201
|
He J, Chen G, Liu M, Xu Z, Chen H, Yang L, Lv Y. Scaffold strategies for modulating immune microenvironment during bone regeneration. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 108:110411. [PMID: 31923946 DOI: 10.1016/j.msec.2019.110411] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 10/21/2019] [Accepted: 11/07/2019] [Indexed: 12/18/2022]
Abstract
Implanted bone scaffolds often fail to successfully integrate with the host tissue because they do not elicit a favorable immune reaction. Properties of bone scaffold not only provide mechanical and chemical signals to support cell adhesion, migration, proliferation and differentiation, but also play a pivotal role in determining the extent of immune response during bone regeneration. Appropriate design parameters of bone scaffold are of great significance in the process of developing a new generation of bone implants. Herein, this article addresses the recent advances in the field of bone scaffolds for immune response, particularly focusing on the physical and chemical properties of bone scaffold in manipulating the host response. Furthermore, incorporation of bioactive molecules and cells with immunoregulatory function in bone scaffolds are also presented. Finally, continuing challenges and future directions of scaffold-based strategies for modulating immune microenvironment are discussed.
Collapse
Affiliation(s)
- Jianhua He
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, PR China; Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing 400044, PR China.
| | - Guobao Chen
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, PR China; Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing 400044, PR China
| | - Mengying Liu
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, PR China; Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing 400044, PR China
| | - Zhiling Xu
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, PR China; Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing 400044, PR China.
| | - Hua Chen
- College of Chemistry and Chemical Engineering, Chongqing University, Chongqing 400044, PR China.
| | - Li Yang
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, PR China; Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing 400044, PR China.
| | - Yonggang Lv
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, PR China; Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing 400044, PR China.
| |
Collapse
|
202
|
Adams S, Wuescher LM, Worth R, Yildirim-Ayan E. Mechano-Immunomodulation: Mechanoresponsive Changes in Macrophage Activity and Polarization. Ann Biomed Eng 2019; 47:2213-2231. [PMID: 31218484 PMCID: PMC7043232 DOI: 10.1007/s10439-019-02302-4] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 06/07/2019] [Indexed: 12/31/2022]
Abstract
In recent years, biomaterial- and scaffold-based immunomodulation strategies were implemented in tissue regeneration efforts for manipulating macrophage polarization (a.k.a. phenotype or lineage commitment, or differentiation). Yet, most of our understanding of macrophage phenotype commitment and phagocytic capacity is limited to how physical cues (extracellular matrix stiffness, roughness, and topography) and soluble chemical cues (cytokines and chemokines released from the scaffold) influence macrophage polarization. In the context of immune response-tissue interaction, the mechanical cues experienced by the residing cells within the tissue also play a critical role in macrophage polarization and inflammatory response. However, there is no compiled study discussing the effect of the dynamic mechanical environment around the tissues on macrophage polarization and the innate immune response. The aim of this comprehensive review paper is 2-fold; (a) to highlight the importance of mechanical cues on macrophage lineage commitment and function and (b) to summarize the important studies dedicated to understand how macrophage polarization changes with different mechanical loading modalities. For the first time, this review paper compiles and compartmentalizes the studies investigating the role of dynamic mechanical loading with various modalities, amplitude, and frequency on macrophage differentiation. A deeper understanding of macrophage phenotype in mechanically dominant tissues (i.e. musculoskeletal tissues, lung tissues, and cardiovascular tissues) provides mechanistic insights into the design of mechano-immunomodulatory tissue scaffold for tissue regeneration.
Collapse
Affiliation(s)
- Sarah Adams
- Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH, 43606, USA
| | - Leah M Wuescher
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA
| | - Randall Worth
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA
| | - Eda Yildirim-Ayan
- Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH, 43606, USA.
- Department of Orthopaedic Surgery, University of Toledo Medical Center, Toledo, OH, 43614, USA.
| |
Collapse
|
203
|
Simon LR, Masters KS. Disease-inspired tissue engineering: Investigation of cardiovascular pathologies. ACS Biomater Sci Eng 2019; 6:2518-2532. [PMID: 32974421 DOI: 10.1021/acsbiomaterials.9b01067] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Once focused exclusively on the creation of tissues to repair or replace diseased or damaged organs, the field of tissue engineering has undergone an important evolution in recent years. Namely, tissue engineering techniques are increasingly being applied to intentionally generate pathological conditions. Motivated in part by the wide gap between 2D cultures and animal models in the current disease modeling continuum, disease-inspired tissue-engineered platforms have numerous potential applications, and may serve to advance our understanding and clinical treatment of various diseases. This review will focus on recent progress toward generating tissue-engineered models of cardiovascular diseases, including cardiac hypertrophy, fibrosis, and ischemia reperfusion injury, atherosclerosis, and calcific aortic valve disease, with an emphasis on how these disease-inspired platforms can be used to decipher disease etiology. Each pathology is discussed in the context of generating both disease-specific cells as well as disease-specific extracellular environments, with an eye toward future opportunities to integrate different tools to yield more complex and physiologically relevant culture platforms. Ultimately, the development of effective disease treatments relies upon our ability to develop appropriate experimental models; as cardiovascular diseases are the leading cause of death worldwide, the insights yielded by improved in vitro disease modeling could have substantial ramifications for public health and clinical care.
Collapse
Affiliation(s)
- LaTonya R Simon
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705
| | - Kristyn S Masters
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705.,Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705
| |
Collapse
|
204
|
Adhikari U, An X, Rijal N, Hopkins T, Khanal S, Chavez T, Tatu R, Sankar J, Little KJ, Hom DB, Bhattarai N, Pixley SK. Embedding magnesium metallic particles in polycaprolactone nanofiber mesh improves applicability for biomedical applications. Acta Biomater 2019; 98:215-234. [PMID: 31059833 DOI: 10.1016/j.actbio.2019.04.061] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 04/28/2019] [Accepted: 04/30/2019] [Indexed: 12/19/2022]
Abstract
Magnesium (Mg) metal is of great interest in biomedical applications, especially in tissue engineering. Mg exhibits excellent in vivo biocompatibility, biodegradability and, during degradation, releases Mg ions (Mg2+) with the potential to improve tissue repair. We used electrospinning technology to incorporate Mg particles into nanofibers. Various ratios of Mg metal microparticles (<44 µm diameter) were incorporated into nanofiber polycaprolactone (PCL) meshes. Physicochemical properties of the meshes were analyzed by scanning electron microscopy (SEM), Fourier-transform infrared spectroscopy (FTIR), mechanical tensile testing, X-ray diffractometry and UV-VIS spectrophotometry. Biological properties of meshes were evaluated in vitro and in vivo. Under mammalian cell culture conditions, Mg-containing meshes released hydrogen gas and relative amounts of free Mg2+ that reflected the Mg/PCL ratios. All meshes were non-cytotoxic for 3T3 fibroblasts and PC-12 pheochromocytoma cells. In vivo implantation under the skin of mice for 3, 8 and 28 days showed that Mg-containing meshes were well vascularized, with improved measures of inflammation and healing compared to meshes without Mg. Evidence included an earlier appearance and infiltration of tissue repairing macrophages and, after 28 days, evidence of more mature tissue remodeling. Thus, these new composite nanofiber meshes have promising material properties that mitigated inflammatory tissue responses to PCL alone and improved tissue healing, thus providing a suitable matrix for use in clinically relevant tissue engineering applications. STATEMENT OF SIGNIFICANCE: The biodegradable metal, magnesium, safely biodegrades in the body, releasing beneficial byproducts. To improve tissue delivery, magnesium metal particles were incorporated into electrospun nanofiber meshes composed of a biodegradable, biocompatible polymer, polycaprolactone (PCL). Magnesium addition, at several concentrations, did not alter PCL chemistry, but did alter physical properties. Under cell culture conditions, meshes released magnesium ions and hydrogen gas and were not cytotoxic for two cell types. After implantation in mice, the mesh with magnesium resulted in earlier appearance of M2-like, reparative macrophages and improved tissue healing versus mesh alone. This is in agreement with other studies showing beneficial effects of magnesium metal and provides a new type of scaffold material that will be useful in clinically relevant tissue engineering applications.
Collapse
|
205
|
Assaying How Phagocytic Success Depends on the Elasticity of a Large Target Structure. Biophys J 2019; 117:1496-1507. [PMID: 31586520 DOI: 10.1016/j.bpj.2019.08.043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 08/14/2019] [Accepted: 08/26/2019] [Indexed: 01/26/2023] Open
Abstract
Biofilm infections can consist of bacterial aggregates that are an order of magnitude larger than neutrophils, phagocytic immune cells that densely surround aggregates but do not enter them. Because a neutrophil is too small to engulf the entire aggregate, it must be able to detach and engulf a few bacteria at a time if it is to use phagocytosis to clear the infection. Current research techniques do not provide a method for determining how the success of phagocytosis, here defined as the complete engulfment of a piece of foreign material, depends on the mechanical properties of a larger object from which the piece must be removed before being engulfed. This article presents a step toward such a method. By varying polymer concentration or cross-linking density, the elastic moduli of centimeter-sized gels are varied over the range that was previously measured for Pseudomonas aeruginosa biofilms grown from clinical bacterial isolates. Human neutrophils are isolated from blood freshly drawn from healthy adult volunteers, exposed to gel containing embedded beads for 1 h, and removed from the gel. The percentage of collected neutrophils that contain beads that had previously been within the gels is used to measure successful phagocytic engulfment. Both increased polymer concentration in agarose gels and increased cross-linking density in alginate gels are associated with a decreased success of phagocytic engulfment. Upon plotting the percentage of neutrophils showing successful engulfment as a function of the elastic modulus of the gel to which they were applied, it is found that data from both alginate and agarose gels collapse onto the same curve. This suggests that gel mechanics may be impacting the success of phagocytosis and demonstrates that this experiment is a step toward realizing methods for measuring how the mechanics of a large target, or a large structure in which smaller targets are embedded, impact the success of phagocytic engulfment.
Collapse
|
206
|
Ai FF, Mao M, Zhang Y, Kang J, Zhu L. The in vivo biocompatibility of titanized polypropylene lightweight mesh is superior to that of conventional polypropylene mesh. Neurourol Urodyn 2019; 39:96-107. [PMID: 31584215 DOI: 10.1002/nau.24159] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 08/23/2019] [Indexed: 01/14/2023]
Abstract
OBJECTIVE To evaluate the histological response to and changes in the biomechanical properties of titanized polypropylene lightweight mesh and conventional polypropylene mesh at 1 and 12 weeks following implantation in the sheep vagina. METHODS We compared a titanized polypropylene lightweight mesh (TiLOOP Mesh) to a conventional polypropylene mesh (Gynemesh PS) in a sheep vagina model. Explants were harvested after 1 and 12 weeks (n = 6/mesh type/time point) for histological observation. After 12 weeks, mesh-tissue complex specimens were biomechanically assessed by a uniaxial tension system. RESULTS One week after implantation, there was no significant difference in the inflammatory response between the two groups. Twelve weeks after implantation, the TiLOOP light mesh elicited a lower inflammatory response than was observed for the Gynemesh PS (1.44 ± 0.61 vs 2.05 ± 0.80, P = .015). Twelve weeks after implantation, the collagen I/III ratio was lower in the TiLOOP light mesh group than in the Gynemesh PS group (9.41 ± 5.06 vs 15.21 ± 8.21, P = .019). The messenger RNA expression levels of the inflammatory factors interleukin 10 and tumor necrosis factor α were lower in the TiLOOP Mesh group than in the Gynemesh PS group at both 1 and 12 weeks (P < .05). There were no significant differences in any of the evaluated biomechanical characteristics between the two meshes (P > .05). CONCLUSION Although the titanized polypropylene lightweight mesh induces slightly less tissue reactivity and has better in vivo biocompatibility, further studies should be conducted including the complications and the success rate of pelvic organ prolapse in patients before recommending it in pelvic floor reconstruction.
Collapse
Affiliation(s)
- Fang-Fang Ai
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.,Department of Obstetrics and Gynecology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Meng Mao
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Ye Zhang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Jia Kang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Lan Zhu
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| |
Collapse
|
207
|
Baldwin M, Snelling S, Dakin S, Carr A. Augmenting endogenous repair of soft tissues with nanofibre scaffolds. J R Soc Interface 2019; 15:rsif.2018.0019. [PMID: 29695606 DOI: 10.1098/rsif.2018.0019] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 04/04/2018] [Indexed: 12/21/2022] Open
Abstract
As our ability to engineer nanoscale materials has developed we can now influence endogenous cellular processes with increasing precision. Consequently, the use of biomaterials to induce and guide the repair and regeneration of tissues is a rapidly developing area. This review focuses on soft tissue engineering, it will discuss the types of biomaterial scaffolds available before exploring physical, chemical and biological modifications to synthetic scaffolds. We will consider how these properties, in combination, can provide a precise design process, with the potential to meet the requirements of the injured and diseased soft tissue niche. Finally, we frame our discussions within clinical trial design and the regulatory framework, the consideration of which is fundamental to the successful translation of new biomaterials.
Collapse
Affiliation(s)
- Mathew Baldwin
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Sarah Snelling
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Stephanie Dakin
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Andrew Carr
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| |
Collapse
|
208
|
Young SA, Riahinezhad H, Amsden BG. In situ-forming, mechanically resilient hydrogels for cell delivery. J Mater Chem B 2019; 7:5742-5761. [PMID: 31531443 DOI: 10.1039/c9tb01398a] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Injectable, in situ-forming hydrogels can improve cell delivery in tissue engineering applications by facilitating minimally invasive delivery to irregular defect sites and improving cell retention and survival. Tissues targeted for cell delivery often undergo diverse mechanical loading including high stress, high strain, and repetitive loading conditions. This review focuses on the development of hydrogel systems that meet the requirements of mechanical resiliency, cytocompatibility, and injectability for such applications. First, we describe the most important design considerations for maintaining the viability and function of encapsulated cells, for reproducing the target tissue morphology, and for achieving degradation profiles that facilitate tissue replacement. Models describing the relationships between hydrogel structure and mechanical properties are described, focusing on design principles necessary for producing mechanically resilient hydrogels. The advantages and limitations of current strategies for preparing cytocompatible, injectable, and mechanically resilient hydrogels are reviewed, including double networks, nanocomposites, and high molecular weight amphiphilic copolymer networks. Finally, challenges and opportunities are outlined to guide future research in this developing field.
Collapse
Affiliation(s)
- Stuart A Young
- Department of Chemical Engineering, Queen's University, Kingston, ON, Canada.
| | - Hossein Riahinezhad
- Department of Chemical Engineering, Queen's University, Kingston, ON, Canada.
| | - Brian G Amsden
- Department of Chemical Engineering, Queen's University, Kingston, ON, Canada.
| |
Collapse
|
209
|
Gonzalez-Fernandez T, Sikorski P, Leach JK. Bio-instructive materials for musculoskeletal regeneration. Acta Biomater 2019; 96:20-34. [PMID: 31302298 PMCID: PMC6717669 DOI: 10.1016/j.actbio.2019.07.014] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 06/26/2019] [Accepted: 07/09/2019] [Indexed: 02/06/2023]
Abstract
The prevalence and cost of disorders affecting the musculoskeletal system are predicted to rise significantly in the coming years due to the aging global population and the increase of associated risk factors. Despite being the second largest cause of disability, the clinical options for therapeutic intervention remain limited. The clinical translation of cell-based therapies for the treatment of musculoskeletal disorders faces many challenges including maintenance of cell survival in the harsh in vivo environment and the lack of control over regulating cell phenotype upon implantation. In order to address these challenges, the development of bio-instructive materials to modulate cell behavior has taken center stage as a strategy to increase the therapeutic potential of various cell populations. However, the determination of the necessary cues for a specific application and how these signals should be presented from a biomaterial remains elusive. This review highlights recent biochemical and physical strategies used to engineer bio-instructive materials for the repair of musculoskeletal tissues. There is a particular emphasis on emerging efforts such as the engineering of immunomodulatory and antibacterial materials, as well as the incorporation of these strategies into biofabrication and organ-on-a-chip approaches. STATEMENT OF SIGNIFICANCE: Disorders affecting the musculoskeletal system affect individuals across the lifespan and have a profound effect on mobility and quality of life. While small defects in many tissues can heal successfully, larger defects are often unable to heal or instead heal with inferior quality fibrous tissue and require clinical intervention. Cell-based therapies are a promising option for clinical translation, yet challenges related to maintaining cell survival and instructing cell phenotype upon implantation have limited the success of this approach. Bio-instructive materials provide an exciting opportunity to modulate cell behavior and enhance the efficacy of cell-based approaches for musculoskeletal repair. However, the identification of critical instructive cues and how to present these stimuli is a focus of intense investigation. This review highlights recent biochemical and physical strategies used to engineer bio-instructive materials for the repair of musculoskeletal tissues, while also considering exciting progress in the engineering of immunomodulatory and antibacterial materials.
Collapse
Affiliation(s)
| | - Pawel Sikorski
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, USA; Department of Physics, Norwegian University of Science and Technology, NTNU, Trondheim, Norway
| | - J Kent Leach
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, USA; Department of Orthopaedic Surgery, School of Medicine, UC Davis Health, Sacramento, CA, USA.
| |
Collapse
|
210
|
Wang K, Shi L, Linthicum W, Man K, He X, Wen Q, Rojanasakul LW, Rojanasakul Y, Yang Y. Substrate Stiffness-Dependent Carbon Nanotube-Induced Lung Fibrogenesis. NANO LETTERS 2019; 19:5443-5451. [PMID: 31369708 PMCID: PMC6724206 DOI: 10.1021/acs.nanolett.9b01943] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Most living tissues exhibit the specific stiffness, which has been known to have profound influence on cell behaviors, yet how the stiffness affects cellular responses to engineered nanomaterials has not been elucidated. Particularly, discrepancies exist between in vitro and in vivo nanotoxicological studies. Here, we investigated the effects of substrate stiffness on the fibrogenic responses of normal human lung fibroblasts (NHLFs) to multiwalled carbon nanotubes (MWCNTs). NHLFs were grown on polyacrylamide (PAAm) hydrogels with the stiffness comparable to that of human normal and fibrotic lung tissues, and treated with MWCNTs for various time. The fibrogenic responses, including cell proliferation, reactive oxygen species production, and collagen I expression, of NHLFs to MWCNTs were observed to be regulated by substrate stiffness in a time-dependent manner. NHLFs generally were rounded on soft hydrogels and required a long treatment time to exhibit fibrogenic responses, while on stiff hydrogels the cells were well-spread with defined stress fibers and short-time MWCNTs treatment sufficiently induced the fibrogenic responses. Mechanistic studies showed that MWCNTs induced fibrogenesis of NHLFs through promoting expression and phosphorylation of focal adhesion kinase (FAK), while attenuating intracellular tension in the cells on stiff gels could increase MWCNTs uptake and thus elevate the induced fibrogenic responses. Moreover, we proposed a time-stiffness superposition principle to describe the equivalent effects of treatment time and substrate stiffness on nanomaterials-induced fibrogenesis, which suggested that increasing substrate stiffness expedited fibrogenesis and shed light on the rational design of in vitro models for nanotoxicological study.
Collapse
Affiliation(s)
- Kai Wang
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Lin Shi
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Will Linthicum
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, Massachusetts 01609, United States
| | - Kun Man
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Xiaoqing He
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, West Virginia 26506, United States
| | - Qi Wen
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, Massachusetts 01609, United States
- Department of Physics, Worcester Polytechnic Institute, Worcester, Massachusetts 01609, United States
| | - Liying Wang Rojanasakul
- Allergy and Clinical Immunology Branch, National Institute for Occupational Safety and Health, Morgantown, West Virginia 26505, United States
| | - Yon Rojanasakul
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, West Virginia 26506, United States
| | - Yong Yang
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| |
Collapse
|
211
|
Meli VS, Veerasubramanian PK, Atcha H, Reitz Z, Downing TL, Liu WF. Biophysical regulation of macrophages in health and disease. J Leukoc Biol 2019; 106:283-299. [PMID: 30861205 PMCID: PMC7001617 DOI: 10.1002/jlb.mr0318-126r] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Macrophages perform critical functions for homeostasis and immune defense in tissues throughout the body. These innate immune cells are capable of recognizing and clearing dead cells and pathogens, and orchestrating inflammatory and healing processes that occur in response to injury. In addition, macrophages are involved in the progression of many inflammatory diseases including cardiovascular disease, fibrosis, and cancer. Although it has long been known that macrophages respond dynamically to biochemical signals in their microenvironment, the role of biophysical cues has only recently emerged. Furthermore, many diseases that involve macrophages are also characterized by changes to the tissue biophysical environment. This review will discuss current knowledge about the effects of biophysical cues including matrix stiffness, material topography, and applied mechanical forces, on macrophage behavior. We will also describe the role of molecules that are known to be important for mechanotransduction, including adhesion molecules, ion channels, as well as nuclear mediators such as transcription factors, scaffolding proteins, and epigenetic regulators. Together, this review will illustrate a developing role of biophysical cues in macrophage biology, and also speculate upon molecular targets that may potentially be exploited therapeutically to treat disease.
Collapse
Affiliation(s)
- Vijaykumar S. Meli
- Department of Biomedical Engineering, University of California Irvine, CA 92697
- The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California Irvine, CA 92697
| | - Praveen K. Veerasubramanian
- Department of Biomedical Engineering, University of California Irvine, CA 92697
- The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California Irvine, CA 92697
| | - Hamza Atcha
- Department of Biomedical Engineering, University of California Irvine, CA 92697
- The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California Irvine, CA 92697
| | - Zachary Reitz
- Department of Biomedical Engineering, University of California Irvine, CA 92697
- The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California Irvine, CA 92697
| | - Timothy L. Downing
- Department of Biomedical Engineering, University of California Irvine, CA 92697
- The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California Irvine, CA 92697
- Department of Microbiology and Molecular Genetics, University of California Irvine, CA 92697
| | - Wendy F. Liu
- Department of Biomedical Engineering, University of California Irvine, CA 92697
- The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California Irvine, CA 92697
- Department of Chemical and Biomolecular Engineering, University of California Irvine, CA 92697
| |
Collapse
|
212
|
Kim TH, Kang MS, Mandakhbayar N, El-Fiqi A, Kim HW. Anti-inflammatory actions of folate-functionalized bioactive ion-releasing nanoparticles imply drug-free nanotherapy of inflamed tissues. Biomaterials 2019; 207:23-38. [DOI: 10.1016/j.biomaterials.2019.03.034] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 03/21/2019] [Accepted: 03/22/2019] [Indexed: 01/04/2023]
|
213
|
Andrechak JC, Dooling LJ, Discher DE. The macrophage checkpoint CD47 : SIRPα for recognition of 'self' cells: from clinical trials of blocking antibodies to mechanobiological fundamentals. Philos Trans R Soc Lond B Biol Sci 2019; 374:20180217. [PMID: 31431181 DOI: 10.1098/rstb.2018.0217] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Immunotherapies against some solid tumour types have recently shown unprecedented, durable cures in the clinic, and the most successful thus far involves blocking inhibitory receptor 'checkpoints' on T cells. A similar approach with macrophages is emerging by blocking the ubiquitously expressed 'marker of self' CD47 from binding the inhibitory receptor SIRPα on macrophages. Here, we first summarize available information on the safety and efficacy of CD47 blockade, which raises some safety concerns with the clearance of 'self' cells but also suggests some success against haematological (liquid) and solid cancers. Checkpoint blockade generally benefits from parallel activation of the immune cell, which can occur for macrophages in multiple ways, such as by combination with a second, tumour-opsonizing antibody and perhaps also via rigidity sensing. Cytoskeletal forces in phagocytosis and inhibitory 'self'-signalling are thus reviewed together with macrophage mechanosensing, which extends to regulating levels of SIRPα and the nuclear protein lamin A, which affects phenotype and cell trafficking. Considerations of such physical factors in cancer and the immune system can inform the design of new immunotherapies and help to refine existing therapies to improve safety and efficacy. This article is part of a discussion meeting issue 'Forces in cancer: interdisciplinary approaches in tumour mechanobiology'.
Collapse
Affiliation(s)
- Jason C Andrechak
- Biophysical Engineering Labs, University of Pennsylvania, Philadelphia, PA, USA.,Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, PA, USA
| | - Lawrence J Dooling
- Biophysical Engineering Labs, University of Pennsylvania, Philadelphia, PA, USA
| | - Dennis E Discher
- Biophysical Engineering Labs, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
214
|
Means AK, Dong P, Clubb FJ, Friedemann MC, Colvin LE, Shrode CA, Coté GL, Grunlan MA. A self-cleaning, mechanically robust membrane for minimizing the foreign body reaction: towards extending the lifetime of sub-Q glucose biosensors. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2019; 30:79. [PMID: 31240399 PMCID: PMC6988489 DOI: 10.1007/s10856-019-6282-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 06/17/2019] [Indexed: 06/09/2023]
Abstract
Long-term, subcutaneously implanted continuous glucose biosensors have the potential to improve diabetes management and reduce associated complications. However, the innate foreign body reaction (FBR) both alters the local glucose concentrations in the surrounding tissues and compromises glucose diffusion to the biosensor due to the recruitment of high-metabolizing inflammatory cells and the formation of a dense, collagenous fibrous capsule. Minimizing the FBR has mainly focused on "passively antifouling" materials that reduce initial cellular attachment, including poly(ethylene glycol) (PEG). Instead, the membrane reported herein utilizes an "actively antifouling" or "self-cleaning" mechanism to inhibit cellular attachment through continuous, cyclic deswelling/reswelling in response to normal temperature fluctuations of the subcutaneous tissue. This thermoresponsive double network (DN) membrane is based on N-isopropylacrylamide (NIPAAm) and 2-acrylamido-2-methylpropane sulfonic acid (AMPS) (75:25 and 100:0 NIPAAm:AMPS in the 1st and 2nd networks, respectively; "DN-25%"). The extent of the FBR reaction of a subcutaneously implanted DN-25% cylindrical membrane was evaluated in rodents in parallel with a PEG-diacrylate (PEG-DA) hydrogel as an established benchmark biocompatible control. Notably, the DN-25% implants were more than 25× stronger and tougher than the PEG-DA implants while maintaining a modulus near that of subcutaneous tissue. From examining the FBR at 7, 30 and 90 days after implantation, the thermoresponsive DN-25% implants demonstrated a rapid healing response and a minimal fibrous capsule (~20-25 µm), similar to the PEG-DA implants. Thus, the dynamic self-cleaning mechanism of the DN-25% membranes represents a new approach to limit the FBR while achieving the durability necessary for long-term implantable glucose biosensors.
Collapse
Affiliation(s)
- A Kristen Means
- Department of Materials Science & Engineering, Texas A&M University, College Station, TX, 77843-3003, USA
| | - Ping Dong
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843-3120, USA
| | - Fred J Clubb
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843-3120, USA
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX, 77843-4467, USA
| | - Molly C Friedemann
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX, 77843-4467, USA
| | - Lydia E Colvin
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843-3120, USA
| | - Courtney A Shrode
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843-3120, USA
| | - Gerard L Coté
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843-3120, USA
- Center for Remote Health Technologies Systems, Texas A&M University, College Station, TX, 77843-3120, USA
| | - Melissa A Grunlan
- Department of Materials Science & Engineering, Texas A&M University, College Station, TX, 77843-3003, USA.
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843-3120, USA.
- Center for Remote Health Technologies Systems, Texas A&M University, College Station, TX, 77843-3120, USA.
- Department of Chemistry, Texas A&M University, College Station, TX, 77843-3255, USA.
| |
Collapse
|
215
|
Xue YZB, Niu YM, Tang B, Wang CM. PCL/EUG scaffolds with tunable stiffness can regulate macrophage secretion behavior. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2019; 148:4-11. [PMID: 31226307 DOI: 10.1016/j.pbiomolbio.2019.05.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Osteoarthritis (OA) is a prevalent joint disorder worldwide. Recent studies suggested that macrophages play an important role in the progression of OA. However, the detailed pathology related to macrophages is still ambiguous, especially where related to mechanotransduction. In this study, polycaprolactone (PCL) and Eucommia Ulmoides Gum (EUG) composite scaffolds were first fabricated by electrospinning. The stiffness of as-fabricated scaffolds was altered by adjusting the PCL-to-EUG ratio. The mechanical properties, structural characteristics and chemical composition of the scaffolds were investigated using various materials characterization techniques. The results show that stiffness of the scaffolds was in the same range as that of cartilage tissues with OA. Confocal microscopy and reverse transcription-polymerase chain reaction (RT-PCR) were performed to investigate the macrophages cultured on the scaffolds. Significant morphological changes of cells were observed on PCL/EUG scaffolds with different stiffness. The expression of inflammatory and fibrosis-related cytokines increases as scaffold stiffness decreases, similar to the trend observed in OA progression.
Collapse
Affiliation(s)
- Y Z B Xue
- Department of Materials Science and Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, China; State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China; Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Y M Niu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - B Tang
- Department of Materials Science and Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, China; Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| | - C M Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China.
| |
Collapse
|
216
|
Gentile P, Garcovich S. Concise Review: Adipose-Derived Stem Cells (ASCs) and Adipocyte-Secreted Exosomal microRNA (A-SE-miR) Modulate Cancer Growth and proMote Wound Repair. J Clin Med 2019; 8:jcm8060855. [PMID: 31208047 PMCID: PMC6616456 DOI: 10.3390/jcm8060855] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 06/05/2019] [Accepted: 06/12/2019] [Indexed: 12/13/2022] Open
Abstract
Adipose-derived stem cells (ASCs) have been routinely used from several years in regenerative surgery without any definitive statement about their potential pro-oncogenic or anti-oncogenic role. ASCs has proven to favor tumor progression in several experimental cancer models, playing a central role in regulating tumor invasiveness and metastatic potential through several mechanisms, such as the paracrine release of exosomes containing pro-oncogenic molecules and the induction of epithelial-mesenchymal transition. However, the high secretory activity and the preferential tumor-targeting make also ASCs a potentially suitable vehicle for delivery of new anti-cancer molecules in tumor microenvironment. Nanotechnologies, viral vectors, drug-loaded exosomes, and micro-RNAs (MiR) represent additional new tools that can be applied for cell-mediated drug delivery in a tumor microenvironment. Recent studies revealed that the MiR play important roles in paracrine actions on adipose-resident macrophages, and their dysregulation has been implicated in the pathogenesis of obesity, diabetes, and diabetic complications as wounds. Numerous MiR are present in adipose tissues, actively participating in the regulation of adipogenesis, adipokine secretion, inflammation, and inter-cellular communications in the local tissues. These results provide important insights into Adipocyte-secreted exosomal microRNA (A-SE-MiR) function and they suggest evaluating the potential role of A-SE-MiR in tumor progression, the mechanisms underlying ASCs-cancer cell interplay and clinical safety of ASCs-based therapies.
Collapse
Affiliation(s)
- Pietro Gentile
- Surgical Science Department, Plastic and Reconstructive Surgery Unit, University of "Tor Vergata", 00133 Rome, Italy.
| | - Simone Garcovich
- Institute of Dermatology, F. Policlinico Gemelli IRCSS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| |
Collapse
|
217
|
Mukherjee S, Darzi S, Paul K, Werkmeister JA, Gargett CE. Mesenchymal stem cell-based bioengineered constructs: foreign body response, cross-talk with macrophages and impact of biomaterial design strategies for pelvic floor disorders. Interface Focus 2019; 9:20180089. [PMID: 31263531 PMCID: PMC6597526 DOI: 10.1098/rsfs.2018.0089] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/07/2019] [Indexed: 02/06/2023] Open
Abstract
An excessive foreign body response (FBR) has contributed to the adverse events associated with polypropylene mesh usage for augmenting pelvic organ prolapse surgery. Consequently, current biomaterial research considers the critical role of the FBR and now focuses on developing better biocompatible biomaterials rather than using inert implants to improve the clinical outcomes of their use. Tissue engineering approaches using mesenchymal stem cells (MSCs) have improved outcomes over traditional implants in other biological systems through their interaction with macrophages, the main cellular player in the FBR. The unique angiogenic, immunomodulatory and regenerative properties of MSCs have a direct impact on the FBR following biomaterial implantation. In this review, we focus on key aspects of the FBR to tissue-engineered MSC-based implants for supporting pelvic organs and beyond. We also discuss the immunomodulatory effects of the recently discovered endometrial MSCs on the macrophage response to new biomaterials designed for use in pelvic floor reconstructive surgery. We conclude with a focus on considerations in biomaterial design that take into account the FBR and will likely influence the development of the next generation of biomaterials for gynaecological applications.
Collapse
Affiliation(s)
- Shayanti Mukherjee
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria 3168, Australia.,CSIRO Manufacturing, Clayton, Victoria 3168, Australia
| | - Saeedeh Darzi
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
| | - Kallyanashis Paul
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria 3168, Australia
| | - Jerome A Werkmeister
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria 3168, Australia.,CSIRO Manufacturing, Clayton, Victoria 3168, Australia
| | - Caroline E Gargett
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria 3168, Australia
| |
Collapse
|
218
|
Markov PA, Khramova DS, Shumikhin KV, Nikitina IR, Beloserov VS, Martinson EA, Litvinets SG, Popov SV. Mechanical properties of the pectin hydrogels and inflammation response to their subcutaneous implantation. J Biomed Mater Res A 2019; 107:2088-2098. [DOI: 10.1002/jbm.a.36721] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 05/06/2019] [Accepted: 05/09/2019] [Indexed: 11/08/2022]
Affiliation(s)
- Pavel A. Markov
- Institute of Physiology, Komi Science CentreThe Urals Branch of the Russian Academy of Sciences Syktyvkar Russia
| | - Darya S. Khramova
- Institute of Physiology, Komi Science CentreThe Urals Branch of the Russian Academy of Sciences Syktyvkar Russia
| | - Konstantin V. Shumikhin
- Institute of Physiology, Komi Science CentreThe Urals Branch of the Russian Academy of Sciences Syktyvkar Russia
| | - Ida R. Nikitina
- Institute of Physiology, Komi Science CentreThe Urals Branch of the Russian Academy of Sciences Syktyvkar Russia
| | | | | | | | - Sergey V. Popov
- Institute of Physiology, Komi Science CentreThe Urals Branch of the Russian Academy of Sciences Syktyvkar Russia
| |
Collapse
|
219
|
Goswami R, Arya RK, Biswas D, Zhu X, Rahaman SO. Transient Receptor Potential Vanilloid 4 Is Required for Foreign Body Response and Giant Cell Formation. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:1505-1512. [PMID: 31121133 DOI: 10.1016/j.ajpath.2019.04.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 04/05/2019] [Accepted: 04/11/2019] [Indexed: 12/11/2022]
Abstract
The presence of biomaterials and devices implanted into soft tissue is associated with development of a foreign body response (FBR), a chronic inflammatory condition that can ultimately lead to implant failure, which may cause harm to or death of the patient. Development of FBR includes activation of macrophages at the tissue-implant interface, generation of destructive foreign body giant cells (FBGCs), and generation of fibrous tissue that encapsulates the implant. However, the mechanisms underlying the FBR remain poorly understood, as neither the materials composing the implants nor their chemical properties can explain triggering of the FBR. Herein, we report that genetic ablation of transient receptor potential vanilloid 4 (TRPV4), a Ca2+-permeable mechanosensitive cation channel in the transient receptor potential vanilloid family, protects TRPV4 knockout mice from FBR-related events. The mice showed diminished collagen deposition along with reduced macrophage accumulation and FBGC formation compared with wild-type mice in a s.c. implantation model. Analysis of macrophage markers in spleen tissues and peritoneal cavity showed that the TRPV4 deficiency did not impair basal macrophage maturation. Furthermore, genetic deficiency or pharmacologic antagonism of TRPV4 blocked cytokine-induced FBGC formation, which was restored by lentivirus-mediated TRPV4 reintroduction. Taken together, these results suggest an important, previously unknown, role for TRPV4 in FBR.
Collapse
Affiliation(s)
- Rishov Goswami
- Department of Nutrition and Food Science, University of Maryland, College Park, Maryland
| | - Rakesh K Arya
- Department of Nutrition and Food Science, University of Maryland, College Park, Maryland
| | - Debabrata Biswas
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland
| | - Xiaoping Zhu
- Department of Veterinary Medicine, University of Maryland, College Park, Maryland
| | - Shaik O Rahaman
- Department of Nutrition and Food Science, University of Maryland, College Park, Maryland.
| |
Collapse
|
220
|
Dynamic fibroblast contractions attract remote macrophages in fibrillar collagen matrix. Nat Commun 2019; 10:1850. [PMID: 31015429 PMCID: PMC6478854 DOI: 10.1038/s41467-019-09709-6] [Citation(s) in RCA: 137] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 03/26/2019] [Indexed: 12/23/2022] Open
Abstract
Macrophage (Mϕ)-fibroblast interactions coordinate tissue repair after injury whereas miscommunications can result in pathological healing and fibrosis. We show that contracting fibroblasts generate deformation fields in fibrillar collagen matrix that provide far-reaching physical cues for Mϕ. Within collagen deformation fields created by fibroblasts or actuated microneedles, Mϕ migrate towards the force source from several hundreds of micrometers away. The presence of a dynamic force source in the matrix is critical to initiate and direct Mϕ migration. In contrast, collagen condensation and fiber alignment resulting from fibroblast remodelling activities or chemotactic signals are neither required nor sufficient to guide Mϕ migration. Binding of α2β1 integrin and stretch-activated channels mediate Mϕ migration and mechanosensing in fibrillar collagen ECM. We propose that Mϕ mechanosense the velocity of local displacements of their substrate, allowing contractile fibroblasts to attract Mϕ over distances that exceed the range of chemotactic gradients. Macrophages play an important role in wound healing but the guidance cues driving macrophages to sites of repair are still not clear. Here the authors discover that macrophages are attracted to contracting fibroblasts by responding to locally sensed displacements of collagen fibres.
Collapse
|
221
|
Sridharan R, Cavanagh B, Cameron AR, Kelly DJ, O'Brien FJ. Material stiffness influences the polarization state, function and migration mode of macrophages. Acta Biomater 2019; 89:47-59. [PMID: 30826478 DOI: 10.1016/j.actbio.2019.02.048] [Citation(s) in RCA: 231] [Impact Index Per Article: 46.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 02/20/2019] [Accepted: 02/27/2019] [Indexed: 01/25/2023]
Abstract
Biomaterial implantation is followed by an inflammatory cascade dominated by macrophages, which determine implant acceptance or rejection through pro- and anti-inflammatory polarization states (Anderson et al., 2008; Brown and Badylak, 2013). It is known that chemical signals such as bacterial endotoxins and cytokines (IL4) can direct macrophage polarization (Mantovani et al., 2004); however, recent evidence implicates biophysical cues in this process (McWhorter et al., 2015; Patel et al., 2012). Here we report that THP-1 derived macrophages cultured on collagen-coated polyacrylamide gels of varying stiffness adapt their polarization state, functional roles and migration mode according to the stiffness of the underlying substrate. Through gene expression and protein secretion analysis, we show that stiff polyacrylamide gels (323 kPa) prime macrophages towards a pro-inflammatory phenotype with impaired phagocytosis in macrophages, while soft (11 kPa) and medium (88 kPa) stiffness gels prime cells towards an anti-inflammatory, highly phagocytic phenotype. Furthermore, we show that stiffness dictates the migration mode of macrophages; on soft and medium stiffness gels, cells display Rho-A kinase (ROCK)-dependent, podosome-independent fast amoeboid migration and on stiff gels they adopt a ROCK-independent, podosome-dependent slow mesenchymal migration mode. We also provide a mechanistic insight into this process by showing that the anti-inflammatory property of macrophages on soft and medium gels is ROCK-dependent and independent of the ligand presented to them. Together, our results demonstrate that macrophages adapt their polarization, function and migration mode in response to the stiffness of the underlying substrate and suggest that biomaterial stiffness is capable of directing macrophage behaviour independent of the biochemical cues being presented to them. The results from this study establish an important role for substrate stiffness in directing macrophage behaviour, and will lead to the design of immuno-informed biomaterials that are capable of modulating the macrophage response after implantation. STATEMENT OF SIGNIFICANCE: Biomaterial implantation is followed by an inflammatory cascade dominated by macrophages, which determine implant acceptance or rejection through pro- and anti-inflammatory polarization states. It is known that chemical signals can direct macrophage polarization; however, recent evidence implicates biophysical cues in this process. Here we report that macrophages cultured on gels of varying stiffness adapt their polarization state, functional roles and migration mode according to the stiffness of the underlying substrate. The results from this study establish an important role for substrate stiffness in directing macrophage behaviour, and will lead to the design of immuno-informed biomaterials that are capable of modulating the macrophage response after implantation.
Collapse
Affiliation(s)
- Rukmani Sridharan
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, Dublin 2, Ireland; Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland; Advanced Materials Bio-Engineering Research (AMBER) Centre, Trinity College Dublin, Dublin 2, Ireland
| | - Brenton Cavanagh
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, Dublin 2, Ireland; Cellular and Molecular Imaging Core, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland
| | - Andrew R Cameron
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, Dublin 2, Ireland; Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland; Advanced Materials Bio-Engineering Research (AMBER) Centre, Trinity College Dublin, Dublin 2, Ireland
| | - Daniel J Kelly
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, Dublin 2, Ireland; Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland; Advanced Materials Bio-Engineering Research (AMBER) Centre, Trinity College Dublin, Dublin 2, Ireland
| | - Fergal J O'Brien
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, Dublin 2, Ireland; Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland; Advanced Materials Bio-Engineering Research (AMBER) Centre, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
222
|
3D type I collagen environment leads up to a reassessment of the classification of human macrophage polarizations. Biomaterials 2019; 208:98-109. [PMID: 31005702 DOI: 10.1016/j.biomaterials.2019.04.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 03/21/2019] [Accepted: 04/11/2019] [Indexed: 12/20/2022]
Abstract
Macrophages have multiple roles in development, tissue homeostasis and repair and present a high degree of phenotypic plasticity embodied in the concept of polarization. One goal of macrophage biology field is to characterize these polarizations at the molecular level. To achieve this task, it is necessary to integrate how physical environment signals are interpreted by macrophages under immune stimulation. In this work, we study how a 3D scaffold obtained from polymerized fibrillar rat type I collagen modulates the polarizations of human macrophages and reveal that some traditionally used markers should be reassessed. We demonstrate that integrin β2 is a regulator of STAT1 phosphorylation in response to IFNγ/LPS as well as responsible for the inhibition of ALOX15 expression in response to IL-4/IL-13 in 3D. Meanwhile, we also find that the CCL19/CCL20 ratio is reverted in 3D under IFNγ/LPS stimulation. 3D also induces the priming of the NLRP3 inflammasome resulting in an increased IL-1β and IL-6 secretion. These results give the molecular basis for assessing collagen induced immunomodulation of human macrophages in various physiological and pathological contexts such as cancer.
Collapse
|
223
|
Pageon SV, Govendir MA, Kempe D, Biro M. Mechanoimmunology: molecular-scale forces govern immune cell functions. Mol Biol Cell 2019; 29:1919-1926. [PMID: 30088799 PMCID: PMC6232972 DOI: 10.1091/mbc.e18-02-0120] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Immune cell recognition of antigens is a pivotal process in initiating immune responses against injury, pathogens, and cancers. Breakthroughs over the past decade support a major role for mechanical forces in immune responses, laying the foundation for the emerging field of mechanoimmunology. In this Perspective, we discuss the mechanical forces acting at the level of ligand–receptor interactions and how they underpin receptor triggering, signal initiation, and immune cell activation. We also highlight the novel biophysical tools and advanced imaging techniques that have afforded us the recent progress in our understanding of the role of forces in immune cell functions.
Collapse
Affiliation(s)
- Sophie V Pageon
- EMBL Australia, Single Molecule Science Node, School of Medical Sciences, and
| | - Matt A Govendir
- EMBL Australia, Single Molecule Science Node, School of Medical Sciences, and
| | - Daryan Kempe
- EMBL Australia, Single Molecule Science Node, School of Medical Sciences, and
| | - Maté Biro
- EMBL Australia, Single Molecule Science Node, School of Medical Sciences, and.,ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
224
|
Kim TH, Ly C, Christodoulides A, Nowell CJ, Gunning PW, Sloan EK, Rowat AC. Stress hormone signaling through β-adrenergic receptors regulates macrophage mechanotype and function. FASEB J 2019; 33:3997-4006. [PMID: 30509116 PMCID: PMC6404566 DOI: 10.1096/fj.201801429rr] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 11/05/2018] [Indexed: 12/11/2022]
Abstract
Critical functions of immune cells require them to rapidly change their shape and generate forces in response to cues from their surrounding environment. However, little is known about how soluble factors that may be present in the microenvironment modulate key aspects of cellular mechanobiology-such as immune cell deformability and force generation-to impact functions such as phagocytosis and migration. Here we show that signaling by soluble stress hormones through β-adrenoceptors (β-AR) reduces the deformability of macrophages; this is dependent on changes in the organization of the actin cytoskeleton and is associated with functional changes in phagocytosis and migration. Pharmacologic interventions reveal that the impact of β-AR signaling on macrophage deformability is dependent on actin-related proteins 2/3, indicating that stress hormone signaling through β-AR shifts actin organization to favor branched structures rather than linear unbranched actin filaments. These findings show that through remodeling of the actin cytoskeleton, β-AR-mediated stress hormone signaling modulates macrophage mechanotype to impact functions that play a critical role in immune response.-Kim, T.-H., Ly, C., Christodoulides, A., Nowell, C. J., Gunning, P. W., Sloan, E. K., Rowat, A. C. Stress hormone signaling through β-adrenergic receptors regulates macrophage mechanotype and function.
Collapse
Affiliation(s)
- Tae-Hyung Kim
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California, USA
- Cousins Center for Psychoneuroimmunology, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, California, USA
| | - Chau Ly
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California, USA
- Department of Bioengineering, University of California, Los Angeles, California, USA
| | - Alexei Christodoulides
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California, USA
| | - Cameron J. Nowell
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Peter W. Gunning
- School of Medical Sciences, University of New South Wales Sydney, Kensington, New South Wales, Australia
| | - Erica K. Sloan
- Cousins Center for Psychoneuroimmunology, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, California, USA
- UCLA Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California, USA
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; and
- UCLA AIDS Institute, University of California, Los Angeles, California, USA
| | - Amy C. Rowat
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California, USA
- Department of Bioengineering, University of California, Los Angeles, California, USA
- UCLA Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California, USA
| |
Collapse
|
225
|
Hsieh JY, Keating MT, Smith TD, Meli VS, Botvinick EL, Liu WF. Matrix crosslinking enhances macrophage adhesion, migration, and inflammatory activation. APL Bioeng 2019; 3:016103. [PMID: 31069336 PMCID: PMC6481736 DOI: 10.1063/1.5067301] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 03/05/2019] [Indexed: 12/13/2022] Open
Abstract
Macrophages are versatile cells of the innate immune system that can adopt a variety of functional phenotypes depending on signals in their environment. In previous work, we found that culture of macrophages on fibrin, the provisional extracellular matrix protein, inhibits their inflammatory activation when compared to cells cultured on polystyrene surfaces. Here, we sought to investigate the role of matrix stiffness in the regulation of macrophage activity by manipulating the mechanical properties of fibrin. We utilize a photo-initiated crosslinking method to introduce dityrosine crosslinks to a fibrin gel and confirm an increase in gel stiffness through active microrheology. We observe that matrix crosslinking elicits distinct changes in macrophage morphology, integrin expression, migration, and inflammatory activation. Macrophages cultured on a stiffer substrate exhibit greater cell spreading and expression of αM integrin. Furthermore, macrophages cultured on crosslinked fibrin exhibit increased motility. Finally, culture of macrophages on photo-crosslinked fibrin enhances their inflammatory activation compared to unmodified fibrin, suggesting that matrix crosslinking regulates the functional activation of macrophages. These findings provide insight into how the physical properties of the extracellular matrix might control macrophage behavior during inflammation and wound healing.
Collapse
Affiliation(s)
- Jessica Y Hsieh
- Department of Biomedical Engineering and The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California Irvine, Irvine, California 92697, USA
| | - Mark T Keating
- Department of Biomedical Engineering and The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California Irvine, Irvine, California 92697, USA
| | - Tim D Smith
- Department of Biomedical Engineering and The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California Irvine, Irvine, California 92697, USA
| | - Vijaykumar S Meli
- Department of Biomedical Engineering and The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California Irvine, Irvine, California 92697, USA
| | - Elliot L Botvinick
- Department of Biomedical Engineering and The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California Irvine, Irvine, California 92697, USA
| | - Wendy F Liu
- Department of Biomedical Engineering and The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California Irvine, Irvine, California 92697, USA
| |
Collapse
|
226
|
Trel'ová D, Salgarella AR, Ricotti L, Giudetti G, Cutrone A, Šrámková P, Zahoranová A, Chorvát D, Haško D, Canale C, Micera S, Kronek J, Menciassi A, Lacík I. Soft Hydrogel Zwitterionic Coatings Minimize Fibroblast and Macrophage Adhesion on Polyimide Substrates. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2019; 35:1085-1099. [PMID: 29792034 DOI: 10.1021/acs.langmuir.8b00765] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Minimizing the foreign body reaction to polyimide-based implanted devices plays a pivotal role in several biomedical applications. In this work, we propose materials exhibiting nonbiofouling properties and a Young's modulus reflecting that of soft human tissues. We describe the synthesis, characterization, and in vitro validation of poly(carboxybetaine) hydrogel coatings covalently attached to polyimide substrates via a photolabile 4-azidophenyl group, incorporated in poly(carboxybetaine) chains at two concentrations of 1.6 and 3.1 mol %. The presence of coatings was confirmed by attenuated total reflectance Fourier transform infrared spectroscopy. White light interferometry was used to evaluate the coating continuity and thickness (between 3 and 6 μm under dry conditions). Confocal laser scanning microscopy allowed us to quantify the thickness of the swollen hydrogel coatings that ranged between 13 and 32 μm. The different hydrogel formulations resulted in stiffness values ranging from 2 to 19 kPa and led to different fibroblast and macrophage responses in vitro. Both cell types showed a minimum adhesion on the softest hydrogel type. In addition, both the overall macrophage activation and cytotoxicity were observed to be negligible for all of the tested material formulations. These results are a promising starting point toward future advanced implantable systems. In particular, such technology paves the way for novel neural interfaces able to minimize the fibrotic reaction, once implanted in vivo, and to maximize their long-term stability and functionality.
Collapse
Affiliation(s)
- Dušana Trel'ová
- Department for Biomaterials Research , Polymer Institute of the Slovak Academy of Sciences , Dúbravská cesta 9 , 845 41 Bratislava , Slovakia
| | - Alice Rita Salgarella
- The BioRobotics Institute, Scuola Superiore Sant'Anna , Viale R. Piaggio 34 , 56025 Pontedera ( PI ), Italy
| | - Leonardo Ricotti
- The BioRobotics Institute, Scuola Superiore Sant'Anna , Viale R. Piaggio 34 , 56025 Pontedera ( PI ), Italy
| | - Guido Giudetti
- The BioRobotics Institute, Scuola Superiore Sant'Anna , Viale R. Piaggio 34 , 56025 Pontedera ( PI ), Italy
| | - Annarita Cutrone
- The BioRobotics Institute, Scuola Superiore Sant'Anna , Viale R. Piaggio 34 , 56025 Pontedera ( PI ), Italy
- SMANIA srl, via G. Volpe 12 , 56121 Pisa , Italy
| | - Petra Šrámková
- Department for Biomaterials Research , Polymer Institute of the Slovak Academy of Sciences , Dúbravská cesta 9 , 845 41 Bratislava , Slovakia
| | - Anna Zahoranová
- Department for Biomaterials Research , Polymer Institute of the Slovak Academy of Sciences , Dúbravská cesta 9 , 845 41 Bratislava , Slovakia
| | - Dušan Chorvát
- International Laser Centre , Ilkovičova 3 , Bratislava 841 04 , Slovak Republic
| | - Daniel Haško
- International Laser Centre , Ilkovičova 3 , Bratislava 841 04 , Slovak Republic
| | - Claudio Canale
- Department of Physics , University of Genova , Via dodecaneso 33 , 16133 Genova , Italy
- Department of Nanophysics , Istituto Italiano di Tecnologia (IIT) , Via Morego 30 , 16163 Genova , Italy
| | - Silvestro Micera
- The BioRobotics Institute, Scuola Superiore Sant'Anna , Viale R. Piaggio 34 , 56025 Pontedera ( PI ), Italy
- Bertarelli Foundation Chair in Translational Neuroengineering, Center for Neuroprosthetics and Institute of Bioengineering , Ecole Polytechnique Federale de Lausanne , 1015 , Lausanne , Switzerland
| | - Juraj Kronek
- Department for Biomaterials Research , Polymer Institute of the Slovak Academy of Sciences , Dúbravská cesta 9 , 845 41 Bratislava , Slovakia
| | - Arianna Menciassi
- The BioRobotics Institute, Scuola Superiore Sant'Anna , Viale R. Piaggio 34 , 56025 Pontedera ( PI ), Italy
| | - Igor Lacík
- Department for Biomaterials Research , Polymer Institute of the Slovak Academy of Sciences , Dúbravská cesta 9 , 845 41 Bratislava , Slovakia
| |
Collapse
|
227
|
Qin XH, Senturk B, Valentin J, Malheiro V, Fortunato G, Ren Q, Rottmar M, Maniura-Weber K. Cell-Membrane-Inspired Silicone Interfaces that Mitigate Proinflammatory Macrophage Activation and Bacterial Adhesion. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2019; 35:1882-1894. [PMID: 30153734 DOI: 10.1021/acs.langmuir.8b02292] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Biofouling on silicone implants causes serious complications such as fibrotic encapsulation, bacterial infection, and implant failure. Here we report the development of antifouling, antibacterial silicones through covalent grafting with a cell-membrane-inspired zwitterionic gel layer composed of 2-methacryolyl phosphorylcholine (MPC). To investigate how substrate properties influence cell adhesion, we cultured human-blood-derived macrophages and Escherichia coli on poly(dimethylsiloxane) (PDMS) and MPC gel surfaces with a range of 0.5-50 kPa in stiffness. Cells attach to glass, tissue culture polystyrene, and PDMS surfaces, but they fail to form stable adhesions on MPC gel surfaces due to their superhydrophilicity and resistance to biofouling. Cytokine secretion assays confirm that MPC gels have a much lower potential to trigger proinflammatory macrophage activation than PDMS. Finally, modification of the PDMS surface with a long-term stable hydrogel layer was achieved by the surface-initiated atom-transfer radical polymerization (SI-ATRP) of MPC and confirmed by the decrease in contact angle from 110 to 20° and the >70% decrease in the attachment of macrophages and bacteria. This study provides new insights into the design of antifouling and antibacterial interfaces to improve the long-term biocompatibility of medical implants.
Collapse
|
228
|
Cellular responses to thermoresponsive stiffness memory elastomer nanohybrid scaffolds by 3D-TIPS. Acta Biomater 2019; 85:157-171. [PMID: 30557696 DOI: 10.1016/j.actbio.2018.12.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Revised: 11/11/2018] [Accepted: 12/13/2018] [Indexed: 12/12/2022]
Abstract
Increasing evidence suggests the contribution of the dynamic mechanical properties of the extracellular matrix (ECM) to regulate tissue remodeling and regeneration. Following our recent study on a family of thermoresponsive 'stiffness memory' elastomeric nanohybrid scaffolds manufactured via an indirect 3D printing guided thermally-induced phase separation process (3D-TIPS), this work reports in vitro and in vivo cellular responses towards these scaffolds with different initial stiffness and hierarchically interconnected porous structure. The viability of mouse embryonic dermal fibroblasts in vitro and the tissue responses during the stiffness softening of the scaffolds subcutaneously implanted in rats for three months were evaluated by immunohistochemistry and histology. Scaffolds with a higher initial stiffness and a hierarchical porous structure outperformed softer ones, providing initial mechanical support to cells and surrounding tissues before promoting cell and tissue growth during stiffness softening. Vascularization was guided throughout the digitally printed interconnected networks. All scaffolds exhibited polarization of the macrophage response from a macrophage phenotype type I (M1) towards a macrophage phenotype type II (M2) and down-regulation of the T-cell proliferative response with increasing implantation time; however, scaffolds with a more pronounced thermo-responsive stiffness memory mechanism exerted higher inflammo-informed effects. These results pave the way for personalized and biologically responsive soft tissue implants and implantable device with better mechanical matches, angiogenesis and tissue integration. Statement of Significance This work reports cellular responses to a family of 3D-TIPS thermoresponsive nanohybrid elastomer scaffolds with different stiffness softening both in vitro and in vivo rat models. The results, for the first time, have revealed the effects of initial stiffness and dynamic stiffness softening of the scaffolds on tissue integration, vascularization and inflammo-responses, without coupling chemical crosslinking processes. The 3D printed, hierarchically interconnected porous structures guide the growth of myofibroblasts, collagen fibers and blood vessels in real 3D scales. In vivo study on those unique smart elastomer scaffolds will help pave the way for personalized and biologically responsive soft tissue implants and implantable devices with better mechanical matches, angiogenesis and tissue integration.
Collapse
|
229
|
Kim JK, Shin YJ, Ha LJ, Kim DH, Kim DH. Unraveling the Mechanobiology of the Immune System. Adv Healthc Mater 2019; 8:e1801332. [PMID: 30614636 PMCID: PMC7700013 DOI: 10.1002/adhm.201801332] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 12/01/2018] [Indexed: 12/20/2022]
Abstract
Cells respond and actively adapt to environmental cues in the form of mechanical stimuli. This extends to immune cells and their critical role in the maintenance of tissue homeostasis. Multiple recent studies have begun illuminating underlying mechanisms of mechanosensation in modulating immune cell phenotypes. Since the extracellular microenvironment is critical to modify cellular physiology that ultimately determines the functionality of the cell, understanding the interactions between immune cells and their microenvironment is necessary. This review focuses on mechanoregulation of immune responses mediated by macrophages, dendritic cells, and T cells, in the context of modern mechanobiology.
Collapse
Affiliation(s)
- Jeong-Ki Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Yu Jung Shin
- Department of Bioengineering, University of Washington, Seattle, WA 98109, USA
| | - Leslie Jaesun Ha
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Deok-Ho Kim
- Department of Bioengineering, University of Washington, Seattle, WA 98109, USA
| | - Dong-Hwee Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| |
Collapse
|
230
|
Kim M, Lee S, Ki CS. Cellular Behavior of RAW264.7 Cells in 3D Poly(ethylene glycol) Hydrogel Niches. ACS Biomater Sci Eng 2019; 5:922-932. [PMID: 33405849 DOI: 10.1021/acsbiomaterials.8b01150] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Although macrophages undergo dynamic cellular responses in diverse extracellular environments, macrophage research has mostly relied on conventional culture methodologies such as two-dimensional and suspension cultures. In contrast, recent efforts have revealed evidence of the characteristic cellular behaviors of macrophages in actual tissues using a three-dimensional (3D) culture matrix. In this work, we exploited a poly(ethylene glycol)-based hydrogel as a macrophage culture matrix and observed cellular behaviors in 3D by manipulating the matrix properties. In the 3D microenvironment, macrophage-like RAW264.7 cells proliferated and formed spherical clusters by degrading the surrounding hydrogel network. Interestingly, we observed the significant upregulation of matrix metalloproteinases (MMPs) (i.e., MMP9 and MMP14) as well as M1 polarization markers (i.e., iNOS, COX2, TNF-α) in 3D, whereas M2 polarization markers (i.e., CD206, Arg1, TGF-β) were downregulated. Specifically, the expressions of both M1 and M2 markers were simultaneously increased in a stiff matrix compared to those of a soft matrix. In addition, matrix degradability significantly influenced the TNF-α secretion of encapsulated RAW264.7 cells. The MMP sensitivity of the hydrogel decreased TNF-α expression in a soft matrix, whereas it upregulated TNF-α in a stiff matrix compared to those of MMP-insensitive hydrogel. These findings suggest that the highly tunable poly(ethylene glycol) hydrogels can dictate macrophage behavior by altering the surrounding 3D microenvironment.
Collapse
|
231
|
Ji Y, Li J, Wei Y, Gao W, Fu X, Wang Y. Substrate stiffness affects the immunosuppressive and trophic function of hMSCs via modulating cytoskeletal polymerization and tension. Biomater Sci 2019; 7:5292-5300. [DOI: 10.1039/c9bm01202h] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Soft substrates improve the immunosuppressive and trophic function of hMSCs via cytoskeleton inhibition.
Collapse
Affiliation(s)
- Yurong Ji
- School of Materials Science and Engineering
- South China University of Technology
- Guangzhou 510640
- PR China
- National Engineering Research Center for Tissue Restoration and Reconstruction
| | - Jing Li
- School of Materials Science and Engineering
- South China University of Technology
- Guangzhou 510640
- PR China
- National Engineering Research Center for Tissue Restoration and Reconstruction
| | - Yingqi Wei
- Key Laboratory of Biomedical Engineering of Guangdong Province and Innovation Center for Tissue Restoration and Reconstruction
- South China University of Technology
- Guangzhou
- PR China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education
| | - Wendong Gao
- Key Laboratory of Biomedical Engineering of Guangdong Province and Innovation Center for Tissue Restoration and Reconstruction
- South China University of Technology
- Guangzhou
- PR China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education
| | - Xiaoling Fu
- School of Materials Science and Engineering
- South China University of Technology
- Guangzhou 510640
- PR China
- National Engineering Research Center for Tissue Restoration and Reconstruction
| | - Yingjun Wang
- Key Laboratory of Biomedical Engineering of Guangdong Province and Innovation Center for Tissue Restoration and Reconstruction
- South China University of Technology
- Guangzhou
- PR China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education
| |
Collapse
|
232
|
Ščigalková I, Bystroňová J, Kovářová L, Pravda M, Velebný V, Riabov V, Klüter H, Kzhyshkowska J, Vrana NE. The effect of healing phenotype-inducing cytokine formulations within soft hydrogels on encapsulated monocytes and incoming immune cells. RSC Adv 2019; 9:21396-21404. [PMID: 35521319 PMCID: PMC9066154 DOI: 10.1039/c9ra02878a] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 06/17/2019] [Indexed: 11/21/2022] Open
Abstract
Hydrogels made from the derivatives of gelatin and hyaluronic acid were used as coatings to control the immune responses.
Collapse
Affiliation(s)
| | | | - Lenka Kovářová
- Contipro a.s
- 561 02 Dolni Dobrouc
- Czech Republic
- Institute of Physical Chemistry
- Faculty of Chemistry
| | | | | | - Vladimir Riabov
- Institute for Transfusion Medicine and Immunology
- Medical Faculty Mannheim
- University of Heidelberg
- 68167 Mannheim
- Germany
| | - Harald Klüter
- Institute for Transfusion Medicine and Immunology
- Medical Faculty Mannheim
- University of Heidelberg
- 68167 Mannheim
- Germany
| | - Julia Kzhyshkowska
- Institute for Transfusion Medicine and Immunology
- Medical Faculty Mannheim
- University of Heidelberg
- 68167 Mannheim
- Germany
| | - Nihal Engin Vrana
- Protip Medical
- 67000 Strasbourg
- France
- Inserm UMR 1121, Biomaterials and Bioengineering
- 67085 Strasbourg
| |
Collapse
|
233
|
Morikawa S, Iribar H, Gutiérrez-Rivera A, Ezaki T, Izeta A. Pericytes in Cutaneous Wound Healing. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1147:1-63. [DOI: 10.1007/978-3-030-16908-4_1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
234
|
Li Z, Bratlie KM. How Cross-Linking Mechanisms of Methacrylated Gellan Gum Hydrogels Alter Macrophage Phenotype. ACS APPLIED BIO MATERIALS 2018; 2:217-225. [DOI: 10.1021/acsabm.8b00562] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Zhuqing Li
- Department of Materials Science & Engineering, Iowa State University, Ames, Iowa 50011, United States
| | - Kaitlin M. Bratlie
- Department of Materials Science & Engineering, Iowa State University, Ames, Iowa 50011, United States
- Department of Chemical & Biological Engineering, Iowa State University, Ames, Iowa 50011, United States
- Division of Materials Sciences & Engineering, Ames National Laboratory, Ames, Iowa 50011, United States
| |
Collapse
|
235
|
Bertucci TB, Dai G. Biomaterial Engineering for Controlling Pluripotent Stem Cell Fate. Stem Cells Int 2018; 2018:9068203. [PMID: 30627175 PMCID: PMC6304878 DOI: 10.1155/2018/9068203] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 10/11/2018] [Indexed: 01/02/2023] Open
Abstract
Pluripotent stem cells (PSCs) represent an exciting cell source for tissue engineering and regenerative medicine due to their self-renewal and differentiation capacities. The majority of current PSC protocols rely on 2D cultures and soluble factors to guide differentiation; however, many other environmental signals are beginning to be explored using biomaterial platforms. Biomaterials offer new opportunities to engineer the stem cell niches and 3D environments for exploring biophysical and immobilized signaling cues to further our control over stem cell fate. Here, we review the biomaterial platforms that have been engineered to control PSC fate. We explore how altering immobilized biochemical cues and biophysical cues such as dimensionality, stiffness, and topography can enhance our control over stem cell fates. Finally, we highlight biomaterial culture systems that assist in the translation of PSC technologies for clinical applications.
Collapse
Affiliation(s)
- Taylor B Bertucci
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| | - Guohao Dai
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| |
Collapse
|
236
|
Sridharan R, Ryan EJ, Kearney CJ, Kelly DJ, O’Brien FJ. Macrophage Polarization in Response to Collagen Scaffold Stiffness Is Dependent on Cross-Linking Agent Used To Modulate the Stiffness. ACS Biomater Sci Eng 2018; 5:544-552. [DOI: 10.1021/acsbiomaterials.8b00910] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Rukmani Sridharan
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, Dublin 2, Ireland
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
- Advanced Materials Bio-Engineering Research Centre (AMBER), Trinity College Dublin & Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Emily J. Ryan
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, Dublin 2, Ireland
- Advanced Materials Bio-Engineering Research Centre (AMBER), Trinity College Dublin & Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Cathal J. Kearney
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, Dublin 2, Ireland
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
- Advanced Materials Bio-Engineering Research Centre (AMBER), Trinity College Dublin & Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Daniel J. Kelly
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, Dublin 2, Ireland
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
- Advanced Materials Bio-Engineering Research Centre (AMBER), Trinity College Dublin & Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Fergal J. O’Brien
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, Dublin 2, Ireland
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
- Advanced Materials Bio-Engineering Research Centre (AMBER), Trinity College Dublin & Royal College of Surgeons in Ireland, Dublin 2, Ireland
| |
Collapse
|
237
|
Affiliation(s)
- Wendy F Liu
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
238
|
Li L, Bae KH, Ng S, Yamashita A, Kurisawa M. Peroxidase-immobilized porous silica particles for in situ formation of peroxidase-free hydrogels with attenuated immune responses. Acta Biomater 2018; 81:103-114. [PMID: 30273747 DOI: 10.1016/j.actbio.2018.09.054] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 08/10/2018] [Accepted: 09/27/2018] [Indexed: 10/28/2022]
Abstract
Enzymatic crosslinking chemistry using horseradish peroxidase (HRP) has been widely utilized as an effective approach to fabricating injectable hydrogels with high efficiency under mild reaction conditions. However, their clinical applications are limited by the immunogenicity of the plant-derived enzyme. Herein we report the design, synthesis and characterization of HRP-immobilized porous silica particles (HRP-particles) and their use for in situ formation of HRP-free hydrogels. HRP was immobilized on aminopropyl-modified porous silica particles of 70-140 µm in diameter via poly(ethylene glycol) spacers of different molecular weights by reductive amination reaction. Two different HRP-free hydrogels based on dextran-tyramine and gelatin-hydroxyphenylpropionic acid (GHPA) conjugates were produced by passing a solution containing the conjugates and H2O2 through a syringe packed with HRP-particles. The storage modulus and gelation rate of both hydrogels were tunable by varying the contact time between the polymer solution and HRP-particles. Our in vitro study revealed that HRP-free GHPA hydrogel was less stimulatory to activated mouse macrophages than HRP-containing GHPA hydrogel with the same stiffness. Furthermore, HRP-free GHPA hydrogel exhibited remarkably lower levels of local and systemic inflammation than HRP-containing one upon subcutaneous injection in immunocompetent C57BL/6J mice. The attenuated immunogenicity of HRP-free GHPA hydrogels makes them an attractive platform for tissue engineering applications. STATEMENT OF SIGNIFICANCE: The immunogenicity of HRP is a significant issue for clinical application of HRP-catalyzed in situ forming hydrogels. HRP-particles are developed to overcome the safety concerns by fabricating HRP-free hydrogels. The porosity of silica particles and molecular weight of poly(ethylene glycol) spacers are discovered as important factors determining the catalytic ability of HRP-particles to induce the in situ crosslinking of polymer-phenol conjugates. Although several articles speculate the potential of HRP to trigger immune responses when administered as a part of hydrogel formulation, no literature has attempted to investigate the immunogenicity of HRP-containing hydrogels in comparison with HRP-free hydrogels. Our findings suggest that the immunogenicity issue should be carefully considered before clinical translation of HRP-containing hydrogels.
Collapse
|
239
|
Ghuman H, Mauney C, Donnelly J, Massensini AR, Badylak SF, Modo M. Biodegradation of ECM hydrogel promotes endogenous brain tissue restoration in a rat model of stroke. Acta Biomater 2018; 80:66-84. [PMID: 30232030 PMCID: PMC6217851 DOI: 10.1016/j.actbio.2018.09.020] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 09/12/2018] [Accepted: 09/14/2018] [Indexed: 12/15/2022]
Abstract
The brain is considered to have a limited capacity to repair damaged tissue and no regenerative capacity following injury. Tissue lost after a stroke is therefore not spontaneously replaced. Extracellular matrix (ECM)-based hydrogels implanted into the stroke cavity can attract endogenous cells. These hydrogels can be formulated at different protein concentrations that govern their rheological and inductive properties. We evaluated histologically 0, 3, 4 and 8 mg/mL of porcine-derived urinary bladder matrix (UBM)-ECM hydrogel concentrations implanted in a 14-day old stroke cavity. Less concentrated hydrogels (3 and 4 mg/mL) were efficiently degraded with a 95% decrease in volume by 90 days, whereas only 32% of the more concentrated and stiffer hydrogel (8 mg/mL) was resorbed. Macrophage infiltration and density within the bioscaffold progressively increased in the less concentrated hydrogels and decreased in the 8 mg/mL hydrogels. The less concentrated hydrogels showed a robust invasion of endothelial cells with neovascularization. No neovascularization occurred with the stiffer hydrogel. Invasion of neural cells increased with time in all hydrogel concentrations. Differentiation of neural progenitors into mature neurons with axonal projections was evident, as well as a robust invasion of oligodendrocytes. However, relatively few astrocytes were present in the ECM hydrogel, although some were present in the newly forming tissue between degrading scaffold patches. Implantation of an ECM hydrogel partially induced neural tissue restoration, but a more complete understanding is required to evaluate its potential therapeutic application. STATEMENT OF SIGNIFICANCE: Extracellular matrix hydrogel promotes tissue regeneration in many peripheral soft tissues. However, the brain has generally been considered to lack the potential for tissue regeneration. We here demonstrate that tissue regeneration in the brain can be achieved using implantation of ECM hydrogel into a tissue cavity. A structure-function relationship is key to promote tissue regeneration in the brain. Specifically, weaker hydrogels that were retained in the cavity underwent an efficient biodegradation within 14 days post-implantation to promote a tissue restoration within the lesion cavity. In contrast, stiffer ECM hydrogel only underwent minor biodegradation and did not lead to a tissue restoration. Inductive hydrogels weaker than brain tissue provide the appropriate condition to promote an endogenous regenerative response that restores tissue in a cavity. This approach offers new avenues for the future treatment of chronic tissue damage caused by stroke and other acute brain injuries.
Collapse
Affiliation(s)
- Harmanvir Ghuman
- University of Pittsburgh, McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA; Department of Bioengineering, Pittsburgh, PA, USA
| | | | | | - Andre R Massensini
- University of Pittsburgh, McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA; Universidade Federal de Minas Gerais, Department of Physiology and Biophysics, Belo Horizonte, Brazil
| | - Stephen F Badylak
- University of Pittsburgh, McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA; Department of Bioengineering, Pittsburgh, PA, USA; Department of Surgery, Pittsburgh, PA, USA
| | - Michel Modo
- University of Pittsburgh, McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA; Department of Bioengineering, Pittsburgh, PA, USA; Department of Radiology, Pittsburgh, PA, USA.
| |
Collapse
|
240
|
Lee D, Lee K, Cha C. Microfluidics‐Assisted Fabrication of Microtissues with Tunable Physical Properties for Developing an In Vitro Multiplex Tissue Model. ACTA ACUST UNITED AC 2018. [DOI: 10.1002/adbi.201800236] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
- Dongjin Lee
- School of Materials Science and EngineeringUlsan National Institute of Science and Technology (UNIST) 50 UNIST‐gil Ulju‐gun Ulsan 44919 Korea
| | - Kangseok Lee
- Department of Biomedical EngineeringSchool of Life SciencesUlsan National Institute of Science and Technology (UNIST) Ulsan 44919 Korea
| | - Chaenyung Cha
- School of Materials Science and EngineeringUlsan National Institute of Science and Technology (UNIST) 50 UNIST‐gil Ulju‐gun Ulsan 44919 Korea
- Department of Biomedical EngineeringSchool of Life SciencesUlsan National Institute of Science and Technology (UNIST) Ulsan 44919 Korea
| |
Collapse
|
241
|
Rios PD, Skoumal M, Liu J, Youngblood R, Kniazeva E, Garcia AJ, Shea LD. Evaluation of encapsulating and microporous nondegradable hydrogel scaffold designs on islet engraftment in rodent models of diabetes. Biotechnol Bioeng 2018; 115:2356-2364. [PMID: 29873059 PMCID: PMC6131066 DOI: 10.1002/bit.26741] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 05/11/2018] [Accepted: 05/22/2018] [Indexed: 02/06/2023]
Abstract
Islet transplantation is a promising therapeutic option for type 1 diabetes mellitus, yet the current delivery into the hepatic portal vasculature is limited by poor engraftment. Biomaterials have been used as a means to promote engraftment and function at extrahepatic sites, with strategies being categorized as encapsulation or microporous scaffolds that can either isolate or integrate islets with the host tissue, respectively. Although these approaches are typically studied separately using distinct material platforms, herein, we developed nondegradable polyethylene glycol (PEG)-based hydrogels for islet encapsulation or as microporous scaffolds for islet seeding to compare the initial engraftment and function of islets in syngeneic diabetic mice. Normoglycemia was restored with transplantation of islets within either encapsulating or microporous hydrogels containing 700 islet equivalents (IEQ), with transplantation on microporous hydrogels producing lower blood glucose levels at earlier times. A glucose challenge test at 1 month after transplant indicated that encapsulated islets had a delay in glucose-stimulated insulin secretion, whereas microporous hydrogels restored normoglycemia in times consistent with native pancreata. Encapsulated islets remained isolated from the host tissue, whereas the microporous scaffolds allowed for revascularization of the islets after transplant. Finally, we compared the inflammatory response after transplantation for the two systems and noted that microporous hydrogels had a substantially increased presence of neutrophils. Collectively, these findings suggest that both encapsulation and microporous PEG scaffold designs allow for stable engraftment of syngeneic islets and the ability to restore normoglycemia, yet the architecture influences islet function and responsiveness after transplantation.
Collapse
Affiliation(s)
- Peter D Rios
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois
| | - Michael Skoumal
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Jeffrey Liu
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois
- Interdisciplinary Biological Sciences Program, Northwestern University, Evanston, Illinois
| | - Richard Youngblood
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Ekaterina Kniazeva
- Department of Obstetrics and Gynecology, Northwestern University, Chicago, Illinois
| | - Andrés J Garcia
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, Georgia
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
242
|
Wu S, Yue H, Wu J, Zhang W, Jiang M, Ma G. The interacting role of physical stiffness and tumor cells on the macrophages polarization. Colloids Surf A Physicochem Eng Asp 2018. [DOI: 10.1016/j.colsurfa.2018.04.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
243
|
Pakshir P, Hinz B. The big five in fibrosis: Macrophages, myofibroblasts, matrix, mechanics, and miscommunication. Matrix Biol 2018; 68-69:81-93. [DOI: 10.1016/j.matbio.2018.01.019] [Citation(s) in RCA: 162] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 01/25/2018] [Accepted: 01/28/2018] [Indexed: 02/07/2023]
|
244
|
Shayan M, Padmanabhan J, Morris AH, Cheung B, Smith R, Schroers J, Kyriakides TR. Nanopatterned bulk metallic glass-based biomaterials modulate macrophage polarization. Acta Biomater 2018; 75:427-438. [PMID: 29859902 PMCID: PMC6119487 DOI: 10.1016/j.actbio.2018.05.051] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 05/16/2018] [Accepted: 05/30/2018] [Indexed: 12/16/2022]
Abstract
Polarization of macrophages by chemical, topographical and mechanical cues presents a robust strategy for designing immunomodulatory biomaterials. Here, we studied the ability of nanopatterned bulk metallic glasses (BMGs), a new class of metallic biomaterials, to modulate murine macrophage polarization. Cytokine/chemokine analysis of IL-4 or IFNγ/LPS-stimulated macrophages showed that the secretion of TNF-α, IL-1α, IL-12, CCL-2 and CXCL1 was significantly reduced after 24-hour culture on BMGs with 55 nm nanorod arrays (BMG-55). Additionally, under these conditions, macrophages increased phagocytic potential and exhibited decreased cell area with multiple actin protrusions. These in vitro findings suggest that nanopatterning can modulate biochemical cues such as IFNγ/LPS. In vivo evaluation of the subcutaneous host response at 2 weeks demonstrated that the ratio of Arg-1 to iNOS increased in macrophages adjacent to BMG-55 implants, suggesting modulation of polarization. In addition, macrophage fusion and fibrous capsule thickness decreased and the number and size of blood vessels increased, which is consistent with changes in macrophage responses. Our study demonstrates that nanopatterning of BMG implants is a promising technique to selectively polarize macrophages to modulate the immune response, and also presents an effective tool to study mechanisms of macrophage polarization and function. STATEMENT OF SIGNIFICANCE Implanted biomaterials elicit a complex series of tissue and cellular responses, termed the foreign body response (FBR), that can be influenced by the polarization state of macrophages. Surface topography can influence polarization, which is broadly characterized as either inflammatory or repair-like. The latter has been linked to improved outcomes of the FBR. However, the impact of topography on macrophage polarization is not fully understood, in part, due to a lack of high moduli biomaterials that can be reproducibly processed at the nanoscale. Here, we studied macrophage interactions with nanopatterned bulk metallic glasses (BMGs), a class of metallic alloys with amorphous microstructure and formability like polymers. We show that nanopatterned BMGs modulate macrophage polarization and transiently induce less fibrotic and more angiogenic responses. Overall, we demonstrate nanopatterning of BMG implants as a technique to polarize macrophages and modulate the FBR.
Collapse
Affiliation(s)
- Mahdis Shayan
- Department of Pathology, Yale University, New Haven, CT 06520, USA
| | | | - Aaron H Morris
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| | - Bettina Cheung
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| | - Ryan Smith
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| | - Jan Schroers
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA; Department of Mechanical Engineering and Materials Science, Yale University, New Haven, CT 06520, USA
| | - Themis R Kyriakides
- Department of Pathology, Yale University, New Haven, CT 06520, USA; Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA.
| |
Collapse
|
245
|
Jansen LE, Amer LD, Chen EYT, Nguyen TV, Saleh LS, Emrick T, Liu WF, Bryant SJ, Peyton SR. Zwitterionic PEG-PC Hydrogels Modulate the Foreign Body Response in a Modulus-Dependent Manner. Biomacromolecules 2018; 19:2880-2888. [PMID: 29698603 PMCID: PMC6190668 DOI: 10.1021/acs.biomac.8b00444] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Reducing the foreign body response (FBR) to implanted biomaterials will enhance their performance in tissue engineering. Poly(ethylene glycol) (PEG) hydrogels are increasingly popular for this application due to their low cost, ease of use, and the ability to tune their compliance via molecular weight and cross-linking densities. PEG hydrogels can elicit chronic inflammation in vivo, but recent evidence has suggested that extremely hydrophilic, zwitterionic materials and particles can evade the immune system. To combine the advantages of PEG-based hydrogels with the hydrophilicity of zwitterions, we synthesized hydrogels with comonomers PEG and the zwitterion phosphorylcholine (PC). Recent evidence suggests that stiff hydrogels elicit increased immune cell adhesion to hydrogels, which we attempted to reduce by increasing hydrogel hydrophilicity. Surprisingly, hydrogels with the highest amount of zwitterionic comonomer elicited the highest FBR. Lowering the hydrogel modulus (165 to 3 kPa), or PC content (20 to 0 wt %), mitigated this effect. A high density of macrophages was found at the surface of implants associated with a high FBR, and mass spectrometry analysis of the proteins adsorbed to these gels implicated extracellular matrix, immune response, and cell adhesion protein categories as drivers of macrophage recruitment. Overall, we show that modulus regulates macrophage adhesion to zwitterionic-PEG hydrogels, and demonstrate that chemical modifications to hydrogels should be studied in parallel with their physical properties to optimize implant design.
Collapse
Affiliation(s)
| | - Luke D Amer
- Department of Chemical and Biological Engineering , University of Colorado Boulder , Boulder , Colorado 80309 , United States
| | - Esther Y-T Chen
- Department of Biomedical Engineering , University of California, Irvine , Irvine , California 92697 , United States
| | | | - Leila S Saleh
- Department of Chemical and Biological Engineering , University of Colorado Boulder , Boulder , Colorado 80309 , United States
| | | | - Wendy F Liu
- Department of Biomedical Engineering , University of California, Irvine , Irvine , California 92697 , United States
| | - Stephanie J Bryant
- Department of Chemical and Biological Engineering , University of Colorado Boulder , Boulder , Colorado 80309 , United States
| | | |
Collapse
|
246
|
Regulation of Macrophages by Extracellular Matrix Composition and Adhesion Geometry. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2018. [DOI: 10.1007/s40883-018-0065-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
247
|
Chen Y, Wang J, Zhu X, Chen X, Yang X, Zhang K, Fan Y, Zhang X. The directional migration and differentiation of mesenchymal stem cells toward vascular endothelial cells stimulated by biphasic calcium phosphate ceramic. Regen Biomater 2018; 5:129-139. [PMID: 29977596 PMCID: PMC6007427 DOI: 10.1093/rb/rbx028] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 09/15/2017] [Accepted: 09/20/2017] [Indexed: 12/12/2022] Open
Abstract
Osteoinductivity of porous calcium phosphate (CaP) ceramics has been widely investigated and confirmed, and it might be attributed to the rapid formation of the vascular networks after in vivo implantation of the ceramics. In this study, to explore the vascularization mechanism within the CaP ceramics, the migration and differentiation of bone marrow-derived mesenchymal stem cells (BMSCs) under the stimulation of porous biphasic calcium phosphate (BCP) ceramic with excellent osteoinductivity were systematically investigated. The results indicated that the directional migration of BMSCs toward BCP ceramic occurred when evaluated by using a transwell model, and the BMSCs migration was enhanced by the seeded macrophages on the ceramic in advance. Besides, by directly culturing BMSCs on BCP ceramic discs under both in vitro and in vivo physiological environment, it was found that the differentiation of BMSCs toward vascular endothelial cells (VECs) happened under the stimulation of BCP ceramic, as was confirmed by the up-regulated gene expressions and protein secretions of VECs-related characteristic factors, including kinase insert domain receptor, von willebrand factor, vascular cell adhesion molecule-1 and cadherin 5 in the BMSCs. This study offered a possibility for explaining the origin of VECs during the rapid vascularization process after in vivo implantation of porous CaP ceramics and could give some useful guidance to reveal the vascularization mechanism of the ceramics.
Collapse
Affiliation(s)
- Ying Chen
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Jing Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Xiangdong Zhu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Xuening Chen
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Xiao Yang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Kai Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Yujiang Fan
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| |
Collapse
|
248
|
Zhu Y, Hideyoshi S, Jiang H, Matsumura Y, Dziki JL, LoPresti ST, Huleihel L, Faria GNF, Fuhrman LC, Lodono R, Badylak SF, Wagner WR. Injectable, porous, biohybrid hydrogels incorporating decellularized tissue components for soft tissue applications. Acta Biomater 2018; 73:112-126. [PMID: 29649634 PMCID: PMC5985206 DOI: 10.1016/j.actbio.2018.04.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 03/07/2018] [Accepted: 04/02/2018] [Indexed: 12/16/2022]
Abstract
Biodegradable injectable hydrogels have been extensively studied and evaluated in various medical applications such as for bulking agents, drug delivery reservoirs, temporary barriers, adhesives, and cell delivery matrices. Where injectable hydrogels are intended to facilitate a healing response, it may be desirable to encourage rapid cellular infiltration into the hydrogel volume from the tissue surrounding the injection site. In this study, we developed a platform technique to rapidly form pores in a thermally responsive injectable hydrogel, poly(NIPAAm-co-VP-co-MAPLA) by using mannitol particles as porogens. In a rat hindlimb muscle injection model, hydrogels incorporating porosity had significantly accelerated cellular infiltration. To influence the inflammatory response to the injected hydrogel, enzymatically digested urinary bladder matrix (UBM) was mixed with the solubilized hydrogel. The presence of UBM was associated with greater polarization of the recruited macrophage population to the M2 phenotype, indicating a more constructive foreign body response. The hybrid hydrogel positively affected the wound healing outcomes of defects in rabbit adipose tissue with negligible inflammation and fibrosis, whereas scar formation and chronic inflammation were observed with autotransplantation and in saline injected groups. These results demonstrate the value of combining the effects of promoting cell infiltration and mediating the foreign body response for improved biomaterials options soft tissue defect filling applications. STATEMENT OF SIGNIFICANCE Our objective was to develop a fabrication process to create porous injectable hydrogels incorporating decellularized tissue digest material. This new hydrogel material was expected to exhibit faster cellular infiltration and a greater extent of pro-M2 macrophage polarization compared to control groups not incorporating each of the functional components. Poly(NIPAAm-co-VP-co-MAPLA) was chosen as the representative thermoresponsive hydrogel, and mannitol particles and digested urinary bladder matrix (UBM) were selected as the porogen and the bioactive decellularized material components respectively. In rat hindlimb intramuscular injection models, this new hydrogel material induced more rapid cellular infiltration and a greater extent of M2 macrophage polarization compared to control groups not incorporating all of the functional components. The hybrid hydrogel positively affected the wound healing outcomes of defects in rabbit adipose tissue with negligible inflammation and fibrosis, whereas scar formation and chronic inflammation were observed with autotransplantation and in saline injected groups. The methodology of this report provides a straightforward and convenient mechanism to promote cell infiltration and mediate foreign body response in injectable hydrogels for soft tissue applications. We believe that the readership of Acta Biomaterialia will find the work of interest both for its specific results and general translatability of the findings.
Collapse
Affiliation(s)
- Yang Zhu
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Sato Hideyoshi
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Hongbin Jiang
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Yasumoto Matsumura
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Jenna L Dziki
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Samuel T LoPresti
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Luai Huleihel
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA; Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Gabriela N F Faria
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Leah C Fuhrman
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Ricardo Lodono
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Stephen F Badylak
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15219, USA; Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - William R Wagner
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15219, USA; Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA; Department of Chemical Engineering, University of Pittsburgh, Pittsburgh, PA 15219, USA.
| |
Collapse
|
249
|
Xue YZB, Niu YM, Tang B, Wang CM. WITHDRAWN: PCL/EUG scaffolds with tunable stiffness can regulate macrophage secretion behavior. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2018:S0079-6107(17)30320-6. [PMID: 29792891 DOI: 10.1016/j.pbiomolbio.2018.05.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 04/14/2018] [Accepted: 05/19/2018] [Indexed: 01/04/2023]
Abstract
The Publisher regrets that this article is an accidental duplication of an article that has already been published, http://dx.doi.org/10.1016/j.pbiomolbio.2018.05.008. The duplicate article has therefore been withdrawn.
Collapse
Affiliation(s)
- Y Z B Xue
- Department of Materials Science and Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, China; State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China; Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Y M Niu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| | - B Tang
- Department of Materials Science and Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, China; Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, China; Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, China.
| | - C M Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China.
| |
Collapse
|
250
|
Saleh LS, Bryant SJ. In Vitro and In Vivo Models for Assessing the Host Response to Biomaterials. ACTA ACUST UNITED AC 2018; 24:13-21. [PMID: 30479632 DOI: 10.1016/j.ddmod.2018.04.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The foreign body response (FBR) occurs ubiquitously to essentially all non-biological materials that are implanted into higher organisms. The FBR is characterized by inflammation followed by fibrosis and is mediated largely by macrophages. While many current medical devices tolerate the FBR, the FBR is responsible for many asceptic device failures and is hindering advancements of new devices that rely on device-host communication to function. To this end, in vitro and in vivo models are critical to studying how a biomaterial, via its chemistry and properties, affect the FBR. This short review highlights the main in vitro and in vivo models that are used to study the FBR. In vitro models that capture macrophage interrogation of a biomaterial and evaluation of macrophage attachment, polarization and fusion are described. In vivo models using rodents, which provide a relatively simple model of the complex FBR process, and human-relevant nonhuman primate models are described. Collectively, the combination of in vitro and in vivo models will help advance our fundmental understanding of the FBR and enable new biomaterials to be developed that can effectively modulate the FBR to achieve a desire device-host outcome.
Collapse
Affiliation(s)
- Leila S Saleh
- Department of Chemical and Biological Engineering, University of Colorado, 3415 Colorado Avenue, Boulder, CO 80303, USA
| | - Stephanie J Bryant
- Department of Chemical and Biological Engineering, University of Colorado, 3415 Colorado Avenue, Boulder, CO 80303, USA.,BioFrontiers Institute, University of Colorado, 3415 Colorado Avenue, Boulder, CO 80303, USA.,Material Science and Engineering Program, University of Colorado, 3415 Colorado Avenue, Boulder, CO 80303, USA
| |
Collapse
|