1
|
Mahanty M, Dutta B, Ou W, Zhu X, Bromberg JS, He X, Rahaman SO. Macrophage microRNA-146a is a central regulator of the foreign body response to biomaterial implants. Biomaterials 2025; 314:122855. [PMID: 39362025 DOI: 10.1016/j.biomaterials.2024.122855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 09/20/2024] [Accepted: 09/26/2024] [Indexed: 10/05/2024]
Abstract
Host recognition and immune-mediated foreign body response (FBR) to biomaterials can adversely affect the functionality of implanted materials. FBR presents a complex bioengineering and medical challenge due to the lack of current treatments, making the detailed exploration of its molecular mechanisms crucial for developing new and effective therapies. To identify key molecular targets underlying the generation of FBR, here we perform analysis of microRNAs (miR) and mRNAs responses to implanted biomaterials. We found that (a) miR-146a levels inversely affect macrophage accumulation, foreign body giant cell (FBGC) formation, and fibrosis in a murine implant model; (b) macrophage-derived miR-146a is a crucial regulator of the FBR and FBGC formation, as confirmed by global and cell-specific knockout of miR-146a; (c) miR-146a modulates genes related to inflammation, fibrosis, and mechanosensing; (d) miR-146a modulates tissue stiffness near the implant during FBR as assessed by atomic force microscopy; and (e) miR-146a is linked to F-actin production and cellular traction force induction as determined by traction force microscopy, which are vital for FBGC formation. These novel findings suggest that targeting macrophage miR-146a could be a selective strategy to inhibit FBR, potentially improving the biocompatibility of biomaterials.
Collapse
Affiliation(s)
- Manisha Mahanty
- University of Maryland, Department of Nutrition and Food Science, College Park, MD, 20742, USA
| | - Bidisha Dutta
- University of Maryland, Department of Nutrition and Food Science, College Park, MD, 20742, USA
| | - Wenquan Ou
- University of Maryland, Fischell Department of Bioengineering, College Park, MD, 20742, USA
| | - Xiaoping Zhu
- University of Maryland, Department of Veterinary Medicine, College Park, MD, 20742, USA
| | | | - Xiaoming He
- University of Maryland, Fischell Department of Bioengineering, College Park, MD, 20742, USA
| | - Shaik O Rahaman
- University of Maryland, Department of Nutrition and Food Science, College Park, MD, 20742, USA.
| |
Collapse
|
2
|
Ahmine AN, Bdiri M, Féréol S, Fodil R. A comprehensive study of AFM stiffness measurements on inclined surfaces: theoretical, numerical, and experimental evaluation using a Hertz approach. Sci Rep 2024; 14:25869. [PMID: 39468207 PMCID: PMC11519481 DOI: 10.1038/s41598-024-75958-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 10/09/2024] [Indexed: 10/30/2024] Open
Abstract
Atomic Force Microscopy (AFM) is a leading nanoscale technique known for its significant advantages in the analysis of soft materials and biological samples. Traditional AFM data analysis is often based on the Hertz model, which assumes perpendicular indentation of a planar sample. However, this assumption is not always valid due to the varying geometries of soft materials, whether natural, synthetic or biological. In this study, we present a new theoretical model that incorporates correction coefficients into Hertz's model to account for cone-like and spherical probes, and to consider local tilt at the probe-sample interface. We validate our model using finite element analysis (FEA) simulations and experimental AFM measurements on tilted polyacrylamide gels. Our results highlight the need to include local tilt at the probe-sample contact to ensure accurate AFM measurements. This represents a step forward in our understanding of the elastic properties at the surface of soft materials in the broadest sense.
Collapse
Affiliation(s)
- Anis Nassim Ahmine
- INSERM, U955 IMRB, "Biology of the Neuromuscular System" Team, Univ Paris-Est Créteil, 94010, Créteil, France
| | - Myriam Bdiri
- CNRS, UMR 7182, Institut de Chimie et des Matériaux Paris-Est (ICMPE), Univ Paris-Est Créteil, 94320, Thiais, France
| | - Sophie Féréol
- INSERM, U955 IMRB, "Biology of the Neuromuscular System" Team, Univ Paris-Est Créteil, 94010, Créteil, France
| | - Redouane Fodil
- INSERM, U955 IMRB, "Biology of the Neuromuscular System" Team, Univ Paris-Est Créteil, 94010, Créteil, France.
| |
Collapse
|
3
|
Ezzo M, Spindler K, Wang JB, Lee D, Pecoraro G, Cowen J, Pakshir P, Hinz B. Acute contact with profibrotic macrophages mechanically activates fibroblasts via αvβ3 integrin-mediated engagement of Piezo1. SCIENCE ADVANCES 2024; 10:eadp4726. [PMID: 39441936 PMCID: PMC11498225 DOI: 10.1126/sciadv.adp4726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 09/18/2024] [Indexed: 10/25/2024]
Abstract
Fibrosis-excessive scarring after injury-causes >40% of disease-related deaths worldwide. In this misguided repair process, activated fibroblasts drive the destruction of organ architecture by accumulating and contracting extracellular matrix. The resulting stiff scar tissue, in turn, enhances fibroblast contraction-bearing the question of how this positive feedback loop begins. We show that direct contact with profibrotic but not proinflammatory macrophages triggers acute fibroblast contractions. The contractile response depends on αvβ3 integrin expression on macrophages and Piezo1 expression on fibroblasts. The touch of macrophages elevates fibroblast cytosolic calcium within seconds, followed by translocation of the transcription cofactors nuclear factor of activated T cells 1 and Yes-associated protein, which drive fibroblast activation within hours. Intriguingly, macrophages induce mechanical stress in fibroblasts on soft matrix that alone suppresses their spontaneous activation. We propose that acute contact with suitable macrophages mechanically kick-starts fibroblast activation in an otherwise nonpermissive soft environment. The molecular components mediating macrophage-fibroblast mechanotransduction are potential targets for antifibrosis strategies.
Collapse
Affiliation(s)
- Maya Ezzo
- Laboratory of Tissue Repair and Regeneration, Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
- Keenan Research Institute for Biomedical Science of the St. Michael’s Hospital, Toronto, Ontario, Canada
| | - Katrin Spindler
- Keenan Research Institute for Biomedical Science of the St. Michael’s Hospital, Toronto, Ontario, Canada
- School of Life Sciences, Reutlingen University, 72762 Reutlingen, Germany
| | - Jun Bo Wang
- Laboratory of Tissue Repair and Regeneration, Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Dahea Lee
- Keenan Research Institute for Biomedical Science of the St. Michael’s Hospital, Toronto, Ontario, Canada
| | - Gilbert Pecoraro
- Laboratory of Tissue Repair and Regeneration, Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
- School of Life Sciences, Reutlingen University, 72762 Reutlingen, Germany
| | - Justin Cowen
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Pardis Pakshir
- Laboratory of Tissue Repair and Regeneration, Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Boris Hinz
- Laboratory of Tissue Repair and Regeneration, Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
- Keenan Research Institute for Biomedical Science of the St. Michael’s Hospital, Toronto, Ontario, Canada
| |
Collapse
|
4
|
Alemifar A, Burnette K, Jandres B, Hurt S, Tse HM, Robinson JL. Electrospun Fiber Surface Roughness Modulates Human Monocyte-Derived Macrophage Phenotype. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.30.610568. [PMID: 39282362 PMCID: PMC11398424 DOI: 10.1101/2024.08.30.610568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Abstract
Injuries to fibrous connective tissues have very little capacity for self-renewal and exhibit poor healing after injury. Phenotypic shifts in macrophages play a vital role in mediating the healing response, creating an opportunity to design immunomodulatory biomaterials which control macrophage polarization and promote regeneration. In this study, electrospun poly(-caprolactone) fibers with increasing surface roughness (SR) were produced by increasing relative humidity and inducing vapor-induced phase separation during the electrospinning process. The impact of surface roughness on macrophage phenotype was assessed using human monocyte-derived macrophages in vitro and in vivo using B6.Cg-Tg(Csf1r-EGFP)1Hume/J (MacGreen) mice. In vitro experiments showed that macrophages cultured on mesh with increasing SR exhibited decreased release of both pro- and anti-inflammatory cytokines potentially driven by increased protein adsorption and biophysical impacts on the cells. Further, increasing SR led to an increase in the expression of the pro-regenerative cell surface marker CD206 relative to the pro-inflammatory marker CD80. Mesh with increasing SR were implanted subcutaneously in MacGreen mice, again showing an increase in the ratio of cells expressing CD206 to those expressing CD80 visualized by immunofluorescence. SR on implanted biomaterials is sufficient to drive macrophage polarization, demonstrating a simple feature to include in biomaterial design to control innate immunity.
Collapse
Affiliation(s)
- Aidan Alemifar
- Department of Orthopaedic Surgery and Sports Medicine, University of Washington
- Bioengineering Graduate Program, University of Kansas
| | - KaLia Burnette
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center
| | - Bryan Jandres
- Department of Biochemistry, University of Washington
| | - Samuel Hurt
- Department of Chemical and Petroleum Engineering, University of Kansas
| | - Hubert M Tse
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center
| | - Jennifer L Robinson
- Department of Orthopaedic Surgery and Sports Medicine, University of Washington
- Department of Mechanical Engineering, University of Washington
| |
Collapse
|
5
|
Huo Z, Yang W, Harati J, Nene A, Borghi F, Piazzoni C, Milani P, Guo S, Galluzzi M, Boraschi D. Biomechanics of Macrophages on Disordered Surface Nanotopography. ACS APPLIED MATERIALS & INTERFACES 2024; 16:27164-27176. [PMID: 38750662 DOI: 10.1021/acsami.4c04330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2024]
Abstract
Macrophages are involved in every stage of the innate/inflammatory immune responses in the body tissues, including the resolution of the reaction, and they do so in close collaboration with the extracellular matrix (ECM). Simplified substrates with nanotopographical features attempt to mimic the structural properties of the ECM to clarify the functional features of the interaction of the ECM with macrophages. We still have a limited understanding of the macrophage behavior upon interaction with disordered nanotopography, especially with features smaller than 10 nm. Here, we combine atomic force microscopy (AFM), finite element modeling (FEM), and quantitative biochemical approaches in order to understand the mechanotransduction from the nanostructured surface into cellular responses. AFM experiments show a decrease of macrophage stiffness, measured with the Young's modulus, as a biomechanical response to a nanostructured (ns-) ZrOx surface. FEM experiments suggest that ZrOx surfaces with increasing roughness represent weaker mechanical boundary conditions. The mechanical cues from the substrate are transduced into the cell through the formation of integrin-regulated focal adhesions and cytoskeletal reorganization, which, in turn, modulate cell biomechanics by downregulating cell stiffness. Surface nanotopography and consequent biomechanical response impact the overall behavior of macrophages by increasing movement and phagocytic ability without significantly influencing their inflammatory behavior. Our study suggests a strong potential of surface nanotopography for the regulation of macrophage functions, which implies a prospective application relative to coating technology for biomedical devices.
Collapse
Affiliation(s)
- Zixin Huo
- Shenzhen Key Laboratory of Smart Sensing and Intelligent Systems, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wenjie Yang
- Laboratory of Inflammation and Vaccines, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Javad Harati
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Ajinkya Nene
- Laboratory of Inflammation and Vaccines, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Francesca Borghi
- CIMaINa and Dipartimento di Fisica "Aldo Pontremoli", Università degli Studi di Milano, Via Celoria 16, 20133 Milan, Italy
| | - Claudio Piazzoni
- CIMaINa and Dipartimento di Fisica "Aldo Pontremoli", Università degli Studi di Milano, Via Celoria 16, 20133 Milan, Italy
| | - Paolo Milani
- CIMaINa and Dipartimento di Fisica "Aldo Pontremoli", Università degli Studi di Milano, Via Celoria 16, 20133 Milan, Italy
| | - Shifeng Guo
- Shenzhen Key Laboratory of Smart Sensing and Intelligent Systems, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Guangdong Provincial Key Lab of Robotics and Intelligent System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- The Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Massimiliano Galluzzi
- Laboratory of Inflammation and Vaccines, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Diana Boraschi
- Laboratory of Inflammation and Vaccines, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Department of Pharmacology, Shenzhen University of Advanced Technology, Shenzhen 518055, China
- China-Italy Joint Laboratory of Pharmacobiotechnology for Medical Immunomodulation, Shenzhen 518055, China
- Institute of Biomolecular Chemistry, National Research Council, 80078 Pozzuoli, Italy
- Stazione Zoologica Anton Dohrn, 80122 Napoli, Italy
| |
Collapse
|
6
|
Mahanty M, Dutta B, Ou W, Zhu X, Bromberg JS, He X, Rahaman SO. Macrophage microRNA-146a is a central regulator of the foreign body response to biomaterial implants. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.03.588018. [PMID: 38617341 PMCID: PMC11014630 DOI: 10.1101/2024.04.03.588018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Host recognition and immune-mediated foreign body response (FBR) to biomaterials can adversely affect the functionality of implanted materials. To identify key targets underlying the generation of FBR, here we perform analysis of microRNAs (miR) and mRNAs responses to implanted biomaterials. We found that (a) miR-146a levels inversely affect macrophage accumulation, foreign body giant cell (FBGC) formation, and fibrosis in a murine implant model; (b) macrophage-derived miR-146a is a crucial regulator of the FBR and FBGC formation, as confirmed by global and cell-specific knockout of miR-146a; (c) miR-146a modulates genes related to inflammation, fibrosis, and mechanosensing; (d) miR-146a modulates tissue stiffness near the implant during FBR; and (e) miR-146a is linked to F-actin production and cellular traction force induction, which are vital for FBGC formation. These novel findings suggest that targeting macrophage miR-146a could be a selective strategy to inhibit FBR, potentially improving the biocompatibility of biomaterials.
Collapse
|
7
|
Xu Y, Ying L, Lang JK, Hinz B, Zhao R. Modeling mechanical activation of macrophages during pulmonary fibrogenesis for targeted anti-fibrosis therapy. SCIENCE ADVANCES 2024; 10:eadj9559. [PMID: 38552026 PMCID: PMC10980276 DOI: 10.1126/sciadv.adj9559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 02/23/2024] [Indexed: 04/01/2024]
Abstract
Pulmonary fibrosis is an often fatal lung disease. Immune cells such as macrophages were shown to accumulate in the fibrotic lung, but their contribution to the fibrosis development is unclear. To recapitulate the involvement of macrophages in the development of pulmonary fibrosis, we developed a fibrotic microtissue model with cocultured human macrophages and fibroblasts. We show that profibrotic macrophages seeded on topographically controlled stromal tissues became mechanically activated. The resulting co-alignment of macrophages, collagen fibers, and fibroblasts promoted widespread fibrogenesis in micro-engineered lung tissues. Anti-fibrosis treatment using pirfenidone disrupts the polarization and mechanical activation of profibrotic macrophages, leading to fibrosis inhibition. Pirfenidone inhibits the mechanical activation of macrophages by suppressing integrin αMβ2 and Rho-associated kinase 2. These results demonstrate a potential pulmonary fibrogenesis mechanism at the tissue level contributed by macrophages. The cocultured microtissue model is a powerful tool to study the immune-stromal cell interactions and the anti-fibrosis drug mechanism.
Collapse
Affiliation(s)
- Ying Xu
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY 14260, USA
| | - Linxuan Ying
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY 14260, USA
| | - Jennifer K. Lang
- Division of Cardiovascular Medicine and the Clinical and Translational Research Center, University at Buffalo, State University of New York; Veterans Affairs Western New York Health Care System, University at Buffalo, State University of New York; Department of Biomedical Engineering, University at Buffalo, State University of New York; Department of Medicine, University at Buffalo, State University of New York; Department of Pharmacology and Toxicology, University at Buffalo, State University of New York, Buffalo, NY, 14260, USA
| | - Boris Hinz
- Laboratory of Tissue Repair and Regeneration, Keenan Research Centre for Biomedical Science of the St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | - Ruogang Zhao
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY 14260, USA
| |
Collapse
|
8
|
Miller C, Sask KN. Fetuin-A adsorption to tunable polydimethylsiloxane and subsequent macrophage response. J Biomed Mater Res A 2023; 111:1096-1109. [PMID: 36592125 DOI: 10.1002/jbm.a.37491] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 12/01/2022] [Accepted: 12/15/2022] [Indexed: 01/03/2023]
Abstract
Surface modifications can be applied to biomaterials to alter the various surface properties that influence protein-material interactions and the cellular response. The plasma protein fetuin-A has been found to adsorb to many biomaterials but details of its interactions with polydimethylsiloxane (PDMS) and roles in regulating the immune response are not clear. Here, PDMS modifications are achieved by altering the ratio of PDMS formulations to control elastic modulus, and by coating PDMS with polydopamine (PDA) to attach fetuin-A. Surface characterization confirmed that altering the PDMS formulation changed the elastic modulus without affecting surface wetting properties. Surface roughness was measured using atomic force microscopy and surface chemistry was determined using X-ray photoelectron spectroscopy, with only minor changes detected on the softest samples. PDA deposition on PDMS was confirmed and contact angle measurements demonstrated an increase in hydrophilicity. Fetuin-A adsorption was influenced by the PDMS formulations, adsorption changed in a competitive plasma environment, and PDA was able to immobilize the greatest amount of fetuin-A. The inflammatory effects of fetuin-A were investigated, and data suggests that the elastic modulus influences cytokine secretion from macrophages at certain timepoints, a result likely due to varied protein amounts and orientations/conformations in response to material stiffness. The addition of a PDA layer demonstrated the potentially cytokine mitigating effect upon fetuin-A immobilization when compared to unmodified PDMS samples. The results provide new insight into the interactions of fetuin-A with PDMS and PDA, and the potential immune regulatory properties of fetuin-A modified materials.
Collapse
Affiliation(s)
- Chelsea Miller
- School of Biomedical Engineering, McMaster University, Hamilton, Ontario, Canada
| | - Kyla N Sask
- School of Biomedical Engineering, McMaster University, Hamilton, Ontario, Canada
- Department of Materials Science and Engineering, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
9
|
Xu Y, Ying L, Lang JK, Hinz B, Zhao R. Modeling Mechanical Activation of Macrophages During Pulmonary Fibrogenesis for Targeted Anti-Fibrosis Therapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.19.549794. [PMID: 37503121 PMCID: PMC10370161 DOI: 10.1101/2023.07.19.549794] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Pulmonary fibrosis, as seen in idiopathic pulmonary fibrosis (IPF) and COVID-induced pulmonary fibrosis, is an often-fatal lung disease. Increased numbers of immune cells such as macrophages were shown to accumulate in the fibrotic lung, but it is unclear how they contribute to the development of fibrosis. To recapitulate the macrophage mechanical activation in the fibrotic lung tissue microenvironment, we developed a fibrotic microtissue model with cocultured human macrophages and fibroblasts. We show that profibrotic macrophages seeded on topographically controlled stromal tissue constructs become mechanically activated. The resulting co-alignment of macrophages, collagen fibers and fibroblasts promote widespread fibrogenesis in micro-engineered lung tissues. Anti-fibrosis treatment using pirfenidone disrupts the polarization and mechanical activation of profibrotic macrophages, leading to fibrosis inhibition. Pirfenidone inhibits the mechanical activation of macrophages by suppressing integrin αMβ2 (CD11b/CD18) and Rho-associated kinase 2, which is a previously unknown mechanism of action of the drug. Together, these results demonstrate a potential pulmonary fibrogenesis mechanism at the tissue level contributed by mechanically activated macrophages. We propose the coculture, force-sensing microtissue model as a powerful tool to study the complex immune-stromal cell interactions and the mechanism of action of anti-fibrosis drugs.
Collapse
|
10
|
Li W, Xu F, Dai F, Deng T, Ai Y, Xu Z, He C, Ai F, Song L. Hydrophilic surface-modified 3D printed flexible scaffolds with high ceramic particle concentrations for immunopolarization-regulation and bone regeneration. Biomater Sci 2023; 11:3976-3997. [PMID: 37115001 DOI: 10.1039/d3bm00362k] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
Bioceramic scaffolds used in bone tissue engineering suffer from a low concentration of ceramic particles (<50 wt%), because the high concentration of ceramic particles increases the brittleness of the composite. 3D printed flexible PCL/HA scaffolds with high ceramic particle concentrations (84 wt%) were successfully fabricated in this study. However, the hydrophobicity of PCL weakens the composite scaffold hydrophilicity, which may limit the osteogenic ability to some extent. Thus, as a less time-consuming, less labour intensive, and more cost-effective treatment method, alkali treatment (AT) was employed to modify the surface hydrophilicity of the PCL/HA scaffold, and its regulation of immune responses and bone regeneration were investigated in vivo and in vitro. Initially, several concentrations of NaOH (0.5, 1, 1.5, 2, 2.5, and 5 mol L-1) were employed in tests to determine the appropriate concentration for AT. Based on the comprehensive consideration of the results of mechanical experiments and hydrophilicity, 2 mol L-1 and 2.5 mol L-1 of NaOH were selected for further investigation in this study. The PCL/HA-AT-2 scaffold dramatically reduced foreign body reactions as compared to the PCL/HA and PCL/HA-AT-2.5 scaffolds, promoted macrophage polarization towards the M2 phenotype and enhanced new bone formation. The Wnt/β-catenin pathway might participate in the signal transduction underlying hydrophilic surface-modified 3D printed scaffold-regulated osteogenesis, according to the results of immunohistochemical staining. In conclusion, hydrophilic surface-modified 3D printed flexible scaffolds with high ceramic particle concentrations can regulate the immune reactions and macrophage polarization to promote bone regeneration, and the PCL/HA-AT-2 scaffold is a potential candidate for bone tissue repair.
Collapse
Affiliation(s)
- Wenfeng Li
- The Center of Stomatology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
- The Institute of Periodontal Disease, Nanchang University, Nanchang, China.
- JXHC Key Laboratory of Periodontology (The Second Affiliated Hospital of Nanchang University), Nanchang, China
| | - Fancheng Xu
- The Center of Stomatology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
- The Institute of Periodontal Disease, Nanchang University, Nanchang, China.
- JXHC Key Laboratory of Periodontology (The Second Affiliated Hospital of Nanchang University), Nanchang, China
| | - Fang Dai
- The Center of Stomatology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
- The Institute of Periodontal Disease, Nanchang University, Nanchang, China.
- JXHC Key Laboratory of Periodontology (The Second Affiliated Hospital of Nanchang University), Nanchang, China
| | - Tian Deng
- The Center of Stomatology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
- The Institute of Periodontal Disease, Nanchang University, Nanchang, China.
- JXHC Key Laboratory of Periodontology (The Second Affiliated Hospital of Nanchang University), Nanchang, China
| | - Yufeng Ai
- The Center of Stomatology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
- The Institute of Periodontal Disease, Nanchang University, Nanchang, China.
- JXHC Key Laboratory of Periodontology (The Second Affiliated Hospital of Nanchang University), Nanchang, China
| | - Zhiyong Xu
- School of Pharmacy, Nanchang University, Nanchang, China
| | - Chenjiang He
- The Center of Stomatology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
- The Institute of Periodontal Disease, Nanchang University, Nanchang, China.
- JXHC Key Laboratory of Periodontology (The Second Affiliated Hospital of Nanchang University), Nanchang, China
| | - Fanrong Ai
- JXHC Key Laboratory of Periodontology (The Second Affiliated Hospital of Nanchang University), Nanchang, China
- School of Advanced Manufacturing, Nanchang University, Nanchang, China.
| | - Li Song
- The Center of Stomatology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
- The Institute of Periodontal Disease, Nanchang University, Nanchang, China.
- JXHC Key Laboratory of Periodontology (The Second Affiliated Hospital of Nanchang University), Nanchang, China
| |
Collapse
|
11
|
Lee M, Du H, Winer DA, Clemente-Casares X, Tsai S. Mechanosensing in macrophages and dendritic cells in steady-state and disease. Front Cell Dev Biol 2022; 10:1044729. [PMID: 36467420 PMCID: PMC9712790 DOI: 10.3389/fcell.2022.1044729] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 11/01/2022] [Indexed: 11/18/2022] Open
Abstract
Macrophages and dendritic cells are myeloid cells that play critical roles in immune responses. Macrophages help to maintain homeostasis through tissue regeneration and the clearance of dead cells, but also mediate inflammatory processes against invading pathogens. As the most potent antigen-presenting cells, dendritic cells are important in connecting innate to adaptive immune responses via activation of T cells, and inducing tolerance under physiological conditions. While it is known that macrophages and dendritic cells respond to biochemical cues in the microenvironment, the role of extracellular mechanical stimuli is becoming increasingly apparent. Immune cell mechanotransduction is an emerging field, where accumulating evidence suggests a role for extracellular physical cues coming from tissue stiffness in promoting immune cell recruitment, activation, metabolism and inflammatory function. Additionally, many diseases such as pulmonary fibrosis, cardiovascular disease, cancer, and cirrhosis are associated with changes to the tissue biophysical environment. This review will discuss current knowledge about the effects of biophysical cues including matrix stiffness, topography, and mechanical forces on macrophage and dendritic cell behavior under steady-state and pathophysiological conditions. In addition, we will also provide insight on molecular mediators and signaling pathways important in macrophage and dendritic cell mechanotransduction.
Collapse
Affiliation(s)
- Megan Lee
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Huixun Du
- Buck Institute for Research on Aging, Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, United States
| | - Daniel A. Winer
- Division of Cellular and Molecular Biology, Diabetes Research Group, Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Department of Pathology, University Health Network, Toronto, ON, Canada
- Buck Institute for Research on Aging, Novato, CA, United States
| | - Xavier Clemente-Casares
- Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton, AB, Canada
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada
| | - Sue Tsai
- Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton, AB, Canada
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
12
|
Bomb K, Pradhan L, Zhang Q, Jarai BM, Bhattacharjee A, Burris DL, Kloxin AM, Fromen CA. Destructive fibrotic teamwork: how both microenvironment stiffness and profibrotic interleukin 13 impair alveolar macrophage phenotype and function. Biomater Sci 2022; 10:5689-5706. [PMID: 36018297 PMCID: PMC9632634 DOI: 10.1039/d2bm00828a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The pulmonary fibrotic microenvironment is characterized by increased stiffness of lung tissue and enhanced secretion of profibrotic soluble cues contributing to a feedback loop that leads to dysregulated wound healing and lung failure. Pinpointing the individual and tandem effects of profibrotic stimuli in impairing immune cell response remains difficult and is needed for improved therapeutic strategies. We utilized a statistical design of experiment (DOE) to investigate how microenvironment stiffness and interleukin 13 (IL13), a profibrotic soluble factor linked with disease severity, contribute to the impaired macrophage response commonly observed in pulmonary fibrosis. We used engineered bioinspired hydrogels of different stiffness, ranging from healthy to fibrotic lung tissue, and cultured murine alveolar macrophages (MH-S cells) with or without IL13 to quantify cell response and analyze independent and synergistic effects. We found that, while both stiffness and IL13 independently influence macrophage morphology, phenotype, phagocytosis and efferocytosis, these factors work synergistically to exacerbate impaired macrophage phenotype and efferocytosis. These unique findings provide insights into how macrophages in fibrotic conditions are not as effective in clearing debris, contributing to fibrosis initiation/progression, and more broadly inform how underlying drivers of fibrosis modulate immune cell response to facilitate therapeutic strategies.
Collapse
Affiliation(s)
- Kartik Bomb
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, USA.
| | - Lina Pradhan
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, USA.
| | - Qi Zhang
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, USA.
| | - Bader M Jarai
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, USA.
| | | | - David L Burris
- Mechanical Engineering, University of Delaware, Newark, DE, USA
| | - April M Kloxin
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, USA.
- Material Science and Engineering, University of Delaware, Newark, DE, USA
| | - Catherine A Fromen
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, USA.
| |
Collapse
|
13
|
Kalashnikov N, Moraes C. Engineering physical microenvironments to study innate immune cell biophysics. APL Bioeng 2022; 6:031504. [PMID: 36156981 PMCID: PMC9492295 DOI: 10.1063/5.0098578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/22/2022] [Indexed: 12/04/2022] Open
Abstract
Innate immunity forms the core of the human body's defense system against infection, injury, and foreign objects. It aims to maintain homeostasis by promoting inflammation and then initiating tissue repair, but it can also lead to disease when dysregulated. Although innate immune cells respond to their physical microenvironment and carry out intrinsically mechanical actions such as migration and phagocytosis, we still do not have a complete biophysical description of innate immunity. Here, we review how engineering tools can be used to study innate immune cell biophysics. We first provide an overview of innate immunity from a biophysical perspective, review the biophysical factors that affect the innate immune system, and then explore innate immune cell biophysics in the context of migration, phagocytosis, and phenotype polarization. Throughout the review, we highlight how physical microenvironments can be designed to probe the innate immune system, discuss how biophysical insight gained from these studies can be used to generate a more comprehensive description of innate immunity, and briefly comment on how this insight could be used to develop mechanical immune biomarkers and immunomodulatory therapies.
Collapse
Affiliation(s)
- Nikita Kalashnikov
- Department of Chemical Engineering, McGill University, Montreal, Quebec H3A 0G4, Canada
| | | |
Collapse
|
14
|
Coronel MM, Martin KE, Hunckler MD, Kalelkar P, Shah RM, García AJ. Hydrolytically Degradable Microgels with Tunable Mechanical Properties Modulate the Host Immune Response. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2106896. [PMID: 35274457 PMCID: PMC10288386 DOI: 10.1002/smll.202106896] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 02/27/2022] [Indexed: 06/14/2023]
Abstract
Hydrogel microparticles (microgels) are an attractive approach for therapeutic delivery because of their modularity, injectability, and enhanced integration with the host tissue. Multiple microgel fabrication strategies and chemistries have been implemented, yet manipulation of microgel degradability and its effect on in vivo tissue responses remains underexplored. Here, the authors report a facile method to synthesize microgels crosslinked with ester-containing junctions to afford tunable degradation kinetics. Monodisperse microgels of maleimide-functionalized poly(ethylene-glycol) are generated using droplet microfluidics crosslinked with thiol-terminated, ester-containing molecules. Tunable mechanics are achievable based on the ratio of degradable to nondegradable crosslinkers in the continuous phase. Degradation in an aqueous medium leads to microgel deformation based on swelling and a decrease in elastic modulus. Furthermore, degradation byproducts are cytocompatible and do not cause monocytic cell activation under noninflammatory conditions. These injectable microgels possess time-dependent degradation on the order of weeks in vivo. Lastly, the evaluation of tissue responses in a subcutaneous dorsal pocket shows a dynamic type-1 like immune response to the synthetic microgels, driven by interferon gamma (IFN-γ ) expression, which can be moderated by tuning the degradation properties. Collectively, this study demonstrates the development of a hydrolytic microgel platform that can be adapted to desired host tissue immune responses.
Collapse
Affiliation(s)
- María M Coronel
- Woodruff School of Mechanical Engineering and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Karen E Martin
- Woodruff School of Mechanical Engineering and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Michael D Hunckler
- Woodruff School of Mechanical Engineering and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Pranav Kalelkar
- Woodruff School of Mechanical Engineering and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Rahul M Shah
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Andrés J García
- Woodruff School of Mechanical Engineering and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| |
Collapse
|
15
|
Wang Y, Shi R, Zhai R, Yang S, Peng T, Zheng F, Shen Y, Li M, Li L. Matrix stiffness regulates macrophage polarization in atherosclerosis. Pharmacol Res 2022; 179:106236. [PMID: 35483516 DOI: 10.1016/j.phrs.2022.106236] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/01/2022] [Accepted: 04/21/2022] [Indexed: 12/12/2022]
Abstract
Atherosclerosis is a chronic inflammatory disease and the pathological basis of many fatal cardiovascular diseases. Macrophages, the main inflammatory cells in atherosclerotic plaque, have a paradox role in disease progression. In response to different microenvironments, macrophages mainly have two polarized directions: pro-inflammatory macrophages and anti-inflammatory macrophages. More and more evidence shows that macrophage is mechanosensitive and matrix stiffness regulate macrophage phenotypes in atherosclerosis. However, the molecular mechanism of matrix stiffness regulating macrophage polarization still lacks in-depth research, which hinders the development of new anti-atherosclerotic therapies. In this review, we discuss the important role of matrix stiffness in regulating macrophage polarization through mechanical signal transduction (Hippo, Piezo, cytoskeleton, and integrin) and epigenetic mechanisms (miRNA, DNA methylation, and histone). We hope to provide a new perspective for atherosclerosis therapy by targeting matrix stiffness and macrophage polarization.
Collapse
Affiliation(s)
- Yin Wang
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Ruotong Shi
- Norman Bethune College of Medicine, Jilin University, Changchun 130021, China
| | - Ran Zhai
- Norman Bethune College of Medicine, Jilin University, Changchun 130021, China
| | - Shiyan Yang
- Norman Bethune College of Medicine, Jilin University, Changchun 130021, China
| | - Tianqi Peng
- Norman Bethune College of Medicine, Jilin University, Changchun 130021, China
| | - Fuwen Zheng
- Norman Bethune College of Medicine, Jilin University, Changchun 130021, China
| | - YanNan Shen
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China.
| | - Meiying Li
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Lisha Li
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| |
Collapse
|
16
|
Acharya TK, Sahu RP, Kumar S, Kumar S, Rokade TP, Chakraborty R, Dubey NK, Shikha D, Chawla S, Goswami C. Function and regulation of thermosensitive ion channel TRPV4 in the immune system. CURRENT TOPICS IN MEMBRANES 2022; 89:155-188. [DOI: 10.1016/bs.ctm.2022.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
|
17
|
Goswami R, Arya RK, Sharma S, Dutta B, Stamov DR, Zhu X, Rahaman SO. Mechanosensing by TRPV4 mediates stiffness-induced foreign body response and giant cell formation. Sci Signal 2021; 14:eabd4077. [PMID: 34726952 PMCID: PMC9976933 DOI: 10.1126/scisignal.abd4077] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Implantation of biomaterials or devices into soft tissue often leads to the development of the foreign body response (FBR), an inflammatory condition that can cause implant failure, tissue injury, and death of the patient. Macrophages accumulate and fuse to generate destructive foreign body giant cells (FBGCs) at the tissue-implant interface, leading to the development of fibrous scar tissue around the implant that is generated by myofibroblasts. We previously showed that the FBR in vivo and FBGC formation in vitro require transient receptor potential vanilloid 4 (TRPV4), a mechanosensitive ion channel. Here, we report that TRPV4 was required specifically for the FBR induced by implant stiffness independently of biochemical cues and for intracellular stiffening that promotes FBGC formation in vitro. TRPV4 deficiency reduced collagen deposition and the accumulation of macrophages, FBGCs, and myofibroblasts at stiff, but not soft, implants in vivo and inhibited macrophage-induced differentiation of wild-type fibroblasts into myofibroblasts in vitro. Atomic force microscopy demonstrated that TRPV4 was required for implant-adjacent tissue stiffening in vivo and for cytoskeletal remodeling and intracellular stiffening induced by fusogenic cytokines in vitro. Together, these data suggest a mechanism whereby a reciprocal functional interaction between TRPV4 and substrate stiffness leads to cytoskeletal remodeling and cellular force generation to promote FBGC formation during the FBR.
Collapse
Affiliation(s)
- Rishov Goswami
- Department of Nutrition and Food Science, University of Maryland, College Park, MD 20742, USA
| | - Rakesh K. Arya
- Department of Nutrition and Food Science, University of Maryland, College Park, MD 20742, USA
| | - Shweta Sharma
- Department of Nutrition and Food Science, University of Maryland, College Park, MD 20742, USA
| | - Bidisha Dutta
- Department of Nutrition and Food Science, University of Maryland, College Park, MD 20742, USA
| | - Dimitar R. Stamov
- JPK BioAFM Business, Nano Surfaces Division, Bruker Nano GmbH, Am Studio 2D, 12489 Berlin, Germany
| | - Xiaoping Zhu
- Department of Veterinary Medicine, University of Maryland, College Park, MD 20742, USA
| | - Shaik O. Rahaman
- Department of Nutrition and Food Science, University of Maryland, College Park, MD 20742, USA.,Corresponding author.:
| |
Collapse
|
18
|
Novak C, Ballinger MN, Ghadiali S. Mechanobiology of Pulmonary Diseases: A Review of Engineering Tools to Understand Lung Mechanotransduction. J Biomech Eng 2021; 143:110801. [PMID: 33973005 PMCID: PMC8299813 DOI: 10.1115/1.4051118] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 05/01/2021] [Indexed: 12/17/2022]
Abstract
Cells within the lung micro-environment are continuously subjected to dynamic mechanical stimuli which are converted into biochemical signaling events in a process known as mechanotransduction. In pulmonary diseases, the abrogated mechanical conditions modify the homeostatic signaling which influences cellular phenotype and disease progression. The use of in vitro models has significantly expanded our understanding of lung mechanotransduction mechanisms. However, our ability to match complex facets of the lung including three-dimensionality, multicellular interactions, and multiple simultaneous forces is limited and it has proven difficult to replicate and control these factors in vitro. The goal of this review is to (a) outline the anatomy of the pulmonary system and the mechanical stimuli that reside therein, (b) describe how disease impacts the mechanical micro-environment of the lung, and (c) summarize how existing in vitro models have contributed to our current understanding of pulmonary mechanotransduction. We also highlight critical needs in the pulmonary mechanotransduction field with an emphasis on next-generation devices that can simulate the complex mechanical and cellular environment of the lung. This review provides a comprehensive basis for understanding the current state of knowledge in pulmonary mechanotransduction and identifying the areas for future research.
Collapse
Affiliation(s)
- Caymen Novak
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Davis Heart and Lung Research Institute, The Ohio State University, Wexner Medical Center, 473 West 12th Avenue, Columbus, OH 43210
| | - Megan N. Ballinger
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Davis Heart and Lung Research Institute, The Ohio State University, Wexner Medical Center, 473 West 12th Avenue, Columbus, OH 43210
| | - Samir Ghadiali
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Davis Heart and Lung Research Institute, The Ohio State University, Wexner Medical Center, 473 West 12th Avenue, Columbus, OH 43210; Department of Biomedical Engineering, The Ohio State University, 2124N Fontana Labs, 140 West 19th Avenue, Columbus, OH 43210
| |
Collapse
|
19
|
The Evolving Roles of Cardiac Macrophages in Homeostasis, Regeneration, and Repair. Int J Mol Sci 2021; 22:ijms22157923. [PMID: 34360689 PMCID: PMC8347787 DOI: 10.3390/ijms22157923] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/21/2021] [Accepted: 07/23/2021] [Indexed: 12/13/2022] Open
Abstract
Macrophages were first described as phagocytic immune cells responsible for maintaining tissue homeostasis by the removal of pathogens that disturb normal function. Historically, macrophages have been viewed as terminally differentiated monocyte-derived cells that originated through hematopoiesis and infiltrated multiple tissues in the presence of inflammation or during turnover in normal homeostasis. However, improved cell detection and fate-mapping strategies have elucidated the various lineages of tissue-resident macrophages, which can derive from embryonic origins independent of hematopoiesis and monocyte infiltration. The role of resident macrophages in organs such as the skin, liver, and the lungs have been well characterized, revealing functions well beyond a pure phagocytic and immunological role. In the heart, recent research has begun to decipher the functional roles of various tissue-resident macrophage populations through fate mapping and genetic depletion studies. Several of these studies have elucidated the novel and unexpected roles of cardiac-resident macrophages in homeostasis, including maintaining mitochondrial function, facilitating cardiac conduction, coronary development, and lymphangiogenesis, among others. Additionally, following cardiac injury, cardiac-resident macrophages adopt diverse functions such as the clearance of necrotic and apoptotic cells and debris, a reduction in the inflammatory monocyte infiltration, promotion of angiogenesis, amelioration of inflammation, and hypertrophy in the remaining myocardium, overall limiting damage extension. The present review discusses the origin, development, characterization, and function of cardiac macrophages in homeostasis, cardiac regeneration, and after cardiac injury or stress.
Collapse
|
20
|
Tan J, Zhang QY, Huang LP, Huang K, Xie HQ. Decellularized scaffold and its elicited immune response towards the host: the underlying mechanism and means of immunomodulatory modification. Biomater Sci 2021; 9:4803-4820. [PMID: 34018503 DOI: 10.1039/d1bm00470k] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The immune response of the host towards a decellularized scaffold is complex. Not only can a number of immune cells influence this process, but also the characteristics, preparation and modification of the decellularized scaffold can significantly impact this reaction. Such factors can, together or alone, trigger immune cells to polarize towards either a pro-healing or pro-inflammatory direction. In this article, we have comprehensively reviewed factors which may influence the immune response of the host towards a decellularized scaffold, including the source of the biomaterial, biophysical properties or modifications of the scaffolds with bioactive peptides, drugs and cytokines. Furthermore, the underlying mechanism has also been recapitulated.
Collapse
Affiliation(s)
- Jie Tan
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Med-X Center for Materials, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China.
| | - Qing-Yi Zhang
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Med-X Center for Materials, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China.
| | - Li-Ping Huang
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Med-X Center for Materials, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China.
| | - Kai Huang
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Med-X Center for Materials, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China.
| | - Hui-Qi Xie
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Med-X Center for Materials, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China.
| |
Collapse
|
21
|
Implant Fibrosis and the Underappreciated Role of Myofibroblasts in the Foreign Body Reaction. Cells 2021; 10:cells10071794. [PMID: 34359963 PMCID: PMC8304203 DOI: 10.3390/cells10071794] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/08/2021] [Accepted: 07/13/2021] [Indexed: 02/06/2023] Open
Abstract
Body implants and implantable medical devices have dramatically improved and prolonged the life of countless patients. However, our body repair mechanisms have evolved to isolate, reject, or destroy any object that is recognized as foreign to the organism and inevitably mounts a foreign body reaction (FBR). Depending on its severity and chronicity, the FBR can impair implant performance or create severe clinical complications that will require surgical removal and/or replacement of the faulty device. The number of review articles discussing the FBR seems to be proportional to the number of different implant materials and clinical applications and one wonders, what else is there to tell? We will here take the position of a fibrosis researcher (which, coincidentally, we are) to elaborate similarities and differences between the FBR, normal wound healing, and chronic healing conditions that result in the development of peri-implant fibrosis. After giving credit to macrophages in the inflammatory phase of the FBR, we will mainly focus on the activation of fibroblastic cells into matrix-producing and highly contractile myofibroblasts. While fibrosis has been discussed to be a consequence of the disturbed and chronic inflammatory milieu in the FBR, direct activation of myofibroblasts at the implant surface is less commonly considered. Thus, we will provide a perspective how physical properties of the implant surface control myofibroblast actions and accumulation of stiff scar tissue. Because formation of scar tissue at the surface and around implant materials is a major reason for device failure and extraction surgeries, providing implant surfaces with myofibroblast-suppressing features is a first step to enhance implant acceptance and functional lifetime. Alternative therapeutic targets are elements of the myofibroblast mechanotransduction and contractile machinery and we will end with a brief overview on such targets that are considered for the treatment of other organ fibroses.
Collapse
|
22
|
Vasse GF, Nizamoglu M, Heijink IH, Schlepütz M, van Rijn P, Thomas MJ, Burgess JK, Melgert BN. Macrophage-stroma interactions in fibrosis: biochemical, biophysical, and cellular perspectives. J Pathol 2021; 254:344-357. [PMID: 33506963 PMCID: PMC8252758 DOI: 10.1002/path.5632] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/18/2020] [Accepted: 01/08/2021] [Indexed: 12/16/2022]
Abstract
Fibrosis results from aberrant wound healing and is characterized by an accumulation of extracellular matrix, impairing the function of an affected organ. Increased deposition of extracellular matrix proteins, disruption of matrix degradation, but also abnormal post-translational modifications alter the biochemical composition and biophysical properties of the tissue microenvironment - the stroma. Macrophages are known to play an important role in wound healing and tissue repair, but the direct influence of fibrotic stroma on macrophage behaviour is still an under-investigated element in the pathogenesis of fibrosis. In this review, the current knowledge on interactions between macrophages and (fibrotic) stroma will be discussed from biochemical, biophysical, and cellular perspectives. Furthermore, we provide future perspectives with regard to how macrophage-stroma interactions can be examined further to ultimately facilitate more specific targeting of these interactions in the treatment of fibrosis. © 2021 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Gwenda F Vasse
- University of Groningen, University Medical Center GroningenBiomedical Engineering Department‐FB40GroningenThe Netherlands
- University of Groningen, University Medical Center Groningen, W.J. Kolff Institute for Biomedical Engineering and Materials ScienceGroningenThe Netherlands
- University of Groningen, Department of Molecular PharmacologyGroningen Research Institute for PharmacyGroningenThe Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC)GroningenThe Netherlands
| | - Mehmet Nizamoglu
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC)GroningenThe Netherlands
- University of Groningen, University Medical Center GroningenDepartment of Pathology and Medical BiologyGroningenThe Netherlands
| | - Irene H Heijink
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC)GroningenThe Netherlands
- University of Groningen, University Medical Center GroningenDepartment of Pathology and Medical BiologyGroningenThe Netherlands
- University of Groningen, University Medical Center GroningenDepartment of PulmonologyGroningenThe Netherlands
| | - Marco Schlepütz
- Immunology & Respiratory Diseases ResearchBoehringer Ingelheim Pharma GmbH & Co KGBiberach an der RissGermany
| | - Patrick van Rijn
- University of Groningen, University Medical Center GroningenBiomedical Engineering Department‐FB40GroningenThe Netherlands
- University of Groningen, University Medical Center Groningen, W.J. Kolff Institute for Biomedical Engineering and Materials ScienceGroningenThe Netherlands
| | - Matthew J Thomas
- Immunology & Respiratory Diseases ResearchBoehringer Ingelheim Pharma GmbH & Co KGBiberach an der RissGermany
| | - Janette K Burgess
- University of Groningen, University Medical Center Groningen, W.J. Kolff Institute for Biomedical Engineering and Materials ScienceGroningenThe Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC)GroningenThe Netherlands
- University of Groningen, University Medical Center GroningenDepartment of Pathology and Medical BiologyGroningenThe Netherlands
| | - Barbro N Melgert
- University of Groningen, Department of Molecular PharmacologyGroningen Research Institute for PharmacyGroningenThe Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC)GroningenThe Netherlands
| |
Collapse
|
23
|
Dudiki T, Mahajan G, Liu H, Zhevlakova I, Bertagnolli C, Nascimento DW, Kothapalli CR, Byzova TV. Kindlin3 regulates biophysical properties and mechanics of membrane to cortex attachment. Cell Mol Life Sci 2021; 78:4003-4018. [PMID: 33783564 PMCID: PMC11071771 DOI: 10.1007/s00018-021-03817-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 03/01/2021] [Accepted: 03/18/2021] [Indexed: 12/19/2022]
Abstract
Kindlin3 (K3), a FERM domain containing protein expressed in hematopoietic cells controls integrin activation and thus hemostatic and inflammatory responses. However, its role in the mechanics of plasma membrane remains unclear. Here, we show that genetic knockout of K3 in microglia and macrophages resulted in defective plasma membrane tension and membrane blebbing. Atomic force microscopy (AFM) of K3-deficient cells revealed a significant loss in membrane-to-cortex attachment (MCA), and consequently reduced membrane tension. This loss in MCA is amplified by the mislocalization of the cell cortex proteins-ezrin, radixin, and moesin (ERM)-to the plasma membrane of microglia and macrophages. Re-expression of K3 in K3-deficient macrophages rescued the defects and localization of ERMs implying a key role for K3 in MCA. Analysis of two K3 mutants, K3int affecting integrin binding and activation, and K3pxn/act disrupting binding to paxillin and actin but not integrin functions, demonstrated that the role of K3 in membrane mechanics is separate from integrin activation. The K3pxn/act mutant substantially diminished both membrane tension and Yes-associated protein (YAP) translocation to the nucleus, while preserving integrin activation, cell spreading, and migration. Together, our results show that K3 coordinates membrane mechanics, ERM protein recruitment to the membrane, and YAP translocation by linking integrin at the membrane to paxillin and actin of the cytoskeleton. This novel function of K3 is distinct from its role in integrin activation.
Collapse
Affiliation(s)
- Tejasvi Dudiki
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Gautam Mahajan
- Chemical and Biomedical Engineering Department, Washkewicz College of Engineering, Cleveland State University, Cleveland, OH, 44115, USA
| | - Huan Liu
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Irina Zhevlakova
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Chase Bertagnolli
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | | | - Chandrasekhar R Kothapalli
- Chemical and Biomedical Engineering Department, Washkewicz College of Engineering, Cleveland State University, Cleveland, OH, 44115, USA.
| | - Tatiana V Byzova
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA.
| |
Collapse
|
24
|
Xu A, Zhou J, Li Y, Qiao L, Jin C, Chen W, Sun L, Wu S, Li X, Zhou D, Jia S, Zhang B, Yao J, Zhang X, You H, Huang J. 14-kDa phosphohistidine phosphatase is a potential therapeutic target for liver fibrosis. Am J Physiol Gastrointest Liver Physiol 2021; 320:G351-G365. [PMID: 33406007 DOI: 10.1152/ajpgi.00334.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Liver fibrosis, a major cause of morbidity and mortality worldwide, leads to liver damage, seriously threatening human health. In our previous study, we demonstrated that 14 kDa phosphohistidine phosphatase (PHP14) was upregulated in fibrotic liver tissue and involved in the migration and lamellipodia formation of hepatic stellate cells (HSCs). In this study, we evaluated PHP14 as a therapeutic target for liver fibrosis and investigated the mechanism by which it mediates liver fibrosis. AAV-shPhpt1 administration significantly attenuates CCl4-induced liver fibrosis in mice. In particular, fibrosis-associated inflammatory infiltration was significantly suppressed after PHP14 knockdown. Mechanistically, PHP14 regulated macrophage recruitment, infiltration, and migration by affecting podosome formation of macrophages. Inhibition of PHP14 decreased the expression of the fibrogenic signature at the early stage of liver fibrogenesis and the activation of HSCs in vivo. Thus, PHP14 can be considered a potential therapeutic target for liver fibrosis.NEW & NOTEWORTHY PHP14 inhibition via adeno-associated virus (AAV)-mediated gene silencing could potently attenuate carbon tetrachloride (CCl4)-induced liver fibrosis. PHP14 could regulate the migration of macrophages to the site of injury in vivo. PHP14 knockdown in vivo influenced the environment of fibrogenesis and relevant signaling pathways, subsequently affecting myofibroblast activation.
Collapse
Affiliation(s)
- Anjian Xu
- Experimental Center, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China.,National Clinical Research Center for Digestive Disease, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Jichao Zhou
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People's Republic of China
| | - Yanmeng Li
- Experimental Center, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China.,National Clinical Research Center for Digestive Disease, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Luyao Qiao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People's Republic of China
| | - Caicai Jin
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People's Republic of China
| | - Wei Chen
- Experimental Center, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Lan Sun
- Department of Pathology, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Shanna Wu
- Clinical Laboratory Center, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Xiaojin Li
- Experimental Center, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Donghu Zhou
- Experimental Center, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Siyu Jia
- Experimental Center, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Bei Zhang
- Experimental Center, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Jingyi Yao
- Experimental Center, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Xiaowei Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People's Republic of China
| | - Hong You
- Experimental Center, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China.,National Clinical Research Center for Digestive Disease, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China.,Liver Research Center, National Clinical Research Center for Digestive Diseases, Beijing, People's Republic of China
| | - Jian Huang
- Experimental Center, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China.,National Clinical Research Center for Digestive Disease, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
25
|
Sridharan R, Kelly DJ, O'Brien FJ. Substrate Stiffness Modulates the Crosstalk Between Mesenchymal Stem Cells and Macrophages. J Biomech Eng 2021; 143:031001. [PMID: 33067618 DOI: 10.1115/1.4048809] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Indexed: 07/25/2024]
Abstract
Upon implantation of a biomaterial, mesenchymal stem cells (MSCs) and macrophages contribute to the wound healing response and the regeneration cascade. Although biomaterial properties are known to direct MSC differentiation and macrophage polarization, the role of biomaterial cues, specifically stiffness, in directing the crosstalk between the two cell types is still poorly understood. This study aimed to elucidate the role of substrate stiffness in modulating the immunomodulatory properties of MSCs and to shed light on their complex interactions with macrophages when presented with diverse biomaterial stiffness cues, a situation analogous to the implant environment where multiple cell types interact with an implanted biomaterial to determine regenerative outcomes. We show that MSCs do not play an immunomodulatory role in the absence of an inflammatory stimulus. Using collagen-coated polyacrylamide gels of varying stiffness values, we demonstrate that the immunomodulatory capability of MSCs in the presence of an inflammatory stimulus is not dependent on the stiffness of the underlying substrate. Moreover, using paracrine and direct contact culture models, we show that a bidirectional crosstalk between MSCs and macrophages is necessary for promoting anti-inflammatory responses and positive immunomodulation, which is dependent on the stiffness of the underlying substrate. We finally show that direct cell-cell contact is not essential for this effect, with paracrine interactions promoting immunomodulatory interactions between MSCs and macrophages. Together, these results demonstrate that biophysical cues such as stiffness that are presented by biomaterials can be tuned to promote positive interactions between MSCs and macrophages which can in turn direct the downstream regenerative response.
Collapse
Affiliation(s)
- Rukmani Sridharan
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin 2 D02 YN77, Ireland; Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2 D02 PN40, Ireland; Advanced Materials Bio-Engineering Research Centre (AMBER), Trinity College Dublin, Dublin 2, D02 PN40, Ireland
| | - Daniel J Kelly
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin 2 D02 YN77, Ireland; Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2 D02 PN40, Ireland; Advanced Materials Bio-Engineering Research Centre (AMBER), Trinity College Dublin, Dublin 2, D02 PN40, Ireland
| | - Fergal J O'Brien
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin 2 D02 YN77, Ireland; Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2 D02 PN40, Ireland; Advanced Materials Bio-Engineering Research Centre (AMBER), Trinity College Dublin, Dublin 2, D02 PN40, Ireland
| |
Collapse
|
26
|
Dutta B, Goswami R, Rahaman SO. TRPV4 Plays a Role in Matrix Stiffness-Induced Macrophage Polarization. Front Immunol 2020; 11:570195. [PMID: 33381111 PMCID: PMC7767862 DOI: 10.3389/fimmu.2020.570195] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 11/04/2020] [Indexed: 12/16/2022] Open
Abstract
Phenotypic polarization of macrophages is deemed essential in innate immunity and various pathophysiological conditions. We have now determined key aspects of the molecular mechanism by which mechanical cues regulate macrophage polarization. We show that Transient Receptor Potential Vanilloid 4 (TRPV4), a mechanosensitive ion channel, mediates substrate stiffness-induced macrophage polarization. Using atomic force microscopy, we showed that genetic ablation of TRPV4 function abrogated fibrosis-induced matrix stiffness generation in skin tissues. We have determined that stiffer skin tissue promotes the M1 macrophage subtype in a TRPV4-dependent manner; soft tissue does not. These findings were further validated by our in vitro results which showed that stiff matrix (50 kPa) alone increased expression of macrophage M1 markers in a TRPV4-dependent manner, and this response was further augmented by the addition of soluble factors; neither of which occurred with soft matrix (1 kPa). A direct requirement for TRPV4 in M1 macrophage polarization spectrum in response to increased stiffness was evident from results of gain-of-function assays, where reintroduction of TRPV4 significantly upregulated the expression of M1 markers in TRPV4 KO macrophages. Together, these data provide new insights regarding the role of TRPV4 in matrix stiffness-induced macrophage polarization spectrum that may be explored in tissue engineering and regenerative medicine and targeted therapeutics.
Collapse
Affiliation(s)
- Bidisha Dutta
- Department of Nutrition and Food Science, University of Maryland, College Park, MD, United States
| | - Rishov Goswami
- Department of Nutrition and Food Science, University of Maryland, College Park, MD, United States
| | - Shaik O Rahaman
- Department of Nutrition and Food Science, University of Maryland, College Park, MD, United States
| |
Collapse
|
27
|
Nanoscale mechanical properties of chitosan hydrogels as revealed by AFM. Prog Biomater 2020; 9:187-201. [PMID: 33156481 DOI: 10.1007/s40204-020-00141-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 10/08/2020] [Indexed: 01/25/2023] Open
Abstract
In the context of tissue engineering, chitosan hydrogels are attractive biomaterials because they represent a family of natural polymers exhibiting several suitable features (cytocompatibility, bioresorbability, wound healing, bacteriostatic and fungistatic properties, structural similarity with glycosaminoglycans), and tunable mechanical properties. Optimizing the design of these biomaterials requires fine knowledge of its physical characteristics prior to assessment of the cell-biomaterial interactions. In this work, using atomic force microscopy (AFM), we report a characterization of mechanical and topographical properties at the submicron range of chitosan hydrogels, depending on physico-chemical parameters such as their polymer concentration (1.5%, 2.5% and 3.5%), their degree of acetylation (4% and 38.5%), and the conditions of the gelation process. Well-known polyacrylamide gels were used to validate the methodology approach for the determination and analysis of elastic modulus (i.e., Young's modulus) distribution at the gel surface. We present elastic modulus distribution and topographical and stiffness maps for different chitosan hydrogels. For each chitosan hydrogel formulation, AFM analyses reveal a specific asymmetric elastic modulus distribution that constitutes a useful hallmark for chitosan hydrogel characterization. Our results regarding the local mechanical properties and the topography of chitosan hydrogels initiate new possibilities for an interpretation of the behavior of cells in contact with such soft materials.
Collapse
|
28
|
Yang J, Pan X, Wang L, Yu G. Alveolar cells under mechanical stressed niche: critical contributors to pulmonary fibrosis. Mol Med 2020; 26:95. [PMID: 33054759 PMCID: PMC7556585 DOI: 10.1186/s10020-020-00223-w] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 10/07/2020] [Indexed: 12/14/2022] Open
Abstract
Pulmonary fibrosis arises from the repeated epithelial mild injuries and insufficient repair lead to over activation of fibroblasts and excessive deposition of extracellular matrix, which result in a mechanical stretched niche. However, increasing mechanical stress likely exists before the establishment of fibrosis since early micro injuries increase local vascular permeability and prompt cytoskeletal remodeling which alter cellular mechanical forces. It is noteworthy that COVID-19 patients with severe hypoxemia will receive mechanical ventilation as supportive treatment and subsequent pathology studies indicate lung fibrosis pattern. At advanced stages, mechanical stress originates mainly from the stiff matrix since boundaries between stiff and compliant parts of the tissue could generate mechanical stress. Therefore, mechanical stress has a significant role in the whole development process of pulmonary fibrosis. The alveoli are covered by abundant capillaries and function as the main gas exchange unit. Constantly subject to variety of damages, the alveolar epithelium injuries were recently recognized to play a vital role in the onset and development of idiopathic pulmonary fibrosis. In this review, we summarize the literature regarding the effects of mechanical stress on the fundamental cells constituting the alveoli in the process of pulmonary fibrosis, particularly on epithelial cells, capillary endothelial cells, fibroblasts, mast cells, macrophages and stem cells. Finally, we briefly review this issue from a more comprehensive perspective: the metabolic and epigenetic regulation.
Collapse
Affiliation(s)
- Juntang Yang
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Outstanding Overseas Scientists Center for Pulmonary Fibrosis of Henan Province, College of Life Science, Institute of Biomedical Science, Henan Normal University, Xinxiang, Henan, China
| | - Xin Pan
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Outstanding Overseas Scientists Center for Pulmonary Fibrosis of Henan Province, College of Life Science, Institute of Biomedical Science, Henan Normal University, Xinxiang, Henan, China
| | - Lan Wang
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Outstanding Overseas Scientists Center for Pulmonary Fibrosis of Henan Province, College of Life Science, Institute of Biomedical Science, Henan Normal University, Xinxiang, Henan, China
| | - Guoying Yu
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Outstanding Overseas Scientists Center for Pulmonary Fibrosis of Henan Province, College of Life Science, Institute of Biomedical Science, Henan Normal University, Xinxiang, Henan, China.
| |
Collapse
|
29
|
Scheraga RG, Southern BD, Grove LM, Olman MA. The Role of TRPV4 in Regulating Innate Immune Cell Function in Lung Inflammation. Front Immunol 2020; 11:1211. [PMID: 32676078 PMCID: PMC7333351 DOI: 10.3389/fimmu.2020.01211] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 05/15/2020] [Indexed: 12/22/2022] Open
Abstract
Ion channels/pumps are essential regulators of innate immune cell function. Macrophages have been increasingly recognized to have phenotypic plasticity and location-specific functions in the lung. Transient receptor potential vanilloid 4 (TRPV4) function in lung injury has been shown to be stimulus- and cell-type specific. In the current review, we discuss the importance of TRPV4 in macrophages and its role in phagocytosis and cytokine secretion in acute lung injury/acute respiratory distress syndrome (ARDS). Furthermore, TRPV4 controls a MAPK molecular switch from predominately c-Jun N-terminal kinase, JNK activation, to that of p38 activation, that mediates phagocytosis and cytokine secretion in a matrix stiffness-dependent manner. Expanding knowledge regarding the downstream mechanisms by which TRPV4 acts to tailor macrophage function in pulmonary inflammatory diseases will allow for formulation of novel therapeutics.
Collapse
Affiliation(s)
- Rachel G. Scheraga
- Respiratory Institute, Cleveland Clinic, Cleveland, OH, United States
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Brian D. Southern
- Respiratory Institute, Cleveland Clinic, Cleveland, OH, United States
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Lisa M. Grove
- Respiratory Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Mitchell A. Olman
- Respiratory Institute, Cleveland Clinic, Cleveland, OH, United States
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| |
Collapse
|
30
|
Fang JY, Yang Z, Han B. Switch of macrophage fusion competency by 3D matrices. Sci Rep 2020; 10:10348. [PMID: 32587271 PMCID: PMC7316750 DOI: 10.1038/s41598-020-67056-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 06/02/2020] [Indexed: 12/13/2022] Open
Abstract
Foreign body reaction reflects the integration between biomaterials and host cells. At the implantation microenvironment, macrophages usually fuse into multinuclear cells, also known as foreign body giant cells, to respond to the biomaterial implants. To understand the biomaterial-induced macrophage fusion, we examined whether biomaterial alone can initiate and control the fusion rate without exogenous cytokines and chemicals. We introduced a collagen-based 3D matrix to embed Raw264.7 cell line and primary rat bone marrow-derived macrophages. We found the biomaterial-stimuli interacted regional macrophages and altered the overall fusogenic protein expressions to regulate the macrophage fusion rate. The fusion rate could be altered by modulating the cell-matrix and cell-cell adhesions. The fused macrophage morphologies, the nuclei number in the fused macrophage, and the fusion rates were matrix dependent. The phenomena were also observed in the in vivo models. These results suggest that the biomaterial-derived stimuli exert similar functions as cytokines to alter the competency of macrophage fusion as well as their drug sensitivity in the biomaterial implanted tissue environment. Furthermore, this in vitro 3D-matrix model has the potential to serve as a toolbox to predict the host tissue response on implanted biomaterials.
Collapse
Affiliation(s)
- Josephine Y Fang
- Nimni-Cordoba Tissue Engineering and Drug Discovery Laboratory, Division of Plastic and Reconstructive Surgery, Departments of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California, United States
- Center of Craniofacial Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, California, United States
| | - Zhi Yang
- Nimni-Cordoba Tissue Engineering and Drug Discovery Laboratory, Division of Plastic and Reconstructive Surgery, Departments of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California, United States
| | - Bo Han
- Nimni-Cordoba Tissue Engineering and Drug Discovery Laboratory, Division of Plastic and Reconstructive Surgery, Departments of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California, United States.
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Souther California, Los Angeles, California, United States.
| |
Collapse
|
31
|
Karavasili C, Tsongas K, Andreadis II, Andriotis EG, Papachristou ET, Papi RM, Tzetzis D, Fatouros DG. Physico-mechanical and finite element analysis evaluation of 3D printable alginate-methylcellulose inks for wound healing applications. Carbohydr Polym 2020; 247:116666. [PMID: 32829794 DOI: 10.1016/j.carbpol.2020.116666] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 06/12/2020] [Accepted: 06/16/2020] [Indexed: 01/08/2023]
Abstract
The present study reports on the comprehensive physico-mechanical evaluation of 3D printable alginate-methylcellulose hydrogels with bioactive components (Manuka honey, aloe vera gel, eucalyptus essential oil) using a combined experimental-numerical approach. The 3D printable carbohydrate inks demonstrated good swelling properties under moist conditions and adequate antimicrobial and antibiofilm efficacy against both Gram positive and negative bacteria. The effect of the bioactive compounds on the viscosity and mechanical properties of the 3D printable hydrogels was assessed with rheological, nanoindentation and shear test measurements. All hydrogel compositions showed good biocompatibility on human dermal fibroblasts, stimulating cell growth as confirmed by an in vitro wound healing assay. Finite element analysis simulation was employed to further advance the calculation accuracy of the nanoindentation tests, concluding that combination of an experimental and a numerical technique may constitute a useful method to characterize the mechanical behavior of composite hydrogel films for use in wound healing applications.
Collapse
Affiliation(s)
- Christina Karavasili
- Laboratory of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki GR-54124, Greece.
| | - Konstantinos Tsongas
- Digital Manufacturing and Materials Characterization Laboratory, School of Science and Technology, International Hellenic University, 14km Thessaloniki - N. Moudania, Thermi GR-57001, Greece
| | - Ioannis I Andreadis
- Laboratory of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki GR-54124, Greece
| | - Eleftherios G Andriotis
- Laboratory of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki GR-54124, Greece
| | - Eleni T Papachristou
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, Thessaloniki GR-54124, Greece
| | - Rigini M Papi
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, Thessaloniki GR-54124, Greece
| | - Dimitrios Tzetzis
- Digital Manufacturing and Materials Characterization Laboratory, School of Science and Technology, International Hellenic University, 14km Thessaloniki - N. Moudania, Thermi GR-57001, Greece
| | - Dimitrios G Fatouros
- Laboratory of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki GR-54124, Greece
| |
Collapse
|
32
|
Xiao J, Huang T, Hu J, Zan F, Liao Z, Gu R, Wu G, Liao H. The Thickness of Surface Grafting Layer on Bio-materials directly Mediates the Immuno-reacitivity of Macrophages in vitro. Open Life Sci 2020; 15:198-208. [PMID: 33987476 PMCID: PMC8114785 DOI: 10.1515/biol-2020-0020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 11/27/2019] [Indexed: 11/15/2022] Open
Abstract
Introducing the surface grafting layers to regulate bio-compatibility and bio-function is an important step in the tissue engineering field. However, whether the thickness variation of the introduced biomimetic layer impacts the behavior of the adhered immune effector cells is yet to be dissected fully. In this study, we used a surface-induced atom transfer radical polymerization (SI-ATRP) method to synthetize and graft poly-phenoxyethyl methacrylate (PHEMA) brushes having different lengths on the glass substrates. Primary murine peritoneal macrophages were collected and cultured on the PHEMA brushes and we investigated the influence of polymer brushes having different lengths on macrophages phenotype and function. Our results demonstrated that the thicker brushes (200 nm and 450 nm) are superior to the thinner layers (50 nm) for macrophages survival, proliferation, cell elongation and migration. Moreover, the thicker brushes are more beneficial for macrophage's activities and functions, presented by the increased production of M1-associated cytokines IL-6 and MCP-1, the elevated cell phagocytosis and the activation molecule F4/80 expression, and the reduced macrophages apoptosis in thicker brushes-sustained macrophages. Our data suggests that the thickness of the substrate grafting layer directly impacts macrophages recruitment and pro-inflammatory function, which is important in determining the intrinsic immuno-compatibilities of the surface modified-biomaterials and mediates material-host interactions in vivo.
Collapse
Affiliation(s)
- JiangWei Xiao
- Department of Anatomy, Guangdong Provincial Key Laboratory of Medical Biomechanics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515 China
| | - Tao Huang
- Department of Anatomy, Guangdong Provincial Key Laboratory of Medical Biomechanics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515 China
| | - JiJie Hu
- Department of Orthopaedics and Traumatology, Nanfang Hospital, Southern Medical University; Guangzhou, 510515 China
| | - Fei Zan
- School of Materials Science and Engineering, South China University of Technology, Guangzhou,510641, China
| | - ZhaoHong Liao
- Department of Anatomy, Guangdong Provincial Key Laboratory of Medical Biomechanics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515 China
| | - RuiCai Gu
- Department of Anatomy, Guangdong Provincial Key Laboratory of Medical Biomechanics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515 China
| | - Gang Wu
- School of Materials Science and Engineering, South China University of Technology, Guangzhou,510641, China
| | - Hua Liao
- Department of Anatomy, Guangdong Provincial Key Laboratory of Medical Biomechanics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515 China
| |
Collapse
|
33
|
Im GI. Biomaterials in orthopaedics: the past and future with immune modulation. Biomater Res 2020; 24:7. [PMID: 32042442 PMCID: PMC7001269 DOI: 10.1186/s40824-020-0185-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 01/28/2020] [Indexed: 02/07/2023] Open
Abstract
Orthopaedics owes its current status of advanced care to the development of biomaterial science more than any other clinical medical specialty. The purpose of this brief review is to introduce the history and present status of biomaterials in orthopaedic field and cast a perspective on the future use of biomaterials to treat musculoskeletal disorders with particular emphasis on immune modulation. While the biomaterials in orthopaedics started from inert materials to replace the function and structure of hard tissue such as bone and cartilage, regenerative medicine will play a greater role in preventing the traumatic loss of tissues, as well as in the earlier stages of diseases. The understanding and modulation of immune response to biomaterials will further lead to the better incorporation of implants into host tissue or the near-perfect regeneration of host tissue.
Collapse
Affiliation(s)
- Gun-Il Im
- Research Institute for Integrative Regenerative Biomedical Engineering, Dongguk University Ilsan Hospital, 814 Siksa-Dong, Goyang, 410-773 Republic of Korea
| |
Collapse
|
34
|
Scheraga RG, Abraham S, Grove LM, Southern BD, Crish JF, Perelas A, McDonald C, Asosingh K, Hasday JD, Olman MA. TRPV4 Protects the Lung from Bacterial Pneumonia via MAPK Molecular Pathway Switching. THE JOURNAL OF IMMUNOLOGY 2020; 204:1310-1321. [PMID: 31969384 DOI: 10.4049/jimmunol.1901033] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 12/22/2019] [Indexed: 12/11/2022]
Abstract
Mechanical cell-matrix interactions can drive the innate immune responses to infection; however, the molecular underpinnings of these responses remain elusive. This study was undertaken to understand the molecular mechanism by which the mechanosensitive cation channel, transient receptor potential vanilloid 4 (TRPV4), alters the in vivo response to lung infection. For the first time, to our knowledge, we show that TRPV4 protects the lung from injury upon intratracheal Pseudomonas aeruginosa in mice. TRPV4 functions to enhance macrophage bacterial clearance and downregulate proinflammatory cytokine secretion. TRPV4 mediates these effects through a novel mechanism of molecular switching of LPS signaling from predominant activation of the MAPK, JNK, to that of p38. This is accomplished through the activation of the master regulator of inflammation, dual-specificity phosphatase 1. Further, TRPV4's modulation of the LPS signal is mechanosensitive in that both upstream activation of p38 and its downstream biological consequences depend on pathophysiological range extracellular matrix stiffness. We further show the importance of TRPV4 on LPS-induced activation of macrophages from healthy human controls. These data are the first, to our knowledge, to demonstrate new roles for macrophage TRPV4 in regulating innate immunity in a mechanosensitive manner through the modulation of dual-specificity phosphatase 1 expression to mediate MAPK activation switching.
Collapse
Affiliation(s)
- Rachel G Scheraga
- Respiratory Institute, Cleveland Clinic, Cleveland, OH 44195; .,Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195; and
| | - Susamma Abraham
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195; and
| | - Lisa M Grove
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195; and
| | - Brian D Southern
- Respiratory Institute, Cleveland Clinic, Cleveland, OH 44195.,Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195; and
| | - James F Crish
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195; and
| | | | - Christine McDonald
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195; and
| | - Kewal Asosingh
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195; and
| | - Jeffrey D Hasday
- Department of Pulmonary and Critical Care, University of Maryland, Baltimore, MD 21201
| | - Mitchell A Olman
- Respiratory Institute, Cleveland Clinic, Cleveland, OH 44195; .,Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195; and
| |
Collapse
|
35
|
Gruber EJ, Leifer CA. Molecular regulation of TLR signaling in health and disease: mechano-regulation of macrophages and TLR signaling. Innate Immun 2020; 26:15-25. [PMID: 31955624 PMCID: PMC6974875 DOI: 10.1177/1753425919838322] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 02/18/2019] [Accepted: 02/19/2019] [Indexed: 12/28/2022] Open
Abstract
Immune cells encounter tissues with vastly different biochemical and physical characteristics. Much of the research emphasis has focused on the role of cytokines and chemokines in regulating immune cell function, but the role of the physical microenvironment has received considerably less attention. The tissue mechanics, or stiffness, of healthy tissues varies dramatically from soft adipose tissue and brain to stiff cartilage and bone. Tissue mechanics also change due to fibrosis and with diseases such as atherosclerosis or cancer. The process by which cells sense and respond to their physical microenvironment is called mechanotransduction. Here we review mechanotransduction in immunologically important diseases and how physical characteristics of tissues regulate immune cell function, with a specific emphasis on mechanoregulation of macrophages and TLR signaling.
Collapse
Affiliation(s)
| | - Cynthia A Leifer
- Department of Microbiology and Immunology, Cornell
University, Ithaca, NY, USA
| |
Collapse
|
36
|
Abstract
Physical stimuli are essential for the function of eukaryotic cells, and changes in physical signals are important elements in normal tissue development as well as in disease initiation and progression. The complexity of physical stimuli and the cellular signals they initiate are as complex as those triggered by chemical signals. One of the most important, and the focus of this review, is the effect of substrate mechanical properties on cell structure and function. The past decade has produced a nearly exponentially increasing number of mechanobiological studies to define how substrate stiffness alters cell biology using both purified systems and intact tissues. Here we attempt to identify common features of mechanosensing in different systems while also highlighting the numerous informative exceptions to what in early studies appeared to be simple rules by which cells respond to mechanical stresses.
Collapse
Affiliation(s)
- Paul A Janmey
- Department of Physiology, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Bioengineering, University of California-Berkeley, Berkeley, California; and Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Daniel A Fletcher
- Department of Physiology, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Bioengineering, University of California-Berkeley, Berkeley, California; and Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Cynthia A Reinhart-King
- Department of Physiology, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Bioengineering, University of California-Berkeley, Berkeley, California; and Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| |
Collapse
|
37
|
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
|
38
|
Shan S, Fang B, Zhang Y, Wang C, Zhou J, Niu C, Gao Y, Zhao D, He J, Wang J, Zhang X, Li Q. Mechanical stretch promotes tumoricidal M1 polarization via the FAK/NF-κB signaling pathway. FASEB J 2019; 33:13254-13266. [PMID: 31539281 DOI: 10.1096/fj.201900799rr] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Macrophages (Mφs) can be used as a part of cell-based cancer immunotherapy. However, they may be hampered by a failure to effectively and stably regulate their polarization state to enhance their tumoricidal effects. In this work, mechanical stretch (MS), as a biology-free modulatory method, was shown to enhance M1 polarization and tumoricidal effects. By using an in vitro Flexcell Tension system, we found that murine Mφ RAW264.7 cells showed higher M1 polarization-related mRNA expression and cytokine release after MS. Further molecular analyses found that focal adhesion kinase and NF-κB activation occurred in the MS-induced M1 polarization. Coculture of MS-preconditioned Mφ with B16F10 skin melanoma cells in vitro showed that the proliferation of B16F10 cells decreased, whereas caspase-3-induced apoptosis increased. Importantly, the injection of MS-preconditioned Mφ into murine skin melanomas in vivo impeded tumor growth; lesions were characterized by increased amounts of M1 Mφ, decreased tumor cell proliferation, and increased tumor cell apoptosis in the tumor microenvironment. Together, our results suggest that MS could be used as a simple preconditioning approach to prepare tumoricidal M1 Mφ for cancer immunotherapy.-Shan, S., Fang, B., Zhang, Y., Wang, C., Zhou, J., Niu, C., Gao, Y., Zhao, D., He, J., Wang, J., Zhang, X., Li, Q. Mechanical stretch promotes tumoricidal M1 polarization via the FAK/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Shengzhou Shan
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bin Fang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yifan Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chuandong Wang
- Department of Orthopedic Surgery, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jia Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chenguang Niu
- Department of Endodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ya Gao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Danyang Zhao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiahao He
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Wang
- Department of Otorhinolaryngology, Head and Neck Surgery, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoling Zhang
- Department of Orthopedic Surgery, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
39
|
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
|
40
|
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
|
41
|
Wang K, Xue Y, Peng Y, Pang X, Zhang Y, Ruiz-Ortega LI, Tian Y, Ngan AHW, Tang B. Elastic modulus and migration capability of drug treated leukemia cells K562. Biochem Biophys Res Commun 2019; 516:177-182. [PMID: 31204049 DOI: 10.1016/j.bbrc.2019.06.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 06/05/2019] [Indexed: 01/25/2023]
Abstract
Leukemia is a commonly seen disease caused by abnormal differentiation of hematopoietic stem cells and blasting in bone marrow. Despite drugs are used to treat the disease clinically, the influence of these drugs on leukemia cells' biomechanical properties, which are closely related to complications like leukostasis or infiltration, is still unclear. Due to non-adherent and viscoelastic nature of leukemia cells, accurate measurement of their elastic modulus is still a challenging issue. In this study, we adopted rate-jump method together with optical tweezers indentation to accurately measure elastic modulus of leukemia cells K562 after phorbol 12-myristate 13-acetate (PMA), all-trans retinoic acid (ATRA), Cytoxan (CTX), and Dexamethasone (DEX) treatment, respectively. We found that compared to control sample, K562 cells treated by PMA showed nearly a threefold increase in elastic modulus. Transwell experiment results suggested that the K562 cells treated with PMA have the lowest migration capability. Besides, it was shown that the cytoskeleton protein gene α-tubulin and vimentin have a significant increase in expression after PMA treatment by qPCR. The results indicate that PMA has a significant influence on protein expression, stiffness, and migration ability of the leukemia cell K562, and may also play an important role in the leukostasis in leukemia.
Collapse
Affiliation(s)
- Kui Wang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, PR China; Department of Mechanical Engineering, University of Hong Kong, Hong Kong, PR China
| | - Yuntian Xue
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, PR China
| | - Ying Peng
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, PR China
| | - Xiangchao Pang
- College of Materials Science and Engineering, Central South University of Forestry and Technology, Changsha, 410004, PR China
| | - Yuanjun Zhang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, PR China
| | - L I Ruiz-Ortega
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, PR China
| | - Ye Tian
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, PR China
| | - A H W Ngan
- Department of Mechanical Engineering, University of Hong Kong, Hong Kong, PR China
| | - Bin Tang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, PR China; Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, PR China.
| |
Collapse
|
42
|
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
|
43
|
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
|
44
|
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
|
45
|
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
|
46
|
Witherel CE, Abebayehu D, Barker TH, Spiller KL. Macrophage and Fibroblast Interactions in Biomaterial-Mediated Fibrosis. Adv Healthc Mater 2019; 8:e1801451. [PMID: 30658015 PMCID: PMC6415913 DOI: 10.1002/adhm.201801451] [Citation(s) in RCA: 166] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 12/07/2018] [Indexed: 01/08/2023]
Abstract
Biomaterial-mediated inflammation and fibrosis remain a prominent challenge in designing materials to support tissue repair and regeneration. Despite the many biomaterial technologies that have been designed to evade or suppress inflammation (i.e., delivery of anti-inflammatory drugs, hydrophobic coatings, etc.), many materials are still subject to a foreign body response, resulting in encapsulation of dense, scar-like extracellular matrix. The primary cells involved in biomaterial-mediated fibrosis are macrophages, which modulate inflammation, and fibroblasts, which primarily lay down new extracellular matrix. While macrophages and fibroblasts are implicated in driving biomaterial-mediated fibrosis, the signaling pathways and spatiotemporal crosstalk between these cell types remain loosely defined. In this review, the role of M1 and M2 macrophages (and soluble cues) involved in the fibrous encapsulation of biomaterials in vivo is investigated, with additional focus on fibroblast and macrophage crosstalk in vitro along with in vitro models to study the foreign body response. Lastly, several strategies that have been used to specifically modulate macrophage and fibroblast behavior in vitro and in vivo to control biomaterial-mediated fibrosis are highlighted.
Collapse
Affiliation(s)
- Claire E. Witherel
- Drexel University, School of Biomedical Engineering, Science and Health Systems, 3141 Chestnut Street, Philadelphia, Pennsylvania 19104, USA
| | - Daniel Abebayehu
- University of Virginia, Department of Biomedical Engineering, School of Engineering & School of Medicine, 415 Lane Road, Charlottesville, Virginia 22904, USA
| | - Thomas H. Barker
- University of Virginia, Department of Biomedical Engineering, School of Engineering & School of Medicine, 415 Lane Road, Charlottesville, Virginia 22904, USA
| | - Kara L. Spiller
- Drexel University, School of Biomedical Engineering, Science and Health Systems, 3141 Chestnut Street, Philadelphia, Pennsylvania 19104, USA,
| |
Collapse
|
47
|
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
|
48
|
Chandrasekharan A, Seong KY, Yim SG, Kim S, Seo S, Yoon J, Yang SY. In situ
photocrosslinkable hyaluronic acid-based surgical glue with tunable mechanical properties and high adhesive strength. ACTA ACUST UNITED AC 2018. [DOI: 10.1002/pola.29290] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- Ajeesh Chandrasekharan
- Department of Biomaterials Science, Life and Industry Convergence Institute; Pusan National University; Miryang 50463 Republic of Korea
| | - Keum-Yong Seong
- Department of Biomaterials Science, Life and Industry Convergence Institute; Pusan National University; Miryang 50463 Republic of Korea
| | - Sang-Gu Yim
- Department of Biomaterials Science, Life and Industry Convergence Institute; Pusan National University; Miryang 50463 Republic of Korea
| | - Sodam Kim
- Department of Biomaterials Science, Life and Industry Convergence Institute; Pusan National University; Miryang 50463 Republic of Korea
| | - Sungbaek Seo
- Department of Biomaterials Science, Life and Industry Convergence Institute; Pusan National University; Miryang 50463 Republic of Korea
| | - Jinhwan Yoon
- Department of Chemistry Education; Graduate Department of Chemical Materials, Pusan National University; Busan 46241 Republic of Korea
| | - Seung Yun Yang
- Department of Biomaterials Science, Life and Industry Convergence Institute; Pusan National University; Miryang 50463 Republic of Korea
| |
Collapse
|
49
|
Cui Y, Zhou F, Bai H, Wei L, Tan J, Zeng Z, Song Q, Chen J, Huang N. Real-time QCM-D monitoring of endothelial cells and macrophages adhering and spreading to SEMA4D/heparin surfaces. Colloids Surf B Biointerfaces 2018; 171:522-529. [DOI: 10.1016/j.colsurfb.2018.07.062] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 07/05/2018] [Accepted: 07/26/2018] [Indexed: 01/25/2023]
|
50
|
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]
|