151
|
Rezaei M, Davani F, Alishahi M, Masjedi F. Updates in immunocompatibility of biomaterials: applications for regenerative medicine. Expert Rev Med Devices 2022; 19:353-367. [PMID: 35531761 DOI: 10.1080/17434440.2022.2075730] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Biomaterials, either metallic, ceramic, or polymeric, can be used in medicine as a part of the implants, dialysis membranes, bone scaffolds, or components of artificial organs. Polymeric biomaterials cover a vast range of biomedical applications. The biocompatibility and immunocompatibility of polymeric materials are of fundamental importance for their possible therapeutic uses, as the immune system can intervene in the materials' performance. Therefore, based on application, different routes can be utilized for immunoregulation. AREAS COVERED As different biomaterials can be modulated by different strategies, this study aims to summarize and evaluate the available methods for the immunocompatibility enhancement of more common polymeric biomaterials based on their nature. Different strategies such as surface modification, physical characterization, and drug incorporation are investigated for the immunomodulation of nanoparticles, hydrogels, sponges, and nanofibers. EXPERT OPINION Recently, strategies for triggering appropriate immune responses by functional biomaterials have been highlighted. As most strategies correspond to the physical and surface properties of biomaterials, specific modulation can be conducted for each biomaterial system. Besides, different applications require different modulations of the immune system. In the future, the selection of novel materials and immune regulators can play a role in tuning the immune system for regenerative medicine.
Collapse
Affiliation(s)
- Mahdi Rezaei
- Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Farideh Davani
- Burn and Wound Healing Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohsen Alishahi
- Burn and Wound Healing Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Masjedi
- Shiraz Nephro-Urology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
152
|
Qazi TH, Blatchley MR, Davidson MD, Yavitt FM, Cooke ME, Anseth KS, Burdick JA. Programming hydrogels to probe spatiotemporal cell biology. Cell Stem Cell 2022; 29:678-691. [PMID: 35413278 PMCID: PMC9081204 DOI: 10.1016/j.stem.2022.03.013] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The recapitulation of complex microenvironments that regulate cell behavior during development, disease, and wound healing is key to understanding fundamental biological processes. In vitro, multicellular morphogenesis, organoid maturation, and disease modeling have traditionally been studied using either non-physiological 2D substrates or 3D biological matrices, neither of which replicate the spatiotemporal biochemical and biophysical complexity of biology. Here, we provide a guided overview of the recent advances in the programming of synthetic hydrogels that offer precise control over the spatiotemporal properties within cellular microenvironments, such as advances in the control of cell-driven remodeling, bioprinting, or user-defined manipulation of properties (e.g., via light irradiation).
Collapse
Affiliation(s)
- Taimoor H Qazi
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael R Blatchley
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA; Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80303, USA
| | - Matthew D Davidson
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA; BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA; Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80303, USA
| | - F Max Yavitt
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA; Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80303, USA
| | - Megan E Cooke
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA; Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80303, USA
| | - Kristi S Anseth
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA; Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80303, USA
| | - Jason A Burdick
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA; BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA; Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80303, USA.
| |
Collapse
|
153
|
Zarubova J, Hasani-Sadrabadi MM, Ardehali R, Li S. Immunoengineering strategies to enhance vascularization and tissue regeneration. Adv Drug Deliv Rev 2022; 184:114233. [PMID: 35304171 PMCID: PMC10726003 DOI: 10.1016/j.addr.2022.114233] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 03/07/2022] [Accepted: 03/11/2022] [Indexed: 12/11/2022]
Abstract
Immune cells have emerged as powerful regulators of regenerative as well as pathological processes. The vast majority of regenerative immunoengineering efforts have focused on macrophages; however, growing evidence suggests that other cells of both the innate and adaptive immune system are as important for successful revascularization and tissue repair. Moreover, spatiotemporal regulation of immune cells and their signaling have a significant impact on the regeneration speed and the extent of functional recovery. In this review, we summarize the contribution of different types of immune cells to the healing process and discuss ways to manipulate and control immune cells in favor of vascularization and tissue regeneration. In addition to cell delivery and cell-free therapies using extracellular vesicles, we discuss in situ strategies and engineering approaches to attract specific types of immune cells and modulate their phenotypes. This field is making advances to uncover the extraordinary potential of immune cells and their secretome in the regulation of vascularization and tissue remodeling. Understanding the principles of immunoregulation will help us design advanced immunoengineering platforms to harness their power for tissue regeneration.
Collapse
Affiliation(s)
- Jana Zarubova
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA; Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Prague 14220, Czech Republic
| | | | - Reza Ardehali
- Division of Cardiology, Department of Internal Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; Eli and Edythe Broad Stem Cell Research Center, University of California, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Song Li
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA; Eli and Edythe Broad Stem Cell Research Center, University of California, Los Angeles, CA 90095, USA; Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA.
| |
Collapse
|
154
|
Brown BN, Chung WL, Lowe J, LoPresti ST, Cheetham J, Almarza AJ, Badylak SF. Inductive Remodeling of Extracellular Matrix Scaffolds in the Temporomandibular Joint of Pigs. Tissue Eng Part A 2022; 28:447-457. [PMID: 34809494 PMCID: PMC9131358 DOI: 10.1089/ten.tea.2021.0123] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 11/04/2021] [Indexed: 11/12/2022] Open
Abstract
The temporomandibular joint (TMJ) disc is a fibrocartilaginous tissue located between the condyle of the mandible and glenoid fossa and articular eminence of the temporal bone. Damage or derangement of the TMJ disc can require surgical removal (discectomy) to restore function. Removal of the TMJ disc, however, leaves the joint space vulnerable to condylar remodeling and degradation, potentially leading to long-term complications. No consistently effective clinical option exists for repair or replacement of the disc following discectomy. This study investigates the use of an acellular scaffold composed of extracellular matrix (ECM) derived from small intestinal submucosa (SIS) as a regenerative template for the TMJ disc in a porcine model. Acellular SIS ECM scaffolds were implanted following discectomy and allowed to remodel for 2, 4, 12, and 24 weeks postimplantation. Remodeling of the implanted device was assessed by longitudinal magnetic resonance imaging (MRI) over the course of 6 months, as well as gross morphologic, histologic, biochemical, and biomechanical analysis (tension and compression) of explanted tissues (disc and condyle) at the time of sacrifice. When the scaffold remained in the joint space, longitudinal MRI demonstrated that the scaffolds promoted new tissue formation within the joint space throughout the study period. The scaffolds were rapidly populated with host-derived cells and remodeled with formation of new, dense, aligned fibrocartilage resembling native tissue as early as 1 month postimplantation. De-novo formation of peripheral muscular and tendinous attachments resembling those in native tissue was also observed. The remodeled scaffolds approached native disc biochemical composition and compressive modulus, and possessed 50% of the tensile modulus within 3 months postimplantation. No degradation of the condylar surface was observed. These results suggest that this acellular bioscaffold fills a medical need for which there is currently no effective treatment and may represent a clinically relevant "off-the-shelf" implant for reconstruction of the TMJ disc.
Collapse
Affiliation(s)
- Bryan N. Brown
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - William L. Chung
- Oral and Maxillofacial Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jesse Lowe
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Samuel T. LoPresti
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jonathan Cheetham
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Clinical Sciences, Cornell University College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Alejandro J. Almarza
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Oral and Craniofacial Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Center for Craniofacial Regeneration, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Stephen F. Badylak
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
155
|
Garzón H, Suárez LJ, Muñoz S, Cardona J, Fontalvo M, Alfonso-Rodríguez CA. Biomaterials Used for Periodontal Disease Treatment: Focusing on Immunomodulatory Properties. Int J Biomater 2022; 2022:7693793. [PMID: 35528847 PMCID: PMC9072036 DOI: 10.1155/2022/7693793] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/23/2022] [Accepted: 03/05/2022] [Indexed: 12/25/2022] Open
Abstract
The growing use of biomaterials with different therapeutic purposes increases the need for their physiological understanding as well as to seek its integration with the human body. Chronic inflammatory local pathologies, generally associated with infectious or autoimmunity processes, have been a current therapeutic target due to the difficulty in their treatment. The recent development of biomaterials with immunomodulatory capacity would then become one of the possible strategies for their management in local pathologies, by intervening in situ, without generating alterations in the systemic immune response. The treatment of periodontal disease as an inflammatory entity has involved the use of different approaches and biomaterials. There is no conclusive, high evidence about the use of these biomaterials in the regeneration of periodontitis sequelae, so the profession keeps looking for other different strategies. The use of biomaterials with immunomodulatory properties could be one, with a promising future. This review of the literature summarizes the scientific evidence about biomaterials used in the treatment of periodontal disease.
Collapse
Affiliation(s)
- H. Garzón
- Grupo de Investigación en Salud Oral, Departamento de Periodoncia, Universidad Antonio Nariño, Bogotá, Colombia
| | - L. J. Suárez
- Departamento de Ciencias Básicas y Medicina Oral, Universidad Nacional de Colombia, Bogotá, Colombia
| | - S. Muñoz
- Grupo de Investigación en Salud Oral, Departamento de Periodoncia, Universidad Antonio Nariño, Bogotá, Colombia
| | - J. Cardona
- Grupo de Investigación en Salud Oral, Departamento de Periodoncia, Universidad Antonio Nariño, Bogotá, Colombia
| | - M. Fontalvo
- Grupo de Investigación en Salud Oral, Departamento de Periodoncia, Universidad Antonio Nariño, Bogotá, Colombia
| | - C. A. Alfonso-Rodríguez
- Grupo de Investigación en Salud Oral, Departamento de Periodoncia, Universidad Antonio Nariño, Bogotá, Colombia
| |
Collapse
|
156
|
Patil P, Russo KA, McCune JT, Pollins AC, Cottam MA, Dollinger BR, DeJulius CR, Gupta MK, D'Arcy R, Colazo JM, Yu F, Bezold MG, Martin JR, Cardwell NL, Davidson JM, Thompson CM, Barbul A, Hasty AH, Guelcher SA, Duvall CL. Reactive oxygen species-degradable polythioketal urethane foam dressings to promote porcine skin wound repair. Sci Transl Med 2022; 14:eabm6586. [PMID: 35442705 PMCID: PMC10165619 DOI: 10.1126/scitranslmed.abm6586] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Porous, resorbable biomaterials can serve as temporary scaffolds that support cell infiltration, tissue formation, and remodeling of nonhealing skin wounds. Synthetic biomaterials are less expensive to manufacture than biologic dressings and can achieve a broader range of physiochemical properties, but opportunities remain to tailor these materials for ideal host immune and regenerative responses. Polyesters are a well-established class of synthetic biomaterials; however, acidic degradation products released by their hydrolysis can cause poorly controlled autocatalytic degradation. Here, we systemically explored reactive oxygen species (ROS)-degradable polythioketal (PTK) urethane (UR) foams with varied hydrophilicity for skin wound healing. The most hydrophilic PTK-UR variant, with seven ethylene glycol (EG7) repeats flanking each side of a thioketal bond, exhibited the highest ROS reactivity and promoted optimal tissue infiltration, extracellular matrix (ECM) deposition, and reepithelialization in porcine skin wounds. EG7 induced lower foreign body response, greater recruitment of regenerative immune cell populations, and resolution of type 1 inflammation compared to more hydrophobic PTK-UR scaffolds. Porcine wounds treated with EG7 PTK-UR foams had greater ECM production, vascularization, and resolution of proinflammatory immune cells compared to polyester UR foam-based NovoSorb Biodegradable Temporizing Matrix (BTM)-treated wounds and greater early vascular perfusion and similar wound resurfacing relative to clinical gold standard Integra Bilayer Wound Matrix (BWM). In a porcine ischemic flap excisional wound model, EG7 PTK-UR treatment led to higher wound healing scores driven by lower inflammation and higher reepithelialization compared to NovoSorb BTM. PTK-UR foams warrant further investigation as synthetic biomaterials for wound healing applications.
Collapse
Affiliation(s)
- Prarthana Patil
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Katherine A Russo
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Joshua T McCune
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Alonda C Pollins
- Department of Plastic Surgery, Vanderbilt University Medical Center, Nashville, TN 37212, USA
| | - Matthew A Cottam
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Bryan R Dollinger
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Carlisle R DeJulius
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Mukesh K Gupta
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Richard D'Arcy
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Juan M Colazo
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Fang Yu
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Mariah G Bezold
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - John R Martin
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Nancy L Cardwell
- Department of Plastic Surgery, Vanderbilt University Medical Center, Nashville, TN 37212, USA
| | - Jeffrey M Davidson
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Callie M Thompson
- Vanderbilt Burn Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Adrian Barbul
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN 37212, USA.,Department of Surgery, Veterans Administration Medical Center, Nashville, TN 37212, USA
| | - Alyssa H Hasty
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.,Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN 37212, USA
| | - Scott A Guelcher
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA.,Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Craig L Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| |
Collapse
|
157
|
von Mentzer U, Corciulo C, Stubelius A. Biomaterial Integration in the Joint: Pathological Considerations, Immunomodulation, and the Extracellular Matrix. Macromol Biosci 2022; 22:e2200037. [PMID: 35420256 DOI: 10.1002/mabi.202200037] [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: 02/27/2022] [Revised: 03/30/2022] [Indexed: 11/08/2022]
Abstract
Defects of articular joints are becoming an increasing societal burden due to a persistent increase in obesity and aging. For some patients suffering from cartilage erosion, joint replacement is the final option to regain proper motion and limit pain. Extensive research has been undertaken to identify novel strategies enabling earlier intervention to promote regeneration and cartilage healing. With the introduction of decellularized extracellular matrix (dECM), researchers have tapped into the potential for increased tissue regeneration by designing biomaterials with inherent biochemical and immunomodulatory signals. Compared to conventional and synthetic materials, dECM-based materials invoke a reduced foreign body response. It is therefore highly beneficial to understand the interplay of how these native tissue-based materials initiate a favorable remodeling process by the immune system. Yet, such an understanding also demands increasing considerations of the pathological environment and remodeling processes, especially for materials designed for early disease intervention. This knowledge would avoid rejection and help predict complications in conditions with inflammatory components such as arthritides. This review outlines general issues facing biomaterial integration and emphasizes the importance of tissue-derived macromolecular components in regulating essential homeostatic, immunological, and pathological processes to increase biomaterial integration for patients suffering from joint degenerative diseases. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Ula von Mentzer
- Division of Chemical Biology, Department of Biology and Biological Engineering, Chalmers University of Technology, Kemivägen 10, Gothenburg, 41296, Sweden
| | - Carmen Corciulo
- Centre for Bone and Arthritis Research, Department of Rheumatology and Inflammation, Sahlgrenska Academy at the University of Gothenburg, Guldhedsgatan 10A, Gothenburg, 41296, Sweden
| | - Alexandra Stubelius
- Division of Chemical Biology, Department of Biology and Biological Engineering, Chalmers University of Technology, Kemivägen 10, Gothenburg, 41296, Sweden
| |
Collapse
|
158
|
Tang W, Qi J, Wang Q, Qu Y, Fu S, Luan J. Investigating the Adipogenic Effects of Different Tissue-Derived Decellularized Matrices. Front Bioeng Biotechnol 2022; 10:872897. [PMID: 35497363 PMCID: PMC9046558 DOI: 10.3389/fbioe.2022.872897] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 03/29/2022] [Indexed: 11/13/2022] Open
Abstract
Objective: Decellularized adipose-derived matrix (DAM) can promote adipogenic differentiation and adipose tissue remodeling, but the biological impact of tissue origin on DAM remains unknown. The present study aimed to investigate the effects of tissue origins on the adipogenic capacity of the decellularized matrix by comparing the cellular and tissue responses of DAM versus acellular dermal matrix (ADM). Methods: The in vitro response of adipose-derived stem/stromal cells (ADSCs) to DAM and ADM was characterized by proliferation and differentiation. The in vivo remodeling response was evaluated in the subcutaneous injection model of immunocompromised mice, using histology, protein expression, and transcriptome analysis. Results: Both DAM and ADM exhibited excellent decellularization effects and cytocompatibility. In the absence of exogenous stimuli, DAM could induce adipogenic differentiation of ADSCs compared with ADM. In the animal model, the levels of PDGF, VEGF, and ACRP30 were higher in the DAM groups than in the ADM group, and more neovascularization and extensive adipose tissue remodeling were observed. The mRNA-seq analysis indicated that the DAM implant regulated tissue remodeling by modulating Lat1/2 expression along with Hippo Signaling pathway in the early stage. Conclusion: Tissue origin can influence the biological response of the decellularized matrix. DAM can retain favorable tissue-specific characteristics after the decellularization process and have unique adipogenic effects in vitro and vivo, which can be fully utilized for soft tissue repair and regeneration.
Collapse
Affiliation(s)
| | | | | | | | - Su Fu
- *Correspondence: Su Fu, ; Jie Luan,
| | - Jie Luan
- *Correspondence: Su Fu, ; Jie Luan,
| |
Collapse
|
159
|
Larouche JA, Fraczek PM, Kurpiers SJ, Yang BA, Davis C, Castor-Macias JA, Sabin K, Anderson S, Harrer J, Hall M, Brooks SV, Jang YC, Willett N, Shea LD, Aguilar CA. Neutrophil and natural killer cell imbalances prevent muscle stem cell-mediated regeneration following murine volumetric muscle loss. Proc Natl Acad Sci U S A 2022; 119:e2111445119. [PMID: 35377804 PMCID: PMC9169656 DOI: 10.1073/pnas.2111445119] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 02/09/2022] [Indexed: 02/06/2023] Open
Abstract
Volumetric muscle loss (VML) overwhelms the innate regenerative capacity of mammalian skeletal muscle (SkM), leading to numerous disabilities and reduced quality of life. Immune cells are critical responders to muscle injury and guide tissue resident stem cell– and progenitor-mediated myogenic repair. However, how immune cell infiltration and intercellular communication networks with muscle stem cells are altered following VML and drive pathological outcomes remains underexplored. Herein, we contrast the cellular and molecular mechanisms of VML injuries that result in the fibrotic degeneration or regeneration of SkM. Following degenerative VML injuries, we observed the heightened infiltration of natural killer (NK) cells as well as the persistence of neutrophils beyond 2 wk postinjury. Functional validation of NK cells revealed an antagonistic role in neutrophil accumulation in part via inducing apoptosis and CCR1-mediated chemotaxis. The persistent infiltration of neutrophils in degenerative VML injuries was found to contribute to impairments in muscle stem cell regenerative function, which was also attenuated by transforming growth factor beta 1 (TGFβ1). Blocking TGFβ signaling reduced neutrophil accumulation and fibrosis and improved muscle-specific force. Collectively, these results enhance our understanding of immune cell–stem cell cross talk that drives regenerative dysfunction and provide further insight into possible avenues for fibrotic therapy exploration.
Collapse
Affiliation(s)
- Jacqueline A. Larouche
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109
| | - Paula M. Fraczek
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109
| | - Sarah J. Kurpiers
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109
| | - Benjamin A. Yang
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109
| | - Carol Davis
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109
| | - Jesus A. Castor-Macias
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109
| | - Kaitlyn Sabin
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109
| | - Shannon Anderson
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332
| | - Julia Harrer
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332
| | - Matthew Hall
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109
| | - Susan V. Brooks
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109
| | - Young C. Jang
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332
| | - Nick Willett
- Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR 97403
| | - Lonnie D. Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109
| | - Carlos A. Aguilar
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109
- Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI 48109
| |
Collapse
|
160
|
Kim Y, Jung HJ, Lee Y, Koo S, Thangam R, Jang WY, Kim SY, Park S, Lee S, Bae G, Patel KD, Wei Q, Lee KB, Paulmurugan R, Jeong WK, Hyeon T, Kim D, Kang H. Manipulating Nanoparticle Aggregates Regulates Receptor-Ligand Binding in Macrophages. J Am Chem Soc 2022; 144:5769-5783. [PMID: 35275625 DOI: 10.1021/jacs.1c08861] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The receptor-ligand interactions in cells are dynamically regulated by modulation of the ligand accessibility. In this study, we utilize size-tunable magnetic nanoparticle aggregates ordered at both nanometer and atomic scales. We flexibly anchor magnetic nanoparticle aggregates of tunable sizes over the cell-adhesive RGD ligand (Arg-Gly-Asp)-active material surface while maintaining the density of dispersed ligands accessible to macrophages at constant. Lowering the accessible ligand dispersity by increasing the aggregate size at constant accessible ligand density facilitates the binding of integrin receptors to the accessible ligands, which promotes the adhesion of macrophages. In high ligand dispersity, distant magnetic manipulation to lift the aggregates (which increases ligand accessibility) stimulates the binding of integrin receptors to the accessible ligands available under the aggregates to augment macrophage adhesion-mediated pro-healing polarization both in vitro and in vivo. In low ligand dispersity, distant control to drop the aggregates (which decreases ligand accessibility) repels integrin receptors away from the aggregates, thereby suppressing integrin receptor-ligand binding and macrophage adhesion, which promotes inflammatory polarization. Here, we present "accessible ligand dispersity" as a novel fundamental parameter that regulates receptor-ligand binding, which can be reversibly manipulated by increasing and decreasing the ligand accessibility. Limitless tuning of nanoparticle aggregate dimensions and morphology can offer further insight into the regulation of receptor-ligand binding in host cells.
Collapse
Affiliation(s)
- Yuri Kim
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Hee Joon Jung
- Department of Materials Science and Engineering, Northwestern University, Evanston, Illinois 60208, United States
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois 60208, United States
- NUANCE Center, Northwestern University, Evanston, Illinois 60208, United States
| | - Yunjung Lee
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Sagang Koo
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Ramar Thangam
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Woo Young Jang
- Department of Orthopedic Surgery, Korea University Anam Hospital, Seoul 02841, Republic of Korea
| | - Seong Yeol Kim
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Sangwoo Park
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Sungkyu Lee
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Gunhyu Bae
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Kapil Dev Patel
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Qiang Wei
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu 610065, China
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, New Jersey 08854, United States
| | - Ramasamy Paulmurugan
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford University, Palo Alto, California 94304, United States
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University School of Medicine, Stanford University, Palo Alto, California 94304, United States
| | - Woong Kyo Jeong
- Department of Orthopedic Surgery, Korea University Anam Hospital, Seoul 02841, Republic of Korea
| | - Taeghwan Hyeon
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Dokyoon Kim
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
- Department of Bionano Engineering and Bionanotechnology, Hanyang University, Ansan 15588, Republic of Korea
| | - Heemin Kang
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
- Department of Biomicrosystem Technology, Korea University, Seoul 02841, Republic of Korea
| |
Collapse
|
161
|
Wei Z, Li F, Pi G. Association Between Gut Microbiota and Osteoarthritis: A Review of Evidence for Potential Mechanisms and Therapeutics. Front Cell Infect Microbiol 2022; 12:812596. [PMID: 35372125 PMCID: PMC8966131 DOI: 10.3389/fcimb.2022.812596] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 02/24/2022] [Indexed: 12/12/2022] Open
Abstract
Osteoarthritis (OA) is a multifactorial joint disease characterized by degeneration of articular cartilage, which leads to joints pain, disability and reduced quality of life in patients with OA. Interpreting the potential mechanisms underlying OA pathogenesis is crucial to the development of new disease modifying treatments. Although multiple factors contribute to the initiation and progression of OA, gut microbiota has gradually been regarded as an important pathogenic factor in the development of OA. Gut microbiota can be regarded as a multifunctional “organ”, closely related to a series of immune, metabolic and neurological functions. This review summarized research evidences supporting the correlation between gut microbiota and OA, and interpreted the potential mechanisms underlying the correlation from four aspects: immune system, metabolism, gut-brain axis and gut microbiota modulation. Future research should focus on whether there are specific gut microbiota composition or even specific pathogens and the corresponding signaling pathways that contribute to the initiation and progression of OA, and validate the potential of targeting gut microbiota for the treatment of patients with OA.
Collapse
Affiliation(s)
| | - Feng Li
- *Correspondence: Feng Li, ; Guofu Pi,
| | - Guofu Pi
- *Correspondence: Feng Li, ; Guofu Pi,
| |
Collapse
|
162
|
Li R, Wan L, Zhang X, Liu W, Rong M, Li X, Lu H. Effect of a neodymium-doped yttrium aluminium garnet laser on the physicochemical properties of contaminated titanium surfaces and macrophage polarization. J Periodontal Res 2022; 57:533-544. [PMID: 35266182 DOI: 10.1111/jre.12982] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 12/10/2021] [Accepted: 01/25/2022] [Indexed: 11/28/2022]
Abstract
AIM(S) The objective of this study was to evaluate the changes in the physical and chemical properties of titanium surfaces contaminated by a Nd:YAG laser with different levels of energy and the regulation of macrophage polarization. MATERIALS AND METHODS The titanium specimens were divided into four groups. The blank control group consisted of the above-mentioned contaminated titanium specimens, and the conditioned control group consisted of sandblasted and acid-etched (SLA) titanium surfaces. The blank control and condition control groups were sealed and preserved in a sterile dark box. There were two experimental groups treated with the Nd:YAG laser-one with 0.5 W and the second with 1.0 W. Surface characteristics were evaluated using scanning electron microscopy, surface profilometry, and contact angle assays. The macrophage viability and proliferation of mouse RAW246.7 were analysed, and the macrophage surface markers, macrophage cytokines, and inflammatory and anti-inflammatory genes were expressed. RESULTS The Nd:YAG laser increased the hydrophilicity and roughness of the titanium surface after decontamination. Fewer RAW264.7 cells were observed on the titanium surface after Nd:YAG decontamination than on the contaminated titanium surface expressing the M1-type macrophage marker CCR7, whereas more cells were observed after decontamination than on the contaminated titanium surface expressing the M2-type macrophage marker CD206. Following Nd:YAG laser treatment, the secretion of the inflammatory cytokines IL-1β and TNF-α by RAW264.7 cells on the titanium surface was decreased, whereas the secretion of the anti-inflammatory cytokines IL-4 and IL-10 was increased. RAW264.7 cells cultured for 3 days on the titanium surface after Nd:YAG decontamination treatment expressed significantly reduced levels of the inflammation-related genes IL-1β, TNF-α, IL-6 and iNOS. The expression of the anti-inflammatory genes Arg-1, IL-4, IL-10 and TGF-β by RAW264.7 cells was significantly up-regulated after 3 days of incubation on the titanium surface after Nd:YAG decontamination treatment. CONCLUSION(S) The Nd:YAG laser increased the hydrophilicity and roughness of the titanium surface after decontamination, and this change inhibited M1-type macrophage polarization and promoted M2-type macrophage polarization.
Collapse
Affiliation(s)
- Ruiqi Li
- Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Lei Wan
- Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Xueyang Zhang
- Stomatological Hospital, Southern Medical University, Guangzhou, China.,Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, China
| | - Wenjing Liu
- Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Mingdeng Rong
- Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoling Li
- Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Haibin Lu
- Stomatological Hospital, Southern Medical University, Guangzhou, China.,Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, China
| |
Collapse
|
163
|
Dolan CP, Motherwell JM, Franco SR, Janakiram NB, Valerio MS, Goldman SM, Dearth CL. Evaluating the potential use of functional fibrosis to facilitate improved outcomes following volumetric muscle loss injury. Acta Biomater 2022; 140:379-388. [PMID: 34843950 DOI: 10.1016/j.actbio.2021.11.032] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 11/04/2021] [Accepted: 11/23/2021] [Indexed: 11/01/2022]
Abstract
Volumetric muscle loss (VML) was defined as the frank loss of skeletal muscle tissue with associated chronic functional deficits. Significant effort has been dedicated to developing approaches for treating VML injuries, most of which have focused on stimulating regeneration of the affected musculature via a variety of approaches (e.g., biomaterials). VML injury induces a prolonged inflammatory response which causes fibrotic tissue deposition and is thought to inhibit de novo myofiber regeneration despite observed improvements in functional outcomes (i.e., functional fibrosis; FF). Recent approaches have sought to attenuate inflammation and/or fibrosis as a means to create a permissive environment for regenerative therapies. However, there are currently no clinically available interventions capable of facilitating full restoration of form and function following VML injury; thus, an unmet clinical need exists for a near-term interventional strategy to treat affected patients. FF could serve as an alternative approach to facilitate improved functional outcomes following VML injuries. We sought to investigate whether intentionally exploiting the concept of FF (i.e., induction of a supraphysiological fibrotic response via the delivery of a polypropylene mesh combined with TGFβ) would enhance the function of the VML affected musculature. We found that FF treatment induces enhanced fibrotic tissue deposition within the VML defect as evidenced by histological and molecular analysis. FF-treated animals exhibit improved in vivo muscle function compared to untreated control animals at 8 weeks post-injury, thus substantiating the concept that FF could serve as an efficacious approach for facilitating improved functional outcomes following VML injury. STATEMENT OF SIGNIFICANCE: VML injuries result in long-term functional impairments and reduced quality of life for affected individuals, namely combat injured US Service members, and no clinical interventions can restore the form and function of the injured limb. Extensive efforts have been aimed at developing therapeutics to address this critical gap; unfortunately, most interventions facilitate only modest regeneration. Interestingly, improved muscle function has been observed in VML studies following treatment with a therapeutic, despite a lack of myogenic tissue formation; a phenomenon termed Functional Fibrosis (FF). Herein we exploited the concept of FF to enhance the function of VML affected musculature. This finding is significant in that the commercially available interventions used to induce FF can be translated into the clinic near-term, thus improving the standard of care for VML injuries.
Collapse
|
164
|
Tajima K, Yagi H, Morisaku T, Nishi K, Kushige H, Kojima H, Higashi H, Kuroda K, Kitago M, Adachi S, Natsume T, Nishimura K, Oya M, Kitagawa Y. An organ-derived extracellular matrix triggers in situ kidney regeneration in a preclinical model. NPJ Regen Med 2022; 7:18. [PMID: 35228532 PMCID: PMC8885654 DOI: 10.1038/s41536-022-00213-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 02/07/2022] [Indexed: 01/20/2023] Open
Abstract
It has not been considered that nephrons regenerate in adult mammals. We present that an organ-derived extracellular matrix in situ induces nephron regeneration in a preclinical model. A porcine kidney-derived extracellular matrix was sutured onto the surface of partial nephrectomy (PN)-treated kidney. Twenty-eight days after implantation, glomeruli, vessels, and renal tubules, characteristic of nephrons, were histologically observed within the matrix. No fibrillogenesis was observed in the matrix nor the matrix-sutured kidney, although this occurred in a PN kidney without the matrix, indicating the structures were newly induced by the matrix. The expression of renal progenitor markers, including Sall1, Six2, and WT-1, within the matrix supported the induction of nephron regeneration by the matrix. Furthermore, active blood flow was observed inside the matrix using computed tomography. The matrix provides structural and functional foundations for the development of cell-free scaffolds with a remarkably low risk of immune rejection and cancerization.
Collapse
|
165
|
Fernandez-Yague MA, Hymel LA, Olingy CE, McClain C, Ogle ME, García JR, Minshew D, Vyshnya S, Lim HS, Qiu P, García AJ, Botchwey EA. Analyzing immune response to engineered hydrogels by hierarchical clustering of inflammatory cell subsets. SCIENCE ADVANCES 2022; 8:eabd8056. [PMID: 35213226 PMCID: PMC8880784 DOI: 10.1126/sciadv.abd8056] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 12/31/2021] [Indexed: 06/14/2023]
Abstract
Understanding the immune response to hydrogel implantation is critical for the design of immunomodulatory biomaterials. To study the progression of inflammation around poly(ethylene glycol) hydrogels presenting Arg-Gly-Asp (RGD) peptides and vascular endothelial growth factor, we used temporal analysis of high-dimensional flow cytometry data paired with intravital imaging, immunohistochemistry, and multiplexed proteomic profiling. RGD-presenting hydrogels created a reparative microenvironment promoting CD206+ cellular infiltration and revascularization in wounded dorsal skin tissue. Unbiased clustering algorithms (SPADE) revealed significant phenotypic transition shifts as a function of the cell-adhesion hydrogel properties. SPADE identified an intermediate macrophage subset functionally regulating in vivo cytokine secretion that was preferentially recruited for RGD-presenting hydrogels, whereas dendritic cell subsets were preferentially recruited to RDG-presenting hydrogels. Last, RGD-presenting hydrogels controlled macrophage functional cytokine secretion to direct polarization and vascularization. Our studies show that unbiased clustering of single-cell data provides unbiased insights into the underlying immune response to engineered materials.
Collapse
Affiliation(s)
- Marc A. Fernandez-Yague
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Lauren A. Hymel
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Claire E. Olingy
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Claire McClain
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Molly E. Ogle
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - José R. García
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Dustin Minshew
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Sofiya Vyshnya
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Hong Seo Lim
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Peng Qiu
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Andrés J. García
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Edward A. Botchwey
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
| |
Collapse
|
166
|
Bu W, Wu Y, Ghaemmaghami AM, Sun H, Mata A. Rational design of hydrogels for immunomodulation. Regen Biomater 2022; 9:rbac009. [PMID: 35668923 PMCID: PMC9160883 DOI: 10.1093/rb/rbac009] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 01/21/2022] [Accepted: 01/30/2022] [Indexed: 11/13/2022] Open
Abstract
Abstract
The immune system protects organisms against endogenous and exogenous harm and plays a key role in tissue development, repair, and regeneration. Traditional immunomodulatory biologics exhibit limitations including degradation by enzymes, short half-life, and lack of targeting ability. Encapsulating or binding these biologics within biomaterials is an effective way to address these problems. Hydrogels are promising immunomodulatory materials because of their prominent biocompatibility, tuneability, and versatility. However, to take advantage of these opportunities and optimize material performance, it is important to more specifically elucidate, and leverage on, how hydrogels affect and control the immune response. Here, we summarize how key physical and chemical properties of hydrogels affect the immune response. We first provide an overview of underlying steps of the host immune response upon exposure to biomaterials. Then, we discuss recent advances in immunomodulatory strategies where hydrogels play a key role through a) physical properties including dimensionality, stiffness, porosity, and topography; b) chemical properties including wettability, electric property, and molecular presentation; and c) the delivery of bioactive molecules via chemical or physical cues. Thus, this review aims to build a conceptual and practical toolkit for the design of immune-instructive hydrogels capable of modulating the host immune response.
Collapse
Affiliation(s)
- Wenhuan Bu
- Liaoning Provincial Key Laboratory of Oral Diseases, School of Stomatology, China Medical University, Shenyang, 110001, China
- Department of Dental Materials, School of Stomatology, China Medical University, Shenyang, 110001, China
- Department of Center Laboratory, School of Stomatology, China Medical University, Shenyang, 110001, China
- School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, UK
- Biodiscovery Institute, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Yuanhao Wu
- School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, UK
- Biodiscovery Institute, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Amir M Ghaemmaghami
- Division of Immunology, School of Life Sciences, University of Nottingham, Nottingham, NG7 2RD, UK
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90024, USA
| | - Hongchen Sun
- Liaoning Provincial Key Laboratory of Oral Diseases, School of Stomatology, China Medical University, Shenyang, 110001, China
| | - Alvaro Mata
- School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, UK
- Biodiscovery Institute, University of Nottingham, Nottingham, NG7 2RD, UK
- Department of Chemical and Environmental Engineering, University of Nottingham, Nottingham, NG7 2RD, UK
| |
Collapse
|
167
|
Chen J, Hou S, Liang Q, He W, Li R, Wang H, Zhu Y, Zhang B, Chen L, Dai X, Zhang T, Ren J, Duan H. Localized Degradation of Neutrophil Extracellular Traps by Photoregulated Enzyme Delivery for Cancer Immunotherapy and Metastasis Suppression. ACS NANO 2022; 16:2585-2597. [PMID: 35080858 DOI: 10.1021/acsnano.1c09318] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Extrusion of neutrophil extracellular traps (NETs), a fundamental host innate immune defense against pathogens, has recently been linked to cancer resistance to immunotherapy and distant metastasis. These findings highlight interesting areas of cancer-elicited inflammation and potential therapeutic strategies. Disrupting existing NETs with DNase I has been proved to enhance the therapeutic efficacy of tumor immunotherapy and attenuate metastatic spread. However, systemic biodistribution of DNase I raises safety issues, potentially impairing host defense against infection. Hence, tumor-specific delivery and metastatic niche-targeted effects are attractive options for localized degradation of NETs. We have engineered a nanoplatform with a plasmonic gold blackbody (AuPB) core with broad-spectrum photo activity and a mesoporous polydopamine (mPDA) shell for efficient loading and photoregulated release of DNase I. The on-demand released DNase I triggered by the second near-infrared (NIR-II) light irradiation breaks the "NET-mediated physical barrier", thereby increasing the contact of immune cytotoxic cells with tumor cells in living mice and sensitizing immune checkpoint therapy of primary colorectal cancer (CRC). Moreover, the deposition and light-controlled cargo release from systemically delivered AuPB@mPDA carriers in liver, the most frequent site of CRC metastasis, abolished NET-mediated capture of circulating tumor cells and hence metastatic seeding. Our findings indicate that the localized, light-regulated release of DNase I by photoactive carriers in the NIR-II window represent a translational route for immune-mediated tumor regression and metastasis inhibition.
Collapse
Affiliation(s)
- Jiayuan Chen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shuai Hou
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 70 Nanyang Drive, Singapore 637457
| | - Qing Liang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Wenshan He
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ruiqi Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Haihong Wang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ying Zhu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Biying Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lingjuan Chen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiaofang Dai
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Tao Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jinghua Ren
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hongwei Duan
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 70 Nanyang Drive, Singapore 637457
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore 636921
| |
Collapse
|
168
|
Andreuzzi E, Fejza A, Polano M, Poletto E, Camicia L, Carobolante G, Tarticchio G, Todaro F, Di Carlo E, Scarpa M, Scarpa M, Paulitti A, Capuano A, Canzonieri V, Maiero S, Fornasarig M, Cannizzaro R, Doliana R, Colombatti A, Spessotto P, Mongiat M. Colorectal cancer development is affected by the ECM molecule EMILIN-2 hinging on macrophage polarization via the TLR-4/MyD88 pathway. J Exp Clin Cancer Res 2022; 41:60. [PMID: 35148799 PMCID: PMC8840294 DOI: 10.1186/s13046-022-02271-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 01/22/2022] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Colorectal cancer is one of the most frequent and deadly tumors. Among the key regulators of CRC growth and progression, the microenvironment has emerged as a crucial player and as a possible route for the development of new therapeutic opportunities. More specifically, the extracellular matrix acts directly on cancer cells and indirectly affecting the behavior of stromal and inflammatory cells, as well as the bioavailability of growth factors. Among the ECM molecules, EMILIN-2 is frequently down-regulated by methylation in CRC and the purpose of this study was to verify the impact of EMILIN-2 loss in CRC development and its possible value as a prognostic biomarker. METHODS The AOM/DSS CRC protocol was applied to Emilin-2 null and wild type mice. Tumor development was monitored by endoscopy, the molecular analyses performed by IHC, IF and WB and the immune subpopulations characterized by flow cytometry. Ex vivo cultures of monocyte/macrophages from the murine models were used to verify the molecular pathways. Publicly available datasets were exploited to determine the CRC patients' expression profile; Spearman's correlation analyses and Cox regression were applied to evaluate the association with the inflammatory response; the clinical outcome was predicted by Kaplan-Meier survival curves. Pearson correlation analyses were also applied to a cohort of patients enrolled in our Institute. RESULTS In preclinical settings, loss of EMILIN-2 associated with an increased number of tumor lesions upon AOM/DSS treatment. In addition, in the early stages of the disease, the Emilin-2 knockout mice displayed a myeloid-derived suppressor cells-rich infiltrate. Instead, in the late stages, lack of EMILIN-2 associated with a decreased number of M1 macrophages, resulting in a higher percentage of the tumor-promoting M2 macrophages. Mechanistically, EMILIN-2 triggered the activation of the Toll-like Receptor 4/MyD88/NF-κB pathway, instrumental for the polarization of macrophages towards the M1 phenotype. Accordingly, dataset and immunofluorescence analyses indicated that low EMILIN-2 expression levels correlated with an increased M2/M1 ratio and with poor CRC patients' prognosis. CONCLUSIONS These novel results indicate that EMILIN-2 is a key regulator of the tumor-associated inflammatory environment and may represent a promising prognostic biomarker for CRC patients.
Collapse
Affiliation(s)
- Eva Andreuzzi
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy.
| | - Albina Fejza
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Maurizio Polano
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Evelina Poletto
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Lucrezia Camicia
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Greta Carobolante
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Giulia Tarticchio
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Federico Todaro
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Emma Di Carlo
- Department of Medicine and Sciences of Aging, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy.,Anatomic Pathology and Immuno-Oncology Unit, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Melania Scarpa
- Ricerca Traslazionale Avanzata, Istituto Oncologico Veneto IOV - IRCCS, Padua, Italy
| | - Marco Scarpa
- Clinica Chirurgica I- Azienda Ospedaliera di Padova, Padua, Italy
| | - Alice Paulitti
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Alessandra Capuano
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Vincenzo Canzonieri
- Pathology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Stefania Maiero
- Division of Oncological Gastroenterology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Mara Fornasarig
- Division of Oncological Gastroenterology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Renato Cannizzaro
- Division of Oncological Gastroenterology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Roberto Doliana
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Alfonso Colombatti
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Paola Spessotto
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Maurizio Mongiat
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy.
| |
Collapse
|
169
|
Rana N, Suliman S, Al-Sharabi N, Mustafa K. Extracellular Vesicles Derived from Primed Mesenchymal Stromal Cells Loaded on Biphasic Calcium Phosphate Biomaterial Exhibit Enhanced Macrophage Polarization. Cells 2022; 11:470. [PMID: 35159282 PMCID: PMC8834243 DOI: 10.3390/cells11030470] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/21/2022] [Accepted: 01/26/2022] [Indexed: 02/04/2023] Open
Abstract
Mesenchymal stromal cells (MSC) loaded on biphasic calcium phosphate biomaterial (MSC + BCP) have been used as an advanced therapy medicinal product to treat complex maxillofacial bone defects in patients. Further, MSC-derived extracellular vesicles (EVs) are established vehicles of paracrine factors, supporting inter-cellular communication between MSC and other interacting cell types, such as monocytes/macrophages. However, the information about the immunomodulatory potential of EVs derived from MSC and biomaterial constructs (MSC + BCP:EV) and inflammatory primed constructs (MSCp + BCP:EV) are scarce. Hence, we isolated and characterized EVs from these different systems, and compared their cytokine contents with plastic-adherent MSC-derived EVs (MSC:EV). When EVs from all three MSC systems were added to the primary blood-derived macrophages in vitro, significantly higher numbers of M0 (naive) macrophages shifted to M2-like (anti-inflammatory) by MSCp + BCP:EV treatment. Further, this treatment led to enhanced switching of M1 polarized macrophages to M2 polarized, and conversely, M2 to M1, as evaluated by determining the M1/M2 ratios after treatment. The enhanced macrophage modulation by MSCp + BCP:EV was attributed to their higher immunomodulatory (TNFα, IL1β, IL5), angiogenic (VEGF), and chemokine-rich (RANTES, MCP1, MIP1β) cytokine cargo. In conclusion, we successfully isolated and characterized EVs from MSC + BCP constructs and demonstrated that, depending upon the tissue microenvironment, these EVs contribute towards modulating the macrophage-mediated inflammation and healing responses. The study offers new insights into the use of biomaterial-induced EVs for MSC secretome delivery, as a step towards future 'cell-free' bone regenerative therapies.
Collapse
Affiliation(s)
| | | | | | - Kamal Mustafa
- Center for Translational Oral Research (TOR), Tissue Engineering Group, Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, 5009 Bergen, Norway; (N.R.); (S.S.); (N.A.-S.)
| |
Collapse
|
170
|
Yu F, Lian R, Liu L, Liu T, Bi C, Hong K, Zhang S, Ren J, Wang H, Ouyang N, Du LJ, Liu Y, Zhou L, Liu Y, Fang B, Li Y, Duan SZ, Xia L. Biomimetic Hydroxyapatite Nanorods Promote Bone Regeneration via Accelerating Osteogenesis of BMSCs through T Cell-Derived IL-22. ACS NANO 2022; 16:755-770. [PMID: 35005890 DOI: 10.1021/acsnano.1c08281] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Manipulations of morphological properties of nanobiomaterials have been demonstrated to modulate the outcome of osteoimmunomodulation and eventually osteogenesis through innate immune response. However, the functions and mechanisms of adaptive immune cells in the process of nanobiomaterials-mediated bone regeneration have remained unknown. Herein, we developed bone-mimicking hydroxyapatite (HAp) nanorods with different aspect ratios as model materials to investigate the impacts of the nanoshape features on osteogenesis and to explore the underlying mechanisms focusing on the functions of T cells and T cell-derived cytokines. HAp nanorods with different aspect ratios (HAp-0, HAp-30, and HAp-100) were implanted into mouse mandibular defect models. Micro-CT and hematoxylin and eosin staining demonstrated that HAp-100 had the best osteogenic effects. Flow cytometry analysis revealed that HAp-100 increased the percentage of T cells in injured mandibles. The osteogenic effects of HAp-100 were significantly blunted in injured mandibles of TCRβ-/- mice. The Luminex xMAP assay and ELISA showed that HAp-100 induced a marked increase of interleukin (IL)-22 in injured mandibles. In cultured T cells, HAp-100 manifested the best capacity to induce the production of IL-22. Conditioned media from HAp-100-primed T cells promoted osteogenesis and JAK1/STAT3 activation in bone marrow stromal cells, all of which were abolished by neutralizing antibodies against IL-22. In summary, bone-mimicking HAp nanorods with different aspect ratios could regulate osteogenesis through modulation of T cells and IL-22 in the bone regeneration process. These findings provided insights for mediation of the immune response of T cells by nanomaterials on osteogenesis and strategies for designing biomaterials with osteoimmunomodulative functions.
Collapse
Affiliation(s)
- Fei Yu
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai 200125, China
| | - Ruixian Lian
- The Key Laboratory for Ultrafine Materials of Ministry of Education, Engineering Research Centre for Biomedical Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Lu Liu
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai 200125, China
| | - Ting Liu
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai 200125, China
| | - Chao Bi
- Department of Stomatology, First Affiliated Hospital, Anhui Medical University, Hefei 230061, China
| | - Kan Hong
- The Key Laboratory for Ultrafine Materials of Ministry of Education, Engineering Research Centre for Biomedical Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Shuiquan Zhang
- The Key Laboratory for Ultrafine Materials of Ministry of Education, Engineering Research Centre for Biomedical Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Jiazi Ren
- CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Haikun Wang
- CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ningjuan Ouyang
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai 200125, China
| | - Lin-Juan Du
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai 200125, China
| | - Yuan Liu
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai 200125, China
| | - Lujun Zhou
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai 200125, China
| | - Yan Liu
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai 200125, China
| | - Bing Fang
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai 200125, China
| | - Yulin Li
- The Key Laboratory for Ultrafine Materials of Ministry of Education, Engineering Research Centre for Biomedical Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
- Wenzhou Institute of Shanghai University, Wenzhou 325000, China
| | - Sheng-Zhong Duan
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai 200125, China
| | - Lunguo Xia
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai 200125, China
| |
Collapse
|
171
|
An immunologically active, adipose-derived extracellular matrix biomaterial for soft tissue reconstruction: concept to clinical trial. NPJ Regen Med 2022; 7:6. [PMID: 35031598 PMCID: PMC8760240 DOI: 10.1038/s41536-021-00197-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 11/19/2021] [Indexed: 11/08/2022] Open
Abstract
Soft tissue reconstruction remains an intractable clinical challenge as current surgical options and synthetic implants may produce inadequate outcomes. Soft tissue deficits may be surgically reconstructed using autologous adipose tissue, but these procedures can lead to donor site morbidity, require multiple procedures, and have highly variable outcomes. To address this clinical need, we developed an "off-the-shelf" adipose extracellular matrix (ECM) biomaterial from allograft human tissue (Acellular Adipose Tissue, AAT). We applied physical and chemical processing methods to remove lipids and create an injectable matrix that mimicked the properties of lipoaspirate. Biological activity was assessed using cell migration and adipogenesis assays. Characterization of regenerative immune properties in a murine muscle injury model revealed that allograft and xenograft AAT induced pro-regenerative CD4+ T cells and macrophages with xenograft AAT additionally attracting eosinophils secreting interleukin 4 (Il4). In immunocompromised mice, AAT injections retained similar volumes as human fat grafts but lacked cysts and calcifications seen in the fat grafts. The combination of AAT with human adipose-derived stem cells (ASCs) resulted in lower implant volumes. However, tissue remodeling and adipogenesis increased significantly in combination with ASCs. Larger injected volumes of porcine-derived AAT demonstrated biocompatibility and greater retention when applied allogeneicly in Yorkshire cross pigs. AAT was implanted in healthy volunteers in abdominal tissue that was later removed by elective procedures. AAT implants were well tolerated in all human subjects. Implants removed between 1 and 18 weeks demonstrated increasing cellular infiltration and immune populations, suggesting continued tissue remodeling and the potential for long-term tissue replacement.
Collapse
|
172
|
Modulation of the Immune System Promotes Tissue Regeneration. Mol Biotechnol 2022; 64:599-610. [PMID: 35022994 DOI: 10.1007/s12033-021-00430-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 11/22/2021] [Indexed: 10/19/2022]
Abstract
The immune system plays an essential role in the angiogenesis, repair, and regeneration of damaged tissues. Therefore, the design of scaffolds that manipulate immune cells and factors in such a way that could accelerate the repair of damaged tissues, following implantation, is one of the main goals of regenerative medicine. However, before manipulating the immune system, the function of the various components of the immune system during the repair process should be well understood and the fabrication conditions of the manipulated scaffolds should be brought closer to the physiological state of the body. In this article, we first review the studies aimed at the role of distinct immune cell populations in angiogenesis and support of damaged tissue repair. In the second part, we discuss the use of strategies that promote tissue regeneration by modulating the immune system. Given that various studies have shown an increase in tissue repair rate with the addition of stem cells and growth factors to the scaffolds, and regarding the limited resources of stem cells, we suggest the design of scaffolds that are capable to develop repair of damaged tissue by manipulating the immune system and create an alternative for repair strategies that use stem cells or growth factors.
Collapse
|
173
|
Vantucci CE, Guyer T, Leguineche K, Chatterjee P, Lin A, Nash KE, Hastings MA, Fulton T, Smith CT, Maniar D, Frey Rubio DA, Peterson K, Harrer JA, Willett NJ, Roy K, Guldberg RE. Systemic Immune Modulation Alters Local Bone Regeneration in a Delayed Treatment Composite Model of Non-Union Extremity Trauma. Front Surg 2022; 9:934773. [PMID: 35874126 PMCID: PMC9300902 DOI: 10.3389/fsurg.2022.934773] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 06/20/2022] [Indexed: 11/25/2022] Open
Abstract
Bone non-unions resulting from severe traumatic injuries pose significant clinical challenges, and the biological factors that drive progression towards and healing from these injuries are still not well understood. Recently, a dysregulated systemic immune response following musculoskeletal trauma has been identified as a contributing factor for poor outcomes and complications such as infections. In particular, myeloid-derived suppressor cells (MDSCs), immunosuppressive myeloid-lineage cells that expand in response to traumatic injury, have been highlighted as a potential therapeutic target to restore systemic immune homeostasis and ultimately improve functional bone regeneration. Previously, we have developed a novel immunomodulatory therapeutic strategy to deplete MDSCs using Janus gold nanoparticles that mimic the structure and function of antibodies. Here, in a preclinical delayed treatment composite injury model of bone and muscle trauma, we investigate the effects of these nanoparticles on circulating MDSCs, systemic immune profiles, and functional bone regeneration. Unexpectedly, treatment with the nanoparticles resulted in depletion of the high side scatter subset of MDSCs and an increase in the low side scatter subset of MDSCs, resulting in an overall increase in total MDSCs. This overall increase correlated with a decrease in bone volume (P = 0.057) at 6 weeks post-treatment and a significant decrease in mechanical strength at 12 weeks post-treatment compared to untreated rats. Furthermore, MDSCs correlated negatively with endpoint bone healing at multiple timepoints. Single cell RNA sequencing of circulating immune cells revealed differing gene expression of the SNAb target molecule S100A8/A9 in MDSC sub-populations, highlighting a potential need for more targeted approaches to MDSC immunomodulatory treatment following trauma. These results provide further insights on the role of systemic immune dysregulation for severe trauma outcomes in the case of non-unions and composite injuries and suggest the need for additional studies on targeted immunomodulatory interventions to enhance healing.
Collapse
Affiliation(s)
- Casey E Vantucci
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States of America.,Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, United States of America
| | - Tyler Guyer
- Knight Campus or Accelerating Scientific Impact, University of Oregon, Eugene, OR, United States of America
| | - Kelly Leguineche
- Knight Campus or Accelerating Scientific Impact, University of Oregon, Eugene, OR, United States of America
| | - Paramita Chatterjee
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, United States of America.,Marcus Center for Therapeutic Cell Characterization and Manufacturing, Georgia Institute of Technology, Atlanta, GA, United States of America
| | - Angela Lin
- Knight Campus or Accelerating Scientific Impact, University of Oregon, Eugene, OR, United States of America
| | - Kylie E Nash
- Knight Campus or Accelerating Scientific Impact, University of Oregon, Eugene, OR, United States of America
| | - Molly Ann Hastings
- Knight Campus or Accelerating Scientific Impact, University of Oregon, Eugene, OR, United States of America
| | - Travis Fulton
- The Atlanta Veterans Affairs Medical Center Atlanta, Decatur, GA, United States of America.,Department of Orthopaedics, Emory University, Atlanta, GA, United States of America
| | - Clinton T Smith
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States of America
| | - Drishti Maniar
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States of America.,Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, United States of America
| | - David A Frey Rubio
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States of America
| | - Kaya Peterson
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States of America
| | - Julia Andraca Harrer
- Knight Campus or Accelerating Scientific Impact, University of Oregon, Eugene, OR, United States of America
| | - Nick J Willett
- Knight Campus or Accelerating Scientific Impact, University of Oregon, Eugene, OR, United States of America.,The Atlanta Veterans Affairs Medical Center Atlanta, Decatur, GA, United States of America.,Department of Orthopaedics, Emory University, Atlanta, GA, United States of America
| | - Krishnendu Roy
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States of America.,Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, United States of America
| | - Robert E Guldberg
- Knight Campus or Accelerating Scientific Impact, University of Oregon, Eugene, OR, United States of America
| |
Collapse
|
174
|
Chen J, Wang B, Caserto JS, Shariati K, Cao P, Pan Y, Xu Q, Ma M. Sustained Delivery of SARS-CoV-2 RBD Subunit Vaccine Using a High Affinity Injectable Hydrogel Scaffold. Adv Healthc Mater 2022; 11:e2101714. [PMID: 34755476 PMCID: PMC8652948 DOI: 10.1002/adhm.202101714] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/26/2021] [Indexed: 12/12/2022]
Abstract
The receptor binding domain (RBD) of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein that mediates viral entry into host cells is a good candidate immunogen for vaccine development against coronavirus disease 2019 (COVID-19). Because of its small size, most preclinical and early clinical efforts have focused on multimerizing RBD on various formats of nanoparticles to increase its immunogenicity. Using an easily administered injectable hydrogel scaffold that is rationally designed for enhanced retainment of RBD, an alternative and facile approach for boosting RBD immunogenicity in mice is demonstrated. Prolonged delivery of poly (I:C) adjuvanted RBD by the hydrogel scaffold results in sustained exposure to lymphoid tissues, which elicits serum IgG titers comparable to those induced by three bolus injections, but more long-lasting and polarized toward TH 1-mediated IgG2b. The hydrogel scaffold induces potent germinal center (GC) reactions, correlating with RBD-specific antibody generation and robust type 1 T cell responses. Besides being an enduring RBD reservoir, the hydrogel scaffold becomes a local inflammatory niche for innate immune cell activation. Collectively, the injectable hydrogel scaffold provides a simple, practical, and inexpensive means to enhance the efficacy of RBD-based subunit vaccines against COVID-19 and may be applicable to other circulating and emerging pathogens.
Collapse
Affiliation(s)
- Jing Chen
- Department of Biological and Environmental EngineeringCornell UniversityIthacaNY14853USA
| | - Bo Wang
- Department of Biological and Environmental EngineeringCornell UniversityIthacaNY14853USA
| | - Julia S. Caserto
- Robert Frederick Smith School of Chemical and Biomolecular EngineeringCornell UniversityIthacaNY14853USA
| | - Kaavian Shariati
- Department of Biological and Environmental EngineeringCornell UniversityIthacaNY14853USA
| | - Peng Cao
- College of PharmacyNanjing University of Chinese MedicineNanjing210023China
| | - Yang Pan
- College of PharmacyNanjing University of Chinese MedicineNanjing210023China
| | - Qixuan Xu
- College of PharmacyNanjing University of Chinese MedicineNanjing210023China
| | - Minglin Ma
- Department of Biological and Environmental EngineeringCornell UniversityIthacaNY14853USA
| |
Collapse
|
175
|
Pan D, Schellhardt L, Acevedo-Cintron JA, Hunter D, Snyder-Warwick AK, Mackinnon SE, Wood MD. IL-4 expressing cells are recruited to nerve after injury and promote regeneration. Exp Neurol 2022; 347:113909. [PMID: 34717939 PMCID: PMC8887027 DOI: 10.1016/j.expneurol.2021.113909] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/18/2021] [Accepted: 10/24/2021] [Indexed: 01/03/2023]
Abstract
Interleukin-4 (IL-4) has garnered interest as a cytokine that mediates regeneration across multiple tissues including peripheral nerve. Within nerve, we previously showed endogenous IL-4 was critical to regeneration across nerve gaps. Here, we determined a generalizable role of IL-4 in nerve injury and regeneration. In wild-type (WT) mice receiving a sciatic nerve crush, IL-4 expressing cells preferentially accumulated within the injured nerve compared to affected sites proximal, such as dorsal root ganglia (DRGs), or distal muscle. Immunohistochemistry and flow cytometry confirmed that eosinophils (CD45+, CD11b+, CD64-, Siglec-F+) were sources of IL-4 expression. Examination of targets for IL-4 within nerve revealed macrophages, as well as subsets of neurons expressed IL-4R, while Schwann cells expressed limited IL-4R. Dorsal root ganglia cultures were exposed to IL-4 and demonstrated an increased proportion of neurons that extended axons compared to cultures without IL-4 (control), as well as longer myelinated axons compared to cultures without IL-4. The role of endogenous IL-4 during nerve injury and regeneration in vivo was assessed following a sciatic nerve crush using IL-4 knockout (KO) mice. Loss of IL-4 affected macrophage accumulation within injured nerve compared to WT mice, as well as shifted macrophage phenotype towards a CD206- phenotype with altered gene expression. Furthermore, this loss of IL-4 delayed initial axon regeneration from the injury crush site and subsequently delayed functional recovery and re-innervation of neuromuscular junctions compared to wild-type mice. Given the role of endogenous IL-4 in nerve regeneration, exogenous IL-4 was administered daily to WT mice following a nerve crush to examine regeneration. Daily IL-4 administration increased early axonal extension and CD206+ macrophage accumulation but did not alter functional recovery compared to untreated mice. Our data demonstrate IL-4 promotes nerve regeneration and recovery after injury.
Collapse
Affiliation(s)
- Deng Pan
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Lauren Schellhardt
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jesús A Acevedo-Cintron
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Daniel Hunter
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Alison K Snyder-Warwick
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Susan E Mackinnon
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Matthew D Wood
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
176
|
Gvaramia D, Kern J, Jakob Y, Tritschler H, Brenner RE, Breiter R, Kzhyshkowska J, Rotter N. Modulation of the inflammatory response to decellularized collagen matrix for cartilage regeneration. J Biomed Mater Res A 2021; 110:1021-1035. [PMID: 34967101 DOI: 10.1002/jbm.a.37349] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 12/13/2021] [Accepted: 12/15/2021] [Indexed: 12/21/2022]
Abstract
Decellularized extracellular matrices (DECM) are among the most common types of materials used in tissue engineering due to their cell instructive properties, biodegradability, and accessibility. Particularly in cartilage, a natural collagen type II matrix can be a promising means to provide the necessary cues and support for chondrogenic stem and progenitor cells (CSPCs). However, efficient remodeling of the transplanted DECM is largely dependent on the host immune response, with macrophages playing the central role in orchestrating both inflammatory and regenerative processes. Here we assessed the reaction of human primary macrophages to the cartilage DECM. Our findings show that the xenogeneic collagen matrix can elicit a mixed response in human macrophages, whereby the inflammatory response (M1) and the activation of remodeling (M2) type of macrophages are both present. Additionally, we demonstrate the inhibitory effect of macrophage response on the migratory capacity of human CSPCs. We further show that the inflammatory reaction of macrophages to the cartilage DECM, as well as the resulting inhibitory effects on CSPC migration, can be attenuated by interleukin-4 (IL-4). Finally, we demonstrate that IL-4 can effectively bind the matrix, thereby modulating macrophage response by reducing the inflammatory reaction and inducing the M2 phenotype.
Collapse
Affiliation(s)
- David Gvaramia
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Johann Kern
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Yvonne Jakob
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Hanna Tritschler
- Division for Biochemistry of Joint and Connective Tissue Diseases, Department of Orthopaedics, University of Ulm, Ulm, Germany
| | - Rolf E Brenner
- Division for Biochemistry of Joint and Connective Tissue Diseases, Department of Orthopaedics, University of Ulm, Ulm, Germany
| | - Roman Breiter
- Institute of Bioprocess Engineering, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Julia Kzhyshkowska
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim of Ruprecht-Karls-University Heidelberg, Mannheim, Germany
| | - Nicole Rotter
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
177
|
Rømer AMA, Thorseth ML, Madsen DH. Immune Modulatory Properties of Collagen in Cancer. Front Immunol 2021; 12:791453. [PMID: 34956223 PMCID: PMC8692250 DOI: 10.3389/fimmu.2021.791453] [Citation(s) in RCA: 113] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 11/22/2021] [Indexed: 12/22/2022] Open
Abstract
During tumor growth the extracellular matrix (ECM) undergoes dramatic remodeling. The normal ECM is degraded and substituted with a tumor-specific ECM, which is often of higher collagen density and increased stiffness. The structure and collagen density of the tumor-specific ECM has been associated with poor prognosis in several types of cancer. However, the reason for this association is still largely unknown. Collagen can promote cancer cell growth and migration, but recent studies have shown that collagens can also affect the function and phenotype of various types of tumor-infiltrating immune cells such as tumor-associated macrophages (TAMs) and T cells. This suggests that tumor-associated collagen could have important immune modulatory functions within the tumor microenvironment, affecting cancer progression as well as the efficacy of cancer immunotherapy. The effects of tumor-associated collagen on immune cells could help explain why a high collagen density in tumors is often correlated with a poor prognosis. Knowledge about immune modulatory functions of collagen could potentially identify targets for improving current cancer therapies or for development of new treatments. In this review, the current knowledge about the ability of collagen to influence T cell activity will be summarized. This includes direct interactions with T cells as well as induction of immune suppressive activity in other immune cells such as macrophages. Additionally, the potential effects of collagen on the efficacy of cancer immunotherapy will be discussed.
Collapse
Affiliation(s)
- Anne Mette Askehøj Rømer
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark.,Department of Science and Environment, Roskilde University, Roskilde, Denmark
| | - Marie-Louise Thorseth
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark.,Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Daniel Hargbøl Madsen
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark.,Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
178
|
Immune-instructive materials as new tools for immunotherapy. Curr Opin Biotechnol 2021; 74:194-203. [PMID: 34959210 DOI: 10.1016/j.copbio.2021.11.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 11/08/2021] [Accepted: 11/11/2021] [Indexed: 12/13/2022]
Abstract
Immune instructive materials, are materials with the ability to modulate or mimic the function of immune cells, provide exciting opportunities for developing new therapies in many areas including medical devices, chronic inflammation, cancer, and autoimmune diseases. In this review we highlight some of the latest research involving material-based strategies for modulating macrophage phenotype and dendritic cell function, as well as a brief description on biomaterial use in T cell and natural killer cell engineering. We highlight studies on material topography, size, shape and surface chemistry to reduce inflammation, along with scaffold and hydrogel delivery systems that are used for modulating DC phenotype and influencing T cell polarization. Artificial antigen presenting cells are also reviewed as a promising approach to cancer immunotherapy.
Collapse
|
179
|
Affiliation(s)
- Jennifer Elisseeff
- From the Translational Tissue Engineering Center, Wilmer Eye Institute, and the Department of Biomedical Engineering, Johns Hopkins University, Baltimore (J.E.); the McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh (S.F.B.); and the Institute for Systems Genetics and the Department of Biochemistry and Molecular Pharmacology, NYU Langone Health (J.D.B.), and the Department of Biomedical Engineering, NYU Tandon School of Engineering (J.D.B.) - both in New York
| | - Stephen F Badylak
- From the Translational Tissue Engineering Center, Wilmer Eye Institute, and the Department of Biomedical Engineering, Johns Hopkins University, Baltimore (J.E.); the McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh (S.F.B.); and the Institute for Systems Genetics and the Department of Biochemistry and Molecular Pharmacology, NYU Langone Health (J.D.B.), and the Department of Biomedical Engineering, NYU Tandon School of Engineering (J.D.B.) - both in New York
| | - Jef D Boeke
- From the Translational Tissue Engineering Center, Wilmer Eye Institute, and the Department of Biomedical Engineering, Johns Hopkins University, Baltimore (J.E.); the McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh (S.F.B.); and the Institute for Systems Genetics and the Department of Biochemistry and Molecular Pharmacology, NYU Langone Health (J.D.B.), and the Department of Biomedical Engineering, NYU Tandon School of Engineering (J.D.B.) - both in New York
| |
Collapse
|
180
|
Regenerative rehabilitation of skeletal muscle damages. КЛИНИЧЕСКАЯ ПРАКТИКА 2021. [DOI: 10.17816/clinpract70873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The article is devoted to the analysis of the current state of regenerative and rehabilitative treatments of skeletal muscles, the possibilities of restoring the functioning of tissue lost due to aging, injuries or diseases. The study of the molecular genetic basis of mechanotransduction and mechanotherapy will allow the identification of genes and molecules, the expression levels of which can serve as biomarkers of the effectiveness of regenerative-rehabilitation measures. These mechanisms are potential therapeutic targets for stimulating of regeneration of skeletal muscles. The focus of the article is on the choice of an individual approach, both when conducting basic scientific research and developing rehabilitation programs. All this will significantly improve patient outcomes.
Collapse
|
181
|
Li M, Li X, Liu B, Lv L, Wang W, Gao D, Zhang Q, Jiang J, Chai M, Yun Z, Tan Y, Gong F, Wu Z, Zhu Y, Ma J, Leng L. Time-Resolved Extracellular Matrix Atlas of the Developing Human Skin Dermis. Front Cell Dev Biol 2021; 9:783456. [PMID: 34901026 PMCID: PMC8661536 DOI: 10.3389/fcell.2021.783456] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 11/11/2021] [Indexed: 11/18/2022] Open
Abstract
Skin aging is a physiological issue that is still relatively poorly understood. Studies have demonstrated that the dermal extracellular matrix (ECM) plays important roles in skin aging. However, the roles of the changes in ECM characteristics and the molecules that are secreted to the extracellular space and are involved in the formation of the dermal matrix from birth to old age remain unclear. To explore the way in which the ECM microenvironment supports the functions of skin development across different age groups is also poorly understood, we used a decellularization method and matrisome analysis to compare the composition, expression, and function of the dermal ECM in toddler, teenager, adult, and elderly skin. We found that the collagens, glycoproteins, proteoglycans, and regulatory factors that support skin development and interact with these core ECM proteins were differentially expressed at different ages. ECM expression markers occurring during the process of skin development were identified. In addition, our results elucidated the characteristics of ECM synthesis, response to skin development, and the features of the ECM that support epidermal stem cell growth via the basement membrane during skin aging.
Collapse
Affiliation(s)
- Mansheng Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Life Omics, Beijing, China
| | - Xiao Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Life Omics, Beijing, China
| | - Binghui Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Life Omics, Beijing, China
| | - Luye Lv
- Institute of NBC Defense, Beijing, China
| | - Wenjuan Wang
- Department of Dermatology, Chinese PLA General Hospital, Beijing, China
| | - Dunqin Gao
- Stem Cell and Regenerative Medicine Lab, State Key Laboratory of Complex Severe and Rare Diseases, Department of Medical Science Research Center, Translational Medicine Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qiyu Zhang
- Stem Cell and Regenerative Medicine Lab, State Key Laboratory of Complex Severe and Rare Diseases, Department of Medical Science Research Center, Translational Medicine Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Junyi Jiang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Life Omics, Beijing, China
| | - Mi Chai
- Department of Plastic and Reconstruction Surgery, Chinese PLA General Hospital, Beijing, China
| | - Zhimin Yun
- Department of Stem Cell and Regenerative Medicine Laboratory, Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Yingxia Tan
- Department of Stem Cell and Regenerative Medicine Laboratory, Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Feng Gong
- Department of Stem Cell and Regenerative Medicine Laboratory, Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Zhihong Wu
- Stem Cell and Regenerative Medicine Lab, State Key Laboratory of Complex Severe and Rare Diseases, Department of Medical Science Research Center, Translational Medicine Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yunping Zhu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Life Omics, Beijing, China.,Basic Medical School, Anhui Medical University, Anhui, China
| | - Jie Ma
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Life Omics, Beijing, China
| | - Ling Leng
- Stem Cell and Regenerative Medicine Lab, State Key Laboratory of Complex Severe and Rare Diseases, Department of Medical Science Research Center, Translational Medicine Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
182
|
Antmen E, Vrana NE, Hasirci V. The role of biomaterials and scaffolds in immune responses in regenerative medicine: macrophage phenotype modulation by biomaterial properties and scaffold architectures. Biomater Sci 2021; 9:8090-8110. [PMID: 34762077 DOI: 10.1039/d1bm00840d] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Scaffolds are an integral part of the regenerative medicine field. The contact of biomaterials with tissue, as was clearly observed over the years, induces immune reactions in a material and patient specific manner, where both surface and bulk properties of scaffolds, together with their 3D architecture, have a significant influence on the outcome. This review presents an overview of the reactions to the biomaterials with a specific focus on clinical complications with the implants in the context of immune reactions and an overview of the studies involving biomaterial properties and interactions with innate immune system cells. We emphasize the impact of these studies on scaffold selection and upscaling of microenvironments created by biomaterials from 2D to 3D using immune cell encapsulation, seeding in a 3D scaffold and co-culture with relevant tissue cells. 3D microenvironments are covered with a specific focus on innate cells since a large proportion of these studies used innate immune cells. Finally, the recent studies on the incorporation of adaptive immune cells in immunomodulatory systems are covered in this review. Biomaterial-immune cell interactions are a critical part of regenerative medicine applications. Current efforts in establishing the ground rules for such interactions following implantation can control immune response during all phases of inflammation. Thus, in the near future for complete functional recovery, tissue engineering and control over biomaterials must be considered at the first step of immune modulation and this review covers these interactions, which have remained elusive up to now.
Collapse
Affiliation(s)
- Ezgi Antmen
- BIOMATEN, Middle East Technical University, Center of Excellence in Biomaterials and Tissue Engineering, Ankara, Turkey.
| | - Nihal Engin Vrana
- SPARTHA Medical, 14B Rue de la Canardiere, Strasbourg Cedex 67100, France. .,INSERM Unité 1121 Biomaterials and Bioengineering, CRBS, 1 Rue Eugène Boeckel, Strasbourg Cedex 67000, France
| | - Vasif Hasirci
- BIOMATEN, Middle East Technical University, Center of Excellence in Biomaterials and Tissue Engineering, Ankara, Turkey. .,Biomaterials A&R Center, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey.,Department of Medical Engineering, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| |
Collapse
|
183
|
Moore EM, Maestas DR, Cherry CC, Garcia JA, Comeau HY, Davenport Huyer L, Kelly SH, Peña AN, Blosser RL, Rosson GD, Elisseeff JH. Biomaterials direct functional B cell response in a material-specific manner. SCIENCE ADVANCES 2021; 7:eabj5830. [PMID: 34851674 PMCID: PMC8635437 DOI: 10.1126/sciadv.abj5830] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 10/13/2021] [Indexed: 05/13/2023]
Abstract
B cells are an adaptive immune target of biomaterials development in vaccine research but, despite their role in wound healing, have not been extensively studied in regenerative medicine. To probe the role of B cells in biomaterial scaffold response, we evaluated the B cell response to biomaterial materials implanted in a muscle wound using a biological extracellular matrix (ECM), as a reference for a naturally derived material, and synthetic polyester polycaprolactone (PCL), as a reference for a synthetic material. In the local muscle tissue, small numbers of B cells are present in response to tissue injury and biomaterial implantation. The ECM materials induced mature B cells in lymph nodes and antigen presentation in the spleen. The synthetic PCL implants resulted in prolonged B cell presence in the wound and induced an antigen-presenting phenotype. In summary, the adaptive B cell immune response to biomaterial induces local, regional, and systemic immunological changes.
Collapse
Affiliation(s)
- Erika M. Moore
- Department of Materials Science and Engineering, University of Florida, Gainesville, FL, USA
- Translational Tissue Engineering Center, Wilmer Eye Institute and the Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - David R. Maestas
- Translational Tissue Engineering Center, Wilmer Eye Institute and the Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Chris C. Cherry
- Translational Tissue Engineering Center, Wilmer Eye Institute and the Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Jordan A. Garcia
- Translational Tissue Engineering Center, Wilmer Eye Institute and the Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Hannah Y. Comeau
- Translational Tissue Engineering Center, Wilmer Eye Institute and the Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Locke Davenport Huyer
- Translational Tissue Engineering Center, Wilmer Eye Institute and the Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Sean H. Kelly
- Translational Tissue Engineering Center, Wilmer Eye Institute and the Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Alexis N. Peña
- Translational Tissue Engineering Center, Wilmer Eye Institute and the Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Richard L. Blosser
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Gedge D. Rosson
- Division of Plastic Surgery, Department of Surgery, Johns Hopkins University, Baltimore, MD, USA
| | - Jennifer H. Elisseeff
- Translational Tissue Engineering Center, Wilmer Eye Institute and the Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
184
|
Ozpinar EW, Frey AL, Cruse G, Freytes DO. Mast Cell-Biomaterial Interactions and Tissue Repair. TISSUE ENGINEERING. PART B, REVIEWS 2021; 27:590-603. [PMID: 33164714 PMCID: PMC8739845 DOI: 10.1089/ten.teb.2020.0275] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 11/03/2020] [Indexed: 12/12/2022]
Abstract
Tissue engineers often use biomaterials to provide structural support along with mechanical and chemical signals to modulate the wound healing process. Biomaterials that are implanted into the body interact with a heterogeneous and dynamic inflammatory environment that is present at the site of injury. Whether synthetically derived, naturally derived, or a combination of both, it is important to assess biomaterials for their ability to modulate inflammation to understand their potential clinical use. One important, but underexplored cell in the context of biomaterials is the mast cell (MC). MCs are granulocytic leukocytes that engage in a variety of events in both the innate and adaptive immune systems. Although highly recognized for their roles in allergic reactions, MCs play an important role in wound healing by recognizing antigens through pattern recognition receptors and the high-affinity immunoglobulin E receptor (FceRI) and releasing granules that affect cell recruitment, fibrosis, extracellular matrix deposition, angiogenesis, and vasculogenesis. MCs also mediate the foreign body response, contributing to the incorporation or rejection of implants. Studies of MC-biomaterial interactions can aid in the elucidation of MC roles during the host tissue response and tissue repair. This review is designed for those in the tissue engineering and biomaterial fields who are interested in exploring the role MCs may play in wound-biomaterial interactions and wound healing. With this review, we hope to inspire more research in the MC-biomaterial space to accelerate the design and construction of optimized implants. Impact statement Mast cells (MCs) are highly specialized inflammatory cells that have crucial, but not fully understood, roles in wound healing and tissue repair. Upon stimulation, they recognize foreign antigens and release granules that help orchestrate the inflammatory response after tissue damage or biomaterial implantation. This review summarizes the current use of MCs in biomaterial research along with literature from the past decade focusing on MC interactions with materials used for tissue repair and regeneration. Studying MC-biomaterial interactions will help (i) further understand the process of inflammation and (ii) design biomaterials and tissue-engineered constructs for optimal repair and regeneration.
Collapse
Affiliation(s)
- Emily W Ozpinar
- The Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina-Chapel Hill, Raleigh, North Carolina, USA
- The Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
| | - Ariana L Frey
- The Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina-Chapel Hill, Raleigh, North Carolina, USA
| | - Glenn Cruse
- The Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - Donald O Freytes
- The Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina-Chapel Hill, Raleigh, North Carolina, USA
- The Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
| |
Collapse
|
185
|
Abstract
Glioblastoma is one of the deadliest forms of primary adult tumors, with median survival of 14.6 months post-diagnosis despite aggressive standard of care treatment. This grim prognosis for glioblastoma patients has changed little in the past two decades, necessitating novel treatment modalities. One potential treatment modality is cancer immunotherapy, which has shown remarkable progress in slowing disease progression or even potentially curing certain solid tumors. However, the transport barriers posed by the blood-brain barrier and the immune privileged status of the central nervous system pose drug delivery obstacles that are unique to brain tumors. In this review, we provide an overview of the various physiological, immunological, and drug delivery barriers that must be overcome for effective glioblastoma treatment. We discuss chemical modification strategies to enable nanomedicines to bypass the blood-brain barrier and reach intracranial tumors. Finally, we highlight recent advances in biomaterial-based strategies for cancer immunotherapy that can be adapted to glioblastoma treatment.
Collapse
Affiliation(s)
- Yuan Rui
- Department of Biomedical Engineering, the Institute for Nanobiotechnology and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jordan J Green
- Department of Biomedical Engineering, the Institute for Nanobiotechnology and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Departments of Neurosurgery, Ophthalmology and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Departments of Materials Science & Engineering and Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA.
- Johns Hopkins University School of Medicine, Bloomberg~Kimmel Institute for Cancer Immunotherapy, Baltimore, MD, USA.
| |
Collapse
|
186
|
Translational considerations for adipose-derived biological scaffolds for soft tissue repair. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2021. [DOI: 10.1016/j.cobme.2021.100321] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
187
|
Karkanitsa M, Fathi P, Ngo T, Sadtler K. Mobilizing Endogenous Repair Through Understanding Immune Reaction With Biomaterials. Front Bioeng Biotechnol 2021; 9:730938. [PMID: 34917594 PMCID: PMC8670074 DOI: 10.3389/fbioe.2021.730938] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 09/10/2021] [Indexed: 12/29/2022] Open
Abstract
With few exceptions, humans are incapable of fully recovering from severe physical trauma. Due to these limitations, the field of regenerative medicine seeks to find clinically viable ways to repair permanently damaged tissue. There are two main approaches to regenerative medicine: promoting endogenous repair of the wound, or transplanting a material to replace the injured tissue. In recent years, these two methods have fused with the development of biomaterials that act as a scaffold and mobilize the body's natural healing capabilities. This process involves not only promoting stem cell behavior, but by also inducing activity of the immune system. Through understanding the immune interactions with biomaterials, we can understand how the immune system participates in regeneration and wound healing. In this review, we will focus on biomaterials that promote endogenous tissue repair, with discussion on their interactions with the immune system.
Collapse
Affiliation(s)
| | | | | | - Kaitlyn Sadtler
- Section on Immuno-Engineering, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
188
|
Mahara A, Kojima K, Yamamoto M, Hirano Y, Yamaoka T. Accelerated tissue regeneration in decellularized vascular grafts with a patterned pore structure. J Mater Chem B 2021; 10:2544-2550. [PMID: 34787632 DOI: 10.1039/d1tb02271g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Decellularized tissue is expected to be utilized as a regenerative scaffold. However, the migration of host cells into the central region of the decellularized tissues is minimal because the tissues are mainly formed with dense collagen and elastin fibers. This results in insufficient tissue regeneration. Herein, it is demonstrated that host cell migration can be accelerated by using decellularized tissue with a patterned pore structure. Patterned pores with inner diameters of 24.5 ± 0.4 μm were fabricated at 100, 250, and 500 μm intervals in the decellularized vascular grafts via laser ablation. The grafts were transplanted into rat subcutaneous tissue for 1, 2, and 4 weeks. All the microporous grafts underwent faster recellularization with macrophages and fibroblast cells than the non-porous control tissue. In the case of non-porous tissue, the cells infiltrated approximately 50% of the area four weeks after transplantation. However, almost the entire area was occupied by the cells after two weeks when the micropores were aligned at a distance of less than 250 μm. These results suggest that host cell infiltration depends on the micropore interval, and a distance shorter than 250 μm can accelerate cell migration into decellularized tissues.
Collapse
Affiliation(s)
- Atsushi Mahara
- Department of Biomedical Engineering, National Cerebral and Cardiovascular Center Research Institute, Kishibe-shin Machi, Suita, Osaka 564-8565, Japan.
| | - Kentaro Kojima
- Department of Biomedical Engineering, National Cerebral and Cardiovascular Center Research Institute, Kishibe-shin Machi, Suita, Osaka 564-8565, Japan. .,Faculty of Chemistry, Materials and Bioengineering, Kansai University, 3-3-35 Yamatecho, Suita, Osaka 565-8680, Japan
| | - Masami Yamamoto
- Department of Biomedical Engineering, National Cerebral and Cardiovascular Center Research Institute, Kishibe-shin Machi, Suita, Osaka 564-8565, Japan. .,Faculty of Medical Engineering, Suzuka University of Medical Science, Suzuka, Mie 510-0293, Japan
| | - Yoshiaki Hirano
- Faculty of Chemistry, Materials and Bioengineering, Kansai University, 3-3-35 Yamatecho, Suita, Osaka 565-8680, Japan
| | - Tetsuji Yamaoka
- Department of Biomedical Engineering, National Cerebral and Cardiovascular Center Research Institute, Kishibe-shin Machi, Suita, Osaka 564-8565, Japan.
| |
Collapse
|
189
|
Bonito V, Koch SE, Krebber MM, Carvajal‐Berrio DA, Marzi J, Duijvelshoff R, Lurier EB, Buscone S, Dekker S, de Jong SMJ, Mes T, Vaessen KRD, Brauchle EM, Bosman AW, Schenke‐Layland K, Verhaar MC, Dankers PYW, Smits AIPM, Bouten CVC. Distinct Effects of Heparin and Interleukin-4 Functionalization on Macrophage Polarization and In Situ Arterial Tissue Regeneration Using Resorbable Supramolecular Vascular Grafts in Rats. Adv Healthc Mater 2021; 10:e2101103. [PMID: 34523263 PMCID: PMC11469141 DOI: 10.1002/adhm.202101103] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 08/12/2021] [Indexed: 12/16/2022]
Abstract
Two of the greatest challenges for successful application of small-diameter in situ tissue-engineered vascular grafts are 1) preventing thrombus formation and 2) harnessing the inflammatory response to the graft to guide functional tissue regeneration. This study evaluates the in vivo performance of electrospun resorbable elastomeric vascular grafts, dual-functionalized with anti-thrombogenic heparin (hep) and anti-inflammatory interleukin 4 (IL-4) using a supramolecular approach. The regenerative capacity of IL-4/hep, hep-only, and bare grafts is investigated as interposition graft in the rat abdominal aorta, with follow-up at key timepoints in the healing cascade (1, 3, 7 days, and 3 months). Routine analyses are augmented with Raman microspectroscopy, in order to acquire the local molecular fingerprints of the resorbing scaffold and developing tissue. Thrombosis is found not to be a confounding factor in any of the groups. Hep-only-functionalized grafts resulted in adverse tissue remodeling, with cases of local intimal hyperplasia. This is negated with the addition of IL-4, which promoted M2 macrophage polarization and more mature neotissue formation. This study shows that with bioactive functionalization, the early inflammatory response can be modulated and affect the composition of neotissue. Nevertheless, variability between graft outcomes is observed within each group, warranting further evaluation in light of clinical translation.
Collapse
Affiliation(s)
- Valentina Bonito
- Department of Biomedical Engineering and Institute for Complex Molecular Systems (ICMS)Eindhoven University of TechnologyEindhoven5600 MBThe Netherlands
| | - Suzanne E. Koch
- Department of Biomedical Engineering and Institute for Complex Molecular Systems (ICMS)Eindhoven University of TechnologyEindhoven5600 MBThe Netherlands
| | - Merle M. Krebber
- Department of Nephrology and HypertensionUniversity Medical Center UtrechtUtrecht3584 CXThe Netherlands
| | - Daniel A. Carvajal‐Berrio
- Department of Biomedical EngineeringResearch Institute of Women's Health and Cluster of Excellence iFIT (EXC 2180) “Image‐Guided and Functionally Instructed Tumor Therapies”Eberhard Karls University TübingenTübingen72076Germany
- NMI Natural and Medical Sciences Institute at the University of TübingenReutlingen72770Germany
| | - Julia Marzi
- Department of Biomedical EngineeringResearch Institute of Women's Health and Cluster of Excellence iFIT (EXC 2180) “Image‐Guided and Functionally Instructed Tumor Therapies”Eberhard Karls University TübingenTübingen72076Germany
- NMI Natural and Medical Sciences Institute at the University of TübingenReutlingen72770Germany
| | - Renee Duijvelshoff
- Department of Biomedical Engineering and Institute for Complex Molecular Systems (ICMS)Eindhoven University of TechnologyEindhoven5600 MBThe Netherlands
- Department of CardiologyIsala Hospitalvan Heesweg 2Zwolle8025 ABThe Netherlands
| | - Emily B. Lurier
- Department of Biomedical Engineering and Institute for Complex Molecular Systems (ICMS)Eindhoven University of TechnologyEindhoven5600 MBThe Netherlands
- School of Biomedical EngineeringScience and Health SystemsDrexel UniversityPhiladelphiaPA19104USA
| | - Serena Buscone
- Department of Biomedical Engineering and Institute for Complex Molecular Systems (ICMS)Eindhoven University of TechnologyEindhoven5600 MBThe Netherlands
| | - Sylvia Dekker
- Department of Biomedical Engineering and Institute for Complex Molecular Systems (ICMS)Eindhoven University of TechnologyEindhoven5600 MBThe Netherlands
| | - Simone M. J. de Jong
- Department of Biomedical Engineering and Institute for Complex Molecular Systems (ICMS)Eindhoven University of TechnologyEindhoven5600 MBThe Netherlands
| | - Tristan Mes
- SupraPolix BVEindhoven5612 AXThe Netherlands
| | - Koen R. D. Vaessen
- Central Laboratory Animal Research Facility (CLARF)Utrecht UniversityUtrecht3584 CXThe Netherlands
| | - Eva M. Brauchle
- Department of Biomedical EngineeringResearch Institute of Women's Health and Cluster of Excellence iFIT (EXC 2180) “Image‐Guided and Functionally Instructed Tumor Therapies”Eberhard Karls University TübingenTübingen72076Germany
- NMI Natural and Medical Sciences Institute at the University of TübingenReutlingen72770Germany
| | | | - Katja Schenke‐Layland
- Department of Biomedical EngineeringResearch Institute of Women's Health and Cluster of Excellence iFIT (EXC 2180) “Image‐Guided and Functionally Instructed Tumor Therapies”Eberhard Karls University TübingenTübingen72076Germany
- NMI Natural and Medical Sciences Institute at the University of TübingenReutlingen72770Germany
| | - Marianne C. Verhaar
- Department of Nephrology and HypertensionUniversity Medical Center UtrechtUtrecht3584 CXThe Netherlands
| | - Patricia Y. W. Dankers
- Department of Biomedical Engineering and Institute for Complex Molecular Systems (ICMS)Eindhoven University of TechnologyEindhoven5600 MBThe Netherlands
| | - Anthal I. P. M. Smits
- Department of Biomedical Engineering and Institute for Complex Molecular Systems (ICMS)Eindhoven University of TechnologyEindhoven5600 MBThe Netherlands
| | - Carlijn V. C. Bouten
- Department of Biomedical Engineering and Institute for Complex Molecular Systems (ICMS)Eindhoven University of TechnologyEindhoven5600 MBThe Netherlands
| |
Collapse
|
190
|
Immuno-regenerative biomaterials for in situ cardiovascular tissue engineering - Do patient characteristics warrant precision engineering? Adv Drug Deliv Rev 2021; 178:113960. [PMID: 34481036 DOI: 10.1016/j.addr.2021.113960] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 08/20/2021] [Accepted: 08/30/2021] [Indexed: 02/07/2023]
Abstract
In situ tissue engineering using bioresorbable material implants - or scaffolds - that harness the patient's immune response while guiding neotissue formation at the site of implantation is emerging as a novel therapy to regenerate human tissues. For the cardiovascular system, the use of such implants, like blood vessels and heart valves, is gradually entering the stage of clinical translation. This opens up the question if and to what extent patient characteristics influence tissue outcomes, necessitating the precision engineering of scaffolds to guide patient-specific neo-tissue formation. Because of the current scarcity of human in vivo data, herein we review and evaluate in vitro and preclinical investigations to predict the potential role of patient-specific parameters like sex, age, ethnicity, hemodynamics, and a multifactorial disease profile, with special emphasis on their contribution to the inflammation-driven processes of in situ tissue engineering. We conclude that patient-specific conditions have a strong impact on key aspects of in situ cardiovascular tissue engineering, including inflammation, hemodynamic conditions, scaffold resorption, and tissue remodeling capacity, suggesting that a tailored approach may be required to engineer immuno-regenerative biomaterials for safe and predictive clinical applicability.
Collapse
|
191
|
Eugenis I, Wu D, Rando TA. Cells, scaffolds, and bioactive factors: Engineering strategies for improving regeneration following volumetric muscle loss. Biomaterials 2021; 278:121173. [PMID: 34619561 PMCID: PMC8556323 DOI: 10.1016/j.biomaterials.2021.121173] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 08/01/2021] [Accepted: 08/14/2021] [Indexed: 12/20/2022]
Abstract
Severe traumatic skeletal muscle injuries, such as volumetric muscle loss (VML), result in the obliteration of large amounts of skeletal muscle and lead to permanent functional impairment. Current clinical treatments are limited in their capacity to regenerate damaged muscle and restore tissue function, promoting the need for novel muscle regeneration strategies. Advances in tissue engineering, including cell therapy, scaffold design, and bioactive factor delivery, are promising solutions for VML therapy. Herein, we review tissue engineering strategies for regeneration of skeletal muscle, development of vasculature and nerve within the damaged muscle, and achievements in immunomodulation following VML. In addition, we discuss the limitations of current state of the art technologies and perspectives of tissue-engineered bioconstructs for muscle regeneration and functional recovery following VML.
Collapse
Affiliation(s)
- Ioannis Eugenis
- Department of Bioengineering, Stanford University, Stanford, CA, USA; Center for Tissue Regeneration, Repair, and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Di Wu
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Center for Tissue Regeneration, Repair, and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Thomas A Rando
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA; Center for Tissue Regeneration, Repair, and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA.
| |
Collapse
|
192
|
Zhen L, Creason SA, Simonovsky FI, Snyder JM, Lindhartsen SL, Mecwan MM, Johnson BW, Himmelfarb J, Ratner BD. Precision-porous polyurethane elastomers engineered for application in pro-healing vascular grafts: Synthesis, fabrication and detailed biocompatibility assessment. Biomaterials 2021; 279:121174. [PMID: 34715636 DOI: 10.1016/j.biomaterials.2021.121174] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 08/16/2021] [Indexed: 01/22/2023]
Abstract
Unmet needs for small diameter, non-biologic vascular grafts and the less-than-ideal performance of medium diameter grafts suggest opportunities for major improvements. Biomaterials that are mechanically matched to native blood vessels, reduce the foreign body capsule (FBC) and demonstrate improved integration and healing are expected to improve graft performance. In this study, we developed biostable, crosslinked polyurethane formulations and used them to fabricate scaffolds with precision-engineered 40 μm pores. We matched the scaffold mechanical properties with those of native blood vessels by optimizing the polyurethane compositions. We hypothesized that such scaffolds promote healing and mitigate the FBC. To test our hypothesis, polyurethanes with 40 μm pores, 100 μm pores, and non-porous slabs were implanted subcutaneously in mice for 3 weeks, and then were examined histologically. Our results show that 40 μm porous scaffolds elicit the highest level of angiogenesis, cellularization, and the least severe foreign body capsule (based on a refined assessment method). This study presents the first biomaterial with tuned mechanical properties and a precision engineered porous structure optimized for healing, thus can be ideal for pro-healing vascular grafts and in situ vascular engineering. In addition, these scaffolds may have wide applications in tissue engineering, drug delivery, and implantable device.
Collapse
Affiliation(s)
- Le Zhen
- Department of Chemical Engineering, University of Washington, Seattle, WA, 98195, USA; Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Sharon A Creason
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Felix I Simonovsky
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Jessica M Snyder
- Department of Comparative Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Sarah L Lindhartsen
- Histology and Imaging Core, University of Washington, Seattle, WA, 98195, USA
| | - Marvin M Mecwan
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Brian W Johnson
- Histology and Imaging Core, University of Washington, Seattle, WA, 98195, USA
| | - Jonathan Himmelfarb
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA; Department of Medicine, Division of Nephrology, University of Washington, Seattle, WA, 98195, USA; Kidney Research Institute, Seattle, WA, 98104, USA; Center for Dialysis Innovation, University of Washington, WA, 98195, USA
| | - Buddy D Ratner
- Department of Chemical Engineering, University of Washington, Seattle, WA, 98195, USA; Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA; Center for Dialysis Innovation, University of Washington, WA, 98195, USA.
| |
Collapse
|
193
|
The immune niche of the liver. Clin Sci (Lond) 2021; 135:2445-2466. [PMID: 34709406 DOI: 10.1042/cs20190654] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 09/17/2021] [Accepted: 10/08/2021] [Indexed: 12/19/2022]
Abstract
The liver is an essential organ that is critical for the removal of toxins, the production of proteins, and the maintenance of metabolic homeostasis. Behind each liver functional unit, termed lobules, hides a heterogeneous, complex, and well-orchestrated system. Despite parenchymal cells being most commonly associated with the liver's primary functionality, it has become clear that it is the immune niche of the liver that plays a central role in maintaining both local and systemic homeostasis by propagating hepatic inflammation and orchestrating its resolution. As such, the immunological processes that are at play in healthy and diseased livers are being investigated thoroughly in order to understand the underpinnings of inflammation and the potential avenues for restoring homeostasis. This review highlights recent advances in our understanding of the immune niche of the liver and provides perspectives for how the implementation of new transcriptomic, multimodal, and spatial technologies can uncover the heterogeneity, plasticity, and location of hepatic immune populations. Findings from these technologies will further our understanding of liver biology and create a new framework for the identification of therapeutic targets.
Collapse
|
194
|
Ma YH, Shi HJ, Wei QS, Deng QW, Sun JH, Liu Z, Lai BQ, Li G, Ding Y, Niu WT, Zeng YS, Zeng X. Developing a mechanically matched decellularized spinal cord scaffold for the in situ matrix-based neural repair of spinal cord injury. Biomaterials 2021; 279:121192. [PMID: 34700225 DOI: 10.1016/j.biomaterials.2021.121192] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 10/15/2021] [Accepted: 10/18/2021] [Indexed: 12/16/2022]
Abstract
Tissue engineering is a promising strategy to repair spinal cord injury (SCI). However, a bioscaffold with mechanical properties that match those of the pathological spinal cord tissue and a pro-regenerative matrix that allows robust neurogenesis for overcoming post-SCI scar formation has yet to be developed. Here, we report that a mechanically enhanced decellularized spinal cord (DSC) scaffold with a thin poly (lactic-co-glycolic acid) (PLGA) outer shell may fulfill the requirements for effective in situ neuroengineering after SCI. Using chemical extraction and electrospinning methods, we successfully constructed PLGA thin shell-ensheathed DSC scaffolds (PLGA-DSC scaffolds) in a way that removed major inhibitory components while preserving the permissive matrix. The DSCs exhibited good cytocompatibility with neural stem cells (NSCs) and significantly enhanced their differentiation toward neurons in vitro. Due to the mechanical reinforcement, the implanted PLGA-DSC scaffolds showed markedly increased resilience to infiltration by myofibroblasts and the deposition of dense collagen matrix, thereby creating a neurogenic niche favorable for the targeted migration, residence and neuronal differentiation of endogenous NSCs after SCI. Furthermore, PLGA-DSC presented a mild immunogenic property but prominent ability to polarize macrophages from the M1 phenotype to the M2 phenotype, leading to significant tissue regeneration and functional restoration after SCI. Taken together, the results demonstrate that the mechanically matched PLGA-DSC scaffolds show promise for effective tissue repair after SCI.
Collapse
Affiliation(s)
- Yuan-Huan Ma
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, 510080, China; Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, Guangdong Province, 510080, China; Guangdong Key Laboratory of Age-Related Cardiocerebral Diseases, Institute of Neurology, Guangdong Medical University, Zhanjiang, Guangdong Province, 524023, China; Guangzhou Institute of Clinical Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong Province, 510180, PR China
| | - Hui-Juan Shi
- Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, Guangdong Province, 510080, China
| | - Qing-Shuai Wei
- Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, Guangdong Province, 510080, China
| | - Qing-Wen Deng
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, 510080, China
| | - Jia-Hui Sun
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, 510080, China
| | - Zhou Liu
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, 510080, China; Guangdong Key Laboratory of Age-Related Cardiocerebral Diseases, Institute of Neurology, Guangdong Medical University, Zhanjiang, Guangdong Province, 524023, China
| | - Bi-Qin Lai
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, 510080, China; Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, Guangdong Province, 510080, China; Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, 510080, China
| | - Ge Li
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, 510080, China; Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, Guangdong Province, 510080, China; Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, 510080, China
| | - Ying Ding
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, 510080, China; Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, Guangdong Province, 510080, China; Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, 510080, China
| | - Wan-Ting Niu
- Department of Orthopedics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Yuan-Shan Zeng
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, 510080, China; Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, Guangdong Province, 510080, China; Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, 510080, China; Institute of Spinal Cord Injury, Sun Yat-sen University, Guangzhou, Guangdong Province, 510120, China
| | - Xiang Zeng
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, 510080, China; Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, Guangdong Province, 510080, China; Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, 510080, China; Institute of Spinal Cord Injury, Sun Yat-sen University, Guangzhou, Guangdong Province, 510120, China.
| |
Collapse
|
195
|
Lafuente-Gracia L, Borgiani E, Nasello G, Geris L. Towards in silico Models of the Inflammatory Response in Bone Fracture Healing. Front Bioeng Biotechnol 2021; 9:703725. [PMID: 34660547 PMCID: PMC8514728 DOI: 10.3389/fbioe.2021.703725] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 09/07/2021] [Indexed: 12/21/2022] Open
Abstract
In silico modeling is a powerful strategy to investigate the biological events occurring at tissue, cellular and subcellular level during bone fracture healing. However, most current models do not consider the impact of the inflammatory response on the later stages of bone repair. Indeed, as initiator of the healing process, this early phase can alter the regenerative outcome: if the inflammatory response is too strongly down- or upregulated, the fracture can result in a non-union. This review covers the fundamental information on fracture healing, in silico modeling and experimental validation. It starts with a description of the biology of fracture healing, paying particular attention to the inflammatory phase and its cellular and subcellular components. We then discuss the current state-of-the-art regarding in silico models of the immune response in different tissues as well as the bone regeneration process at the later stages of fracture healing. Combining the aforementioned biological and computational state-of-the-art, continuous, discrete and hybrid modeling technologies are discussed in light of their suitability to capture adequately the multiscale course of the inflammatory phase and its overall role in the healing outcome. Both in the establishment of models as in their validation step, experimental data is required. Hence, this review provides an overview of the different in vitro and in vivo set-ups that can be used to quantify cell- and tissue-scale properties and provide necessary input for model credibility assessment. In conclusion, this review aims to provide hands-on guidance for scientists interested in building in silico models as an additional tool to investigate the critical role of the inflammatory phase in bone regeneration.
Collapse
Affiliation(s)
- Laura Lafuente-Gracia
- Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium.,Prometheus: Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| | - Edoardo Borgiani
- Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium.,Prometheus: Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.,Biomechanics Research Unit, GIGA in silico Medicine, University of Liège, Liège, Belgium
| | - Gabriele Nasello
- Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium.,Prometheus: Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.,Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| | - Liesbet Geris
- Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium.,Prometheus: Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.,Biomechanics Research Unit, GIGA in silico Medicine, University of Liège, Liège, Belgium.,Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| |
Collapse
|
196
|
Control of satellite cell function in muscle regeneration and its disruption in ageing. Nat Rev Mol Cell Biol 2021; 23:204-226. [PMID: 34663964 DOI: 10.1038/s41580-021-00421-2] [Citation(s) in RCA: 231] [Impact Index Per Article: 57.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/01/2021] [Indexed: 12/19/2022]
Abstract
Skeletal muscle contains a designated population of adult stem cells, called satellite cells, which are generally quiescent. In homeostasis, satellite cells proliferate only sporadically and usually by asymmetric cell division to replace myofibres damaged by daily activity and maintain the stem cell pool. However, satellite cells can also be robustly activated upon tissue injury, after which they undergo symmetric divisions to generate new stem cells and numerous proliferating myoblasts that later differentiate to muscle cells (myocytes) to rebuild the muscle fibre, thereby supporting skeletal muscle regeneration. Recent discoveries show that satellite cells have a great degree of population heterogeneity, and that their cell fate choices during the regeneration process are dictated by both intrinsic and extrinsic mechanisms. Extrinsic cues come largely from communication with the numerous distinct stromal cell types in their niche, creating a dynamically interactive microenvironment. This Review discusses the role and regulation of satellite cells in skeletal muscle homeostasis and regeneration. In particular, we highlight the cell-intrinsic control of quiescence versus activation, the importance of satellite cell-niche communication, and deregulation of these mechanisms associated with ageing. The increasing understanding of how satellite cells are regulated will help to advance muscle regeneration and rejuvenation therapies.
Collapse
|
197
|
Al-Hejailan RS, Bakheet RH, Al-Saud MM, Al-Jufan MB, Al-Hindas HM, Al-Qattan SM, Al-Muhanna MK, Parhar RS, Conca W, Hansmann J, Collison KS, Walles H, Al-Mohanna FA. Toward allogenizing a xenograft: Xenogeneic cardiac scaffolds recellularized with human-induced pluripotent stem cells do not activate human naïve neutrophils. J Biomed Mater Res B Appl Biomater 2021; 110:691-701. [PMID: 34619017 DOI: 10.1002/jbm.b.34948] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 08/31/2021] [Accepted: 09/22/2021] [Indexed: 11/12/2022]
Abstract
The limited availability of human donor organs suitable for transplantation has resulted in ever-increasing patient waiting lists globally. Xenotransplantation is considered a potential option, but is yet to reach clinical practice. Although remarkable progress has been made in overcoming immunological rejection, issues with functionality are still to be resolved. Bioengineering approaches have been used to create cardiac tissues with optimized functions. The use of decellularized xenogeneic cardiac tissues seeded with donor-derived cardiac cells may prove to be a viable strategy as supporting structures of the native tissue such as vasculature can be utilized. Here we used sequential perfusion to decellularize adult rat hearts. The acellular scaffolds were reseeded with human endothelial cells, human fibroblasts, human mesenchymal stem cells, and cardiac cells derived from human-induced pluripotent stem cells. The ability of the resultant recellularized rat scaffolds to activate human naïve neutrophils in vitro was investigated to measure xenogeneic recognition. Our results demonstrate that in contrast to cadaveric xenogeneic hearts, acellular and recellularized xenogeneic scaffolds did not activate human naïve neutrophils and suggest that decellularization removes the xenogeneic antigens that lead to human naïve neutrophil activation thus allowing human cells to populate the now "allogenized" xenogeneic scaffolds.
Collapse
Affiliation(s)
- Reem S Al-Hejailan
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia.,Department of Heart Centre, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Razan H Bakheet
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Mashael M Al-Saud
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | | | - Hussain M Al-Hindas
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Somaya M Al-Qattan
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Muhanna K Al-Muhanna
- Tissue Engineering and Regenerative Medicine (TERM), Würzburg University, Würzburg, Germany
| | - Ranjit S Parhar
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Walter Conca
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Jan Hansmann
- Department of Heart Centre, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Kate S Collison
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Heike Walles
- Department of Heart Centre, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Futwan A Al-Mohanna
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| |
Collapse
|
198
|
Seo BR, Payne CJ, McNamara SL, Freedman BR, Kwee BJ, Nam S, de Lázaro I, Darnell M, Alvarez JT, Dellacherie MO, Vandenburgh HH, Walsh CJ, Mooney DJ. Skeletal muscle regeneration with robotic actuation-mediated clearance of neutrophils. Sci Transl Med 2021; 13:eabe8868. [PMID: 34613813 DOI: 10.1126/scitranslmed.abe8868] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Bo Ri Seo
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA.,Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Christopher J Payne
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA.,Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA.,Viam Inc., New York, NY 10023, USA
| | - Stephanie L McNamara
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA.,Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Benjamin R Freedman
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA.,Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Brian J Kwee
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA.,Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Sungmin Nam
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA.,Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Irene de Lázaro
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA.,Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Max Darnell
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA.,Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Jonathan T Alvarez
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA.,Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Maxence O Dellacherie
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA.,Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Herman H Vandenburgh
- Department of Pathology and Lab Medicine, Brown University, Providence, RI 02912, USA
| | - Conor J Walsh
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA.,Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - David J Mooney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA.,Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| |
Collapse
|
199
|
Hosseini M, Shafiee A. Engineering Bioactive Scaffolds for Skin Regeneration. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2101384. [PMID: 34313003 DOI: 10.1002/smll.202101384] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 03/24/2021] [Indexed: 06/13/2023]
Abstract
Large skin wounds pose a major clinical challenge. Scarcity of donor site and postsurgical scarring contribute to the incomplete or partial loss of function and aesthetic concerns in skin wound patients. Currently, a wide variety of skin grafts are being applied in clinical settings. Scaffolds are used to overcome the issues related to the misaligned architecture of the repaired skin tissues. The current review summarizes the contribution of biomaterials to wound healing and skin regeneration and addresses the existing limitations in skin grafting. Then, the clinically approved biologic and synthetic skin substitutes are extensively reviewed. Next, the techniques for modification of skin grafts aiming for enhanced tissue regeneration are outlined, and a summary of different growth factor delivery systems using biomaterials is presented. Considering the significant progress in biomaterial science and manufacturing technologies, the idea of biomaterial-based skin grafts with the ability for scarless wound healing and reconstructing full skin organ is more achievable than ever.
Collapse
Affiliation(s)
- Motaharesadat Hosseini
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Kelvin Grove, Brisbane, QLD, 4059, Australia
| | - Abbas Shafiee
- Herston Biofabrication Institute, Metro North Hospital and Health Service, Brisbane, QLD, 4029, Australia
- Royal Brisbane and Women's Hospital, Metro North Hospital and Health Service, Brisbane, QLD, 4029, Australia
- UQ Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, QLD, 4102, Australia
| |
Collapse
|
200
|
T lymphocytes as critical mediators in tissue regeneration, fibrosis, and the foreign body response. Acta Biomater 2021; 133:17-33. [PMID: 33905946 DOI: 10.1016/j.actbio.2021.04.023] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 03/23/2021] [Accepted: 04/13/2021] [Indexed: 12/16/2022]
Abstract
Research on the foreign body response (FBR) to biomaterial implants has been focused on the roles that the innate immune system has on mediating tolerance or rejection of implants. However, the immune system also involves the adaptive immune response and it must be included in order to form a complete picture of the response to biomaterials and medical implants. In this review, we explore recent understanding about the roles of adaptive immune cells, specifically T cells, in modulating the immune response to biomaterial implants. The immune response to implants elicits a delicate balance between tissue repair and fibrosis that is mainly regulated by three types of T helper cell responses -T helper type 1, T helper type 2, and T helper type 17- and their crosstalk with innate immune cells. Interestingly, many T cell response mechanisms to implants overlap with the process of fibrosis or repair in different tissues. This review explores the fibrotic and regenerative T cell biology and draws parallels to T cell responses to biomaterials. Additionally, we also explore the biomedical engineering advancements in biomaterial applications in designing particle and scaffold systems to modulate T cell activity for therapeutics and devices. Not only do the deliberate engineering design of physical and chemical material properties and the direct genetic modulation of T cells not only offer insights to T cell biology, but they also present different platforms to develop immunomodulatory biomaterials. Thus, an in-depth understanding of T cells' roles can help to navigate the biomaterial-immune interactions and reconsider the long-lasting adaptive immune response to implants, which, in the end, contribute to the design of immunomodulatory medical implants that can advance the next generation of regenerative therapy. STATEMENT OF SIGNIFICANCE: This review article integrates knowledge of adaptive immune responses in tissue damage, wound healing, and medical device implantation. These three fields, often not discussed in conjunction, are important to consider when evaluating and designing biomaterials. Through incorporation of basic biological research alongside engineering research, we provide an important lens through which to evaluate adaptive immune contributions to regenerative medicine and medical device development.
Collapse
|