limited, and the data of clinical researches are unstable. To optimize tthe effectiveness of stem cells, engineering approaches have been developed to enhance their inherent abilities and impart them with new functionalities, paving the way for the next generation of stem cell therapies. This review offers a detailed analysis of engineered stem cells, including their clinical applications and potential for future development. We begin by briefly introducing the recent advances in the production of stem cells (induced pluripotent stem cells (iPSCs), embryonic stem cells (ESCs), mesenchymal stem cells (MSCs) and hematopoietic stem cells (HSCs)). Furthermore, we present the latest developments of engineered strategies in stem cells, including engineered methods in molecular biology and biomaterial fields, and their application in biomedical research. Finally, we summarize the current obstacles and suggest future prospects for engineered stem cells in clinical translations and biomedical applications.
Collapse
Affiliation(s)
- Jinglin Wang
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China; Division of Hepatobiliary Surgery and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Xiaoxuan Zhang
- Division of Hepatobiliary Surgery and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Hanxu Chen
- Division of Hepatobiliary Surgery and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Haozhen Ren
- Division of Hepatobiliary Surgery and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
| | - Min Zhou
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China.
| | - Yuanjin Zhao
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China; Division of Hepatobiliary Surgery and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China; Shenzhen Research Institute, Southeast University, Shenzhen 518038, China.
| |
Collapse
|
54
|
Wang Y, Wang K, Wang X, Luo Y, Chen H. Hydrogel-Composited Laminate for Islet Immune-Isolation to Treat Type 1 Diabetes. ACS APPLIED MATERIALS & INTERFACES 2024; 16:3042-3055. [PMID: 38215348 DOI: 10.1021/acsami.3c12359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2024]
Abstract
Challenges remain to be solved for the clinical translation of β-cell encapsulation technology in the treatment of type 1 diabetes (T1D). Successful delivery of β cells urgently needs the development of an encapsulation device with a thin dimension and rapid mass transport that offers stable immune isolation and complete retrieval. In this study, we focus on a laminate in which an islet-embedding alginate hydrogel layer (Alg) is sandwiched between two polymer layers (polyether sulfone, PES). Mechanical support by the PES layer protects the alginate from disintegrating after implantation and allows complete retrieval. The multilayered device has a thin membrane configuration (∼1 mm), and the edge of the laminate and the gaps between Alg and PES offer a semiopen structure that could be more permeable to molecules compared with the closed pocket of conventional macroencapsulation. Islets are suspended in the alginate solution and then encapsulated in the hydrogel layer in the middle of the laminate after gelation. Encapsulating syngeneic or xenogeneic islets in the laminate device corrected chemically induced T1D in mice for over 90 days in both the intraperitoneal space and the epididymal fat pad. The multilayered membrane system may therefore provide a translatable solution in β cell-transplantation therapy in T1D.
Collapse
Affiliation(s)
- Yi Wang
- Department of Biomedical Engineering, College of Future Technology, Peking University, Haidian District, Beijing 100871, China
| | - Kai Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Xi Wang
- State key Laboratory of Female Fertility Promotion, Peking University Third Hospital, Beijing 100191, China
| | - Ying Luo
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Haifeng Chen
- Department of Biomedical Engineering, College of Future Technology, Peking University, Haidian District, Beijing 100871, China
| |
Collapse
|
55
|
Liu Z, Zhou X, Chen Y, Ni Y, Zhu Z, Cao W, Chen K, Yan Y, Ji J, Zhang P. Fibrous capsule-resistant, controllably degradable and functionalizable zwitterion-albumin hybrid hydrogels. Biomater Sci 2024; 12:468-478. [PMID: 38086632 DOI: 10.1039/d3bm01783d] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2024]
Abstract
Foreign body response (FBR) represents an immune-mediated cascade reaction capable of inducing the rejection of foreign implants, thereby compromising their in vivo performance. Pure zwitterionic hydrogels have demonstrated the ability to resist long-term FBR, owing to their outstanding antifouling capabilities. However, achieving such a robust anti-FBR effect necessitates stringent requirements concerning the purity of zwitterionic materials, which constrains their broader functional applications. Herein, we present a biocompatible, controllably degradable, and functionalizable zwitterion-albumin hybrid hydrogel. The zwitterionic hydrogel crosslinked with serum albumin exhibits controllable degradation and excels in preventing the adsorption of various proteins and adhesion of cells and bacteria. Moreover, the hydrogel significantly alleviates the host's FBR compared with PEG hydrogels and particularly outperforms PEG-based cross-linker crosslinked zwitterionic hydrogels in reducing collagen encapsulation when subcutaneously implanted into mice. The zwitterion-albumin hybrid hydrogel shows potential as a functionalizable anti-FBR material in the context of implantable materials and biomedical devices.
Collapse
Affiliation(s)
- Zuolong Liu
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, P. R. China.
| | - Xianchi Zhou
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, P. R. China.
| | - Yongcheng Chen
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, P. R. China.
| | - Yanwen Ni
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, P. R. China.
| | - Zihao Zhu
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, P. R. China.
| | - Wenzhong Cao
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, P. R. China.
| | - Kexin Chen
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, P. R. China.
| | - Yu Yan
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, P. R. China.
| | - Jian Ji
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, P. R. China.
- International Research Center for X Polymers, International Campus, Zhejiang University, Haining, P. R. China
| | - Peng Zhang
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, P. R. China.
- International Research Center for X Polymers, International Campus, Zhejiang University, Haining, P. R. China
| |
Collapse
|
56
|
Mittal RK, Mishra R, Uddin R, Sharma V. Hydrogel Breakthroughs in Biomedicine: Recent Advances and Implications. Curr Pharm Biotechnol 2024; 25:1436-1451. [PMID: 38288792 DOI: 10.2174/0113892010281021231229100228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/20/2023] [Accepted: 11/28/2023] [Indexed: 07/23/2024]
Abstract
OBJECTIVE The objective of this review is to present a succinct summary of the latest advancements in the utilization of hydrogels for diverse biomedical applications, with a particular focus on their revolutionary impact in augmenting the delivery of drugs, tissue engineering, along with diagnostic methodologies. METHODS Using a meticulous examination of current literary works, this review systematically scrutinizes the nascent patterns in applying hydrogels for biomedical progress, condensing crucial discoveries to offer a comprehensive outlook on their ever-changing importance. RESULTS The analysis presents compelling evidence regarding the growing importance of hydrogels in biomedicine. It highlights their potential to significantly enhance drug delivery accuracy, redefine tissue engineering strategies, and advance diagnostic techniques. This substantiates their position as a fundamental element in the progress of modern medicine. CONCLUSION In summary, the constantly evolving advancement of hydrogel applications in biomedicine calls for ongoing investigation and resources, given their diverse contributions that can revolutionize therapeutic approaches and diagnostic methods, thereby paving the way for improved patient well-being.
Collapse
Affiliation(s)
- Ravi K Mittal
- Galgotias College of Pharmacy, Greater Noida, 201310, Uttar Pradesh, India
| | - Raghav Mishra
- Lloyd School of Pharmacy, Knowledge Park II, Greater Noida-201306, Uttar Pradesh, India
- GLA University, Mathura-281406, Uttar Pradesh, India
| | - Rehan Uddin
- Sir Madanlal Institute of Pharmacy, Etawah-206001 Uttar Pradesh, India
| | - Vikram Sharma
- Galgotias College of Pharmacy, Greater Noida, 201310, Uttar Pradesh, India
| |
Collapse
|
57
|
Sanjanwala D, Londhe V, Trivedi R, Bonde S, Sawarkar S, Kale V, Patravale V. Polysaccharide-based hydrogels for medical devices, implants and tissue engineering: A review. Int J Biol Macromol 2024; 256:128488. [PMID: 38043653 DOI: 10.1016/j.ijbiomac.2023.128488] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 11/10/2023] [Accepted: 11/27/2023] [Indexed: 12/05/2023]
Abstract
Hydrogels are highly biocompatible biomaterials composed of crosslinked three-dimensional networks of hydrophilic polymers. Owing to their natural origin, polysaccharide-based hydrogels (PBHs) possess low toxicity, high biocompatibility and demonstrate in vivo biodegradability, making them great candidates for use in various biomedical devices, implants, and tissue engineering. In addition, many polysaccharides also show additional biological activities such as antimicrobial, anticoagulant, antioxidant, immunomodulatory, hemostatic, and anti-inflammatory, which can provide additional therapeutic benefits. The porous nature of PBHs allows for the immobilization of antibodies, aptamers, enzymes and other molecules on their surface, or within their matrix, potentiating their use in biosensor devices. Specific polysaccharides can be used to produce transparent hydrogels, which have been used widely to fabricate ocular implants. The ability of PBHs to encapsulate drugs and other actives has been utilized for making neural implants and coatings for cardiovascular devices (stents, pacemakers and venous catheters) and urinary catheters. Their high water-absorption capacity has been exploited to make superabsorbent diapers and sanitary napkins. The barrier property and mechanical strength of PBHs has been used to develop gels and films as anti-adhesive formulations for the prevention of post-operative adhesion. Finally, by virtue of their ability to mimic various body tissues, they have been explored as scaffolds and bio-inks for tissue engineering of a wide variety of organs. These applications have been described in detail, in this review.
Collapse
Affiliation(s)
- Dhruv Sanjanwala
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Nathalal Parekh Marg, Matunga (E), Mumbai 400019, Maharashtra, India; Department of Pharmaceutical Sciences, College of Pharmacy, 428 Church Street, University of Michigan, Ann Arbor, MI 48109, United States.
| | - Vaishali Londhe
- SVKM's NMIMS, Shobhaben Pratapbhai College of Pharmacy and Technology Management, V.L. Mehta Road, Vile Parle (W), Mumbai 400056, Maharashtra, India
| | - Rashmi Trivedi
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur 441002, Maharashtra, India
| | - Smita Bonde
- SVKM's NMIMS, School of Pharmacy and Technology Management, Shirpur Campus, Maharashtra, India
| | - Sujata Sawarkar
- Department of Pharmaceutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, University of Mumbai, Mumbai 400056, Maharashtra, India
| | - Vinita Kale
- Department of Pharmaceutics, Gurunanak College of Pharmacy, Kamptee Road, Nagpur 440026, Maharashtra, India
| | - Vandana Patravale
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Nathalal Parekh Marg, Matunga (E), Mumbai 400019, Maharashtra, India.
| |
Collapse
|
58
|
Li H, He W, Feng Q, Chen J, Xu X, Lv C, Zhu C, Dong H. Engineering superstable islets-laden chitosan microgels with carboxymethyl cellulose coating for long-term blood glucose regulation in vivo. Carbohydr Polym 2024; 323:121425. [PMID: 37940297 DOI: 10.1016/j.carbpol.2023.121425] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/07/2023] [Accepted: 09/19/2023] [Indexed: 11/10/2023]
Abstract
Islet transplantation to restore endogenous insulin secretion is a promising therapy for type 1 diabetes in clinic. However, host immune rejection seriously limits the survival of transplanted islets. Despite of the various encapsulation strategies and materials developed so far to provide immune isolation for transplanted islets, long-term blood glucose regulation is still difficult due to the inherent defects of the encapsulation materials. Herein, a novel islet-encapsulation composite material with low immunogenicity, good biocompatibility and excellent stability is reported. Specifically, chitosan (CS) microgels (diameter: ∼302 μm) are prepared via Michael addition reaction between maleimide grafted chitosan (CS-Mal) and thiol grafted chitosan (CS-NAC) in droplet-based microfluidic device, and then zwitterionic surface layer is constructed on CS microgel surface by covalent binding between maleimide groups on CS and thiol groups on thiol modified carboxymethyl cellulose (CMC-SH). The as-formed carboxymethyl cellulose coated chitosan (CS@CMC) microgels show not only long-term stability in vivo owing to the non-biodegradability of CMC, but also fantastic anti-adsorption and antifibrosis because of the stable zwitterionic surface layer. As a result, islets encapsulated in the CS@CMC microgels exhibit high viability and good insulin secretion function in vivo, and long-term blood glucose regulation is achieved for 180 days in diabetic mice post-transplantation.
Collapse
Affiliation(s)
- Haofei Li
- Department of Biomaterials, School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, China; National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), Guangzhou 510006, China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, China
| | - Weijun He
- Department of Biomaterials, School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, China; National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), Guangzhou 510006, China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, China
| | - Qi Feng
- Department of Biomaterials, School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, China; National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), Guangzhou 510006, China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, China
| | - Junlin Chen
- Department of Biomaterials, School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, China; National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), Guangzhou 510006, China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, China
| | - Xinbin Xu
- Department of Biomaterials, School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, China; National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), Guangzhou 510006, China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, China
| | - Chuhan Lv
- Department of Biomaterials, School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, China; National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), Guangzhou 510006, China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, China
| | - Changchun Zhu
- Department of Biomaterials, School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, China; National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), Guangzhou 510006, China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, China
| | - Hua Dong
- Department of Biomaterials, School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, China; National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), Guangzhou 510006, China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, China; Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510641, China.
| |
Collapse
|
59
|
Zhang Z, He C, Chen X. Designing Hydrogels for Immunomodulation in Cancer Therapy and Regenerative Medicine. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2308894. [PMID: 37909463 DOI: 10.1002/adma.202308894] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/26/2023] [Indexed: 11/03/2023]
Abstract
The immune system not only acts as a defense against pathogen and cancer cells, but also plays an important role in homeostasis and tissue regeneration. Targeting immune systems is a promising strategy for efficient cancer treatment and regenerative medicine. Current systemic immunomodulation therapies are usually associated with low persistence time, poor targeting to action sites, and severe side effects. Due to their extracellular matrix-mimetic nature, tunable properties and diverse bioactivities, hydrogels are intriguing platforms to locally deliver immunomodulatory agents and cells, as well as provide an immunomodulatory microenvironment to recruit, activate, and expand host immune cells. In this review, the design considerations, including polymer backbones, crosslinking mechanisms, physicochemical nature, and immunomodulation-related components, of the hydrogel platforms, are focused on. The immunomodulatory effects and therapeutic outcomes in cancer therapy and tissue regeneration of different hydrogel systems are emphasized, including hydrogel depots for delivery of immunomodulatory agents, hydrogel scaffolds for cell delivery, and immunomodulatory hydrogels depending on the intrinsic properties of materials. Finally, the remained challenges in current systems and future development of immunomodulatory hydrogels are discussed.
Collapse
Affiliation(s)
- Zhen Zhang
- CAS Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
| | - Chaoliang He
- CAS Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Xuesi Chen
- CAS Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, China
| |
Collapse
|
60
|
Dooley M, Luckett J, Alexander MR, Matousek P, Dehghani H, Ghaemmaghami AM, Notingher I. Optimization of diffuse Raman spectroscopy for in-vivo quantification of foreign body response in a small animal model. BIOMEDICAL OPTICS EXPRESS 2023; 14:6592-6606. [PMID: 38420302 PMCID: PMC10898571 DOI: 10.1364/boe.512118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 11/13/2023] [Indexed: 03/02/2024]
Abstract
Diffuse Raman spectroscopy (DRS) allows subsurface molecular analysis of optically turbid samples. Numerical modeling of light propagation was used as a method for improving the design of an DRS instrument to maximize the signal to noise ratio (SNR) while ensuring safe laser exposure parameters required for in-vivo measurements. Experimental validation of the model was performed on both phantom samples and disks implanted postmortem to mimic the typical response to foreign bodies (formation of a fibrotic capsule around an implant). A reduction of laser exposure of over 1500-fold was achieved over previous studies whilst maintaining the same Raman collection rates and reaching the safe power density of 3 mW/mm2. The validation of this approach in a subcutaneous implant in a mouse cadaver showed a further improvement of 1.5-fold SNR, with a thickness limit of detection for the fibrotic layer of 23 µm, under the same acquisition times. In the animal body, a thickness limit of detection of 16 µm was achieved. These results demonstrate the feasibility of numerical model-based optimization for DRS, and that the technique can be improved sufficiently to be used for in-vivo measurement of collagenous capsule formation as a result of the foreign body response in murine models.
Collapse
Affiliation(s)
- Max Dooley
- School of Physics and Astronomy, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Jeni Luckett
- School of Life Sciences, University of Nottingham, University Park, Nottingham NG7 2RD, UK
| | - Morgan R. Alexander
- School of Pharmacy, University of Nottingham, University Park, Nottingham NG7 2RD, UK
| | - Pavel Matousek
- STFC Rutherford Appleton Laboratory, Central Laser Facility, Research Complex at Harwell, UK Research and Innovation (UKRI), Harwell Oxford OX11 0QX, UK
| | - Hamid Dehghani
- School of Computer Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Amir M. Ghaemmaghami
- School of Life Sciences, University of Nottingham, University Park, Nottingham NG7 2RD, UK
| | - Ioan Notingher
- School of Physics and Astronomy, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| |
Collapse
|
61
|
Wright MA, Miller AJ, Dong X, Karinja SJ, Samadi A, Lara DO, Mukherjee S, Veiseh O, Spector JA. Reducing Peri-implant Capsule Thickness in Submuscular Rodent Model of Breast Reconstruction With Delayed Radiotherapy. J Surg Res 2023; 291:158-166. [PMID: 37421826 DOI: 10.1016/j.jss.2023.04.015] [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: 08/24/2022] [Revised: 04/05/2023] [Accepted: 04/29/2023] [Indexed: 07/10/2023]
Abstract
INTRODUCTION Capsular contracture remains the most common complication following device-based breast reconstruction, occurring in up to 50% of women who also undergo adjuvant radiotherapy either before or after device-based reconstruction. While certain risk factors for capsular contracture have been identified, there remains no clinically effective method of prevention. The purpose of the present study is to determine the effect of coating the implant with the novel small molecule Met-Z2-Y12, with and without delayed, targeted radiotherapy, on capsule thickness and morphologic change around smooth silicone implants placed under the latissimus dorsi in a rodent model. METHODS Twenty-four female Sprague Dawley rats each had 2 mL smooth round silicone breast implants implanted bilaterally under the latissimus dorsi muscle. Twelve received uncoated implants and twelve received implants coated with Met-Z2-Y12. Half of the animals from each group received targeted radiotherapy (20 Gray) on postoperative day ten. At three and 6 months after implantation, the tissue surrounding the implants was harvested for analysis of capsular histology including capsule thickness. Additionally, microCT scans were qualitatively analyzed for morphologic change. RESULTS Capsules surrounding Met-Z2-Y12-coated implants were significantly thinner (P = 0.006). The greatest difference in capsule thickness was seen in the irradiated 6-month groups, where mean capsule thickness was 79.1 ± 27.3 μm for uncoated versus 50.9 ± 9.6 μm for Met-Z2-Y12-coated implants (P = 0.038). At the time of explant, there were no capsular morphologic differences between the groups either grossly or per microCT. CONCLUSIONS Met-Z2-Y12 coating of smooth silicone breast implants significantly reduces capsule thickness in a rodent model of submuscular breast reconstruction with delayed radiotherapy.
Collapse
Affiliation(s)
- Matthew A Wright
- Laboratory of Bioregenerative Medicine and Surgery, Division of Plastic Surgery, Department of Surgery, Weill Cornell Medicine, New York, New York
| | - Andrew J Miller
- Laboratory of Bioregenerative Medicine and Surgery, Division of Plastic Surgery, Department of Surgery, Weill Cornell Medicine, New York, New York
| | - Xue Dong
- Laboratory of Bioregenerative Medicine and Surgery, Division of Plastic Surgery, Department of Surgery, Weill Cornell Medicine, New York, New York
| | - Sarah J Karinja
- Laboratory of Bioregenerative Medicine and Surgery, Division of Plastic Surgery, Department of Surgery, Weill Cornell Medicine, New York, New York
| | - Arash Samadi
- Laboratory of Bioregenerative Medicine and Surgery, Division of Plastic Surgery, Department of Surgery, Weill Cornell Medicine, New York, New York
| | - Daniel O Lara
- Laboratory of Bioregenerative Medicine and Surgery, Division of Plastic Surgery, Department of Surgery, Weill Cornell Medicine, New York, New York
| | - Sudip Mukherjee
- Veiseh Lab, Department of Bioengineering, Rice University, Houston, Texas
| | - Omid Veiseh
- Veiseh Lab, Department of Bioengineering, Rice University, Houston, Texas
| | - Jason A Spector
- Laboratory of Bioregenerative Medicine and Surgery, Division of Plastic Surgery, Department of Surgery, Weill Cornell Medicine, New York, New York; Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York.
| |
Collapse
|
62
|
Padmanabhan J, Chen K, Sivaraj D, Henn D, Kuehlmann BA, Kussie HC, Zhao ET, Kahn A, Bonham CA, Dohi T, Beck TC, Trotsyuk AA, Stern-Buchbinder ZA, Than PA, Hosseini HS, Barrera JA, Magbual NJ, Leeolou MC, Fischer KS, Tigchelaar SS, Lin JQ, Perrault DP, Borrelli MR, Kwon SH, Maan ZN, Dunn JCY, Nazerali R, Januszyk M, Prantl L, Gurtner GC. Allometrically scaling tissue forces drive pathological foreign-body responses to implants via Rac2-activated myeloid cells. Nat Biomed Eng 2023; 7:1419-1436. [PMID: 37749310 PMCID: PMC10651488 DOI: 10.1038/s41551-023-01091-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 08/02/2023] [Indexed: 09/27/2023]
Abstract
Small animals do not replicate the severity of the human foreign-body response (FBR) to implants. Here we show that the FBR can be driven by forces generated at the implant surface that, owing to allometric scaling, increase exponentially with body size. We found that the human FBR is mediated by immune-cell-specific RAC2 mechanotransduction signalling, independently of the chemistry and mechanical properties of the implant, and that a pathological FBR that is human-like at the molecular, cellular and tissue levels can be induced in mice via the application of human-tissue-scale forces through a vibrating silicone implant. FBRs to such elevated extrinsic forces in the mice were also mediated by the activation of Rac2 signalling in a subpopulation of mechanoresponsive myeloid cells, which could be substantially reduced via the pharmacological or genetic inhibition of Rac2. Our findings provide an explanation for the stark differences in FBRs observed in small animals and humans, and have implications for the design and safety of implantable devices.
Collapse
Affiliation(s)
- Jagannath Padmanabhan
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Kellen Chen
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Surgery, University of Arizona College of Medicine, Tucson, AZ, USA.
| | - Dharshan Sivaraj
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Surgery, University of Arizona College of Medicine, Tucson, AZ, USA.
| | - Dominic Henn
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Department of Plastic Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Britta A Kuehlmann
- Department of Plastic and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Hudson C Kussie
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Department of Surgery, University of Arizona College of Medicine, Tucson, AZ, USA
| | - Eric T Zhao
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Anum Kahn
- Cell Sciences Imaging Facility (CSIF), Beckman Center, Stanford University, Stanford, CA, USA
| | - Clark A Bonham
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Teruyuki Dohi
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Thomas C Beck
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Artem A Trotsyuk
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Department of Surgery, University of Arizona College of Medicine, Tucson, AZ, USA
| | - Zachary A Stern-Buchbinder
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Peter A Than
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Hadi S Hosseini
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Janos A Barrera
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Noah J Magbual
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Melissa C Leeolou
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Katharina S Fischer
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Seth S Tigchelaar
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - John Q Lin
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - David P Perrault
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Mimi R Borrelli
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Sun Hyung Kwon
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Zeshaan N Maan
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - James C Y Dunn
- Division of Pediatric Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Rahim Nazerali
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael Januszyk
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Lukas Prantl
- Department of Plastic and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Geoffrey C Gurtner
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Surgery, University of Arizona College of Medicine, Tucson, AZ, USA.
| |
Collapse
|
63
|
Xu J, Zhu X, Zhao J, Ling G, Zhang P. Biomedical applications of supramolecular hydrogels with enhanced mechanical properties. Adv Colloid Interface Sci 2023; 321:103000. [PMID: 37839280 DOI: 10.1016/j.cis.2023.103000] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 09/02/2023] [Accepted: 09/16/2023] [Indexed: 10/17/2023]
Abstract
Supramolecular hydrogels bound by hydrogen bonding, host-guest, hydrophobic, and other non-covalent interactions are among the most attractive biomaterials available. Supramolecular hydrogels have attracted extensive attention due to their inherent dynamic reversibility, self-healing, stimuli-response, excellent biocompatibility, and near-physiological environment. However, the inherent contradiction between non-covalent interactions and mechanical strength makes the practical application of supramolecular hydrogels a great challenge. This review describes the mechanical strength of hydrogels mediated by supramolecular interactions, and focuses on the potential strategies for enhancing the mechanical strength of supramolecular hydrogels and illustrates their applications in related fields, such as flexible electronic sensors, wound dressings, and three-dimensional (3D) scaffolds. Finally, the current problems and future research prospects of supramolecular hydrogels are discussed. This review is expected to provide insights that will motivate more advanced research on supramolecular hydrogels.
Collapse
Affiliation(s)
- Jiaqi Xu
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Xiaoguang Zhu
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Jiuhong Zhao
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Guixia Ling
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China..
| | - Peng Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China..
| |
Collapse
|
64
|
Campbell S, Preciado Rivera N, Said S, Lam A, Weir L, Gour J, Smeets NMB, Hoare T. Injectable On-Demand Pulsatile Drug Delivery Hydrogels Using Alternating Magnetic Field-Triggered Polymer Glass Transitions. ACS APPLIED MATERIALS & INTERFACES 2023; 15:48892-48902. [PMID: 37816152 DOI: 10.1021/acsami.3c09299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2023]
Abstract
Remote-controlled pulsatile or staged release has significant potential in a wide range of therapeutic treatments. However, most current approaches are hindered by the low resolution between the on- and off-states of drug release and the need for surgical implantation of larger controlled-release devices. Herein, we describe a method that addresses these limitations by combining injectable hydrogels, superparamagnetic iron oxide nanoparticles (SPIONs) that heat when exposed to an alternating magnetic field (AMF), and polymeric nanoparticles with a glass transition temperature (Tg) just above physiological temperature. Miniemulsion polymerization was used to fabricate poly(methyl methacrylate-co-butyl methacrylate) (p(MMA-co-BMA)) nanoparticles loaded with a model hydrophobic drug and tuned to have a Tg value just above physiological temperature (∼43 °C). Co-encapsulation of these drug-loaded nanoparticles with SPIONs inside a carbohydrate-based injectable hydrogel matrix (formed by rapid hydrazone cross-linking chemistry) enables injection and immobilization of the nanoparticles at the target site. Temperature cycling facilitated a 2.5:1 to 6:1 on/off rhodamine release ratio when the nanocomposites were switched between 37 and 45 °C; release was similarly enhanced by exposing the nanocomposite hydrogel to an AMF to drive heating, with enhanced release upon pulsing observed even 1 week after injection. Coupled with the apparent cytocompatibility of all of the nanocomposite components, these injectable nanocomposite hydrogels are promising as minimally invasive but remotely actuated release delivery vehicles capable of complex release kinetics with high on-off resolution.
Collapse
Affiliation(s)
- Scott Campbell
- Department of Chemical Engineering, McMaster University, 1280 Main St. W., Hamilton L8S 4L7, Ontario, Canada
| | - Nahieli Preciado Rivera
- Department of Chemical Engineering, McMaster University, 1280 Main St. W., Hamilton L8S 4L7, Ontario, Canada
| | - Somiraa Said
- Department of Chemical Engineering, McMaster University, 1280 Main St. W., Hamilton L8S 4L7, Ontario, Canada
- Department of Pharmaceutics, Alexandria University, Alexandria 21521, Egypt
| | - Angus Lam
- Department of Chemical Engineering, McMaster University, 1280 Main St. W., Hamilton L8S 4L7, Ontario, Canada
| | - Lauren Weir
- Department of Chemical Engineering, McMaster University, 1280 Main St. W., Hamilton L8S 4L7, Ontario, Canada
| | - Jared Gour
- Department of Chemical Engineering, McMaster University, 1280 Main St. W., Hamilton L8S 4L7, Ontario, Canada
| | - Niels M B Smeets
- Department of Chemical Engineering, McMaster University, 1280 Main St. W., Hamilton L8S 4L7, Ontario, Canada
| | - Todd Hoare
- Department of Chemical Engineering, McMaster University, 1280 Main St. W., Hamilton L8S 4L7, Ontario, Canada
| |
Collapse
|
65
|
Wang K, Chen D, Wang Z, Yang J, Liu W. An Injectable and Antifouling Supramolecular Polymer Hydrogel with Microenvironment-Regulatory Function to Prevent Peritendinous Adhesion and Promote Tendon Repair. Macromol Biosci 2023; 23:e2300142. [PMID: 37317041 DOI: 10.1002/mabi.202300142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 06/12/2023] [Indexed: 06/16/2023]
Abstract
The imbalance of extrinsic and intrinsic healing of tendon is thought to be the main cause of peritendinous adhesions. In this work, an injectable supramolecular poly(N-(2-hydroxypropyl) acrylamide) (PHPAm) hydrogel is prepared merely via side chain hydrogen-bonding crosslinks. This PHPAm exhibits good antifouling and self-healing properties. The supramolecular hydrogel simultaneously loaded with Prussian blue (PB) nanoparticles and platelet lysate (PL) is explored as a functional physical barrier, which can significantly resist the adhesion of fibrin and fibroblasts, attenuate the local inflammatory response, and enhance the tenocytes activity, thus balancing extrinsic and intrinsic healing. The PHPAm hydrogel is shown to prevent peritendinous adhesions considerably by inhibiting NF-κB inflammatory pathway and TGF-β1/Smad3-mediated fibrosis pathway, thereby significantly improving tendon repair by releasing bioactive factors to regulate the tenocytes behavior. This work provides a new strategy for developing physical barriers to prevent peritendinous adhesions and promote tissue repair effectively.
Collapse
Affiliation(s)
- Kuan Wang
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin, 300350, China
| | - Danyang Chen
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin, 300350, China
| | - Zhuoya Wang
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin, 300350, China
| | - Jianhai Yang
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin, 300350, China
| | - Wenguang Liu
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin, 300350, China
| |
Collapse
|
66
|
Falcone N, Ermis M, Tamay DG, Mecwan M, Monirizad M, Mathes TG, Jucaud V, Choroomi A, de Barros NR, Zhu Y, Vrana NE, Kraatz HB, Kim HJ, Khademhosseini A. Peptide Hydrogels as Immunomaterials and Their Use in Cancer Immunotherapy Delivery. Adv Healthc Mater 2023; 12:e2301096. [PMID: 37256647 PMCID: PMC10615713 DOI: 10.1002/adhm.202301096] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/15/2023] [Indexed: 06/01/2023]
Abstract
Peptide-based hydrogel biomaterials have emerged as an excellent strategy for immune system modulation. Peptide-based hydrogels are supramolecular materials that self-assemble into various nanostructures through various interactive forces (i.e., hydrogen bonding and hydrophobic interactions) and respond to microenvironmental stimuli (i.e., pH, temperature). While they have been reported in numerous biomedical applications, they have recently been deemed promising candidates to improve the efficacy of cancer immunotherapies and treatments. Immunotherapies seek to harness the body's immune system to preemptively protect against and treat various diseases, such as cancer. However, their low efficacy rates result in limited patient responses to treatment. Here, the immunomaterial's potential to improve these efficacy rates by either functioning as immune stimulators through direct immune system interactions and/or delivering a range of immune agents is highlighted. The chemical and physical properties of these peptide-based materials that lead to immuno modulation and how one may design a system to achieve desired immune responses in a controllable manner are discussed. Works in the literature that reports peptide hydrogels as adjuvant systems and for the delivery of immunotherapies are highlighted. Finally, the future trends and possible developments based on peptide hydrogels for cancer immunotherapy applications are discussed.
Collapse
Affiliation(s)
- Natashya Falcone
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, CA, 90034, USA
| | - Menekse Ermis
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, CA, 90034, USA
- BIOMATEN, Center of Excellence in Biomaterials and Tissue Engineering, Middle East Technical University, Ankara, 06800, Turkey
| | - Dilara Goksu Tamay
- BIOMATEN, Center of Excellence in Biomaterials and Tissue Engineering, Middle East Technical University, Ankara, 06800, Turkey
- Department of Biotechnology, Middle East Technical University, Ankara, 06800, Turkey
| | - Marvin Mecwan
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, CA, 90034, USA
| | - Mahsa Monirizad
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, CA, 90034, USA
| | - Tess Grett Mathes
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, CA, 90034, USA
| | - Vadim Jucaud
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, CA, 90034, USA
| | - Auveen Choroomi
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, CA, 90034, USA
| | - Natan Roberto de Barros
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, CA, 90034, USA
| | - Yangzhi Zhu
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, CA, 90034, USA
| | - Nihal Engin Vrana
- SPARTHA Medical, CRBS 1 Rue Eugene Boeckel, Strasbourg, 67000, France
| | - Heinz-Bernhard Kraatz
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, M5S 3E5, Canada
- Department of Physical and Environmental Sciences, University of Toronto Scarborough, Toronto, ON, M1C 1A4, Canada
| | - Han-Jun Kim
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, CA, 90034, USA
- College of Pharmacy, Korea University, Sejong, 30019, Republic of Korea
| | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, CA, 90034, USA
| |
Collapse
|
67
|
Qiu D, Cao C, Prasopthum A, Sun Z, Zhang S, Yang H, Xu Z, Tao J, Ai F, Yang J. Elucidating osseointegration in vivo in 3D printed scaffolds eliciting different foreign body responses. Mater Today Bio 2023; 22:100771. [PMID: 37674779 PMCID: PMC10477687 DOI: 10.1016/j.mtbio.2023.100771] [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: 05/31/2023] [Revised: 08/10/2023] [Accepted: 08/12/2023] [Indexed: 09/08/2023] Open
Abstract
Osseointegration between biomaterial and bone is critical for the clinical success of many orthopaedic and dental implants. However, the mechanisms of in vivo interfacial bonding formation and the role of immune cells in this process remain unclear. In this study, we investigated the bone-scaffold material interfaces in two different 3D printed porous scaffolds (polymer/hydroxyapatite and sintered hydroxyapatite) that elicited different levels of foreign body response (FBR). The polymer/hydroxyapatite composite scaffolds elicited more intensive FBR, which was evidenced by more FBR components, such as macrophages/foreign body giant cells and fibrous tissue, surrounding the material surface. Sintered hydroxyapatite scaffolds showed less intensive FBR compared to the composite scaffolds. The interfacial bonding appeared to form via new bone first forming within the pores of the scaffolds followed by growing towards strut surfaces. In contrast, it was previously thought that bone regeneration starts at biomaterial surfaces via osteogenic stem/progenitor cells first attaching to them. The material-bone interface of the less immunogenic hydroxyapatite scaffolds was heterogenous across all samples, evidenced by the coexistence of osseointegration and FBR components. The presence of FBR components appeared to inhibit osseointegration. Where FBR components were present there was no osseointegration. Our results offer new insight on the in vivo formation of bone-material interface, which highlights the importance of minimizing FBR to facilitate osseointegration for the development of better orthopaedic and dental biomaterials.
Collapse
Affiliation(s)
- Dewei Qiu
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Nanchang University, Jiangxi, China
| | - Chuanliang Cao
- School of Advanced Manufacturing, Nanchang University, Jiangxi, China
| | | | - Zhenchang Sun
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Henan, China
| | - Shan Zhang
- School of Advanced Manufacturing, Nanchang University, Jiangxi, China
| | - Hanwen Yang
- School of Advanced Manufacturing, Nanchang University, Jiangxi, China
| | - Zhiyong Xu
- School of Advanced Manufacturing, Nanchang University, Jiangxi, China
| | - Jun Tao
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Nanchang University, Jiangxi, China
| | - Fanrong Ai
- School of Advanced Manufacturing, Nanchang University, Jiangxi, China
| | - Jing Yang
- School of Pharmacy, University of Nottingham, UK
- Biodiscovery Institute, University of Nottingham, UK
| |
Collapse
|
68
|
Gupta P, Alheib O, Shin JW. Towards single cell encapsulation for precision biology and medicine. Adv Drug Deliv Rev 2023; 201:115010. [PMID: 37454931 PMCID: PMC10798218 DOI: 10.1016/j.addr.2023.115010] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 07/11/2023] [Accepted: 07/13/2023] [Indexed: 07/18/2023]
Abstract
The primary impetus of therapeutic cell encapsulation in the past several decades has been to broaden the options for donor cell sources by countering against immune-mediated rejection. However, another significant advantage of encapsulation is to provide donor cells with physiologically relevant cues that become compromised in disease. The advances in biomaterial design have led to the fundamental insight that cells sense and respond to various signals encoded in materials, ranging from biochemical to mechanical cues. The biomaterial design for cell encapsulation is becoming more sophisticated in controlling specific aspects of cellular phenotypes and more precise down to the single cell level. This recent progress offers a paradigm shift by designing single cell-encapsulating materials with predefined cues to precisely control donor cells after transplantation.
Collapse
Affiliation(s)
- Prerak Gupta
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Omar Alheib
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães 4805-017, Portugal
| | - Jae-Won Shin
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60607, USA.
| |
Collapse
|
69
|
Karinja SJ, Bernstein JL, Mukherjee S, Jin J, Lin A, Abadeer A, Kaymakcalan O, Veiseh O, Spector JA. An Antifibrotic Breast Implant Surface Coating Significantly Reduces Periprosthetic Capsule Formation. Plast Reconstr Surg 2023; 152:775-785. [PMID: 36847657 DOI: 10.1097/prs.0000000000010323] [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: 03/01/2023]
Abstract
BACKGROUND The body responds to prosthetic materials with an inflammatory foreign body response and deposition of a fibrous capsule, which may be deleterious to the function of the device and cause significant discomfort for the patient. Capsular contracture (CC) is the most common complication of aesthetic and reconstructive breast surgery. The source of significant patient morbidity, it can result in pain, suboptimal aesthetic outcomes, implant failure, and increased costs. The underlying mechanism remains unknown. Treatment is limited to reoperation and capsule excision, but recurrence rates remain high. In this study, the authors altered the surface chemistry of silicone implants with a proprietary anti-inflammatory coating to reduce capsule formation. METHODS Silicone implants were coated with Met-Z2-Y12, a biocompatible, anti-inflammatory surface modification. Uncoated and Met-Z2-Y12-coated implants were implanted in C57BL/6 mice. After 21, 90, or 180 days, periprosthetic tissue was removed for histologic analysis. RESULTS The authors compared mean capsule thickness at three time points. At 21, 90, and 180 days, there was a statistically significant reduction in capsule thickness of Met-Z2-Y12-coated implants compared with uncoated implants ( P < 0.05). CONCLUSIONS Coating the surface of silicone implants with Met-Z2-Y12 significantly reduced acute and chronic capsule formation in a mouse model for implant-based breast augmentation and reconstruction. As capsule formation obligatorily precedes CC, these results suggest contracture itself may be significantly attenuated. Furthermore, as periprosthetic capsule formation is a complication without anatomical boundaries, this chemistry may have additional applications beyond breast implants, to a myriad of other implantable medical devices. CLINICAL RELEVANCE STATEMENT Coating of the silicone implant surface with Met-Z2-Y12 alters the periprosthetic capsule architecture and significantly reduces capsule thickness for at least 6 months postoperatively in a murine model. This is a promising step forward in the development of a therapy to prevent capsular contracture.
Collapse
Affiliation(s)
- Sarah J Karinja
- From the Laboratory of Bioregenerative Medicine & Surgery, Weill Cornell Medical College
- Department of Surgery, Division of Plastic Surgery
| | - Jaime L Bernstein
- From the Laboratory of Bioregenerative Medicine & Surgery, Weill Cornell Medical College
- Department of Surgery, Division of Plastic Surgery
| | | | - Julia Jin
- From the Laboratory of Bioregenerative Medicine & Surgery, Weill Cornell Medical College
- Department of Surgery, Division of Plastic Surgery
| | - Alexandra Lin
- From the Laboratory of Bioregenerative Medicine & Surgery, Weill Cornell Medical College
- Department of Surgery, Division of Plastic Surgery
| | - Andrew Abadeer
- From the Laboratory of Bioregenerative Medicine & Surgery, Weill Cornell Medical College
- Department of Surgery, Division of Plastic Surgery
| | - Omer Kaymakcalan
- From the Laboratory of Bioregenerative Medicine & Surgery, Weill Cornell Medical College
- Department of Surgery, Division of Plastic Surgery
| | - Omid Veiseh
- Department of Bioengineering, Rice University
- Sigilon Therapeutics
| | - Jason A Spector
- From the Laboratory of Bioregenerative Medicine & Surgery, Weill Cornell Medical College
- Department of Surgery, Division of Plastic Surgery
- Meinig School of Biomedical Engineering, Cornell University
| |
Collapse
|
70
|
Aghlara-Fotovat S, Musteata E, Doerfert MD, Baruch M, Levitan M, Tabor JJ, Veiseh O. Hydrogel-encapsulation to enhance bacterial diagnosis of colon inflammation. Biomaterials 2023; 301:122246. [PMID: 37481834 PMCID: PMC10792543 DOI: 10.1016/j.biomaterials.2023.122246] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 06/13/2023] [Accepted: 07/12/2023] [Indexed: 07/25/2023]
Abstract
Bacteria can be genetically programmed to sense and report the presence of disease biomarkers in the gastrointestinal (GI) tract. However, diagnostic bacteria are typically delivered via oral administration of liquid cultures, resulting in poor survival and high dispersal in vivo. These limitations confound recovery and analysis of engineered bacteria from GI or stool samples. Here, we demonstrate that encapsulating bacteria inside of alginate core-shell particles enables robust survival, containment, and diagnostic function in vivo. We demonstrate these benefits by encapsulating a strain engineered to report the presence of the biomarker thiosulfate via fluorescent protein expression in order to diagnose dextran sodium sulfate-induced colitis in rats. Hydrogel-encapsulated bacteria engineered to sense and respond to physiological stimuli should enable minimally invasive monitoring of a wide range of diseases and have applications as next-generation smart therapeutics.
Collapse
Affiliation(s)
| | - Elena Musteata
- Systems Synthetic and Physical Biology, Rice University, Houston, TX, USA
| | | | - Moshe Baruch
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Maya Levitan
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Jeffrey J Tabor
- Department of Bioengineering, Rice University, Houston, TX, USA; Systems Synthetic and Physical Biology, Rice University, Houston, TX, USA; Department of Biosciences, Rice University, Houston, TX, USA.
| | - Omid Veiseh
- Department of Bioengineering, Rice University, Houston, TX, USA.
| |
Collapse
|
71
|
Abbaszadeh S, Nosrati-Siahmazgi V, Musaie K, Rezaei S, Qahremani M, Xiao B, Santos HA, Shahbazi MA. Emerging strategies to bypass transplant rejection via biomaterial-assisted immunoengineering: Insights from islets and beyond. Adv Drug Deliv Rev 2023; 200:115050. [PMID: 37549847 DOI: 10.1016/j.addr.2023.115050] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/14/2023] [Accepted: 08/04/2023] [Indexed: 08/09/2023]
Abstract
Novel transplantation techniques are currently under development to preserve the function of impaired tissues or organs. While current technologies can enhance the survival of recipients, they have remained elusive to date due to graft rejection by undesired in vivo immune responses despite systemic prescription of immunosuppressants. The need for life-long immunomodulation and serious adverse effects of current medicines, the development of novel biomaterial-based immunoengineering strategies has attracted much attention lately. Immunomodulatory 3D platforms can alter immune responses locally and/or prevent transplant rejection through the protection of the graft from the attack of immune system. These new approaches aim to overcome the complexity of the long-term administration of systemic immunosuppressants, including the risks of infection, cancer incidence, and systemic toxicity. In addition, they can decrease the effective dose of the delivered drugs via direct delivery at the transplantation site. In this review, we comprehensively address the immune rejection mechanisms, followed by recent developments in biomaterial-based immunoengineering strategies to prolong transplant survival. We also compare the efficacy and safety of these new platforms with conventional agents. Finally, challenges and barriers for the clinical translation of the biomaterial-based immunoengineering transplants and prospects are discussed.
Collapse
Affiliation(s)
- Samin Abbaszadeh
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, Netherlands
| | - Vahideh Nosrati-Siahmazgi
- Department of Pharmaceutical Biomaterials, School of Pharmacy, Zanjan University of Medical Science, 45139-56184 Zanjan, Iran
| | - Kiyan Musaie
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, Netherlands
| | - Saman Rezaei
- Department of Pharmaceutical Biomaterials, School of Pharmacy, Zanjan University of Medical Science, 45139-56184 Zanjan, Iran
| | - Mostafa Qahremani
- Department of Pharmaceutical Biomaterials, School of Pharmacy, Zanjan University of Medical Science, 45139-56184 Zanjan, Iran
| | - Bo Xiao
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715 China.
| | - Hélder A Santos
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, Netherlands; Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland; W.J. Kolff Institute for Biomedical Engineering and Materials Science, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, the Netherlands.
| | - Mohammad-Ali Shahbazi
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, Netherlands; W.J. Kolff Institute for Biomedical Engineering and Materials Science, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, the Netherlands.
| |
Collapse
|
72
|
Cai J, Wang W, Cai P, Cao B. Immune response to foreign materials in spinal fusion surgery. Heliyon 2023; 9:e19950. [PMID: 37810067 PMCID: PMC10559558 DOI: 10.1016/j.heliyon.2023.e19950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 06/22/2023] [Accepted: 09/06/2023] [Indexed: 10/10/2023] Open
Abstract
Spinal fusion surgery is a common procedure used to stabilize the spine and treat back pain. The procedure involves the use of foreign materials such as screws, rods, or cages, which can trigger a foreign body reaction, an immune response that involves the activation of immune cells such as macrophages and lymphocytes. The foreign body reaction can impact the success of spinal fusion, as it can interfere with bone growth and fusion. This review article provides an overview of the cellular and molecular events in the foreign body reaction, the impact of the immune response on spinal fusion, and strategies to minimize its impact. By carefully considering the use of foreign materials and optimizing surgical techniques, the impact of the foreign body reaction can be reduced, leading to better outcomes for patients.
Collapse
Affiliation(s)
| | | | - Peng Cai
- Department of Orthopedics, Chengdu Seventh People's Hospital (Chengdu Tumor Hospital), 51 Zhimin Rd, Wuhou District, 610041, Chengdu, Sichuan, China
| | - Bo Cao
- Department of Orthopedics, Chengdu Seventh People's Hospital (Chengdu Tumor Hospital), 51 Zhimin Rd, Wuhou District, 610041, Chengdu, Sichuan, China
| |
Collapse
|
73
|
Beatty R, Mendez KL, Schreiber LHJ, Tarpey R, Whyte W, Fan Y, Robinson ST, O'Dwyer J, Simpkin AJ, Tannian J, Dockery P, Dolan EB, Roche ET, Duffy GP. Soft robot-mediated autonomous adaptation to fibrotic capsule formation for improved drug delivery. Sci Robot 2023; 8:eabq4821. [PMID: 37647382 DOI: 10.1126/scirobotics.abq4821] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 08/02/2023] [Indexed: 09/01/2023]
Abstract
The foreign body response impedes the function and longevity of implantable drug delivery devices. As a dense fibrotic capsule forms, integration of the device with the host tissue becomes compromised, ultimately resulting in device seclusion and treatment failure. We present FibroSensing Dynamic Soft Reservoir (FSDSR), an implantable drug delivery device capable of monitoring fibrotic capsule formation and overcoming its effects via soft robotic actuations. Occlusion of the FSDSR porous membrane was monitored over 7 days in a rodent model using electrochemical impedance spectroscopy. The electrical resistance of the fibrotic capsule correlated to its increase in thickness and volume. Our FibroSensing membrane showed great sensitivity in detecting changes at the abiotic/biotic interface, such as collagen deposition and myofibroblast proliferation. The potential of the FSDSR to overcome fibrotic capsule formation and maintain constant drug dosing over time was demonstrated in silico and in vitro. Controlled closed loop release of methylene blue into agarose gels (with a comparable fold change in permeability relating to 7 and 28 days in vivo) was achieved by adjusting the magnitude and frequency of pneumatic actuations after impedance measurements by the FibroSensing membrane. By sensing fibrotic capsule formation in vivo, the FSDSR will be capable of probing and adapting to the foreign body response through dynamic actuation changes. Informed by real-time sensor signals, this device offers the potential for long-term efficacy and sustained drug dosing, even in the setting of fibrotic capsule formation.
Collapse
Affiliation(s)
- Rachel Beatty
- Anatomy and Regenerative Medicine Institute (REMEDI), School of Medicine, University of Galway, Galway, Ireland
- SFI Centre for Advanced Materials and BioEngineering Research (AMBER), Trinity College Dublin, Dublin, Ireland
| | - Keegan L Mendez
- Harvard-MIT Program in Health Sciences and Technology, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Lucien H J Schreiber
- Anatomy and Regenerative Medicine Institute (REMEDI), School of Medicine, University of Galway, Galway, Ireland
| | - Ruth Tarpey
- Anatomy and Regenerative Medicine Institute (REMEDI), School of Medicine, University of Galway, Galway, Ireland
- CÚRAM, Centre for Research in Medical Devices, University of Galway, Galway, Ireland
- Biomedical Engineering, School of Engineering, University of Galway, Galway, Ireland
| | - William Whyte
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yiling Fan
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Scott T Robinson
- Anatomy and Regenerative Medicine Institute (REMEDI), School of Medicine, University of Galway, Galway, Ireland
- SFI Centre for Advanced Materials and BioEngineering Research (AMBER), Trinity College Dublin, Dublin, Ireland
| | - Joanne O'Dwyer
- Anatomy and Regenerative Medicine Institute (REMEDI), School of Medicine, University of Galway, Galway, Ireland
| | - Andrew J Simpkin
- School of Mathematical and Statistical Sciences, University of Galway, Galway, Ireland
| | - Joseph Tannian
- Anatomy and Regenerative Medicine Institute (REMEDI), School of Medicine, University of Galway, Galway, Ireland
| | - Peter Dockery
- Anatomy and Regenerative Medicine Institute (REMEDI), School of Medicine, University of Galway, Galway, Ireland
| | - Eimear B Dolan
- Anatomy and Regenerative Medicine Institute (REMEDI), School of Medicine, University of Galway, Galway, Ireland
- CÚRAM, Centre for Research in Medical Devices, University of Galway, Galway, Ireland
- Biomedical Engineering, School of Engineering, University of Galway, Galway, Ireland
| | - Ellen T Roche
- Harvard-MIT Program in Health Sciences and Technology, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Garry P Duffy
- Anatomy and Regenerative Medicine Institute (REMEDI), School of Medicine, University of Galway, Galway, Ireland
- SFI Centre for Advanced Materials and BioEngineering Research (AMBER), Trinity College Dublin, Dublin, Ireland
- CÚRAM, Centre for Research in Medical Devices, University of Galway, Galway, Ireland
| |
Collapse
|
74
|
Wang L, Li H, Wang X, Yang X, Tian C, Sun D, Liu L, Li J. Modification of Low-Energy Surfaces Using Bicyclic Peptides Discovered by Phage Display. J Am Chem Soc 2023; 145:17613-17620. [PMID: 37531461 DOI: 10.1021/jacs.3c02943] [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: 08/04/2023]
Abstract
Solid-binding peptides are a simple and versatile tool for the non-covalent modification of solid material surfaces, and a variety of peptides have been developed by reference to natural proteins or de novo design. Here, for the first time, we report the discovery of a bicyclic peptide targeting the heterogeneous material polypropylene by combining phage display technology and next-generation sequencing. We find that the enrichment properties of bicyclic peptides capable of binding to polypropylene are distinct from linear peptides, as reflected in amino acid abundance and a trend toward negative net charges and high hydrophobicity. The selected bicyclic peptide has a higher binding affinity for polypropylene compared with a previously reported linear peptide, enabling the hydrophilic and adhesive properties of the polypropylene to be more effectively enhanced. Our work paves the way for the exploration and utilization of conformational-restricted cyclic peptides as a new family of functionally evolvable agents for material surface modification.
Collapse
Affiliation(s)
- Lingxiao Wang
- Department of Chemistry, Center for BioAnalytical Chemistry, Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Haodong Li
- Department of Chemistry, Center for BioAnalytical Chemistry, Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Xinyan Wang
- Tsinghua-Peking Center for Life Sciences, Ministry of Education Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Center for Synthetic and Systems Biology, Department of Chemistry, Tsinghua University, Beijing 100084, China
| | - Xichu Yang
- Center for BioAnalytical Chemistry, Hefei National Laboratory of Physical Science at Microscale, University of Science and Technology of China, Hefei 230026, China
| | - Changlin Tian
- Center for BioAnalytical Chemistry, Hefei National Laboratory of Physical Science at Microscale, University of Science and Technology of China, Hefei 230026, China
| | - Demeng Sun
- Center for BioAnalytical Chemistry, Hefei National Laboratory of Physical Science at Microscale, University of Science and Technology of China, Hefei 230026, China
| | - Lei Liu
- Tsinghua-Peking Center for Life Sciences, Ministry of Education Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Center for Synthetic and Systems Biology, Department of Chemistry, Tsinghua University, Beijing 100084, China
| | - Jinghong Li
- Department of Chemistry, Center for BioAnalytical Chemistry, Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
- New Cornerstone Science Laboratory, Shenzhen 518054, China
| |
Collapse
|
75
|
Lu K, Brauns T, Sluder AE, Poznansky MC, Dogan F. Combinatorial islet protective therapeutic approaches in β-cell transplantation: Rationally designed solutions using a target product profile. FASEB Bioadv 2023; 5:287-304. [PMID: 37415930 PMCID: PMC10320848 DOI: 10.1096/fba.2023-00029] [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: 03/19/2023] [Revised: 05/03/2023] [Accepted: 05/17/2023] [Indexed: 07/08/2023] Open
Abstract
While progress has been made in the development of islet cell transplantation (ICT) as a viable alternative to the use of exogenous insulin therapy in the treatment of type 1 diabetes, it has not yet achieved its full potential in clinical studies. Ideally, ICT would enable lifelong maintenance of euglycemia without the need for exogenous insulin, blood glucose monitoring or systemic immune suppression. To achieve such an optimal result, therapeutic approaches should simultaneously promote long-term islet viability, functionality, and localized immune protection. In practice, however, these factors are typically tackled individually. Furthermore, while the requirements of optimal ICT are implicitly acknowledged across numerous publications, the literature contains few comprehensive articulations of the target product profile (TPP) for an optimal ICT product, including key characteristics of safety and efficacy. This review aims to provide a novel TPP for ICT and presents promising tried and untried combinatorial approaches that could be used to achieve the target product profile. We also highlight regulatory barriers to the development and adoption of ICT, particularly in the United States, where ICT is only approved for use in academic clinical trials and is not reimbursed by insurance carriers. Overall, this review argues that the clear definition of a TPP in addition to the use of combinatorial approaches could help to overcome the clinical barriers to the widespread adoption of ICT for the treatment of type 1 diabetes.
Collapse
Affiliation(s)
- Katie Lu
- Vaccine and Immunotherapy CenterMassachusetts General HospitalBostonMassachusettsUSA
- Department of BiologyStanford UniversityStanfordCaliforniaUSA
| | - Timothy Brauns
- Vaccine and Immunotherapy CenterMassachusetts General HospitalBostonMassachusettsUSA
| | - Ann E. Sluder
- Vaccine and Immunotherapy CenterMassachusetts General HospitalBostonMassachusettsUSA
| | - Mark C. Poznansky
- Vaccine and Immunotherapy CenterMassachusetts General HospitalBostonMassachusettsUSA
| | - Fatma Dogan
- Vaccine and Immunotherapy CenterMassachusetts General HospitalBostonMassachusettsUSA
| |
Collapse
|
76
|
Bressler EM, Adams S, Liu R, Colson YL, Wong WW, Grinstaff MW. Boolean logic in synthetic biology and biomaterials: Towards living materials in mammalian cell therapeutics. Clin Transl Med 2023; 13:e1244. [PMID: 37386762 PMCID: PMC10310979 DOI: 10.1002/ctm2.1244] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 04/06/2023] [Accepted: 04/14/2023] [Indexed: 07/01/2023] Open
Abstract
BACKGROUND The intersection of synthetic biology and biomaterials promises to enhance safety and efficacy in novel therapeutics. Both fields increasingly employ Boolean logic, which allows for specific therapeutic outputs (e.g., drug release, peptide synthesis) in response to inputs such as disease markers or bio-orthogonal stimuli. Examples include stimuli-responsive drug delivery devices and logic-gated chimeric antigen receptor (CAR) T cells. In this review, we explore recent manuscripts highlighting the potential of synthetic biology and biomaterials with Boolean logic to create novel and efficacious living therapeutics. MAIN BODY Collaborations in synthetic biology and biomaterials have led to significant advancements in drug delivery and cell therapy. Borrowing from synthetic biology, researchers have created Boolean-responsive biomaterials sensitive to multiple inputs including pH, light, enzymes and more to produce functional outputs such as degradation, gel-sol transition and conformational change. Biomaterials also enhance synthetic biology, particularly CAR T and adoptive T cell therapy, by modulating therapeutic immune cells in vivo. Nanoparticles and hydrogels also enable in situ generation of CAR T cells, which promises to drive down production costs and expand access to these therapies to a larger population. Biomaterials are also used to interface with logic-gated CAR T cell therapies, creating controllable cellular therapies that enhance safety and efficacy. Finally, designer cells acting as living therapeutic factories benefit from biomaterials that improve biocompatibility and stability in vivo. CONCLUSION By using Boolean logic in both cellular therapy and drug delivery devices, researchers have achieved better safety and efficacy outcomes. While early projects show incredible promise, coordination between these fields is ongoing and growing. We expect that these collaborations will continue to grow and realize the next generation of living biomaterial therapeutics.
Collapse
Affiliation(s)
- Eric M. Bressler
- Department of Biomedical Engineering and Biological Design CenterBoston UniversityBostonMassachusettsUSA
| | - Sarah Adams
- Department of Biomedical Engineering and Biological Design CenterBoston UniversityBostonMassachusettsUSA
| | - Rong Liu
- Division of Thoracic SurgeryDepartment of SurgeryMassachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Yolonda L. Colson
- Division of Thoracic SurgeryDepartment of SurgeryMassachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Wilson W. Wong
- Department of Biomedical Engineering and Biological Design CenterBoston UniversityBostonMassachusettsUSA
| | - Mark W. Grinstaff
- Department of Biomedical Engineering and Biological Design CenterBoston UniversityBostonMassachusettsUSA
- Department of Chemistry and Department of Biomedical EngineeringBoston UniversityBostonMassachusettsUSA
| |
Collapse
|
77
|
Mukherjee S, Kim B, Cheng LY, Doerfert MD, Li J, Hernandez A, Liang L, Jarvis MI, Rios PD, Ghani S, Joshi I, Isa D, Ray T, Terlier T, Fell C, Song P, Miranda RN, Oberholzer J, Zhang DY, Veiseh O. Screening hydrogels for antifibrotic properties by implanting cellularly barcoded alginates in mice and a non-human primate. Nat Biomed Eng 2023; 7:867-886. [PMID: 37106151 PMCID: PMC10593184 DOI: 10.1038/s41551-023-01016-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 02/27/2023] [Indexed: 04/29/2023]
Abstract
Screening implantable biomaterials for antifibrotic properties is constrained by the need for in vivo testing. Here we show that the throughput of in vivo screening can be increased by cellularly barcoding a chemically modified combinatorial library of hydrogel formulations. The method involves the implantation of a mixture of alginate formulations, each barcoded with human umbilical vein endothelial cells from different donors, and the association of the identity and performance of each formulation by genotyping single nucleotide polymorphisms of the cells via next-generation sequencing. We used the method to screen 20 alginate formulations in a single mouse and 100 alginate formulations in a single non-human primate, and identified three lead hydrogel formulations with antifibrotic properties. Encapsulating human islets with one of the formulations led to long-term glycaemic control in a mouse model of diabetes, and coating medical-grade catheters with the other two formulations prevented fibrotic overgrowth. High-throughput screening of barcoded biomaterials in vivo may help identify formulations that enhance the long-term performance of medical devices and of biomaterial-encapsulated therapeutic cells.
Collapse
Affiliation(s)
- Sudip Mukherjee
- Department of Bioengineering, Rice University, Houston, TX, USA
- School of Biomedical Engineering, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, India
| | - Boram Kim
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Lauren Y Cheng
- Department of Bioengineering, Rice University, Houston, TX, USA
| | | | - Jiaming Li
- Department of Bioengineering, Rice University, Houston, TX, USA
| | | | - Lily Liang
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Maria I Jarvis
- Department of Bioengineering, Rice University, Houston, TX, USA
| | | | | | | | | | - Trisha Ray
- Department of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Tanguy Terlier
- SIMS Laboratory, Shared Equipment Authority, Rice University, Houston, TX, USA
| | - Cody Fell
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Ping Song
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Roberto N Miranda
- Department of Hematopathology, Division of Pathology/Lab Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jose Oberholzer
- Division of Transplant Surgery, University of Virginia, Charlottesville, VA, USA
| | - David Yu Zhang
- Department of Bioengineering, Rice University, Houston, TX, USA.
- NuProbe USA, Houston, TX, USA.
| | - Omid Veiseh
- Department of Bioengineering, Rice University, Houston, TX, USA.
| |
Collapse
|
78
|
Hao H, Wu S, Lin J, Zheng Z, Zhou Y, Zhang Y, Guo Q, Tian F, Zhao M, Chen Y, Xu X, Hou L, Wang X, Tang R. Immunization against Zika by entrapping live virus in a subcutaneous self-adjuvanting hydrogel. Nat Biomed Eng 2023; 7:928-942. [PMID: 36959404 DOI: 10.1038/s41551-023-01014-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 02/20/2023] [Indexed: 03/25/2023]
Abstract
The threat of new viral outbreaks has heightened the need for ready-to-use vaccines that are safe and effective. Here we show that a subcutaneous vaccine consisting of live Zika virus electrostatically entrapped in a self-adjuvanting hydrogel recruited immune cells at the injection site and provided mice with effective protection against a lethal viral challenge. The hydrogel prevented the escape of the viral particles and upregulated pattern recognition receptors that activated innate antiviral immunity. The local inflammatory niche facilitated the engulfment of the virus by immune cells infiltrating the hydrogel, the processing and cross-presentation of antigens and the expansion of germinal centre B cells and induced robust antigen-specific adaptive responses and immune memory. Inflammatory immune niches entrapping live viruses may facilitate the rapid development of safe and efficacious vaccines.
Collapse
Affiliation(s)
- Haibin Hao
- Department of Chemistry, Zhejiang University, Hangzhou, China
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou, China
| | - Shipo Wu
- Beijing Institute of Biotechnology, Beijing, China
| | - Jiake Lin
- Department of Chemistry, Zhejiang University, Hangzhou, China
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou, China
| | - Zitong Zheng
- Department of Chemistry, Zhejiang University, Hangzhou, China
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou, China
| | - Yuemin Zhou
- Department of Chemistry, Zhejiang University, Hangzhou, China
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou, China
| | - Ying Zhang
- Department of Chemistry, Zhejiang University, Hangzhou, China
| | - Qiang Guo
- Beijing Institute of Biotechnology, Beijing, China
| | - Fengchao Tian
- Department of Chemistry, Zhejiang University, Hangzhou, China
| | - Mengsu Zhao
- Beijing Institute of Biotechnology, Beijing, China
| | - Yi Chen
- Beijing Institute of Biotechnology, Beijing, China
| | - Xurong Xu
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou, China
| | - Lihua Hou
- Beijing Institute of Biotechnology, Beijing, China.
| | - Xiaoyu Wang
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou, China.
- Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| | - Ruikang Tang
- Department of Chemistry, Zhejiang University, Hangzhou, China.
- Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
79
|
Qin T, Smink AM, de Vos P. Enhancing longevity of immunoisolated pancreatic islet grafts by modifying both the intracapsular and extracapsular environment. Acta Biomater 2023:S1742-7061(23)00362-8. [PMID: 37392934 DOI: 10.1016/j.actbio.2023.06.038] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 06/02/2023] [Accepted: 06/26/2023] [Indexed: 07/03/2023]
Abstract
Type 1 diabetes mellitus (T1DM) is a chronic metabolic disease characterized by autoimmune destruction of pancreatic β cells. Transplantation of immunoisolated pancreatic islets might treat T1DM in the absence of chronic immunosuppression. Important advances have been made in the past decade as capsules can be produced that provoke minimal to no foreign body response after implantation. However, graft survival is still limited as islet dysfunction may occur due to chronic damage to islets during islet isolation, immune responses induced by inflammatory cells, and nutritional issues for encapsulated cells. This review summarizes the current challenges for promoting longevity of grafts. Possible strategies for improving islet graft longevity are also discussed, including supplementation of the intracapsular milieu with essential survival factors, promotion of vascularization and oxygenation near capsules, modulation of biomaterials, and co-transplantation of accessory cells. Current insight is that both the intracapsular as well as the extracapsular properties should be improved to achieve long-term survival of islet-tissue. Some of these approaches reproducibly induce normoglycemia for more than a year in rodents. Further development of the technology requires collective research efforts in material science, immunology, and endocrinology. STATEMENT OF SIGNIFICANCE: Islet immunoisolation allows for transplantation of insulin producing cells in absence of immunosuppression and might facilitate the use of xenogeneic cell sources or grafting of cells obtained from replenishable cell sources. However, a major challenge to date is to create a microenvironment that supports long-term graft survival. This review provides a comprehensive overview of the currently identified factors that have been demonstrated to be involved in either stimulating or reducing islet graft survival in immunoisolating devices and discussed current strategies to enhance the longevity of encapsulated islet grafts as treatment for type 1 diabetes. Although significant challenges remain, interdisciplinary collaboration across fields may overcome obstacles and facilitate the translation of encapsulated cell therapy from the laboratory to clinical application.
Collapse
Affiliation(s)
- Tian Qin
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen and University Medical Center Groningen, Hanzeplein 1, EA 11, 9713 GZ, Groningen, The Netherlands.
| | - Alexandra M Smink
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen and University Medical Center Groningen, Hanzeplein 1, EA 11, 9713 GZ, Groningen, The Netherlands
| | - Paul de Vos
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen and University Medical Center Groningen, Hanzeplein 1, EA 11, 9713 GZ, Groningen, The Netherlands
| |
Collapse
|
80
|
Tuch BE, Cheng IS, Dang HP, Chen H, Dargaville TR. Pluripotent stem cells as a therapy for type 1 diabetes. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 199:363-378. [PMID: 37678980 DOI: 10.1016/bs.pmbts.2023.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/09/2023]
Affiliation(s)
- Bernard E Tuch
- Department Diabetes, Central Clinical School, Faculty of Medicine, Nursing & Health Sciences, Monash University, VIC, Australia; Australian Foundation for Diabetes Research, Sydney, NSW, Australia.
| | - Iris S Cheng
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Hoang Phuc Dang
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia.
| | - Hui Chen
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia.
| | - Tim R Dargaville
- School of Chemistry and Physics and Centre for Materials Science, Queensland University of Technology, Brisbane, QLD, Australia.
| |
Collapse
|
81
|
Doloff JC, Ma M, Sadraei A, Tam HH, Farah S, Hollister-Lock J, Vegas AJ, Veiseh O, Quiroz VM, Rakoski A, Aresta-DaSilva S, Bader AR, Griffin M, Weir GC, Brehm MA, Shultz LD, Langer R, Greiner DL, Anderson DG. Identification of a humanized mouse model for functional testing of immune-mediated biomaterial foreign body response. SCIENCE ADVANCES 2023; 9:eade9488. [PMID: 37327334 PMCID: PMC10275594 DOI: 10.1126/sciadv.ade9488] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 05/05/2023] [Indexed: 06/18/2023]
Abstract
Biomedical devices comprise a major component of modern medicine, however immune-mediated fibrosis and rejection can limit their function over time. Here, we describe a humanized mouse model that recapitulates fibrosis following biomaterial implantation. Cellular and cytokine responses to multiple biomaterials were evaluated across different implant sites. Human innate immune macrophages were verified as essential to biomaterial rejection in this model and were capable of cross-talk with mouse fibroblasts for collagen matrix deposition. Cytokine and cytokine receptor array analysis confirmed core signaling in the fibrotic cascade. Foreign body giant cell formation, often unobserved in mice, was also prominent. Last, high-resolution microscopy coupled with multiplexed antibody capture digital profiling analysis supplied spatial resolution of rejection responses. This model enables the study of human immune cell-mediated fibrosis and interactions with implanted biomaterials and devices.
Collapse
Affiliation(s)
- Joshua C. Doloff
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main St., Cambridge, MA 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, 300 Longwood Ave., Boston, MA 02115, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, MA 02139, USA
- Department of Biomedical Engineering, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Minglin Ma
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main St., Cambridge, MA 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, 300 Longwood Ave., Boston, MA 02115, USA
| | - Atieh Sadraei
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main St., Cambridge, MA 02139, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, MA 02139, USA
| | - Hok Hei Tam
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main St., Cambridge, MA 02139, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, MA 02139, USA
| | - Shady Farah
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main St., Cambridge, MA 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, 300 Longwood Ave., Boston, MA 02115, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, MA 02139, USA
| | - Jennifer Hollister-Lock
- Section on Islet Cell and Regenerative Biology, Research Division, Joslin Diabetes Center, One Joslin Place, Boston, MA 02215, USA
| | - Arturo J. Vegas
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main St., Cambridge, MA 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, 300 Longwood Ave., Boston, MA 02115, USA
| | - Omid Veiseh
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main St., Cambridge, MA 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, 300 Longwood Ave., Boston, MA 02115, USA
| | - Victor M. Quiroz
- Department of Biomedical Engineering, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Amanda Rakoski
- Department of Biomedical Engineering, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Stephanie Aresta-DaSilva
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main St., Cambridge, MA 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, 300 Longwood Ave., Boston, MA 02115, USA
| | - Andrew R. Bader
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main St., Cambridge, MA 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, 300 Longwood Ave., Boston, MA 02115, USA
| | - Marissa Griffin
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main St., Cambridge, MA 02139, USA
| | - Gordon C. Weir
- Section on Islet Cell and Regenerative Biology, Research Division, Joslin Diabetes Center, One Joslin Place, Boston, MA 02215, USA
| | - Michael A. Brehm
- Program in Molecular Medicine, Diabetes Centre of Excellence, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | | | - Robert Langer
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main St., Cambridge, MA 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, 300 Longwood Ave., Boston, MA 02115, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, MA 02139, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, MA 02139, USA
- Harvard-MIT Division of Health Science and Technology, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, MA 02139, USA
| | - Dale L. Greiner
- Program in Molecular Medicine, Diabetes Centre of Excellence, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Daniel G. Anderson
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main St., Cambridge, MA 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, 300 Longwood Ave., Boston, MA 02115, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, MA 02139, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, MA 02139, USA
- Harvard-MIT Division of Health Science and Technology, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, MA 02139, USA
| |
Collapse
|
82
|
Nash A, Lokhorst N, Veiseh O. Localized immunomodulation technologies to enable cellular and organoid transplantation. Trends Mol Med 2023:S1471-4914(23)00097-7. [PMID: 37301656 DOI: 10.1016/j.molmed.2023.05.008] [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: 03/06/2023] [Revised: 05/16/2023] [Accepted: 05/17/2023] [Indexed: 06/12/2023]
Abstract
Localized immunomodulation technologies are rapidly emerging as a new modality with the potential to revolutionize transplantation of cells and organs. In the past decade, cell-based immunomodulation therapies saw clinical success in the treatment of cancer and autoimmune diseases. In this review, we describe recent advances in engineering solutions for the development of localized immunomodulation techniques focusing on cellular and organoid transplantation. We begin by describing cell transplantation and highlighting notable clinical successes, particularly in the areas of stem cell therapy, chimeric antigen receptor (CAR)-T cell therapy, and islet transplantation. Next, we detail recent preclinical studies centered on genome editing and biomaterials to enhance localized immunomodulation. We close by discussing future opportunities to improve clinical and commercial success using these approaches to facilitate long-term immunomodulation technologies.
Collapse
Affiliation(s)
- Amanda Nash
- Department of Bioengineering, Rice University, Houston, TX 77030, USA
| | - Nienke Lokhorst
- Department of Pharmaceutical Sciences, Utrecht University, Utrecht 3584, CG, The Netherlands
| | - Omid Veiseh
- Department of Bioengineering, Rice University, Houston, TX 77030, USA.
| |
Collapse
|
83
|
Li W, Wu Y, Zhang X, Wu T, Huang K, Wang B, Liao J. Self-healing hydrogels for bone defect repair. RSC Adv 2023; 13:16773-16788. [PMID: 37283866 PMCID: PMC10240173 DOI: 10.1039/d3ra01700a] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 05/24/2023] [Indexed: 06/08/2023] Open
Abstract
Severe bone defects can be caused by various factors, such as tumor resection, severe trauma, and infection. However, bone regeneration capacity is limited up to a critical-size defect, and further intervention is required. Currently, the most common clinical method to repair bone defects is bone grafting, where autografts are the "gold standard." However, the disadvantages of autografts, including inflammation, secondary trauma and chronic disease, limit their application. Bone tissue engineering (BTE) is an attractive strategy for repairing bone defects and has been widely researched. In particular, hydrogels with a three-dimensional network can be used as scaffolds for BTE owing to their hydrophilicity, biocompatibility, and large porosity. Self-healing hydrogels respond rapidly, autonomously, and repeatedly to induced damage and can maintain their original properties (i.e., mechanical properties, fluidity, and biocompatibility) following self-healing. This review focuses on self-healing hydrogels and their applications in bone defect repair. Moreover, we discussed the recent progress in this research field. Despite the significant existing research achievements, there are still challenges that need to be addressed to promote clinical research of self-healing hydrogels in bone defect repair and increase the market penetration.
Collapse
Affiliation(s)
- Weiwei Li
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University Chengdu 610041 China
| | - Yanting Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University Chengdu 610041 China
| | - Xu Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University Chengdu 610041 China
| | - Tingkui Wu
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University Chengdu 610041 China
| | - Kangkang Huang
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University Chengdu 610041 China
| | - Beiyu Wang
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University Chengdu 610041 China
| | - Jinfeng Liao
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University Chengdu 610041 China
| |
Collapse
|
84
|
Schluck M, Weiden J, Verdoes M, Figdor CG. Insights in the host response towards biomaterial-based scaffolds for cancer therapy. Front Bioeng Biotechnol 2023; 11:1149943. [PMID: 37342507 PMCID: PMC10277494 DOI: 10.3389/fbioe.2023.1149943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 05/23/2023] [Indexed: 06/23/2023] Open
Abstract
Immunotherapeutic strategies have shown promising results in the treatment of cancer. However, not all patients respond, and treatments can have severe side-effects. Adoptive cell therapy (ACT) has shown remarkable therapeutic efficacy across different leukaemia and lymphoma types. But the treatment of solid tumours remains a challenge due to limited persistence and tumour infiltration. We believe that biomaterial-based scaffolds are promising new tools and may address several of the challenges associated with cancer vaccination and ACT. In particular, biomaterial-based scaffold implants allow for controlled delivery of activating signals and/or functional T cells at specific sites. One of the main challenges for their application forms the host response against these scaffolds, which includes unwanted myeloid cell infiltration and the formation of a fibrotic capsule around the scaffold, thereby limiting cell traffic. In this review we provide an overview of several of the biomaterial-based scaffolds designed for cancer therapy to date. We will discuss the host responses observed and we will highlight design parameters that influence this response and their potential impact on therapeutic outcome.
Collapse
Affiliation(s)
- Marjolein Schluck
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Nijmegen, Netherlands
- Oncode Institute, Nijmegen, Netherlands
- Institute for Chemical Immunology, Nijmegen, Netherlands
| | - Jorieke Weiden
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Nijmegen, Netherlands
- Oncode Institute, Nijmegen, Netherlands
- Institute for Chemical Immunology, Nijmegen, Netherlands
| | - Martijn Verdoes
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Nijmegen, Netherlands
- Institute for Chemical Immunology, Nijmegen, Netherlands
| | - Carl G. Figdor
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Nijmegen, Netherlands
- Oncode Institute, Nijmegen, Netherlands
- Institute for Chemical Immunology, Nijmegen, Netherlands
| |
Collapse
|
85
|
Cober ND, Rowe K, Deng Y, Benavente‐Babace A, Courtman DW, Godin M, Stewart DJ. Targeting extracellular vesicle delivery to the lungs by microgel encapsulation. JOURNAL OF EXTRACELLULAR BIOLOGY 2023; 2:e94. [PMID: 38938918 PMCID: PMC11080904 DOI: 10.1002/jex2.94] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 04/29/2023] [Accepted: 05/17/2023] [Indexed: 06/29/2024]
Abstract
Extracellular vesicles (EVs) secreted by stem and progenitor cells have significant potential as cell-free 'cellular' therapeutics. Yet, small EVs (<200 nm) are rapidly cleared after systemic administration, mainly by the liver, presenting challenges targeting EVs to a specific organ or tissue. Microencapsulation using natural nano-porous hydrogels (microgels) has been shown to enhance engraftment and increase the survival of transplanted cells. We sought to encapsulate EVs within microgels to target their delivery to the lung by virtue of their size-based retention within the pulmonary microcirculation. Mesenchymal stromal cell (MSC) derived EVs were labelled with the lipophilic dye (DiR) and encapsulated within agarose-gelatin microgels. Endothelial cells and bone marrow derived macrophages were able to take up EVs encapsulated in microgels in vitro, but less efficiently than the uptake of free EVs. Following intrajugular administration, microgel encapsulated EVs were selectively retained within the lungs for 72h, while free EVs were rapidly cleared by the liver. Furthermore, microgel-loaded EVs demonstrated greater uptake by lung cells, in particular CD45+ immune cells, as assessed by flow cytometry compared to free EVs. Microencapsulation of EVs may be a novel tool for enhancing the targeted delivery of EVs for future therapeutic applications.
Collapse
Affiliation(s)
- Nicholas D. Cober
- Sinclair Centre for Regenerative MedicineOttawa Hospital Research InstituteOttawaOntarioCanada
- Faculty of MedicineDepartment of Cellular and Molecular MedicineUniversity of OttawaOttawaOntarioCanada
| | - Katelynn Rowe
- Sinclair Centre for Regenerative MedicineOttawa Hospital Research InstituteOttawaOntarioCanada
| | - Yupu Deng
- Sinclair Centre for Regenerative MedicineOttawa Hospital Research InstituteOttawaOntarioCanada
| | | | - David W. Courtman
- Sinclair Centre for Regenerative MedicineOttawa Hospital Research InstituteOttawaOntarioCanada
| | - Michel Godin
- Faculty of Science, Department of PhysicsUniversity of OttawaOttawaOntarioCanada
| | - Duncan J. Stewart
- Sinclair Centre for Regenerative MedicineOttawa Hospital Research InstituteOttawaOntarioCanada
- Faculty of MedicineDepartment of Cellular and Molecular MedicineUniversity of OttawaOttawaOntarioCanada
| |
Collapse
|
86
|
Liu B, Zhang C, Zhao H, Gao J, Hu J. Chitosan Hydrogel-Delivered ABE8e Corrects PAX9 Mutant in Dental Pulp Stem Cells. Gels 2023; 9:436. [PMID: 37367107 DOI: 10.3390/gels9060436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 05/19/2023] [Accepted: 05/23/2023] [Indexed: 06/28/2023] Open
Abstract
Hypodontia (dental agenesis) is a genetic disorder, and it has been identified that the mutation C175T in PAX9 could lead to hypodontia. Cas9 nickase (nCas9)-mediated homology-directed repair (HDR) and base editing were used for the correction of this mutated point. This study aimed to investigate the effect of HDR and the base editor ABE8e in editing PAX9 mutant. It was found that the chitosan hydrogel was efficient in delivering naked DNA into dental pulp stem cells (DPSCs). To explore the influence of the C175T mutation in PAX9 on the proliferation of DPSCs, hydrogel was employed to deliver PAX9 mutant vector into DPSCs, finding that the PAX9-containing C175T mutation failed to promote the proliferation of DPSCs. Firstly, DPSCs stably carrying PAX9 mutant were constructed. Either an HDR or ABE8e system was delivered into the above-mentioned stable DPSCs, and then the correction efficiency using Sanger sequencing and Western blotting was determined. Meanwhile, the ABE8e presented significantly higher efficiency in correcting C175T compared with HDR. Furthermore, the corrected PAX9 presented enhanced viability and differentiation capacity for osteogenic and neurogenic lineages; the corrected PAX9 even possessed extremely enhanced transcriptional activation ability. In summary, this study has powerful implications for studies into base editors, chitosan hydrogel, and DPSCs in treating hypodontia.
Collapse
Affiliation(s)
- Bowen Liu
- Outpatient Department of Oral and Maxillofacial Surgery, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Tian Tan Xi Li No. 4, Beijing 100050, China
| | - Chenjiao Zhang
- Department of General, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Tian Tan Xi Li No. 4, Beijing 100050, China
| | - Han Zhao
- Multi-Disciplinary Treatment Center, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Tian Tan Xi Li No. 4, Beijing 100050, China
| | - Jian Gao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Jingchao Hu
- Department of Periodontics, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Tian Tan Xi Li No. 4, Beijing 100050, China
| |
Collapse
|
87
|
Hogrebe NJ, Ishahak M, Millman JR. Developments in stem cell-derived islet replacement therapy for treating type 1 diabetes. Cell Stem Cell 2023; 30:530-548. [PMID: 37146579 PMCID: PMC10167558 DOI: 10.1016/j.stem.2023.04.002] [Citation(s) in RCA: 74] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/20/2023] [Accepted: 04/05/2023] [Indexed: 05/07/2023]
Abstract
The generation of islet-like endocrine clusters from human pluripotent stem cells (hPSCs) has the potential to provide an unlimited source of insulin-producing β cells for the treatment of diabetes. In order for this cell therapy to become widely adopted, highly functional and well-characterized stem cell-derived islets (SC-islets) need to be manufactured at scale. Furthermore, successful SC-islet replacement strategies should prevent significant cell loss immediately following transplantation and avoid long-term immune rejection. This review highlights the most recent advances in the generation and characterization of highly functional SC-islets as well as strategies to ensure graft viability and safety after transplantation.
Collapse
Affiliation(s)
- Nathaniel J Hogrebe
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, 660 South Euclid Avenue, St. Louis, MO 63130, USA.
| | - Matthew Ishahak
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, 660 South Euclid Avenue, St. Louis, MO 63130, USA
| | - Jeffrey R Millman
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, 660 South Euclid Avenue, St. Louis, MO 63130, USA; Department of Biomedical Engineering, Washington University in St. Louis, 1 Brookings Drive, St. Louis, MO 63130, USA.
| |
Collapse
|
88
|
Morrison RA, Brookes S, Puls TJ, Cox A, Gao H, Liu Y, Voytik-Harbin SL. Engineered collagen polymeric materials create noninflammatory regenerative microenvironments that avoid classical foreign body responses. Biomater Sci 2023; 11:3278-3296. [PMID: 36942875 PMCID: PMC10152923 DOI: 10.1039/d3bm00091e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 02/26/2023] [Indexed: 03/23/2023]
Abstract
The efficacy and longevity of medical implants and devices is largely determined by the host immune response, which extends along a continuum from pro-inflammatory/pro-fibrotic to anti-inflammatory/pro-regenerative. Using a rat subcutaneous implantation model, along with histological and transcriptomics analyses, we characterized the tissue response to a collagen polymeric scaffold fabricated from polymerizable type I oligomeric collagen (Oligomer) in comparison to commercial synthetic and collagen-based products. In contrast to commercial biomaterials, no evidence of an immune-mediated foreign body reaction, fibrosis, or bioresorption was observed with Oligomer scaffolds for beyond 60 days. Oligomer scaffolds were noninflammatory, eliciting minimal innate inflammation and immune cell accumulation similar to sham surgical controls. Genes associated with Th2 and regulatory T cells were instead upregulated, implying a novel pathway to immune tolerance and regenerative remodeling for biomaterials.
Collapse
Affiliation(s)
- Rachel A Morrison
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA.
| | - Sarah Brookes
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA.
| | | | - Abigail Cox
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN 47907, USA
| | - Hongyu Gao
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Yunlong Liu
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Sherry L Voytik-Harbin
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA.
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
89
|
Schreib CC, Jarvis MI, Terlier T, Goell J, Mukherjee S, Doerfert MD, Wilson TA, Beauregard M, Martins KN, Lee J, Solis LS, Vazquez E, Oberli MA, Hanak BW, Diehl M, Hilton I, Veiseh O. Lipid Deposition Profiles Influence Foreign Body Responses. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2205709. [PMID: 36871193 PMCID: PMC10309593 DOI: 10.1002/adma.202205709] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 12/09/2022] [Indexed: 05/26/2023]
Abstract
Fibrosis remains a significant cause of failure in implanted biomedical devices and early absorption of proteins on implant surfaces has been shown to be a key instigating factor. However, lipids can also regulate immune activity and their presence may also contribute to biomaterial-induced foreign body responses (FBR) and fibrosis. Here it is demonstrated that the surface presentation of lipids on implant affects FBR by influencing reactions of immune cells to materials as well as their resultant inflammatory/suppressive polarization. Time-of-flight secondary ion mass spectroscopy (ToF-SIMS) is employed to characterize lipid deposition on implants that are surface-modified chemically with immunomodulatory small molecules. Multiple immunosuppressive phospholipids (phosphatidylcholine, phosphatidylinositol, phosphatidylethanolamine, and sphingomyelin) are all found to deposit preferentially on implants with anti-FBR surface modifications in mice. Significantly, a set of 11 fatty acids is enriched on unmodified implanted devices that failed in both mice and humans, highlighting relevance across species. Phospholipid deposition is also found to upregulate the transcription of anti-inflammatory genes in murine macrophages, while fatty acid deposition stimulated the expression of pro-inflammatory genes. These results provide further insights into how to improve the design of biomaterials and medical devices to mitigate biomaterial material-induced FBR and fibrosis.
Collapse
Affiliation(s)
- Christian C. Schreib
- Department of Bioengineering, Rice University, 6500 Main Street, Houston, TX 77030
| | - Maria I. Jarvis
- Department of Bioengineering, Rice University, 6500 Main Street, Houston, TX 77030
- Present address: Lonza Inc. 14905 Kirby Drive, Houston, TX 77047
| | - Tanguy Terlier
- SIMS laboratory, Shared Equipment Authority, Rice University, 6500 Main Street, Houston, TX 77030
| | - Jacob Goell
- Department of Bioengineering, Rice University, 6500 Main Street, Houston, TX 77030
| | - Sudip Mukherjee
- Department of Bioengineering, Rice University, 6500 Main Street, Houston, TX 77030
- Present address: School of Biomedial Engineering, ITT (BHU), Varanasi 221005, Uttar Pradesh, India
| | - Michael D. Doerfert
- Department of Bioengineering, Rice University, 6500 Main Street, Houston, TX 77030
| | - Taylor Anne Wilson
- Department of Neurosurgery, Loma Linda University Health, 11234 Anderson St, Loma Linda, CA 92354
| | - Michael Beauregard
- Department of Bioengineering, Rice University, 6500 Main Street, Houston, TX 77030
| | - Kevin N. Martins
- Department of Neurosurgery, Loma Linda University Health, 11234 Anderson St, Loma Linda, CA 92354
| | - Jared Lee
- Department of Chemistry, Rice University, 6100 Main St, Houston, TX 77005
| | - Leo Sanchez Solis
- Department of Bioengineering, Rice University, 6500 Main Street, Houston, TX 77030
| | - Esperanza Vazquez
- Department of Biomedical Engineering, University of Houston, 3517 Cullen Blvd, Houston, TX 77204
| | - Matthias A. Oberli
- Sigilon Therapeutics, 200 Dexter Avenue, Watertown, MA 02472
- Present address: Xibus systems Inc. 200 Dexter Avenue, Watertown, MA 02472
| | - Brian W. Hanak
- Department of Neurosurgery, Loma Linda University Health, 11234 Anderson St, Loma Linda, CA 92354
| | - Michael Diehl
- Department of Bioengineering, Rice University, 6500 Main Street, Houston, TX 77030
| | - Isaac Hilton
- Department of Bioengineering, Rice University, 6500 Main Street, Houston, TX 77030
- Program of Synthetic, Systems and Physical Biology, Rice University, 6500 Main Street, Houston, TX 77030
| | - Omid Veiseh
- Department of Bioengineering, Rice University, 6500 Main Street, Houston, TX 77030
- Program of Synthetic, Systems and Physical Biology, Rice University, 6500 Main Street, Houston, TX 77030
| |
Collapse
|
90
|
Maity D, Guha Ray P, Buchmann P, Mansouri M, Fussenegger M. Blood-Glucose-Powered Metabolic Fuel Cell for Self-Sufficient Bioelectronics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2300890. [PMID: 36893359 DOI: 10.1002/adma.202300890] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/28/2023] [Indexed: 05/26/2023]
Abstract
Currently available bioelectronic devices consume too much power to be continuously operated on rechargeable batteries, and are often powered wirelessly, with attendant issues regarding reliability, convenience, and mobility. Thus, the availability of a robust, self-sufficient, implantable electrical power generator that works under physiological conditions would be transformative for many applications, from driving bioelectronic implants and prostheses to programing cellular behavior and patients' metabolism. Here, capitalizing on a new copper-containing, conductively tuned 3D carbon nanotube composite, an implantable blood-glucose-powered metabolic fuel cell is designed that continuously monitors blood-glucose levels, converts excess glucose into electrical power during hyperglycemia, and produces sufficient energy (0.7 mW cm-2 , 0.9 V, 50 mm glucose) to drive opto- and electro-genetic regulation of vesicular insulin release from engineered beta cells. It is shown that this integration of blood-glucose monitoring with elimination of excessive blood glucose by combined electro-metabolic conversion and insulin-release-mediated cellular consumption enables the metabolic fuel cell to restore blood-glucose homeostasis in an automatic, self-sufficient, and closed-loop manner in an experimental model of type-1 diabetes.
Collapse
Affiliation(s)
- Debasis Maity
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, Basel, CH-4058, Switzerland
| | - Preetam Guha Ray
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, Basel, CH-4058, Switzerland
| | - Peter Buchmann
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, Basel, CH-4058, Switzerland
| | - Maysam Mansouri
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, Basel, CH-4058, Switzerland
| | - Martin Fussenegger
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, Basel, CH-4058, Switzerland
- Faculty of Science, University of Basel, Mattenstrasse 26, Basel, CH-4058, Switzerland
| |
Collapse
|
91
|
Meyer T, Ramirez C, Tamasi MJ, Gormley AJ. A User's Guide to Machine Learning for Polymeric Biomaterials. ACS POLYMERS AU 2023; 3:141-157. [PMID: 37065715 PMCID: PMC10103193 DOI: 10.1021/acspolymersau.2c00037] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 10/27/2022] [Accepted: 10/27/2022] [Indexed: 11/18/2022]
Abstract
The development of novel biomaterials is a challenging process, complicated by a design space with high dimensionality. Requirements for performance in the complex biological environment lead to difficult a priori rational design choices and time-consuming empirical trial-and-error experimentation. Modern data science practices, especially artificial intelligence (AI)/machine learning (ML), offer the promise to help accelerate the identification and testing of next-generation biomaterials. However, it can be a daunting task for biomaterial scientists unfamiliar with modern ML techniques to begin incorporating these useful tools into their development pipeline. This Perspective lays the foundation for a basic understanding of ML while providing a step-by-step guide to new users on how to begin implementing these techniques. A tutorial Python script has been developed walking users through the application of an ML pipeline using data from a real biomaterial design challenge based on group's research. This tutorial provides an opportunity for readers to see and experiment with ML and its syntax in Python. The Google Colab notebook can be easily accessed and copied from the following URL: www.gormleylab.com/MLcolab.
Collapse
Affiliation(s)
- Travis
A. Meyer
- Department of Biomedical
Engineering, Rutgers, The State University
of New Jersey, Piscataway, New Jersey 08854, United States
| | - Cesar Ramirez
- Department of Biomedical
Engineering, Rutgers, The State University
of New Jersey, Piscataway, New Jersey 08854, United States
| | - Matthew J. Tamasi
- Department of Biomedical
Engineering, Rutgers, The State University
of New Jersey, Piscataway, New Jersey 08854, United States
| | - Adam J. Gormley
- Department of Biomedical
Engineering, Rutgers, The State University
of New Jersey, Piscataway, New Jersey 08854, United States
| |
Collapse
|
92
|
Ji Y, Wang C. Magnetic iron oxide nanoparticle-loaded hydrogels for photothermal therapy of cancer cells. Front Bioeng Biotechnol 2023; 11:1130523. [PMID: 37008029 PMCID: PMC10060874 DOI: 10.3389/fbioe.2023.1130523] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 03/09/2023] [Indexed: 03/18/2023] Open
Abstract
Introduction: Non-invasive photothermal therapy (PTT) is a competitive treatment for solid tumors, while the efficacy is largely dependent on the effective retention of photothermal converters in tumor tissues.Methods: Herein, the development of iron oxide (Fe3O4) nanoparticle-loaded alginate (ALG) hydrogel platform for PTT of colorectal cancer cells is reported. Fe3O4 nanoparticles synthesized via coprecipitation method after reaction of 30 min have a small size (61.3 nm) and more suitable surface potential, and can mediate PTT under near-infrared (NIR) laser irradiation. The premix of Fe3O4 nanoparticles and ALG hydrogel precursors can be gelatinized by Ca2+-mediated cross-linking to form this therapeutic hydrogel platform.Results: The formed Fe3O4 nanoparticles can be effectively taken up by CT26 cancer cells and induce the death of CT26 cells in vitro under NIR laser irradiation because of their excellent photothermal property. In addition, Fe3O4 nanoparticle-loaded ALG hydrogels show negligible cytotoxicity at the studied concentration range, but can significantly kill cancer cells after PTT effect.Conclusion: This ALG-based hydrogel platform provides a valuable reference for subsequent in vivo studies and other related studies on Fe3O4 nanoparticle-loaded hydrogels.
Collapse
Affiliation(s)
- Yunfei Ji
- Department of Critical Care Medicine, Chengde Central Hospital, Chengde, Hebei, China
| | - Chunpu Wang
- Department of Cardiothoracic Surgery, Chengde Central Hospital, Chengde, Hebei, China
- *Correspondence: Chunpu Wang,
| |
Collapse
|
93
|
Wang Y, Wang K, Luo Y, Chen H. Hydrogel-Bondable Asymmetric Planar Membranes with Hierarchical Pore Structures for Cell Scaffolding and Encapsulation. ACS Biomater Sci Eng 2023; 9:1706-1719. [PMID: 36795620 DOI: 10.1021/acsbiomaterials.2c01415] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
Biomaterials for cell replacement therapy could facilitate the delivery, function, and retrieval of transplanted therapeutic cells. However, the limited capacity to accommodate a sufficient quantity of cells in biomedical devices has hindered the success of clinical application, resulting from the suboptimal spatial organization of cells and insufficient permeation of nutrients in the materials. Herein, through the immersion-precipitation phase transfer (IPPT) process from polyether sulfone (PES), we develop planar asymmetric membranes with a hierarchical pore architecture spanning from nanopores (∼20 nm) in the dense skin and open-ended microchannel arrays with gradient pore size increasing vertically from microns to ∼100 μm. The nanoporous skin would be an ultrathin diffusion barrier, while the microchannels could support high-density cell loading by acting as separate chambers allowing uniform distribution of cells in the scaffold. Alginate hydrogel could permeate into the channels and form a sealing layer after gelation, which could slow down the invasion of host immune cells into the scaffold. The hybrid thin-sheet encapsulation system (∼400 μm thick) could protect allogeneic cells over half-year after intraperitoneal (IP) implantation in immune-competent mice. Such structural membranes and plastic-hydrogel hybrids of thin dimensions could find important applications in cell delivery therapy.
Collapse
Affiliation(s)
- Yi Wang
- Department of Biomedical Engineering, College of Future Technology, Peking University, Haidian District, Beijing 100871, China
| | - Kai Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China
| | - Ying Luo
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Haifeng Chen
- Department of Biomedical Engineering, College of Future Technology, Peking University, Haidian District, Beijing 100871, China
| |
Collapse
|
94
|
Parker JB, Griffin MF, Spielman AF, Wan DC, Longaker MT. Exploring the Overlooked Roles and Mechanisms of Fibroblasts in the Foreign Body Response. Adv Wound Care (New Rochelle) 2023; 12:85-96. [PMID: 35819293 PMCID: PMC10081717 DOI: 10.1089/wound.2022.0066] [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: 04/06/2022] [Accepted: 07/06/2022] [Indexed: 11/12/2022] Open
Abstract
Significance: Foreign body response (FBR), wherein a fibrotic capsule forms around an implanted structure, is a common surgical complication that often leads to pain, discomfort, and eventual revision surgeries. Although believed to have some mechanistic overlap with normal wound healing, much remains to be discovered about the specific mechanism by which this occurs. Recent Advances: Current understanding of FBR has focused on the roles of the immune system and the biomaterial, both major contributors to FBR. However, another key player, the fibroblast, is often overlooked. This review summarizes key contributors of FBR, focusing on the roles of fibroblasts. As much remains to be discovered about fibroblasts' specific roles in FBR, we draw on current knowledge of fibroblast subpopulations and functions during wound healing. We also provide an overview on candidate biomaterials and signaling pathways involved in FBR. Critical Issues and Future Directions: While the global implantable medical devices market is considerable and continues to appreciate in value, FBR remains one of the most common surgical implant complications. In parallel with the continued development of candidate biomaterials, further exploration of potential fibroblast subpopulations at a transcriptional level would provide key insights into further understanding the underlying mechanisms by which fibrous encapsulation occurs, and unveil novel directions for antifibrotic and regenerative therapies in the future.
Collapse
Affiliation(s)
- Jennifer B. Parker
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford, California, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Michelle F. Griffin
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford, California, USA
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Amanda F. Spielman
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford, California, USA
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Derrick C. Wan
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford, California, USA
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Michael T. Longaker
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford, California, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
95
|
Geng Z, Zhang Q, Li T, Huang T, Wang H, Zhou Q, Deng S, Zhao Y, Li Y, Cheng C, Gonelle-Gispert C, Buhler LH, Wang Y. Advantages of the retroperitoneal retrocolic space as the transplant site for encapsulated xenogeneic islets. Xenotransplantation 2023; 30:e12787. [PMID: 36454040 DOI: 10.1111/xen.12787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 10/14/2022] [Accepted: 10/18/2022] [Indexed: 12/03/2022]
Abstract
OBJECTIVE Islet allotransplantation has demonstrated improved clinical outcomes using the hepatic portal vein as the standard infusion method. However, the current implantation site is not ideal due to the short-term thrombotic and long-term immune destruction. Meanwhile, the shortage of human organ donors further limits its application. To find a new strategy, we tested a new polymer combination for islet encapsulation and transplantation. Meanwhile, we explored a new site for xenogeneic islet transplantation in mice. METHOD We synthesized a hydrogel combining alginate plus poly-ethylene-imine (Alg/PEI) for the encapsulation of rat, neonatal porcine, and human islets. Transplantation was performed into the retroperitoneal retro-colic space of diabetic mice. Control mice received free islets under the kidney capsule or encapsulated islets into the peritoneum. The biochemical indexes were measured, and the transplanted islets were harvested for immunohistochemical staining of insulin and glucagon. RESULTS Mice receiving encapsulated rat, porcine and human islets transplanted into the retroperitoneal space maintained normoglycemia for a median of 275, 145.5, and 146 days, respectively. In contrast, encapsulated xenogeneic islets transplanted into the peritoneum, maintained function for a median of 61, 95.5, and 82 days, respectively. Meanwhile, xenogeneic islets transplanted free into the kidney capsule lost their function within 3 days after transplantation. Immunohistochemical staining of encapsulated rat, porcine and human islets, retrieved from the retroperitoneal space, allowed to distinguish morphological normal insulin expressing β- and glucagon expressing α-cells at 70, 60, and 100 days post-transplant, respectively. CONCLUSION Transplantation of Alg/PEI encapsulated xenogeneic islets into the retroperitoneal space provides a valuable new implantation strategy for the treatment of type 1 diabetes.
Collapse
Affiliation(s)
- Zhen Geng
- Health Management Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Qi Zhang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Ting Li
- Department of Rheumatology, Wenjiang District People's Hospital, Chengdu, China
| | - Ting Huang
- Department of Breast Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Hailian Wang
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Institute of Organ Transplantation, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Qiao Zhou
- Department of Rheumatology and Immunology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Shaoping Deng
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Institute of Organ Transplantation, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yanshuang Zhao
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yanjiao Li
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Chunming Cheng
- Department of Radiation Oncology, James Comprehensive Cancer Center and College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | | | - Leo H Buhler
- Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Yi Wang
- Department of Critical Care Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 610072, China
| |
Collapse
|
96
|
Volpatti LR, Bochenek MA, Facklam AA, Burns DM, MacIsaac C, Morgart A, Walters B, Langer R, Anderson DG. Partially Oxidized Alginate as a Biodegradable Carrier for Glucose-Responsive Insulin Delivery and Islet Cell Replacement Therapy. Adv Healthc Mater 2023; 12:e2201822. [PMID: 36325648 PMCID: PMC9840661 DOI: 10.1002/adhm.202201822] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 10/18/2022] [Indexed: 11/06/2022]
Abstract
Self-regulated insulin delivery that mimics native pancreas function has been a long-term goal for diabetes therapies. Two approaches towards this goal are glucose-responsive insulin delivery and islet cell transplantation therapy. Here, biodegradable, partially oxidized alginate carriers for glucose-responsive nanoparticles or islet cells are developed. Material composition and formulation are tuned in each of these contexts to enable glycemic control in diabetic mice. For injectable, glucose-responsive insulin delivery, 0.5 mm 2.5% oxidized alginate microgels facilitate repeat dosing and consistently provide 10 days of glycemic control. For islet cell transplantation, 1.5 mm capsules comprised of a blend of unoxidized and 2.5% oxidized alginate maintain cell viability and glycemic control over a period of more than 2 months while reducing the volume of nondegradable material implanted. These data show the potential of these biodegradable carriers for controlled drug and cell delivery for the treatment of diabetes with limited material accumulation in the event of multiple doses.
Collapse
Affiliation(s)
- Lisa R. Volpatti
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Matthew A. Bochenek
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Amanda A. Facklam
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Delaney M. Burns
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Corina MacIsaac
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Harvard–Massachusetts Institute of Technology Division of Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Alexander Morgart
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Benjamin Walters
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Robert Langer
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Harvard–Massachusetts Institute of Technology Division of Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Daniel G. Anderson
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, Boston, MA 02115, USA
- Harvard–Massachusetts Institute of Technology Division of Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
97
|
Engineering Strategies of Islet Product for Endocrine Regeneration. ENGINEERED REGENERATION 2023. [DOI: 10.1016/j.engreg.2023.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
|
98
|
Khazaei M, Khazaei F, Niromand E, Ghanbari E. Tissue engineering approaches and generation of insulin-producing cells to treat type 1 diabetes. J Drug Target 2023; 31:14-31. [PMID: 35896313 DOI: 10.1080/1061186x.2022.2107653] [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: 01/05/2023]
Abstract
Tissue engineering (TE) has become a new effective solution to a variety of medical problems, including diabetes. Mesenchymal stem cells (MSCs), which have the ability to differentiate into endodermal and mesodermal cells, appear to be appropriate for this function. The purpose of this review was to evaluate the outcomes of various researches on the insulin-producing cells (IPCs) generation from MSCs with TE approaches to increase efficacy of type 1 diabetes treatments. The search was performed in PubMed/Medline, Scopus and Embase databases until 2021. Studies revealed that MSCs could also differentiate into IPCs under certain conditions. Therefore, a wide range of protocols have been used for this differentiation, but their effectiveness is very different. Scaffolds can provide a microenvironment that enhances the MSCs to IPCs differentiation, improves their metabolic activity and up-regulate pancreatic-specific transcription factors. They also preserve IPCs architecture and enhance insulin production as well as protect against cell death. This systematic review offers a framework for prospective research based on data. In vitro and in vivo evidence suggests that scaffold-based TE can improve the viability and function of IPCs.
Collapse
Affiliation(s)
- Mozafar Khazaei
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Department of Tissue Engineering, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Fatemeh Khazaei
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Elham Niromand
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Elham Ghanbari
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Department of Tissue Engineering, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
99
|
Salthouse D, Novakovic K, Hilkens CMU, Ferreira AM. Interplay between biomaterials and the immune system: Challenges and opportunities in regenerative medicine. Acta Biomater 2023; 155:1-18. [PMID: 36356914 DOI: 10.1016/j.actbio.2022.11.003] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 10/17/2022] [Accepted: 11/01/2022] [Indexed: 11/09/2022]
Abstract
The use of biomaterials for tissue engineering and regenerative medicine applications has increased dramatically over recent years. However, the clinical uptake of a wide variety of biomaterials remains limited due to adverse effects commonly exhibited by patients, which are caused by the host immune response. Despite this, current in vitro evaluation standards (ISO-10993) for assessing the host response to biomaterials have limitations in predicting the likelihood of in vivo biomaterial acceptance. Furthermore, endotoxin contamination of biomaterials is rarely considered, despite its ability to significantly affect the performance of biomaterials and engineered tissues. This review highlights the importance of the immune response to biomaterials and discusses existing challenges and opportunities in the development and standardised assessment of the immune response to biomaterials, including the importance of endotoxin levels. In addition, the properties of biomaterials that impact the host immune response and the exploitation of immunomodulatory biomaterials in regenerative medicine are explored. Finally, a standardised in vitro pathway of evaluating the immune response to biomaterials (hydrogels) and their regenerative potential is proposed, aiming to ensure safety and consistency, while reducing costs and the use of animals in the biomaterials research for tissue engineering and regenerative medicine. STATEMENT OF SIGNIFICANCE: This review presents a critical analysis of the role of the interactions between the immune system and biomaterials in determining the therapeutic success of biomaterial-based approaches. No such review addressing the lack of understanding of biomaterial-immune system interactions during the developmental and pre-clinical stages of biomaterials, including the impact of the endotoxin levels of biomaterials on the immune response, is published. As there is a lack of in vitro regulations to evaluate the immune response to biomaterials, a standardised in vitro pathway to evaluate the immune response to biomaterials (hydrogels) and their immunomodulatory and regenerative potential for use in tissue engineering/regenerative medicine applications is presented. The aim of the proposed pathway of biomaterial evaluation is to ensure safety and consistency in the biomaterials research community, while reducing costs and animal use (through the concept of the 3R's - reduction, refinement, and replacement of animals).
Collapse
Affiliation(s)
- Daniel Salthouse
- School of Engineering, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Katarina Novakovic
- School of Engineering, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Catharien M U Hilkens
- Translational & Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Ana Marina Ferreira
- School of Engineering, Newcastle University, Newcastle Upon Tyne, United Kingdom.
| |
Collapse
|
100
|
Parlani M, Bedell ML, Mikos AG, Friedl P, Dondossola E. Dissecting the recruitment and self-organization of αSMA-positive fibroblasts in the foreign body response. SCIENCE ADVANCES 2022; 8:eadd0014. [PMID: 36542704 PMCID: PMC9770965 DOI: 10.1126/sciadv.add0014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 11/05/2022] [Indexed: 06/17/2023]
Abstract
The foreign body response (FBR) is a clinically relevant issue that can cause malfunction of implanted medical devices by fibrotic encapsulation. Whereas inflammatory aspects of the FBR have been established, underlying fibroblast-dependent mechanisms remain unclear. We here combine multiphoton microscopy with ad hoc reporter mice expressing α-smooth muscle actin (αSMA) protein to determine the locoregional fibroblast dynamics, activation, and fibrotic encapsulation of polymeric materials. Fibroblasts invaded as individual cells and established a multicellular network, which transited to a two-compartment fibrotic response displaying an αSMA cold external capsule and a long-lasting, inner αSMA hot environment. The recruitment of fibroblasts and extent of fibrosis were only incompletely inhibited after depletion of macrophages, implicating coexistence of macrophage-dependent and macrophage-independent mediators. Furthermore, neither altering material type or porosity modulated αSMA+ cell recruitment and distribution. This identifies fibroblast activation and network formation toward a two-compartment FBR as a conserved, self-organizing process partially independent of macrophages.
Collapse
Affiliation(s)
- Maria Parlani
- Department of Genitourinary Medical Oncology and David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Radboud University Medical Center, Nijmegen, Netherlands
| | - Matthew L. Bedell
- Department of Bioengineering, Rice University, Houston, TX 77030, USA
| | - Antonios G. Mikos
- Department of Bioengineering, Rice University, Houston, TX 77030, USA
| | - Peter Friedl
- Department of Genitourinary Medical Oncology and David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Radboud University Medical Center, Nijmegen, Netherlands
- Cancer Genomics Centre (CGC.nl), 3584 Utrecht, Netherlands
| | - Eleonora Dondossola
- Department of Genitourinary Medical Oncology and David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|