401
|
Aguilar CA, Greising SM, Watts A, Goldman SM, Peragallo C, Zook C, Larouche J, Corona BT. Multiscale analysis of a regenerative therapy for treatment of volumetric muscle loss injury. Cell Death Discov 2018; 4:33. [PMID: 29531830 PMCID: PMC5841404 DOI: 10.1038/s41420-018-0027-8] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 12/22/2017] [Indexed: 01/01/2023] Open
Abstract
Skeletal muscle possesses a remarkable capacity to regenerate when injured, but when confronted with major traumatic injury resulting in volumetric muscle loss (VML), the regenerative process consistently fails. The loss of muscle tissue and function from VML injury has prompted development of a suite of therapeutic approaches but these strategies have proceeded without a comprehensive understanding of the molecular landscape that drives the injury response. Herein, we administered a VML injury in an established rodent model and monitored the evolution of the healing phenomenology over multiple time points using muscle function testing, histology, and expression profiling by RNA sequencing. The injury response was then compared to a regenerative medicine treatment using orthotopic transplantation of autologous minced muscle grafts (~1 mm3 tissue fragments). A chronic inflammatory and fibrotic response was observed at all time points following VML. These results suggest that the pathological response to VML injury during the acute stage of the healing response overwhelms endogenous and therapeutic regenerative processes. Overall, the data presented delineate key molecular characteristics of the pathobiological response to VML injury that are critical effectors of effective regenerative treatment paradigms.
Collapse
Affiliation(s)
- Carlos A. Aguilar
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI USA
| | - Sarah M. Greising
- Extremity Trauma and Regenerative Medicine, United States Army Institute of Surgical Research, Fort Sam Houston, San Antonio, TX USA
| | - Alain Watts
- Massachusetts Institute of Technology - Lincoln Laboratory, Lexington, MA USA
| | - Stephen M. Goldman
- Extremity Trauma and Regenerative Medicine, United States Army Institute of Surgical Research, Fort Sam Houston, San Antonio, TX USA
| | - Chelsea Peragallo
- Massachusetts Institute of Technology - Lincoln Laboratory, Lexington, MA USA
| | - Christina Zook
- Massachusetts Institute of Technology - Lincoln Laboratory, Lexington, MA USA
| | - Jacqueline Larouche
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI USA
| | - Benjamin T. Corona
- Extremity Trauma and Regenerative Medicine, United States Army Institute of Surgical Research, Fort Sam Houston, San Antonio, TX USA
| |
Collapse
|
402
|
Carpenter RA, Kwak JG, Peyton SR, Lee J. Implantable pre-metastatic niches for the study of the microenvironmental regulation of disseminated human tumour cells. Nat Biomed Eng 2018; 2:915-929. [PMID: 30906645 PMCID: PMC6424369 DOI: 10.1038/s41551-018-0307-x] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 09/06/2018] [Indexed: 02/07/2023]
Abstract
Cancer survivors often carry disseminated tumour cells (DTCs), yet owing to DTC dormancy they do not relapse from treatment. Understanding how the local microenvironment regulates the transition of DTCs from a quiescent state to active proliferation could suggest new therapeutic strategies to prevent or delay the formation of metastases. Here, we show that implantable biomaterial microenvironments incorporating human stromal cells, immune cells and cancer cells can be used to examine the post-dissemination phase of the evolution of the tumour microenvironment. After subdermal implantation in mice, porous hydrogel scaffolds seeded with human bone marrow stromal cells form a vascularized niche and recruit human circulating tumour cells released from an orthotopic prostate tumour xenograft. Systemic injection of human peripheral blood mononuclear cells slowed the evolution of the active metastatic niches but did not change the rate of overt metastases, as the ensuing inflammation promoted the formation of DTC colonies. Implantable pre-metastatic niches might enable the study of DTC colonization and proliferation, and facilitate the development of effective anti-metastatic therapies.
Collapse
Affiliation(s)
- Ryan A Carpenter
- Department of Chemical Engineering, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, USA
| | - Jun-Goo Kwak
- Department of Chemical Engineering, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, USA
| | - Shelly R Peyton
- Department of Chemical Engineering, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, USA
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, MA, USA
| | - Jungwoo Lee
- Department of Chemical Engineering, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, USA.
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, MA, USA.
| |
Collapse
|
403
|
Richardson RJ. Parallels between vertebrate cardiac and cutaneous wound healing and regeneration. NPJ Regen Med 2018; 3:21. [PMID: 30416753 PMCID: PMC6220283 DOI: 10.1038/s41536-018-0059-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 10/12/2018] [Indexed: 12/11/2022] Open
Abstract
The cellular events that contribute to tissue healing of non-sterile wounds to the skin and ischaemic injury to internal organs such as the heart share remarkable similarities despite the differences between these injury types and organs. In adult vertebrates, both injuries are characterised by a complex series of overlapping events involving multiple different cell types and cellular interactions. In adult mammals both tissue-healing processes ultimately lead to the permanent formation of a fibrotic, collagenous scar, which can have varying effects on tissue function depending on the site and magnitude of damage. Extensive scarring in the heart as a result of a severe myocardial infarction contributes to ventricular dysfunction and the progression of heart failure. Some vertebrates such as adult zebrafish, however, retain a more embryonic capacity for scar-free tissue regeneration in many tissues including the skin and heart. In this review, the similarities and differences between these different types of wound healing are discussed, with special attention on recent advances in regenerative, non-scarring vertebrate models such as the zebrafish.
Collapse
Affiliation(s)
- Rebecca J Richardson
- School of Physiology, Pharmacology and Neuroscience, Faculty of Biomedical Sciences, University of Bristol, Bristol, UK
| |
Collapse
|
404
|
Aktuelle pathophysiologische Entwicklungen bei fibrosierenden Erkrankungen: Ansatzpunkte für neue Konzepte in der Therapie. Hautarzt 2018; 69:885-891. [DOI: 10.1007/s00105-018-4256-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
405
|
He Y, Mu C, Shen X, Yuan Z, Liu J, Chen W, Lin C, Tao B, Liu B, Cai K. Peptide LL-37 coating on micro-structured titanium implants to facilitate bone formation in vivo via mesenchymal stem cell recruitment. Acta Biomater 2018; 80:412-424. [PMID: 30266635 DOI: 10.1016/j.actbio.2018.09.036] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 09/02/2018] [Accepted: 09/24/2018] [Indexed: 01/06/2023]
Abstract
Titanium (Ti) and Ti-alloys were widely used in clinic orthopedics, however, the insufficient bone formation surrounding Ti-based implants still limited their biological performances. Surface modification of Ti substrates is essential to improve their interactions with bone-forming cells and bone tissue. In this study, we modified Ti substrates by coating peptide LL-37 onto micro-structured Ti substrates and aimed to (i) induce mesenchymal stem cells (MSCs) migration both in vitro and in vivo, (ii) facilitate osteogenic differentiation of MSCs and new bone formation. The surface micro-structured Ti substrates with hydroxyapatite deposition were fabricated by a two-step method including micro-arc oxidation (MAO) and hydrothermal treatment. LL-37 was loaded on micro-structured Ti substrates with the assistance of polydopamine coating. We confirmed that surface-modified Ti substrates benefited viability, adhesion, migration and osteogenic differentiation of MSCs in vitro. In a femur-defect rat model, the surface-modified Ti implants effectively induced CD29+/CD90+ positive cells migration in one week after implantation. According to the results of H&E, Masson's trichrome staining and immunohistochemical staining of OCN, OPN and collagen I, the targeted Ti implants exhibited significant new bone formation after implantation for 4 weeks. These results indicate that the surface modification of Ti samples facilitated bone formation through MSCs recruitment. STATEMENT OF SIGNIFICANCE: The inherent surface bioinertness of titanium (Ti) and Ti-alloys still limits their biological performances in clinical applications. Recently, the strategy of mesenchymal stem cells (MSCs) recruitment has been proposed to improve the osteointegration of bone implants. Herein, we reports the surface modification of Ti implants from the point of MSCs recruitment. Peptide LL-37 was coated on micro-structured Ti substrates to (i) recruit MSCs, (ii) regulate bio-physiological performance of MSCs, and (iii) facilitate bone formation in vivo. Our results improve the understanding of the interaction between Ti implants and MSCs, and provide a promising strategy of MSCs recruitment in the design of bone repair related biomaterials.
Collapse
Affiliation(s)
- Ye He
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, PR China
| | - Caiyun Mu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, PR China
| | - Xinkun Shen
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, PR China; School of Life Science, Chongqing University, Chongqing 400044, PR China
| | - Zhang Yuan
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, PR China
| | - Ju Liu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, PR China
| | - Weizhen Chen
- First Affiliated Hospital, College of Medicine, Zhejiang University, Key Laboratory of Clinical In Vitro Diagnostic Techniques of Zhejiang Province, Hangzhou, Zhejiang 310003, PR China
| | - Chuanchuan Lin
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, PR China
| | - Bailong Tao
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, PR China
| | - Bin Liu
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, School of Life Science, Southwest University, Chongqing 400715, PR China
| | - Kaiyong Cai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, PR China.
| |
Collapse
|
406
|
He XT, Wang J, Li X, Yin Y, Sun HH, Chen FM. The Critical Role of Cell Homing in Cytotherapeutics and Regenerative Medicine. ADVANCED THERAPEUTICS 2018. [DOI: 10.1002/adtp.201800098] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Xiao-Tao He
- State Key Laboratory of Military Stomatology; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
- National Clinical Research Center for Oral Diseases; Department of Periodontology; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
- Shaanxi Engineering Research Center for Dental Materials, and Advanced Manufacture; Biomaterials Unit; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
| | - Jia Wang
- State Key Laboratory of Military Stomatology; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
- Shaanxi Engineering Research Center for Dental Materials, and Advanced Manufacture; Biomaterials Unit; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
| | - Xuan Li
- State Key Laboratory of Military Stomatology; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
- National Clinical Research Center for Oral Diseases; Department of Periodontology; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
- Shaanxi Engineering Research Center for Dental Materials, and Advanced Manufacture; Biomaterials Unit; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
| | - Yuan Yin
- State Key Laboratory of Military Stomatology; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
- Shaanxi Engineering Research Center for Dental Materials, and Advanced Manufacture; Biomaterials Unit; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
| | - Hai-Hua Sun
- National Clinical Research Center for Oral Diseases; Department of Periodontology; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
| | - Fa-Ming Chen
- State Key Laboratory of Military Stomatology; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
- National Clinical Research Center for Oral Diseases; Department of Periodontology; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
- Shaanxi Engineering Research Center for Dental Materials, and Advanced Manufacture; Biomaterials Unit; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
| |
Collapse
|
407
|
Estrellas KM, Chung L, Cheu LA, Sadtler K, Majumdar S, Mula J, Wolf MT, Elisseeff JH, Wagner KR. Biological scaffold-mediated delivery of myostatin inhibitor promotes a regenerative immune response in an animal model of Duchenne muscular dystrophy. J Biol Chem 2018; 293:15594-15605. [PMID: 30139748 DOI: 10.1074/jbc.ra118.004417] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 08/16/2018] [Indexed: 01/16/2023] Open
Abstract
Recent studies have reported that the immune system significantly mediates skeletal muscle repair and regeneration. Additionally, biological scaffolds have been shown to play a role in polarizing the immune microenvironment toward pro-myogenic outcomes. Moreover, myostatin inhibitors are known to promote muscle regeneration and ameliorate fibrosis in animal models of Duchenne muscular dystrophy (DMD), a human disease characterized by chronic muscle degeneration. Biological scaffolds and myostatin inhibition can potentially influence immune-mediated regeneration in the dystrophic environment, but have not been evaluated together. Toward this end, here we created an injectable biological scaffold composed of hyaluronic acid and processed skeletal muscle extracellular matrix. This material formed a cytocompatible hydrogel at physiological temperatures in vitro When injected subfascially above the tibialis anterior muscles of both WT and dystrophic mdx-5Cv mice, a murine model of DMD, the hydrogel spreads across the entire muscle before completely degrading at 3 weeks in vivo We found that the hydrogel is associated with CD206+ pro-regenerative macrophage polarization and elevated anti-inflammatory cytokine expression in both WT and dystrophic mice. Co-injection of both hydrogel and myostatin inhibitor significantly increased FoxP3+ regulatory T cell modulation and Foxp3 gene expression in the scaffold immune microenvironment. Finally, delivery of myostatin inhibitor with the hydrogel increased its bioactivity in vivo, and transplantation of immortalized human myoblasts with the hydrogel promoted their survival in vivo This study identifies a key role for biological scaffolds and myostatin inhibitors in modulating a pro-regenerative immune microenvironment in dystrophic muscle.
Collapse
Affiliation(s)
- Kenneth M Estrellas
- From the Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, Maryland 21205.,the Translational Tissue Engineering Center and
| | - Liam Chung
- the Translational Tissue Engineering Center and.,Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231
| | - Lindsay A Cheu
- the Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611
| | - Kaitlyn Sadtler
- the David H. Koch Institute for Integrative Cancer Research, Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142.,the Department of Anesthesiology, Boston Children's Hospital, Boston, Massachusetts 02115
| | | | - Jyothi Mula
- the NCI at Frederick, National Institutes of Health, Frederick, Maryland 21702, and
| | - Matthew T Wolf
- the Translational Tissue Engineering Center and.,Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231
| | - Jennifer H Elisseeff
- the Translational Tissue Engineering Center and .,Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231
| | - Kathryn R Wagner
- From the Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, Maryland 21205, .,the Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287
| |
Collapse
|
408
|
Macrophage response to hydrophilic biomaterials regulates MSC recruitment and T-helper cell populations. Biomaterials 2018; 182:202-215. [PMID: 30138783 DOI: 10.1016/j.biomaterials.2018.08.029] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 08/04/2018] [Accepted: 08/10/2018] [Indexed: 12/20/2022]
Abstract
Successful biomaterial implantation can be achieved by controlling the activation of the immune system. The innate immune system is typically the focus on synthetic material compatibility, but this study shows an effect of surface properties in the innate as well as the adaptive systems. These studies look at how macrophages respond to the implanted materials by releasing factors to regulate the microenvironment and recruit additional cells. Our research demonstrates how macrophage response to material surface properties can create changes in the adaptive immune response by altering T-helper cell populations and stem cell recruitment. Titanium (Ti) implants of varying wettability (rough, and rough-hydrophilic) were placed in the femur of 10-week-old male C57Bl/6, or macrophage ablated clodronate liposome injected and transgenic MaFIA (C57BL/6-Tg(Csf1r-EGFP-NGFR/FKBP1A/TNFRSF6)2Bck/J) mice. The microenvironment surrounding Ti implants was assessed using custom PCR arrays at 3 and 7 days following implantation. Changes in specific T-helper, macrophage and stem cell populations were evaluated locally at the implant surface and systemically in the contralateral leg bone marrow and spleen by flow cytometry at 1, 3 and 7 days. Macrophage importance in T-helper and stem cell population changes with metallic surfaces was examined in both in vitro and in vivo with macrophage ablation models. We demonstrate that surface modifications applied to titanium implants to increase surface roughness and wettability can polarize the adaptive immune response towards a Th2, pro-wound healing phenotype, leading to faster resolution of inflammation and increased stem cell recruitment around rough hydrophilic implants with macrophages present.
Collapse
|
409
|
Juillerat-Jeanneret L, Aubert JD, Mikulic J, Golshayan D. Fibrogenic Disorders in Human Diseases: From Inflammation to Organ Dysfunction. J Med Chem 2018; 61:9811-9840. [DOI: 10.1021/acs.jmedchem.8b00294] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Lucienne Juillerat-Jeanneret
- Transplantation Center and Transplantation Immunopathology Laboratory, Department of Medicine, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - John-David Aubert
- Pneumology Division and Transplantation Center, Centre Hospitalier Universitaire Vaudois (CHUV), CH1011 Lausanne, Switzerland
| | - Josip Mikulic
- Transplantation Center and Transplantation Immunopathology Laboratory, Department of Medicine, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Dela Golshayan
- Transplantation Center and Transplantation Immunopathology Laboratory, Department of Medicine, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| |
Collapse
|
410
|
Chen S, Li R, Li X, Xie J. Electrospinning: An enabling nanotechnology platform for drug delivery and regenerative medicine. Adv Drug Deliv Rev 2018; 132:188-213. [PMID: 29729295 DOI: 10.1016/j.addr.2018.05.001] [Citation(s) in RCA: 222] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 04/03/2018] [Accepted: 05/01/2018] [Indexed: 02/06/2023]
Abstract
Electrospinning provides an enabling nanotechnology platform for generating a rich variety of novel structured materials in many biomedical applications including drug delivery, biosensing, tissue engineering, and regenerative medicine. In this review article, we begin with a thorough discussion on the method of producing 1D, 2D, and 3D electrospun nanofiber materials. In particular, we emphasize on how the 3D printing technology can contribute to the improvement of traditional electrospinning technology for the fabrication of 3D electrospun nanofiber materials as drug delivery devices/implants, scaffolds or living tissue constructs. We then highlight several notable examples of electrospun nanofiber materials in specific biomedical applications including cancer therapy, guiding cellular responses, engineering in vitro 3D tissue models, and tissue regeneration. Finally, we finish with conclusions and future perspectives of electrospun nanofiber materials for drug delivery and regenerative medicine.
Collapse
|
411
|
Sun G, Shen YI, Harmon JW. Engineering Pro-Regenerative Hydrogels for Scarless Wound Healing. Adv Healthc Mater 2018; 7:e1800016. [PMID: 29663707 DOI: 10.1002/adhm.201800016] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Revised: 02/17/2018] [Indexed: 12/21/2022]
Abstract
Skin and skin appendages protect the body from harmful environment and prevent internal organs from dehydration. Superficial epidermal wounds usually heal without scarring, however, deep dermal wound healing commonly ends up with nonfunctioning scar formation with substantial loss of skin appendage. Wound healing is one of the most complex dynamic biological processes, during which a cascade of biomolecules combine with stem cell influx and matrix synthesis and synergistically contribute to wound healing at all levels. Although many approaches have been investigated to restore complete skin, the clinically effective therapy is still unavailable and the regeneration of perfect skin still remains a significant challenge. The complete mechanism behind scarless skin regeneration still requires further investigation. Fortunately, recent advancement in regenerative medicine empowers us more than ever to restore tissue in a regenerative manner. Many studies have elucidated and reviewed the contribution of stem cells and growth factors to scarless wound healing. This article focuses on recent advances in scarless wound healing, especially strategies to engineer pro-regenerative scaffolds to restore damaged skin in a regenerative manner.
Collapse
Affiliation(s)
- Guoming Sun
- Sunogel Biotechnologies Inc.; 9 W Ridgely Road Ste 270 Lutherville Timonium MD 21093 USA
| | - Yu-I Shen
- Sunogel Biotechnologies Inc.; 9 W Ridgely Road Ste 270 Lutherville Timonium MD 21093 USA
| | - John W. Harmon
- Department of Surgery and the Hendrix Burn Lab; Johns Hopkins University School of Medicine; Baltimore MD 21224 USA
| |
Collapse
|
412
|
Chen W, Xu K, Tao B, Dai L, Yu Y, Mu C, Shen X, Hu Y, He Y, Cai K. Multilayered coating of titanium implants promotes coupled osteogenesis and angiogenesis in vitro and in vivo. Acta Biomater 2018; 74:489-504. [PMID: 29702291 DOI: 10.1016/j.actbio.2018.04.043] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 03/27/2018] [Accepted: 04/22/2018] [Indexed: 12/20/2022]
Abstract
We used surface-modified titanium (Ti) substrates with a multilayered structure composed of chitosan-catechol (Chi-C), gelatin (Gel) and hydroxyapatite (HA) nanofibers, which were previously shown to improve osteogenesis, as a platform to investigate the interaction of osteogenesis and angiogenesis during bone healing. Combined techniques of Transwell co-culture, wound healing assay, enzyme linked immunosorbent assay (ELISA), quantitative real-time polymerase chain reaction (qRT-PCR), western blotting and immunohistochemical staining were used to evaluate adhesion, morphology and migration of adipose-derived mesenchymal stem cells (Ad-MSCs) and human umbilical vein endothelial cells (HUVECs) grown on different Ti substrates. We investigated the effect of substrates on the osteogenic differentiation of Ad-MSCs and reciprocal paracrine effects of Ad-MSCs on HUVECs or vice versa. The multilayered Ti substrates directly regulated the cellular functions of Ad-MSCs and angiogenic HUVECs and mediated communication between them by enhancing paracrine effects via cell-matrix interactions in vitro. The in vivo results showed that the change of microenvironment induced by surface-modified Ti implants promoted the adhesion, recruitment and proliferation of MSCs and facilitated coupled osteogenesis and angiogenesis in bone healing. The study proved that multilayer-film-coated Ti substrates positively mediated cellular biological function in vitro and improved bone healing in vivo. STATEMENT OF SIGNIFICANCE Recent studies have revealed that osteogenesis and angiogenesis are coupled, and that communication between osteoblasts and endothelial cells is essential for bone healing and remodeling processes; however, these conclusions only result from in vitro studies or in vivo studies using transgenic murine models. Relatively little is known about the communication between osteoblasts and endothelial cells in peri-implants during bone healing processes. Our results revealed the cellular/molecular mechanism of how multilayered Ti substrates mediate reciprocal paracrine effects between adipose-derived mesenchymal stem cells and human umbilical vein endothelial cells; moreover, the interactions between the cell-matrix and peri-implant was proven in vivo with enhanced bone healing. This study contributes to our understanding of the fundamental mechanisms of angiogenesis and osteogenesis that affect peri-implantation, and thus, provides new insights into the design of future high-quality orthopedic implants.
Collapse
|
413
|
Kang H, Jung HJ, Kim SK, Wong DSH, Lin S, Li G, Dravid VP, Bian L. Magnetic Manipulation of Reversible Nanocaging Controls In Vivo Adhesion and Polarization of Macrophages. ACS NANO 2018; 12:5978-5994. [PMID: 29767957 DOI: 10.1021/acsnano.8b02226] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Macrophages are key immune cells that perform various physiological functions, such as the maintenance of homeostasis, host defense, disease progression, and tissue regeneration. Macrophages adopt distinctly polarized phenotypes, such as pro-inflammatory M1 phenotype or anti-inflammatory (pro-healing) M2 phenotype, to execute disparate functions. The remotely controlled reversible uncaging of bioactive ligands, such as Arg-Gly-Asp (RGD) peptide, is an appealing approach for temporally regulating the adhesion and resultant polarization of macrophages on implants in vivo. Here, we utilize physical and reversible uncaging of RGD by a magnetic field that allows facile tissue penetration. We first conjugated a RGD-bearing gold nanoparticle (GNP) to the substrate and then a magnetic nanocage (MNC) to the GNP via a flexible linker to form the heterodimeric nanostructure. We magnetically manipulated nanoscale displacement of MNC and thus its proximity to the GNP to reversibly uncage and cage RGD. The uncaging of RGD temporally promoted the adhesion and subsequent M2 polarization of macrophages while inhibiting their M1 polarization both in vitro and in vivo. The RGD uncaging-mediated adhesion and M2 polarization of macrophages involved rho-associated protein kinase signaling. This study demonstrates physical and reversible uncaging of RGD to regulate the adhesion and polarization of host macrophages in vivo. This approach of magnetically regulating the heterodimer conformation for physical and reversible uncaging of RGD offers the promising potential to manipulate inflammatory or tissue-regenerative immune responses to the implants in vivo.
Collapse
Affiliation(s)
- Heemin Kang
- Department of Biomedical Engineering , The Chinese University of Hong Kong , Ma Liu Shui , Hong Kong, China
| | - Hee Joon Jung
- International Institute for Nanotechnology , Evanston , Illinois 60208 , United States
| | - Sung Kyu Kim
- International Institute for Nanotechnology , Evanston , Illinois 60208 , United States
| | - Dexter Siu Hong Wong
- Department of Biomedical Engineering , The Chinese University of Hong Kong , Ma Liu Shui , Hong Kong, China
| | - Sien Lin
- Department of Pharmacology, Guangdong Key Laboratory for Research and Development of Natural Drugs , Guangdong Medical University , Zhanjiang , Guangdong 510000 , China
| | - Gang Li
- The CUHK-ACC Space Medicine Centre on Health Maintenance of Musculoskeletal System , The Chinese University of Hong Kong Shenzhen Research Institute , Shenzhen 518172 , China
| | - Vinayak P Dravid
- International Institute for Nanotechnology , Evanston , Illinois 60208 , United States
| | - Liming Bian
- Department of Biomedical Engineering , The Chinese University of Hong Kong , Ma Liu Shui , Hong Kong, China
- Department of Pharmacology, Guangdong Key Laboratory for Research and Development of Natural Drugs , Guangdong Medical University , Zhanjiang , Guangdong 510000 , China
- Translational Research Centre of Regenerative Medicine and 3D Printing Technologies of Guangzhou Medical University , The Third Affiliated Hospital of Guangzhou Medical University , Guangzhou , Guangdong 510000 , China
- Shenzhen Research Institute, The Chinese University of Hong Kong , Shenzhen 518172 , China
- China Orthopedic Regenerative Medicine Group (CORMed) , Hangzhou , Zhejiang 310058 , China
| |
Collapse
|
414
|
Sivasubramanian S, Mohana S, Maheswari P, Victoria V, Thangam R, Mahalingam J, Chandrasekar-Janebjer G, Savariar V, Madhan B, Gunasekaran P, Kitambi SS. Leprosy-associated Chronic Wound Management Using Biomaterials. J Glob Infect Dis 2018; 10:99-107. [PMID: 29910571 PMCID: PMC5987379 DOI: 10.4103/jgid.jgid_79_17] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Background: Deformities and neuropathic chronic ulcers are the common features associated with leprosy-cured individuals that impact their quality of life and impair rehabilitation efforts. The challenging aspects for treatment of chronic wounds are the factors that inhibit healing. We reasoned that limited success of various therapeutic interventions could be due to the fact that leprosy-cured individual's physiology gets acclimatized to having a chronic wound that any therapeutic intervention is counterbalanced to maintain status quo at the wound site. Therefore, an alternative strategy would be to use biomaterials that gradually alter the wound site allowing the individual's physiology to participate in the healing process. Aims: Developing the human amnion (Amn)-derived biomaterial scaffolds and evaluating its use to heal chronic wounds in leprosy-cured but deformed persons (LCDPs). Materials and Methods: Using an enzymatic protocol, we have developed a rapid method to generate biomaterial scaffolds from discarded human Amn. A clinical trial on 26 LCDPs was performed with the biomaterial, and its wound-healing potential was then compared with LCDPs undergoing standard treatment procedure. Results: Biomaterial-based treatment of chronic wounds on LCDP displayed a higher efficiency in healing when compared to standard treatment. Conclusions: This study exemplifies that biomaterial-based treatment of leprosy-wounds offers an excellent affordable alternative for wound management. This study underlines the importance of involving both local wound environment and systemic effects for healing. In addition, we highlight wound healing as a necessity for successful rehabilitation and reintegration of leprosy-cured person into the society.
Collapse
Affiliation(s)
| | - Sambasivam Mohana
- Department of Virology, King Institute of Preventive Medicine and Research, Chennai, Tamil Nadu, India
| | - Paulraj Maheswari
- Department of Virology, King Institute of Preventive Medicine and Research, Chennai, Tamil Nadu, India
| | - Victor Victoria
- Center for Environmental Research and Development, LIFE, Loyola College, Chennai, Tamil Nadu, India
| | - Ramar Thangam
- Department of Virology, King Institute of Preventive Medicine and Research, Chennai, Tamil Nadu, India.,CSIR-Central Leather Research Institute, Chennai, Tamil Nadu, India
| | - Jayashri Mahalingam
- Department of Virology, King Institute of Preventive Medicine and Research, Chennai, Tamil Nadu, India.,Institute for Healthcare Education and Translational Sciences (IHETS), Hyderabad, Telengana, India
| | | | - Vincent Savariar
- Center for Environmental Research and Development, LIFE, Loyola College, Chennai, Tamil Nadu, India
| | - Balaraman Madhan
- CSIR-Central Leather Research Institute, Chennai, Tamil Nadu, India
| | - Palani Gunasekaran
- Department of Virology, King Institute of Preventive Medicine and Research, Chennai, Tamil Nadu, India
| | - Satish S Kitambi
- Institute for Healthcare Education and Translational Sciences (IHETS), Hyderabad, Telengana, India.,Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Sweden
| |
Collapse
|
415
|
Rotter N, Zenobi-Wong M. [Regeneration - A New Therapeutic Dimension in Otorhinolaryngology]. Laryngorhinootologie 2018; 97:S185-S213. [PMID: 29905357 PMCID: PMC6290928 DOI: 10.1055/s-0043-122309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Regeneration as a therapeutic priniciple and regenerative medicine in general are promising new strategies to add new therapeutic dimensions to our current treatment options. Today, reconstructive surgery, drugs and implants such as the cochlear implant can replace the functions of damaged tissues. In contrast, regenerative therapies aim at the replacement of the damaged tissues themselves while at the same time replacing their lost tissue function. In this review article new technologies such as 3D-bioprinting and the application of decellularised tissues as biomaterials are introduced and explained. A summary of current preclinical and clinical regenerative studies in otorhinolaryngology is complementing these basic aspects.
Collapse
Affiliation(s)
- Nicole Rotter
- Klinik für Hals-Nasen-Ohrenheilkunde, Universitätsmedizin Mannheim, Universitätsklinikum Mannheim
| | | |
Collapse
|
416
|
Loye AM, Kinser ER, Bensouda S, Shayan M, Davis R, Wang R, Chen Z, Schwarz UD, Schroers J, Kyriakides TR. Regulation of Mesenchymal Stem Cell Differentiation by Nanopatterning of Bulk Metallic Glass. Sci Rep 2018; 8:8758. [PMID: 29884812 PMCID: PMC5993820 DOI: 10.1038/s41598-018-27098-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 05/24/2018] [Indexed: 01/08/2023] Open
Abstract
Mesenchymal stem cell (MSC) differentiation is regulated by surface modification including texturing, which is applied to materials to enhance tissue integration. Here, we used Pt57.5Cu14.7Ni5.3P22.5 bulk metallic glass (Pt-BMG) with nanopatterned surfaces achieved by thermoplastic forming to influence differentiation of human MSCs. Pt-BMGs are a unique class of amorphous metals with high strength, elasticity, corrosion resistance, and an unusual plastic-like processability. It was found that flat and nanopattened Pt-BMGs induced osteogenic and adipogenic differentiation, respectively. In addition, osteogenic differentiation on flat BMG exceeded that observed on medical grade titanium and was associated with increased formation of focal adhesions and YAP nuclear localization. In contrast, cells on nanopatterned BMGs exhibited rounded morphology, formed less focal adhesions and had mostly cytoplasmic YAP. These changes were preserved on nanopatterns made of nanorods with increased stiffness due to shorter aspect ratios, suggesting that MSC differentiation was primarily influenced by topography. These observations indicate that both elemental composition and nanotopography can modulate biochemical cues and influence MSCs. Moreover, the processability and highly tunable nature of Pt-BMGs enables the creation of a wide range of surface topographies that can be reproducibly and systematically studied, leading to the development of implants capable of engineering MSC functions.
Collapse
Affiliation(s)
- Ayomiposi M Loye
- Center for Research on Interface Structures and Phenomena, Yale University, New Haven, CT, 06520, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06520, USA
| | - Emily R Kinser
- Center for Research on Interface Structures and Phenomena, Yale University, New Haven, CT, 06520, USA
- Department of Mechanical Engineering and Materials Science, Yale University, New Haven, CT, 06520, USA
- IBM Thomas J, Watson Research Center, New York, NY, 10598, USA
| | - Sabrine Bensouda
- Center for Research on Interface Structures and Phenomena, Yale University, New Haven, CT, 06520, USA
| | - Mahdis Shayan
- Center for Research on Interface Structures and Phenomena, Yale University, New Haven, CT, 06520, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06520, USA
| | - Rose Davis
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, 06520, USA
| | - Rui Wang
- IBM Thomas J, Watson Research Center, New York, NY, 10598, USA
| | - Zheng Chen
- Center for Research on Interface Structures and Phenomena, Yale University, New Haven, CT, 06520, USA
- Department of Mechanical Engineering and Materials Science, Yale University, New Haven, CT, 06520, USA
| | - Udo D Schwarz
- Center for Research on Interface Structures and Phenomena, Yale University, New Haven, CT, 06520, USA
- Department of Mechanical Engineering and Materials Science, Yale University, New Haven, CT, 06520, USA
- Department of Chemical and Enviromental Engineering, Yale University, P.O. Box 208089, New Haven, CT, 06520, USA
| | - Jan Schroers
- Center for Research on Interface Structures and Phenomena, Yale University, New Haven, CT, 06520, USA
- Department of Mechanical Engineering and Materials Science, Yale University, New Haven, CT, 06520, USA
| | - Themis R Kyriakides
- Center for Research on Interface Structures and Phenomena, Yale University, New Haven, CT, 06520, USA.
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06520, USA.
- Department of Pathology, Yale University, P.O. Box 208089, New Haven, CT, 06520, USA.
| |
Collapse
|
417
|
He C, Yang Z, Jin Y, Qi X, Chu J, Deng X. ADM Scaffolds Generate a Pro-regenerative Microenvironment During Full-Thickness Cutaneous Wound Healing Through M2 Macrophage Polarization via Lamtor1. Front Physiol 2018; 9:657. [PMID: 29915541 PMCID: PMC5994424 DOI: 10.3389/fphys.2018.00657] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 05/14/2018] [Indexed: 12/21/2022] Open
Abstract
Adult mammalian skin has a defective regenerative capacity following full-thickness cutaneous injury; this defect overshadows the complete physiological functions of the skin. Immune-mediated skin reconstruction driven by biological scaffolds is a recently developed innovative repair strategy to support regenerative wound healing. However, to date, little is known about how biological scaffolds orchestrate the immune response to promote regeneration. Here, using acellular dermal matrix (ADM) scaffolds, we discovered that the default pro-inflammatory response was altered in response to a pro-regenerative response characterized by specific M2 polarization. M2 macrophages subsequently produced a series of wound healing factors, including matrix metalloproteinases (Mmps), and growth factors which promoted cell proliferation, stabilized angiogenesis, and remodeled the extracellular matrix. Our investigations further revealed that the M2 polarization of macrophages arose from an ADM scaffold-derived amino acid sufficiency signal by collagen degradation via macrophage phagocytosis, which activated the acid-sensing pathway (v-ATPase, Lamtor1, and mTORC1). Lamtor1, the acid-sensing pathway-associated lysosomal adaptor protein was critical for inducing M2 polarization, while with the presence of extracellular interleukin 4 (IL4). Our results suggest that ADM scaffolds generate a pro-regenerative microenvironment during full-thickness cutaneous wound healing through M2 macrophage polarization via Lamtor1.
Collapse
Affiliation(s)
- Chengmin He
- MOE Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China
| | - Zhi Yang
- MOE Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China
| | - Ying Jin
- MOE Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China
| | - Xiaoyang Qi
- MOE Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China
| | - Jin Chu
- MOE Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China
| | - Xiaoyuan Deng
- MOE Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China
| |
Collapse
|
418
|
|
419
|
|
420
|
Decoration of RGD-mimetic porous scaffolds with engineered and devitalized extracellular matrix for adipose tissue regeneration. Acta Biomater 2018; 73:154-166. [PMID: 29684623 DOI: 10.1016/j.actbio.2018.04.039] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 03/22/2018] [Accepted: 04/19/2018] [Indexed: 12/16/2022]
Abstract
Fat grafting is emerging as a promising alternative to silicon implants in breast reconstruction surgery. Unfortunately, this approach does not provide a proper mechanical support and is affected by drawbacks such as tissue resorption and donor site morbidity. Synthetic scaffolds can offer a valuable alternative to address these challenges, but poorly recapitulate the biochemical stimuli needed for tissue regeneration. Here, we aim at combining the positive features of a structural, synthetic polymer to an engineered, devitalized extracellular matrix (ECM) to generate a hybrid construct that can provide a mix of structural and biological stimuli needed for adipose tissue regeneration. A RGD-mimetic synthetic scaffold OPAAF, designed for soft tissue engineering, was decorated with ECM deposited by human adipose stromal cells (hASCs). The adipoinductive potential of the hybrid ECM-OPAAF construct was validated in vitro, by culture with hASC in a perfusion bioreactor system, and in vivo, by subcutaneous implantation in nude mouse. Our findings demonstrate that the hybrid ECM-OPAAF provides proper mechanical support and adipoinductive stimuli, with potential applicability as off-the-shelf material for adipose tissue reconstruction. STATEMENT OF SIGNIFICANCE In this study we combined the functionalities of a synthetic polymer with those of an engineered and subsequently devitalized extracellular matrix (ECM) to generate a hybrid material for adipose tissue regeneration. The developed hybrid ECM-OPAAF was demonstrated to regulate human adipose stromal cells adipogenic commitment in vitro and adipose tissue infiltration in vivo. Our findings demonstrate that the hybrid ECM-OPAAF provide proper mechanical support and adipoinductive stimuli and represents a promising off-the-shelf material for adipose tissue reconstruction. We believe that our approach could offer an alternative strategy for adipose tissue reconstruction in case of mastectomy or congenital abnormalities, overcoming the current limitations of autologous fat based strategies such as volume resorption and donor site morbidity.
Collapse
|
421
|
Gosselin EA, Eppler HB, Bromberg JS, Jewell CM. Designing natural and synthetic immune tissues. NATURE MATERIALS 2018; 17:484-498. [PMID: 29784994 PMCID: PMC6283404 DOI: 10.1038/s41563-018-0077-6] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 04/11/2018] [Indexed: 05/10/2023]
Abstract
Vaccines and immunotherapies have provided enormous improvements for public health, but there are fundamental disconnects between where most studies are performed-in cell culture and animal models-and the ultimate application in humans. Engineering immune tissues and organs, such as bone marrow, thymus, lymph nodes and spleen, could be instrumental in overcoming these hurdles. Fundamentally, designed immune tissues could serve as in vitro tools to more accurately study human immune function and disease, while immune tissues engineered for implantation as next-generation vaccines or immunotherapies could enable direct, on-demand control over generation and regulation of immune function. In this Review, we discuss recent interdisciplinary strategies that are merging materials science and immunology to create engineered immune tissues in vitro and in vivo. We also highlight the hurdles facing these approaches and the need for comparison to existing clinical options, relevant animal models, and other emerging technologies.
Collapse
Affiliation(s)
- Emily A Gosselin
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | - Haleigh B Eppler
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
- Molecular and Cellular Biology, Biological Sciences Training Program, University of Maryland, College Park, MD, USA
| | - Jonathan S Bromberg
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
- Marlene and Stewart Greenebaum Cancer Center, Baltimore, MD, USA
| | - Christopher M Jewell
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA.
- Molecular and Cellular Biology, Biological Sciences Training Program, University of Maryland, College Park, MD, USA.
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA.
- Marlene and Stewart Greenebaum Cancer Center, Baltimore, MD, USA.
- Robert E. Fischell Institute for Biomedical Devices, College Park, MD, USA.
- United States Department of Veterans Affairs, Maryland VA Health Care System, Baltimore, MD, USA.
| |
Collapse
|
422
|
Smits AI, Bouten CV. Tissue engineering meets immunoengineering: Prospective on personalized in situ tissue engineering strategies. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2018. [DOI: 10.1016/j.cobme.2018.02.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
423
|
Bouten CVC, Smits AIPM, Baaijens FPT. Can We Grow Valves Inside the Heart? Perspective on Material-based In Situ Heart Valve Tissue Engineering. Front Cardiovasc Med 2018; 5:54. [PMID: 29896481 PMCID: PMC5987128 DOI: 10.3389/fcvm.2018.00054] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 05/09/2018] [Indexed: 12/14/2022] Open
Abstract
In situ heart valve tissue engineering using cell-free synthetic, biodegradable scaffolds is under development as a clinically attractive approach to create living valves right inside the heart of a patient. In this approach, a valve-shaped porous scaffold "implant" is rapidly populated by endogenous cells that initiate neo-tissue formation in pace with scaffold degradation. While this may constitute a cost-effective procedure, compatible with regulatory and clinical standards worldwide, the new technology heavily relies on the development of advanced biomaterials, the processing thereof into (minimally invasive deliverable) scaffolds, and the interaction of such materials with endogenous cells and neo-tissue under hemodynamic conditions. Despite the first positive preclinical results and the initiation of a small-scale clinical trial by commercial parties, in situ tissue formation is not well understood. In addition, it remains to be determined whether the resulting neo-tissue can grow with the body and preserves functional homeostasis throughout life. More important yet, it is still unknown if and how in situ tissue formation can be controlled under conditions of genetic or acquired disease. Here, we discuss the recent advances of material-based in situ heart valve tissue engineering and highlight the most critical issues that remain before clinical application can be expected. We argue that a combination of basic science - unveiling the mechanisms of the human body to respond to the implanted biomaterial under (patho)physiological conditions - and technological advancements - relating to the development of next generation materials and the prediction of in situ tissue growth and adaptation - is essential to take the next step towards a realistic and rewarding translation of in situ heart valve tissue engineering.
Collapse
Affiliation(s)
- Carlijn V. C. Bouten
- Soft Tissue Engineering and Mechanobiology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, Netherlands
| | - Anthal I. P. M. Smits
- Soft Tissue Engineering and Mechanobiology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, Netherlands
| | - Frank P. T. Baaijens
- Soft Tissue Engineering and Mechanobiology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, Netherlands
| |
Collapse
|
424
|
Early rehabilitation for volumetric muscle loss injury augments endogenous regenerative aspects of muscle strength and oxidative capacity. BMC Musculoskelet Disord 2018; 19:173. [PMID: 29843673 PMCID: PMC5975473 DOI: 10.1186/s12891-018-2095-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 05/16/2018] [Indexed: 02/08/2023] Open
Abstract
Background Volumetric muscle loss (VML) injuries occur due to orthopaedic trauma or the surgical removal of skeletal muscle and result in debilitating long-term functional deficits. Current treatment strategies do not promote significant restoration of function; additionally appropriate evidenced-based practice physical therapy paradigms have yet to be established. The objective of this study was to develop and evaluate early rehabilitation paradigms of passive range of motion and electrical stimulation in isolation or combination to understand the genetic and functional response in the tissue remaining after a multi-muscle VML injury. Methods Adult male mice underwent an ~ 20% multi-muscle VML injury to the posterior compartment (gastrocnemius, soleus, and plantaris muscle) unilaterally and were randomized to rehabilitation paradigm twice per week beginning 2 days post-injury or no treatment. Results The most salient findings of this work are: 1) that the remaining muscle tissue after VML injury was adaptable in terms of improved muscle strength and mitigation of stiffness; but 2) not adaptable to improvements in metabolic capacity. Furthermore, biochemical (i.e., collagen content) and gene (i.e., gene arrays) assays suggest that functional adaptations may reflect changes in the biomechanical properties of the remaining tissue due to the cellular deposition of non-contractile tissue in the void left by the VML injury and/or differentiation of gene expression with early rehabilitation. Conclusions Collectively this work provides evidence of genetic and functional plasticity in the remaining skeletal muscle with early rehabilitation approaches, which may facilitate future evidenced-based practice of early rehabilitation at the clinical level. Electronic supplementary material The online version of this article (10.1186/s12891-018-2095-6) contains supplementary material, which is available to authorized users.
Collapse
|
425
|
Chu C, Liu L, Wang Y, Wei S, Wang Y, Man Y, Qu Y. Macrophage phenotype in the epigallocatechin-3-gallate (EGCG)-modified collagen determines foreign body reaction. J Tissue Eng Regen Med 2018; 12:1499-1507. [PMID: 29704322 DOI: 10.1002/term.2687] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 03/16/2018] [Accepted: 04/16/2018] [Indexed: 02/05/2023]
Affiliation(s)
- Chenyu Chu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology; Sichuan University; Chengdu China
- Department of Oral Implantology, West China Hospital of Stomatology; Sichuan University; Chengdu China
| | - Li Liu
- State key laboratory of Biotherapy, West China Hospital; Sichuan University and Collaborative Innovation Center for Biotherapy; Chengdu Sichuan China
| | - Yufei Wang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology; Sichuan University; Chengdu China
| | - Shimin Wei
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology; Sichuan University; Chengdu China
| | - Yuanjing Wang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology; Sichuan University; Chengdu China
| | - Yi Man
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology; Sichuan University; Chengdu China
- Department of Oral Implantology, West China Hospital of Stomatology; Sichuan University; Chengdu China
| | - Yili Qu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology; Sichuan University; Chengdu China
| |
Collapse
|
426
|
Kang H, Zhang K, Wong DSH, Han F, Li B, Bian L. Near-infrared light-controlled regulation of intracellular calcium to modulate macrophage polarization. Biomaterials 2018; 178:681-696. [PMID: 29705000 DOI: 10.1016/j.biomaterials.2018.03.007] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Revised: 02/06/2018] [Accepted: 03/04/2018] [Indexed: 12/15/2022]
Abstract
Macrophages are multifunctional immune cells with diverse physiological functions such as fighting against infection, influencing progression of pathologies, maintaining homeostasis, and regenerating tissues. Macrophages can be induced to adopt distinct polarized phenotypes, such as classically activated pro-inflammatory (M1) phenotypes or alternatively activated anti-inflammatory and pro-healing (M2), to execute diverse and dynamic immune functions. However, unbalanced polarizations of macrophage can lead to various pathologies, such as atherosclerosis, obesity, tumor, and asthma. Thus, the capability to remotely control macrophage phenotypes is important to the success of treating many pathological conditions involving macrophages. In this study, we developed an upconversion nanoparticle (UCNP)-based photoresponsive nanocarrier for near-infrared (NIR) light-mediated control of intracellular calcium levels to regulate macrophage polarization. UCNP was coated with mesoporous silica (UCNP@mSiO2), into which loaded calcium regulators that can either supply or deplete calcium ions. UCNP@mSiO2 was chemically modified through serial coupling of photocleavable linker and Arg-Gly-Asp (RGD) peptide-bearing molecular cap via cyclodextrin-adamantine host-guest complexation. The RGD-bearing cap functioned as the photolabile gating structure to control the release of calcium regulators and facilitated the cellular uptake of UCNP@mSiO2 nanocarrier. The upconverted UV light emission from the UCNP@mSiO2 under NIR light excitation triggered the cleavage of cap and intracellular release of calcium regulators, thereby allowing temporal regulation on the intracellular calcium levels. Application of NIR light through skin tissue promoted M1 or M2 polarization of macrophages, by elevating or depleting intracellular calcium levels, respectively. To the best of our knowledge, this is the first demonstration of NIR light-mediated remote control on macrophage polarization. This photoresponsive nanocarrier offers the potential to remotely manipulate in vivo immune functions, such as inflammation or tissue regeneration, via NIR light-controlled macrophage polarization.
Collapse
Affiliation(s)
- Heemin Kang
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong, China
| | - Kunyu Zhang
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong, China
| | - Dexter Siu Hong Wong
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong, China
| | - Fengxuan Han
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, Jiangsu, China; Orthopaedic Institute, Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Bin Li
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, Jiangsu, China; Orthopaedic Institute, Medical College, Soochow University, Suzhou, Jiangsu, China; China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, Zhejiang, China.
| | - Liming Bian
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong, China; Shun Hing Institute of Advanced Engineering, The Chinese University of Hong Kong, Hong Kong, China; Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China; Centre for Novel Biomaterials, The Chinese University of Hong Kong, Hong Kong, China; China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, Zhejiang, China; Department of Pharmacology, Guangdong Key Laboratory for Research and Development of Natural Drugs, Guangdong Medical University, Zhanjiang, Guangdong, China.
| |
Collapse
|
427
|
Bonito V, Smits A, Goor O, Ippel B, Driessen-Mol A, Münker T, Bosman A, Mes T, Dankers P, Bouten C. Modulation of macrophage phenotype and protein secretion via heparin-IL-4 functionalized supramolecular elastomers. Acta Biomater 2018. [PMID: 29518556 DOI: 10.1016/j.actbio.2018.02.032] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Hallmark of the in situ tissue engineering approach is the use of bioresorbable, synthetic, acellular scaffolds, which are designed to modulate the inflammatory response and actively trigger tissue regeneration by the body itself at the site of implantation. Much research is devoted to the design of synthetic materials modulating the polarization of macrophages, which are essential mediators of the early stages of the inflammatory response. Here, we present a novel method for the functionalization of elastomers based on synthetic peptide chemistry, supramolecular self-assembly, and immobilization of heparin and interleukin 4 (IL-4), which is known to skew the polarization of macrophages into the wound healing "M2" phenotype. Ureido-pyrimidinone (UPy)-modified chain extended polycaprolactone (CE-UPy-PCL) was mixed with a UPy-modified heparin binding peptide (UPy-HBP) to allow for immobilization of heparin, and further functionalization with IL-4 via its heparin binding domain. As a first proof of principle, CE-UPy-PCL and UPy-HBP were premixed in solution, dropcast and exposed to primary human monocyte-derived macrophages, in the presence or absence of IL-4-heparin functionalization. It was demonstrated that the supramolecular IL-4-heparin functionalization effectively promoted macrophage polarization into an anti-inflammatory phenotype, in terms of morphology, immunohistochemistry and cytokine secretion. Moreover, the supramolecular functionalization approach used was successfully translated to 3D electrospun scaffolds for in situ tissue engineering purposes, where UPy-HBP retention, and heparin and IL-4 attachment to the supramolecular scaffolds were proven over 7 days. Lastly, human monocyte-derived macrophages were cultured on 3D scaffolds, which, in case of IL-4-heparin functionalization, were proven to promote of an anti-inflammatory environment on protein level. This study presents a novel method in designing a versatile class of functionalized elastomers that effectively harness the anti-inflammatory behavior of macrophages in vitro, and as such, may be instrumental for the development of a new class of synthetic materials for in situ tissue engineering purposes. STATEMENT OF SIGNIFICANCE Macrophages and their phenotypic and functional plasticity play a pivotal role in metabolic homeostasis and tissue repair. Based on this notion, bioactivated materials modulating macrophage polarization were extensively investigated in the past. Here, we designed immunomodulating, synthetic materials based on supramolecular immobilization of a heparin binding peptide, and further bioactivation with heparin and IL-4, an anti-inflammatory cytokine responsible for M2 activation and polarization. Human monocyte-derived macrophages cultured on heparin-IL-4 bioactivated materials displayed an elongated morphology and an anti-inflammatory phenotype, with downregulation of pro-inflammatory cytokines and promotion of anti-inflammatory cytokines over time. This study represents the first step in designing a novel class of synthetic, bioactivated materials that harness the regenerative behavior of host macrophages towards in situ tissue regeneration.
Collapse
|
428
|
Qiu X, Liu S, Zhang H, Zhu B, Su Y, Zheng C, Tian R, Wang M, Kuang H, Zhao X, Jin Y. Mesenchymal stem cells and extracellular matrix scaffold promote muscle regeneration by synergistically regulating macrophage polarization toward the M2 phenotype. Stem Cell Res Ther 2018; 9:88. [PMID: 29615126 PMCID: PMC5883419 DOI: 10.1186/s13287-018-0821-5] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 02/14/2018] [Accepted: 02/26/2018] [Indexed: 02/08/2023] Open
Abstract
Background Skeletal muscle plays an important role in the body’s physiology but there are still no effective treatments for volumetric muscle loss (VML) resulting from severe traumatic injury or tumor excision. Recent studies show that a tissue engineering strategy using a compound containing mesenchymal stem cells (MSCs) and decellularized extracellular matrix (ECM) scaffold generates significant regenerative effects on VML injury, but the underlying mechanisms are not fully understood. Methods The characteristics of human umbilical cord MSCs, including multiplication capacity and multidifferentiation ability, were determined. We constructed a compound containing MSCs and decellularized ECM scaffold which was used for tissue regeneration in a VML model. Results We found that MSCs and decellularized ECM scaffold generated synergistic effects on promoting skeletal muscle tissue regeneration. Interestingly, both MSCs and decellularized ECM scaffold could promote macrophage polarization toward the M2 phenotype and suppress macrophage polarization toward the M1 phenotype, which is widely regarded as an important promoting factor in tissue regeneration. More importantly, MSCs and decellularized ECM scaffold generate synergistic promoting effects on macrophage polarization toward the M2 phenotype, not just an additive effect. Conclusions Our findings uncover a previously unknown mechanism that MSCs and decellularized ECM scaffold promote tissue regeneration via collaboratively regulating macrophage polarization. Electronic supplementary material The online version of this article (10.1186/s13287-018-0821-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xinyu Qiu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.,Research and Development Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Shiyu Liu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.,Research and Development Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Hao Zhang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.,Research and Development Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.,Department of Orthodontics, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Bin Zhu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.,Research and Development Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.,Department of Stomatology, General Hospital of Tibet Military Region, Lhasa, Tibet, 850007, China
| | - Yuting Su
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.,Research and Development Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Chenxi Zheng
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.,Research and Development Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Rong Tian
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.,Research and Development Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Miao Wang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.,Research and Development Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Huijuan Kuang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.,Research and Development Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Xinyi Zhao
- State Key Laboratory of Military Stomatology, Department of Dental Materials, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China. .,Department of Dental Materials, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
| | - Yan Jin
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China. .,Research and Development Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
| |
Collapse
|
429
|
Jeon OH, David N, Campisi J, Elisseeff JH. Senescent cells and osteoarthritis: a painful connection. J Clin Invest 2018; 128:1229-1237. [PMID: 29608139 PMCID: PMC5873863 DOI: 10.1172/jci95147] [Citation(s) in RCA: 234] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Senescent cells (SnCs) are associated with age-related pathologies. Osteoarthritis is a chronic disease characterized by pain, loss of cartilage, and joint inflammation, and its incidence increases with age. For years, the presence of SnCs in cartilage isolated from patients undergoing total knee artificial implants has been noted, but these cells' relevance to disease was unclear. In this Review, we summarize current knowledge of SnCs in the multiple tissues that constitute the articular joint. New evidence for the causative role of SnCs in the development of posttraumatic and age-related arthritis is reviewed along with the therapeutic benefit of SnC clearance. As part of their senescence-associated secretory phenotype, SnCs secrete cytokines that impact the immune system and its response to joint tissue trauma. We present concepts of the immune response to tissue trauma as well as the interactions with SnCs and the local tissue environment. Finally, we discuss therapeutic implications of targeting SnCs in treating osteoarthritis.
Collapse
Affiliation(s)
- Ok Hee Jeon
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, USA
- Buck Institute for Research on Aging, Novato, California, USA
| | | | - Judith Campisi
- Buck Institute for Research on Aging, Novato, California, USA
| | - Jennifer H. Elisseeff
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
430
|
Lin J, Zhou W, Han S, Bunpetch V, Zhao K, Liu C, Yin Z, Ouyang H. Cell-material interactions in tendon tissue engineering. Acta Biomater 2018; 70:1-11. [PMID: 29355716 DOI: 10.1016/j.actbio.2018.01.012] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Revised: 12/11/2017] [Accepted: 01/10/2018] [Indexed: 12/19/2022]
Abstract
The interplay between cells and materials is a fundamental topic in biomaterial-based tissue regeneration. One of the principles for biomaterial development in tendon regeneration is to stimulate tenogenic differentiation of stem cells. To this end, efforts have been made to optimize the physicochemical and bio-mechanical properties of biomaterials for tendon tissue engineering. However, recent progress indicated that innate immune cells, especially macrophages, can also respond to the material cues and undergo phenotypical changes, which will either facilitate or hinder tissue regeneration. This process has been, to some extent, neglected by traditional strategies and may partially explain the unsatisfactory outcomes of previous studies; thus, more researchers have turned their focus on developing and designing immunoregenerative biomaterials to enhance tendon regeneration. In this review, we will first summarize the effects of material cues on tenogenic differentiation and paracrine secretion of stem cells. A brief introduction will also be made on how material cues can be manipulated for the regeneration of tendon-to-bone interface. Then, we will discuss the characteristics and influences of macrophages on the repair process of tendon healing and how they respond to different materials cues. These principles may benefit the development of novel biomaterials provided with combinative bioactive cues to activate tenogenic differentiation of stem cells and pro-resolving macrophage phenotype. STATEMENT OF SIGNIFICANCE The progress achieved with the rapid development of biomaterial-based strategies for tendon regeneration has not yielded broad benefits to clinical patients. In addition to the interplay between stem cells and biomaterials, the innate immune response to biomaterials also plays a determinant role in tissue regeneration. Here, we propose that fine-tuning of stem cell behaviors and alternative activation of macrophages through material cues may lead to effective tendon/ligament regeneration. We first review the characteristics of key material cues that have been manipulated to promote tenogenic differentiation and paracrine secretion of stem cells in tendon regeneration. Then, we discuss the potentiality of corresponding material cues in activating macrophages toward a pro-resolving phenotype to promote tissue repair.
Collapse
Affiliation(s)
- Junxin Lin
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University-University of Edinburgh Institute, Zhejiang University, China; Zhejiang Provincial Key Laboratory of Tissue Engineering and Regenerative Medicine, Zhejiang University, China
| | - Wenyan Zhou
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University-University of Edinburgh Institute, Zhejiang University, China; Zhejiang Provincial Key Laboratory of Tissue Engineering and Regenerative Medicine, Zhejiang University, China
| | - Shan Han
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University-University of Edinburgh Institute, Zhejiang University, China; Zhejiang Provincial Key Laboratory of Tissue Engineering and Regenerative Medicine, Zhejiang University, China
| | - Varitsara Bunpetch
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University-University of Edinburgh Institute, Zhejiang University, China; Zhejiang Provincial Key Laboratory of Tissue Engineering and Regenerative Medicine, Zhejiang University, China
| | - Kun Zhao
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University-University of Edinburgh Institute, Zhejiang University, China; Zhejiang Provincial Key Laboratory of Tissue Engineering and Regenerative Medicine, Zhejiang University, China; Department of Sports Medicine, School of Medicine, Zhejiang University, China
| | - Chaozhong Liu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University-University of Edinburgh Institute, Zhejiang University, China; Zhejiang Provincial Key Laboratory of Tissue Engineering and Regenerative Medicine, Zhejiang University, China
| | - Zi Yin
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University-University of Edinburgh Institute, Zhejiang University, China; Zhejiang Provincial Key Laboratory of Tissue Engineering and Regenerative Medicine, Zhejiang University, China
| | - Hongwei Ouyang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University-University of Edinburgh Institute, Zhejiang University, China; Zhejiang Provincial Key Laboratory of Tissue Engineering and Regenerative Medicine, Zhejiang University, China; Department of Sports Medicine, School of Medicine, Zhejiang University, China; China Orthopedic Regenerative Medicine Group (CORMed), China; State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, China.
| |
Collapse
|
431
|
Haumer A, Bourgine PE, Occhetta P, Born G, Tasso R, Martin I. Delivery of cellular factors to regulate bone healing. Adv Drug Deliv Rev 2018; 129:285-294. [PMID: 29357301 DOI: 10.1016/j.addr.2018.01.010] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 01/08/2018] [Accepted: 01/13/2018] [Indexed: 02/06/2023]
Abstract
Bone tissue has a strong intrinsic regenerative capacity, thanks to a delicate and complex interplay of cellular and molecular processes, which tightly involve the immune system. Pathological settings of anatomical, biomechanical or inflammatory nature may lead to impaired bone healing. Innovative strategies to enhance bone repair, including the delivery of osteoprogenitor cells or of potent cytokines/morphogens, indicate the potential of 'orthobiologics', but are not fully satisfactory. Here, we review different approaches based on the delivery of regenerative cues produced by cells but in cell-free, possibly off-the-shelf configurations. Such strategies exploit the paracrine effect of the secretome of mesenchymal stem/stromal cells, presented in soluble form, shuttled through extracellular vesicles, or embedded within the network of extracellular matrix molecules. In addition to osteoinductive molecules, attention is given to factors targeting the resident immune cells, to reshape inflammatory and immunity processes from scarring to regenerative patterns.
Collapse
Affiliation(s)
- Alexander Haumer
- Department of Biomedicine, University Hospital Basel, University of Basel, Switzerland; Department of Biomedical Engineering, University of Basel, Switzerland.
| | - Paul Emile Bourgine
- Department of Biomedicine, University Hospital Basel, University of Basel, Switzerland; Department of Biomedical Engineering, University of Basel, Switzerland.
| | - Paola Occhetta
- Department of Biomedicine, University Hospital Basel, University of Basel, Switzerland; Department of Biomedical Engineering, University of Basel, Switzerland.
| | - Gordian Born
- Department of Biomedicine, University Hospital Basel, University of Basel, Switzerland; Department of Biomedical Engineering, University of Basel, Switzerland.
| | - Roberta Tasso
- Ospedale Policlinico San Martino-IST, IRCCS per l'Oncologia, Genova, Italy
| | - Ivan Martin
- Department of Biomedicine, University Hospital Basel, University of Basel, Switzerland; Department of Biomedical Engineering, University of Basel, Switzerland.
| |
Collapse
|
432
|
Keane TJ, Horejs CM, Stevens MM. Scarring vs. functional healing: Matrix-based strategies to regulate tissue repair. Adv Drug Deliv Rev 2018; 129:407-419. [PMID: 29425770 DOI: 10.1016/j.addr.2018.02.002] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Revised: 12/23/2017] [Accepted: 02/05/2018] [Indexed: 12/11/2022]
Abstract
All vertebrates possess mechanisms to restore damaged tissues with outcomes ranging from regeneration to scarring. Unfortunately, the mammalian response to tissue injury most often culminates in scar formation. Accounting for nearly 45% of deaths in the developed world, fibrosis is a process that stands diametrically opposed to functional tissue regeneration. Strategies to improve wound healing outcomes therefore require methods to limit fibrosis. Wound healing is guided by precise spatiotemporal deposition and remodelling of the extracellular matrix (ECM). The ECM, comprising the non-cellular component of tissues, is a signalling depot that is differentially regulated in scarring and regenerative healing. This Review focuses on the importance of the native matrix components during mammalian wound healing alongside a comparison to scar-free healing and then presents an overview of matrix-based strategies that attempt to exploit the role of the ECM to improve wound healing outcomes.
Collapse
|
433
|
Genipin-Cross-Linked Chitosan Nerve Conduits Containing TNF-α Inhibitors for Peripheral Nerve Repair. Ann Biomed Eng 2018; 46:1013-1025. [PMID: 29603044 DOI: 10.1007/s10439-018-2011-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 03/21/2018] [Indexed: 01/23/2023]
Abstract
Tissue engineered nerve grafts (TENGs) are considered a promising alternative to autologous nerve grafting, which is considered the "gold standard" clinical strategy for peripheral nerve repair. Here, we immobilized tumor necrosis factor-α (TNF-α) inhibitors onto a nerve conduit, which was introduced into a chitosan (CS) matrix scaffold utilizing genipin (GP) as the crosslinking agent, to fabricate CS-GP-TNF-α inhibitor nerve conduits. The in vitro release kinetics of TNF-α inhibitors from the CS-GP-TNF-α inhibitor nerve conduits were investigated using high-performance liquid chromatography. The in vivo continuous release profile of the TNF-α inhibitors released from the CS-GP-TNF-α inhibitor nerve conduits was measured using an enzyme-linked immunosorbent assay over 14 days. We found that the amount of TNF-α inhibitors released decreased with time after the bridging of the sciatic nerve defects in rats. Moreover, 4 and 12 weeks after surgery, histological analyses and functional evaluations were carried out to assess the influence of the TENG on regeneration. Immunochemistry performed 4 weeks after grafting to assess early regeneration outcomes revealed that the TENG strikingly promoted axonal outgrowth. Twelve weeks after grafting, the TENG accelerated myelin sheath formation, as well as functional restoration. In general, the regenerative outcomes following TENG more closely paralleled findings observed with autologous grafting than the use of the CS matrix scaffold. Collectively, our data indicate that the CS-GP-TNF-α inhibitor nerve conduits comprised an elaborate system for sustained release of TNF-α inhibitors in vitro, while studies in vivo demonstrated that the TENG could accelerate regenerating axonal outgrowth and functional restoration. The introduction of CS-GP-TNF-α-inhibitor nerve conduits into a scaffold may contribute to an efficient and adaptive immune microenvironment that can be used to facilitate peripheral nerve repair.
Collapse
|
434
|
Larouche J, Greising SM, Corona BT, Aguilar CA. Robust inflammatory and fibrotic signaling following volumetric muscle loss: a barrier to muscle regeneration. Cell Death Dis 2018. [PMID: 29540673 PMCID: PMC5851980 DOI: 10.1038/s41419-018-0455-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Jacqueline Larouche
- Dept. of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.,Australian Regenerative Medicine Institute, Monash University, Melbourne, Australia
| | - Sarah M Greising
- School of Kinesiology, University of Minnesota, Minneapolis, MN, USA
| | | | - Carlos A Aguilar
- Dept. of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
435
|
Abstract
Stem cells and tissue-derived stromal cells stimulate the repair of degenerated and injured tissues, motivating a growing number of cell-based interventions in the musculoskeletal field. Recent investigations have indicated that these cells are critical for their trophic and immunomodulatory role in controlling endogenous cells. This Review presents recent clinical advances where stem cells and stromal cells have been used to stimulate musculoskeletal tissue repair, including delivery strategies to improve cell viability and retention. Emerging bioengineering strategies are highlighted, particularly toward the development of biomaterials for capturing aspects of the native tissue environment, altering the healing niche, and recruiting endogenous cells.
Collapse
Affiliation(s)
- Claudia Loebel
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jason A Burdick
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
436
|
|
437
|
Montgomery M, Davenport Huyer L, Bannerman D, Mohammadi MH, Conant G, Radisic M. Method for the Fabrication of Elastomeric Polyester Scaffolds for Tissue Engineering and Minimally Invasive Delivery. ACS Biomater Sci Eng 2018; 4:3691-3703. [DOI: 10.1021/acsbiomaterials.7b01017] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
| | | | | | | | | | - Milica Radisic
- Toronto General Research Institute, University Health Network, Toronto, Ontario M5G 2C4, Canada
| |
Collapse
|
438
|
Lu L, Parmar MB, Kulka M, Kwan P, Unsworth LD. Self-Assembling Peptide Nanoscaffold That Activates Human Mast Cells. ACS APPLIED MATERIALS & INTERFACES 2018; 10:6107-6117. [PMID: 29308881 DOI: 10.1021/acsami.7b14560] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Engineering biomaterials to manipulate the immune response to elicit specific therapeutic outcomes is a burgeoning field of research. Mast cells play a distinct and central role in the innate immune response, and are characterized by their rapid release of a myriad of proinflammatory mediators in response to stimulation. These mediators are central to protective actions such as wound healing, angiogenesis, and host defense against pathogens and animal venoms. Considering that mast cells are widely distributed in tissues that interface with the external environment, and are loaded with large amounts of preformed protective compounds, they are ideal targets for novel immunotherapies. Here we report that, by using an engineered nanoscaffold, human mast cells can be contact activated in cell and primary human skin tissue culture using a specific receptor-ligand mechanism. The IgE independent PAMP-12 peptide activates human mast cells through the recently identified Mas-related G-protein coupled receptor member X2 (MRGPRX2) receptor. The PAMP-12 motif was conjugated, via a glycine spacer, with the self-assembling peptide (RADA)4 and mixed with unmodified (RADA)4 to form a nanofiber matrix; mast cell activation was influenced directly by this ratio. Moreover, conjugating the PAMP-12 motif within the matrix was shown to only activate local, tissue-resident mast cells. The result of ex vivo human skin tissue tests confirmed that the engineered nanoscaffold successfully activated skin-resident mast cells by contact. Thus, this nanoscaffold design may provide a new platform to modulate localized mast cell functions thereby facilitating their protective role in the skin.
Collapse
Affiliation(s)
- Lei Lu
- Department of Chemical and Materials Engineering, University of Alberta , 9211-116 Street NW, Edmonton, Alberta T6G 1H9, Canada
- National Institute for Nanotechnology , 11421 Saskatchewan Drive NW, Edmonton, Alberta T6G 2M9, Canada
| | - Manoj B Parmar
- Faculty of Pharmacy & Pharmaceutical Sciences, University of Alberta , Edmonton, Alberta T6G 1E2, Canada
| | - Marianna Kulka
- National Institute for Nanotechnology , 11421 Saskatchewan Drive NW, Edmonton, Alberta T6G 2M9, Canada
- Department of Medical Microbiology and Immunology, University of Alberta , Edmonton, Alberta T6G 2E1, Canada
| | - Peter Kwan
- Wound Healing Research Group, Division of Plastic and Reconstructive Surgery, University of Alberta , 2D2.28 WMC, 8440-112 Street, Edmonton, Alberta T6G 2B7, Canada
| | - Larry D Unsworth
- Department of Chemical and Materials Engineering, University of Alberta , 9211-116 Street NW, Edmonton, Alberta T6G 1H9, Canada
- National Institute for Nanotechnology , 11421 Saskatchewan Drive NW, Edmonton, Alberta T6G 2M9, Canada
| |
Collapse
|
439
|
Yang Z, He C, He J, Chu J, Liu H, Deng X. Curcumin-mediated bone marrow mesenchymal stem cell sheets create a favorable immune microenvironment for adult full-thickness cutaneous wound healing. Stem Cell Res Ther 2018; 9:21. [PMID: 29386050 PMCID: PMC5793416 DOI: 10.1186/s13287-018-0768-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 12/13/2017] [Accepted: 01/02/2018] [Indexed: 02/06/2023] Open
Abstract
Background Adult full-thickness cutaneous wound repair suffers from an imbalanced immune response, leading to nonfunctional reconstructed tissue and fibrosis. Although various treatments have been reported, the immune-mediated tissue regeneration driven by biomaterial offers an attractive regenerative strategy for damaged tissue repair. Methods In this research, we investigated a specific bone marrow-derived mesenchymal stem cell (BMSC) sheet that was induced by the Traditional Chinese Medicine curcumin (CS-C) and its immunomodulatory effects on wound repair. Comparisons were made with the BMSC sheet induced without curcumin (CS-N) and control (saline). Results In vitro cultured BMSC sheets (CS-C) showed that curcumin promoted the proliferation of BMSCs and modified the features of produced extracellular matrix (ECM) secreted by BMSCs, especially the contents of ECM structural proteins such as fibronectin (FN) and collagen I and III, as well as the ratio of collagen III/I. Two-photon fluorescence (TPF) and second-harmonic generation (SHG) imaging of mouse implantation revealed superior engraftment of BMSCs, maintained for 35 days in the CS-C group. Most importantly, CS-C created a favorable immune microenvironment. The chemokine stromal cell-derived factor 1 (SDF1) was abundantly produced by CS-C, thus facilitating a mass migration of leukocytes from which significantly increased expression of signature TH1 cells (interferon gamma) and M1 macrophages (tumor necrosis factor alpha) genes were confirmed at 7 days post-operation. The number of TH1 cells and associated pro-inflammatory M1 macrophages subsequently decreased sharply after 14 days post-operation, suggesting a rapid type I immune regression. Furthermore, the CS-C group showed an increased trend towards M2 macrophage polarization in the early phase. CS-C led to an epidermal thickness and collagen deposition that was closer to that of normal skin. Conclusions Curcumin has a good regulatory effect on BMSCs and this promising CS-C biomaterial creates a pro-regenerative immune microenvironment for cutaneous wound healing.
Collapse
Affiliation(s)
- Zhi Yang
- MOE Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, No. 55 Zhongshan Avenue West, Tianhe District, Guangzhou, 510631, China
| | - Chengmin He
- MOE Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, No. 55 Zhongshan Avenue West, Tianhe District, Guangzhou, 510631, China
| | - Jinyang He
- Tropical Medicine Institute, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jing Chu
- MOE Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, No. 55 Zhongshan Avenue West, Tianhe District, Guangzhou, 510631, China
| | - Hanping Liu
- MOE Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, No. 55 Zhongshan Avenue West, Tianhe District, Guangzhou, 510631, China.
| | - Xiaoyuan Deng
- MOE Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, No. 55 Zhongshan Avenue West, Tianhe District, Guangzhou, 510631, China.
| |
Collapse
|
440
|
Aguado BA, Grim JC, Rosales AM, Watson-Capps JJ, Anseth KS. Engineering precision biomaterials for personalized medicine. Sci Transl Med 2018; 10:eaam8645. [PMID: 29343626 PMCID: PMC6079507 DOI: 10.1126/scitranslmed.aam8645] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 06/24/2017] [Accepted: 12/21/2017] [Indexed: 12/21/2022]
Abstract
As the demand for precision medicine continues to rise, the "one-size-fits-all" approach to designing medical devices and therapies is becoming increasingly outdated. Biomaterials have considerable potential for transforming precision medicine, but individual patient complexity often necessitates integrating multiple functions into a single device to successfully tailor personalized therapies. Here, we introduce an engineering strategy based on unit operations to provide a unified vocabulary and contextual framework to aid the design of biomaterial-based devices and accelerate their translation.
Collapse
Affiliation(s)
- Brian A Aguado
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO 80309, USA
- BioFrontiers Institute, University of Colorado, Boulder, CO 80309, USA
- Howard Hughes Medical Institute, University of Colorado, Boulder, CO 80309, USA
| | - Joseph C Grim
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO 80309, USA
- BioFrontiers Institute, University of Colorado, Boulder, CO 80309, USA
- Howard Hughes Medical Institute, University of Colorado, Boulder, CO 80309, USA
| | - Adrianne M Rosales
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO 80309, USA
- BioFrontiers Institute, University of Colorado, Boulder, CO 80309, USA
| | | | - Kristi S Anseth
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO 80309, USA.
- BioFrontiers Institute, University of Colorado, Boulder, CO 80309, USA
- Howard Hughes Medical Institute, University of Colorado, Boulder, CO 80309, USA
| |
Collapse
|
441
|
Krieger JR, Sok MCP, Turner TC, Botchwey EA. Delivery of Immunomodulatory Microparticles in a Murine Model of Rotator Cuff Tear. MRS ADVANCES 2018; 3:1341-1346. [PMID: 30002922 PMCID: PMC6039128 DOI: 10.1557/adv.2018.50] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Full thickness rotator cuff tears (RCT) and the associated muscle degeneration that results due to this injury presents a significant clinical burden. The prevention or recovery from this degeneration requires the synchronized behavior of many cells that participate in regeneration. Strategies that tune the inflammatory cascade that is initiated after injury serves as a powerful way to influence tissue repair. Here, we use the local, sustained delivery of the immunomodulatory small molecule FTY720 to examine whether the recruitment of pro-regenerative myeloid cells affects the healing outcome. We find that PLGA microparticles have an atrophic effect on the muscle that is ameliorated with the release of FTY720. However, the inability of FTY720 delivery to induce pro-regenerative monocyte and macrophage recruitment and our findings demonstrating enrichment of CD4+ T cells suggest that effects of this small molecule are context dependent and that the underlying mechanisms behind this RCT associated muscle degeneration require further studies.
Collapse
Affiliation(s)
- Jack R Krieger
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Drive, Atlanta, Georgia 30332, United States
| | - Mary Caitlin P Sok
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Drive, Atlanta, Georgia 30332, United States
| | - Thomas C Turner
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Drive, Atlanta, Georgia 30332, United States
| | - Edward A Botchwey
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Drive, Atlanta, Georgia 30332, United States
| |
Collapse
|
442
|
Goldman SM, Henderson BEP, Walters TJ, Corona BT. Co-delivery of a laminin-111 supplemented hyaluronic acid based hydrogel with minced muscle graft in the treatment of volumetric muscle loss injury. PLoS One 2018; 13:e0191245. [PMID: 29329332 PMCID: PMC5766229 DOI: 10.1371/journal.pone.0191245] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 01/02/2018] [Indexed: 12/21/2022] Open
Abstract
Minced muscle autografting mediates de novo myofiber regeneration and promotes partial recovery of neuromuscular strength after volumetric muscle loss injury (VML). A major limitation of this approach is the availability of sufficient donor tissue for the treatment of relatively large VMLs without inducing donor site morbidity. This study evaluated a laminin-111 supplemented hyaluronic acid based hydrogel (HA+LMN) as a putative myoconductive scaffolding to be co-delivered with minced muscle grafts. In a rat tibialis anterior muscle VML model, delivery of a reduced dose of minced muscle graft (50% of VML defect) within HA+LMN resulted in a 42% improvement of peak tetanic torque production over unrepaired VML affected limbs. However, the improvement in strength was not improved compared to a 50% minced graft-only control group. Moreover, histological analysis revealed that the improvement in in vivo functional capacity mediated by minced grafts in HA+LMN was not accompanied by a particularly robust graft mediated regenerative response as determined through donor cell tracking of the GFP+ grafting material. Characterization of the spatial distribution and density of macrophage and satellite cell populations indicated that the combination therapy damps the heightened macrophage response while re-establishing satellite content 14 days after VML to a level consistent with an endogenously healing ischemia-reperfusion induced muscle injury. Moreover, regional analysis revealed that the combination therapy increased satellite cell density mostly in the remaining musculature, as opposed to the defect area. Based on the results, the following salient conclusions were drawn: 1) functional recovery mediated by the combination therapy is likely due to a superposition of de novo muscle fiber regeneration and augmented repair of muscle fibers within the remaining musculature, and 2) The capacity for VML therapies to augment regeneration and repair within the remaining musculature may have significant clinical impact and warrants further exploration.
Collapse
Affiliation(s)
- Stephen M. Goldman
- United States Army Institute of Surgical Research, JBSA Fort Sam Houston, Texas, United States of America
| | - Beth E. P. Henderson
- United States Army Institute of Surgical Research, JBSA Fort Sam Houston, Texas, United States of America
| | - Thomas J. Walters
- United States Army Institute of Surgical Research, JBSA Fort Sam Houston, Texas, United States of America
| | - Benjamin T. Corona
- United States Army Institute of Surgical Research, JBSA Fort Sam Houston, Texas, United States of America
| |
Collapse
|
443
|
Baghdadi MB, Tajbakhsh S. Regulation and phylogeny of skeletal muscle regeneration. Dev Biol 2018; 433:200-209. [DOI: 10.1016/j.ydbio.2017.07.026] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 07/30/2017] [Accepted: 07/31/2017] [Indexed: 12/31/2022]
|
444
|
Dziki JL, Badylak SF. Extracellular Matrix for Myocardial Repair. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1098:151-171. [PMID: 30238370 DOI: 10.1007/978-3-319-97421-7_8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Multiple strategies have been investigated to restore functional myocardium following injury or disease including the local administration of cytokines or chemokines, stem/progenitor cell therapy, mechanical circulatory support, pharmacologic use, and the use of inductive biomaterials. The use of xenogeneic biologic scaffolds composed of extracellular matrix (ECM) has been shown to facilitate functional restoration of several tissues and organs including the esophagus, skeletal muscle, skin, and myocardium, among others. The present chapter describes the current understanding of specific components of biologic scaffolds composed of ECM, the mechanisms by which ECM bioscaffolds promote constructive cardiac remodeling after injury, determinants of remodeling outcome, and the versatility of ECM as a potential cardiac therapeutic.
Collapse
Affiliation(s)
- Jenna L Dziki
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Stephen F Badylak
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA. .,Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA. .,Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
445
|
Lin J, Cai R, Sun B, Dong J, Zhao Y, Miao Q, Chen C. Gd@C82(OH)22 harnesses inflammatory regeneration for osteogenesis of mesenchymal stem cells through JNK/STAT3 signaling pathway. J Mater Chem B 2018; 6:5802-5811. [DOI: 10.1039/c8tb01097h] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Gd@C82(OH)22 dose-dependently manipulates osteogenesis of MSCs in inflammatory microenvironment, which is capable for bone tissue engineering as an immunomodulatory nanoparticle.
Collapse
Affiliation(s)
- Jiao Lin
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience
- National Center for Nanoscience and Technology
- Chinese Academy of Science
- Beijing 100190
- P. R. China
| | - Rong Cai
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience
- National Center for Nanoscience and Technology
- Chinese Academy of Science
- Beijing 100190
- P. R. China
| | - Baoyun Sun
- University of Chinese Academy of Sciences
- Beijing 100049
- P. R. China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and Key Laboratory for Nuclear Techniques, Institute of High Energy Physics
- Beijing
| | - Jinquan Dong
- University of Chinese Academy of Sciences
- Beijing 100049
- P. R. China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and Key Laboratory for Nuclear Techniques, Institute of High Energy Physics
- Beijing
| | - Yuliang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience
- National Center for Nanoscience and Technology
- Chinese Academy of Science
- Beijing 100190
- P. R. China
| | - Qing Miao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience
- National Center for Nanoscience and Technology
- Chinese Academy of Science
- Beijing 100190
- P. R. China
| | - Chunying Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience
- National Center for Nanoscience and Technology
- Chinese Academy of Science
- Beijing 100190
- P. R. China
| |
Collapse
|
446
|
Wang J, Lin C, Gao X, Zheng Z, Lv M, Sun J, Zhang Z. The enhanced osteogenesis and osteointegration of 3-DP PCL scaffolds via structural and functional optimization using collagen networks. RSC Adv 2018; 8:32304-32316. [PMID: 35547520 PMCID: PMC9086255 DOI: 10.1039/c8ra05615c] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Accepted: 09/02/2018] [Indexed: 11/21/2022] Open
Abstract
Optimal balance between biological activity and mechanical stability should be meticulously considered during scaffold design for bone tissue engineering applications. To fabricate an individualized construct with biomechanical and biological functionality for bone tissue regeneration, a polycaprolactone–collagen (PCL–COL) composite construct was developed through the combination of three-dimensional printing (3-DP) technology and biomimetic collagen matrix incorporation, with a 3-DP PCL framework maintaining the mechanical stability and a porous collagen matrix improving the biological activity. The results indicate that the compressive modulus of the composite constructs increased synergistically (over 40 MPa), providing sufficient mechanical support during new bone formation. On the other hand, the collagen matrix with a micro-porous architecture structurally increased scaffold areas and provided cellular adhesion sites, allowing for the functional construction of a favorable 3D microenvironment for BMSC adhesion, proliferation and extracellular matrix production. Moreover, critical-sized long bone defect (CSD) implantation demonstrated that the optimized composite constructs could promote bone tissue regeneration (5.5-fold) and bone-material osteointegration (4.7-fold), and decrease fibrosis encapsulation, compared to pristine PCL. The results indicate that these biomimetically ornamented PCL–COL constructs exhibit favorable mechanical properties and biological functionality, demonstrating great potential as an effective bone graft substitute for bone defect treatment. Meanwhile, they can also harness the advantages of 3-DP technology and a collagen-based functionalized strategy, facilitating the creation of customized and functional PCL–COL constructs for clinical translation. Optimal balance between biological activity and mechanical stability should be meticulously considered during scaffold design for bone tissue engineering applications.![]()
Collapse
Affiliation(s)
- Jinbing Wang
- Department of Oral and Maxillofacial-Head and Neck Oncology
- Shanghai Ninth People's Hospital
- College of Stomatology
- Shanghai Jiao Tong University School of Medicine
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology
| | - Chucheng Lin
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure
- Shanghai Institute of Ceramics
- Chinese Academy of Sciences
- Shanghai 200050
- PR China
| | - Xin Gao
- Department of Oral Implantology
- Shanghai Stomatological Hospital
- Fu Dan University
- Shanghai 200001
- PR China
| | - Zhiwei Zheng
- Department of Oral and Maxillofacial-Head and Neck Oncology
- Shanghai Ninth People's Hospital
- College of Stomatology
- Shanghai Jiao Tong University School of Medicine
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology
| | - Mimgming Lv
- Department of Oral and Maxillofacial-Head and Neck Oncology
- Shanghai Ninth People's Hospital
- College of Stomatology
- Shanghai Jiao Tong University School of Medicine
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology
| | - Jian Sun
- Department of Oral and Maxillofacial-Head and Neck Oncology
- Shanghai Ninth People's Hospital
- College of Stomatology
- Shanghai Jiao Tong University School of Medicine
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology
| | - Zhiyong Zhang
- Shanghai Key Laboratory of Tissue Engineering
- Ninth People's Hospital
- Shanghai Jiao Tong University School of Medicine
- Shanghai 200011
- PR China
| |
Collapse
|
447
|
Corradetti B, Taraballi F, Corbo C, Cabrera F, Pandolfi L, Minardi S, Wang X, Van Eps J, Bauza G, Weiner B, Tasciotti E. Immune tuning scaffold for the local induction of a pro-regenerative environment. Sci Rep 2017; 7:17030. [PMID: 29208986 PMCID: PMC5717048 DOI: 10.1038/s41598-017-16895-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 11/10/2017] [Indexed: 11/10/2022] Open
Abstract
In mammals, tissue regeneration is accomplished through a well-regulated, complex cascade of events. The disruption of the cellular and molecular processes involved in tissue healing might lead to scar formation. Most tissue engineering approaches have tried to improve the regenerative outcome following an injury, through the combination of biocompatible materials, stem cells and bioactive factors. However, implanted materials can cause further healing impairments due to the persistent inflammatory stimuli that trigger the onset of chronic inflammation. Here, it is described at the molecular, cellular and tissue level, the body response to a functionalized biomimetic collagen scaffold. The grafting of chondroitin sulfate on the surface of the scaffold is able to induce a pro-regenerative environment at the site of a subcutaneous implant. The early in situ recruitment, and sustained local retention of anti-inflammatory macrophages significantly reduced the pro-inflammatory environment and triggered a different healing cascade, ultimately leading to collagen fibril re-organization, blood vessel formation, and scaffold integration with the surrounding native tissue.
Collapse
Affiliation(s)
- Bruna Corradetti
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX, 77030, USA
- Department of Life and Environmental Sciences, Polytechnic University of Marche, via Brecce Bianche, 60131, Ancona, Italy
| | - Francesca Taraballi
- Center for Biomimetic Medicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX, 77030, USA
- Houston Methodist Orthopedics and Sports Medicine, Houston, Texas, U.S.A.,, Houston, TX, 77030, USA
| | - Claudia Corbo
- Center for Biomimetic Medicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX, 77030, USA
- Center for Nanomedicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Fernando Cabrera
- Center for Biomimetic Medicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX, 77030, USA
| | - Laura Pandolfi
- Center for Biomimetic Medicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX, 77030, USA
| | - Silvia Minardi
- Center for Biomimetic Medicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX, 77030, USA
| | - Xin Wang
- Center for Biomimetic Medicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX, 77030, USA
| | - Jeffrey Van Eps
- Center for Biomimetic Medicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX, 77030, USA
| | - Guillermo Bauza
- Center for Biomimetic Medicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX, 77030, USA
- Houston Methodist Orthopedics and Sports Medicine, Houston, Texas, U.S.A.,, Houston, TX, 77030, USA
- Center for NanoHealth, Swansea University Medical School, Swansea University Bay, Singleton Park, SA2 8PP, Wales, UK
| | - Bradley Weiner
- Houston Methodist Orthopedics and Sports Medicine, Houston, Texas, U.S.A.,, Houston, TX, 77030, USA
| | - Ennio Tasciotti
- Center for Biomimetic Medicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX, 77030, USA.
- Houston Methodist Orthopedics and Sports Medicine, Houston, Texas, U.S.A.,, Houston, TX, 77030, USA.
- Center for NanoHealth, Swansea University Medical School, Swansea University Bay, Singleton Park, SA2 8PP, Wales, UK.
| |
Collapse
|
448
|
Sun G. Pro-Regenerative Hydrogel Restores Scarless Skin during Cutaneous Wound Healing. Adv Healthc Mater 2017; 6. [PMID: 28945013 DOI: 10.1002/adhm.201700659] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 08/09/2017] [Indexed: 11/06/2022]
Abstract
The transformation of fibrotic healing process to regenerative one has great potential to fully restore wounded skin. The M2 macrophage phenotype promotes constructive tissue remodeling and instructs tissue repair in a regenerative manner. It is hypothesized that hydrogels that can establish robustness of endogenous cells to regulate M2 phenotype will promote constructive dermal remodeling. Toward this end, a series of dextran-based bioabsorbable hydrogels are developed and self-crosslinkable dextran-isocyanatoethyl methacrylate-ethylamine (DexIEME) is identified as the potential scaffold. The initial screening study revealed that DexIEME has superior biocompatibility in varying concentrations. Although DexIEME brings about low proinflammatory responses, it promotes M2 macrophage phenotype. Then the optimized hydrogel formulation is tested for acute skin injuries using both murine and porcine models. Preliminary data demonstrated that the innovative DexIEME hydrogel promotes complete skin regeneration with hair regrowth on pre-existing scars, while untreated scars remain intact. Preclinical studies further demonstrated that the DexIEME hydrogel regenerated perfect skin during deep porcine wound healing. Overall, the approach to investigate immune-modulated hydrogels yields pro-regenerative DexIEME hydrogel, which may lead to greater clinical success in treating deep dermal injury and attenuating scar formation.
Collapse
Affiliation(s)
- Guoming Sun
- Sunogel Biotechnologies Inc.; 9 W Ridgely Road Ste 270 Lutherville Timonium MD 21093 USA
| |
Collapse
|
449
|
Macrophage-based therapeutic strategies in regenerative medicine. Adv Drug Deliv Rev 2017; 122:74-83. [PMID: 28526591 DOI: 10.1016/j.addr.2017.05.010] [Citation(s) in RCA: 221] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 05/12/2017] [Accepted: 05/15/2017] [Indexed: 12/17/2022]
Abstract
Mounting evidence suggests that therapeutic cell and drug delivery strategies designed to actively harness the regenerative potential of the inflammatory response have great potential in regenerative medicine. In particular, macrophages have emerged as a primary target because of their critical roles in regulating multiple phases of tissue repair through their unique ability to rapidly shift phenotypes. Herein, we review macrophage-based therapies, focusing on the translational potential for cell delivery of ex vivo-activated macrophages and delivery of molecules and biomaterials to modulate accumulation and phenotype of endogenous macrophages. We also review current obstacles to progress in translating basic findings to therapeutic applications, including the need for improved understanding of context-dependent macrophage functions and the myriad factors that regulate macrophage phenotype; potential species-specific differences (e.g. humans versus mice); quality control issues; and the lack of standardized procedures and nomenclature for characterizing macrophages. Looking forward, the inherent plasticity of macrophages represents a daunting challenge for harnessing these cells in regenerative medicine therapies but also great opportunity for improving patient outcomes in a variety of pathological conditions.
Collapse
|
450
|
Chen Z, Bachhuka A, Wei F, Wang X, Liu G, Vasilev K, Xiao Y. Nanotopography-based strategy for the precise manipulation of osteoimmunomodulation in bone regeneration. NANOSCALE 2017; 9:18129-18152. [PMID: 29143002 DOI: 10.1039/c7nr05913b] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Immune cells play vital roles in regulating bone dynamics. Successful bone regeneration requires a favourable osteo-immune environment. The high plasticity and diversity of immune cells make it possible to manipulate the osteo-immune response of immune cells, thus modulating the osteoimmune environment and regulating bone regeneration. With the advancement in nanotechnology, nanotopographies with different controlled surface properties can be fabricated. On tuning the surface properties, the osteo-immune response can be precisely modulated. This highly tunable characteristic and immunomodulatory effects make nanotopography a promising strategy to precisely manipulate osteoimmunomdulation for bone tissue engineering applications. This review first summarises the effects of the immune response during bone healing to show the importance of regulating the immune response for the bone response. The plasticity of immune cells is then reviewed to provide rationales for manipulation of the osteoimmune response. Subsequently, we highlight the current types of nanotopographies applied in bone biomaterials and their fabrication techniques, and explain how these nanotopographies modulate the immune response and the possible underlying mechanisms. The effects of immune cells on nanotopography-mediated osteogenesis are emphasized, and we propose the concept of "nano-osteoimmunomodulation" to provide a valuable strategy for the development of nanotopographies with osteoimmunomodulatory properties that can precisely regulate bone dynamics.
Collapse
Affiliation(s)
- Zetao Chen
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, Guangdong, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|