Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Biomacromolecules ; 19(7): 2880-2888, 2018 07 09.
Article in English | MEDLINE | ID: mdl-29698603

ABSTRACT

Reducing the foreign body response (FBR) to implanted biomaterials will enhance their performance in tissue engineering. Poly(ethylene glycol) (PEG) hydrogels are increasingly popular for this application due to their low cost, ease of use, and the ability to tune their compliance via molecular weight and cross-linking densities. PEG hydrogels can elicit chronic inflammation in vivo, but recent evidence has suggested that extremely hydrophilic, zwitterionic materials and particles can evade the immune system. To combine the advantages of PEG-based hydrogels with the hydrophilicity of zwitterions, we synthesized hydrogels with comonomers PEG and the zwitterion phosphorylcholine (PC). Recent evidence suggests that stiff hydrogels elicit increased immune cell adhesion to hydrogels, which we attempted to reduce by increasing hydrogel hydrophilicity. Surprisingly, hydrogels with the highest amount of zwitterionic comonomer elicited the highest FBR. Lowering the hydrogel modulus (165 to 3 kPa), or PC content (20 to 0 wt %), mitigated this effect. A high density of macrophages was found at the surface of implants associated with a high FBR, and mass spectrometry analysis of the proteins adsorbed to these gels implicated extracellular matrix, immune response, and cell adhesion protein categories as drivers of macrophage recruitment. Overall, we show that modulus regulates macrophage adhesion to zwitterionic-PEG hydrogels, and demonstrate that chemical modifications to hydrogels should be studied in parallel with their physical properties to optimize implant design.


Subject(s)
Foreign-Body Reaction/prevention & control , Hydrogels/chemistry , Phosphorylcholine/analogs & derivatives , Polyethylene Glycols/chemistry , Animals , Cell Adhesion , Cells, Cultured , Hydrogels/pharmacology , Hydrophobic and Hydrophilic Interactions , Macrophages/drug effects , Macrophages/physiology , Male , Mice , Mice, Inbred C57BL
2.
Article in English | MEDLINE | ID: mdl-30479632

ABSTRACT

The foreign body response (FBR) occurs ubiquitously to essentially all non-biological materials that are implanted into higher organisms. The FBR is characterized by inflammation followed by fibrosis and is mediated largely by macrophages. While many current medical devices tolerate the FBR, the FBR is responsible for many asceptic device failures and is hindering advancements of new devices that rely on device-host communication to function. To this end, in vitro and in vivo models are critical to studying how a biomaterial, via its chemistry and properties, affect the FBR. This short review highlights the main in vitro and in vivo models that are used to study the FBR. In vitro models that capture macrophage interrogation of a biomaterial and evaluation of macrophage attachment, polarization and fusion are described. In vivo models using rodents, which provide a relatively simple model of the complex FBR process, and human-relevant nonhuman primate models are described. Collectively, the combination of in vitro and in vivo models will help advance our fundmental understanding of the FBR and enable new biomaterials to be developed that can effectively modulate the FBR to achieve a desire device-host outcome.

3.
Gels ; 9(2)2023 Feb 20.
Article in English | MEDLINE | ID: mdl-36826339

ABSTRACT

Biologic therapies have revolutionized treatment options for rheumatoid arthritis (RA) but their continuous administration at high doses may lead to adverse events. Thus, the development of improved drug delivery systems that can sense and respond commensurately to disease flares represents an unmet medical need. Toward this end, we generated induced pluripotent stem cells (iPSCs) that express interleukin-1 receptor antagonist (IL-1Ra, an inhibitor of IL-1) in a feedback-controlled manner driven by the macrophage chemoattractant protein-1 (Ccl2) promoter. Cells were seeded in agarose hydrogel constructs made from 3D printed molds that can be injected subcutaneously via a blunt needle, thus simplifying implantation of the constructs, and the translational potential. We demonstrated that the subcutaneously injected agarose hydrogels containing genome-edited Ccl2-IL1Ra iPSCs showed significant therapeutic efficacy in the K/BxN model of inflammatory arthritis, with nearly complete abolishment of disease severity in the front paws. These implants also exhibited improved implant longevity as compared to the previous studies using 3D woven scaffolds, which require surgical implantation. This minimally invasive cell-based drug delivery strategy may be adapted for the treatment of other autoimmune or chronic diseases, potentially accelerating translation to the clinic.

4.
Adv Healthc Mater ; 11(9): e2102209, 2022 05.
Article in English | MEDLINE | ID: mdl-34967497

ABSTRACT

Poly(ethylene glycol) (PEG) hydrogels hold promise for in vivo applications but induce a foreign body response (FBR). While macrophages are key in the FBR, many questions remain. This study investigates temporal changes in the transcriptome of implant-associated monocytes and macrophages. Proinflammatory pathways are upregulated in monocytes compared to control monocytes but subside by day 28. Macrophages are initially proinflammatory but shift to a profibrotic state by day 14, coinciding with fibrous capsule emergence. Next, this study assesses the origin of macrophages responsible for fibrous encapsulation using wildtype, C-C Motif Chemokine Receptor 2 (CCR2)-/- mice that lack recruited macrophages, and Macrophage Fas-Induced Apoptosis (MaFIA) mice that enable macrophage ablation. Subpopulations of recruited and tissue-resident macrophages are identified. Fibrous encapsulation proceeds in CCR2-/- mice similar to wildtype mice. However, studies in MaFIA mice indicate that macrophages are necessary for fibrous capsule formation. These findings suggest that macrophage origin impacts the FBR progression and provides evidence that tissue-resident macrophages and not the recruited macrophages may drive fibrosis in the FBR to PEG hydrogels. This study demonstrates that implant-associated monocytes and macrophages have temporally distinct transcriptomes in the FBR and that profibrotic pathways associated with macrophages may be enriched in tissue-resident macrophages.


Subject(s)
Foreign Bodies , Macrophage Activation , Animals , Biocompatible Materials/metabolism , Fibrosis , Foreign Bodies/metabolism , Hydrogels/metabolism , Hydrogels/pharmacology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Polyethylene Glycols/metabolism , Polyethylene Glycols/pharmacology
5.
ACS Biomater Sci Eng ; 6(5): 2668-2681, 2020 05 11.
Article in English | MEDLINE | ID: mdl-33463295

ABSTRACT

The foreign body response (FBR) has impaired progress of new implantable medical devices through its hallmark of chronic inflammation and foreign body giant cell (FBGC) formation leading to fibrous encapsulation. Macrophages are known to drive the FBR, but efforts to control macrophage polarization remain challenging. The goal for this study was to investigate whether prostaglandin E2 (PGE2), and specifically its receptors EP2 and/or EP4, attenuate classically activated (i.e., inflammatory) macrophages and macrophage fusion into FBGCs in vitro. Lipopolysaccharide (LPS)-stimulated macrophages exhibited a dose-dependent decrease in gene expression and protein production of tumor necrosis factor alpha (TNF-α) when treated with PGE2. This attenuation was primarily by the EP4 receptor, as the addition of the EP2 antagonist PF 04418948 to PGE2-treated LPS-stimulated cells did not recover TNF-α production while the EP4 antagonist ONO AE3 208 did. However, direct stimulation of EP2 with the agonist butaprost to LPS-stimulated macrophages resulted in a ∼60% decrease in TNF-α secretion after 4 h and corresponded with an increase in gene expression for Cebpb and Il10, suggesting a polarization shift toward alternative activation through EP2 alone. Further, fusion of macrophages into FBGCs induced by interleukin-4 (IL-4) and granulocyte-macrophage colony-stimulating factor (GM-CSF) was inhibited by PGE2 via EP2 signaling and by an EP2 agonist, but not an EP4 agonist. The attenuation by PGE2 was confirmed to be primarily by the EP2 receptor. Mrc1, Dcstamp, and Retlna expressions increased upon IL-4/GM-CSF stimulation, but only Retnla expression with the EP2 agonist returned to levels that were not different from controls. This study identified that PGE2 attenuates classically activated macrophages and macrophage fusion through distinct EP receptors, while targeting EP2 is able to attenuate both. In summary, this study identified EP2 as a potential therapeutic target for reducing the FBR to biomaterials.


Subject(s)
Dinoprostone , Receptors, Prostaglandin E, EP2 Subtype , Lipopolysaccharides/pharmacology , Macrophage Activation , Receptors, Prostaglandin E, EP4 Subtype
6.
Acta Biomater ; 100: 105-117, 2019 12.
Article in English | MEDLINE | ID: mdl-31568879

ABSTRACT

Synthetic hydrogels, such as poly(ethylene glycol) (PEG), are promising for a range of in vivo applications. However, like all non-biological biomaterials, synthetic hydrogels including PEG elicit a foreign body response (FBR). The FBR is thought to be initiated by adsorbed protein that is recognized by and subsequently activates inflammatory cells, notably macrophages, and culminates with fibrotic encapsulation. However, the molecular mechanisms that drive the FBR are not well understood. Toll-like receptors (TLRs) are key receptors that recognize pathogens, but also recognize altered host proteins that display damage-associated molecular patterns (DAMPs). Thus TLRs may play a role in the FBR. Here, we investigated myeloid differentiation primary response gene 88 (MyD88), a signaling adaptor protein that mediates inflammatory cytokine production induced by most TLRs. An in vitro model was used consisting of macrophages cultured on the surface of synthetic hydrogels, specifically PEG, with pre-adsorbed serum proteins. Our in vitro findings demonstrate that MyD88-dependent signaling is the predominant inflammatory pathway in macrophage activation to synthetic hydrogels. When stimulated with TLR agonists to mimic additional DAMPs present in vivo, MyD88-dependent signaling was also the predominant pathway in macrophage activation. An in vivo model of PEG hydrogels implanted subcutaneously in wild-type and MyD88-/- mice also demonstrated that MyD88 is the key contributor to the recruitment of inflammatory cells and formation of the fibrous capsule surrounding the implanted hydrogel. Taken together, findings from this study identify MyD88-mediated inflammation as being a critical pathway involved not only in the inflammatory response, but in formation of the fibrous capsule to PEG hydrogels. STATEMENT OF SIGNIFICANCE: Synthetic hydrogels are promising for in vivo applications but, like all non-biological biomaterials, synthetic hydrogels elicit a foreign body response (FBR). The molecular mechanisms that drive the FBR are not well understood. This work identifies the myeloid differentiation primary response gene 88 (MyD88) as a central mediator to macrophage activation in response to a poly(ethylene glycol) hydrogel with pre-adsorbed proteins in vitro. Moreover, MyD88 was also central to the recruitment of inflammatory cells, which included neutrophils, monocytes, and macrophages, to implanted PEG hydrogels and to fibrous encapsulation. These findings demonstrate that MyD88-mediated inflammation is responsible in part for the formation of the fibrous capsule of the FBR.


Subject(s)
Hydrogels/adverse effects , Implants, Experimental/adverse effects , Inflammation/pathology , Myeloid Differentiation Factor 88/metabolism , Polyethylene Glycols/adverse effects , Signal Transduction , Alarmins/metabolism , Animals , Fibrosis , Foreign-Body Reaction/chemically induced , Foreign-Body Reaction/pathology , Macrophage Activation , Male , Mice, Inbred C57BL
7.
ACS Appl Bio Mater ; 2(11): 4698-4702, 2019 Nov 18.
Article in English | MEDLINE | ID: mdl-35021468

ABSTRACT

We show that protein unfolding on biomaterials may be dramatically reduced via tuning the chemical heterogeneity of the protein-material interface. Specifically, using dynamic single-molecule methods, we confirmed that the transient structure and dynamics of fibronectin (FN) may be mediated through varying the composition of random copolymer brushes. The brushes, which themselves represent an intriguing biomaterial, were composed of oligoethylene glycol and sulfobetaine methacrylate and presumably stabilized FN through partitioning and/or segregation of the copolymers. We further showed that, by controlling the transient structure and dynamics of FN, the secretion of TNF-α and IL-6 by RAW 264.7 was markedly diminished.

8.
Curr Opin Biomed Eng ; 6: 58-65, 2018 Jun.
Article in English | MEDLINE | ID: mdl-30374467

ABSTRACT

Implantation of cell-laden scaffolds is a promising strategy for regenerating tissue that has been damaged due to injury or disease. However, the act of implantation initiates an acute inflammatory response. If the scaffold is non-biologic (i.e., a modified biologic scaffold or synthetic-based scaffold), inflammation will be prolonged through the foreign body response (FBR), which eventually forms a fibrous capsule and walls off the implant from the surrounding host tissue. This host response, from a cellular perspective, can create a harsh environment leading to long-lasting effects on the tissue engineering outcome. At the same time, cells embedded within the scaffold can respond to this environment and influence the interrogating immune cells (e.g., macrophages). This crosstalk, depending on the type of cell, can dramatically influence the host response. This review provides an overview of the FBR and highlights important and recent advancements in the host response to cell-laden scaffolds with a focus on the impact of the communication between immune cells and cells embedded within a scaffold. Understanding this complex interplay between the immune cells, notably macrophages, and the tissue engineering cells is a critically important component to a successful in vivo tissue engineering therapy.

9.
Acta Biomater ; 71: 37-48, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29505890

ABSTRACT

Poly(ethylene glycol) PEG-based hydrogels are promising for cell encapsulation and tissue engineering, but are known to elicit a foreign body response (FBR) in vivo. The goal of this study was to investigate the impact of the FBR, and specifically the presence of inflammatory macrophages, on encapsulated cells and their ability to synthesize new extracellular matrix. This study employed an in vitro co-culture system with murine macrophages and MC3T3-E1 pre-osteoblasts encapsulated in a bone-mimetic hydrogel, which were cultured in transwell inserts, and exposed to an inflammatory stimulant, lipopolysaccharide (LPS). The co-culture was compared to mono-cultures of the cell-laden hydrogels alone and with LPS over 28 days. Two macrophage cell sources, RAW 264.7 and primary derived, were investigated. The presence of LPS-stimulated primary macrophages led to significant changes in the cell-laden hydrogel by a 5.3-fold increase in percent apoptotic osteoblasts at day 28, 4.2-fold decrease in alkaline phosphatase activity at day 10, and 7-fold decrease in collagen deposition. The presence of LPS-stimulated RAW macrophages led to significant changes in the cell-laden hydrogel by 5-fold decrease in alkaline phosphatase activity at day 10 and 4-fold decrease in collagen deposition. Mineralization, as measured by von Kossa stain or quantified by calcium content, was not sensitive to macrophages or LPS. Elevated interleukin-6 and tumor necrosis factor-α secretion were detected in mono-cultures with LPS and co-cultures. Overall, primary macrophages had a more severe inhibitory effect on osteoblast differentiation than the macrophage cell line, with greater apoptosis and collagen I reduction. In summary, this study highlights the detrimental effects of macrophages on encapsulated cells for bone tissue engineering. STATEMENT OF SIGNIFICANCE: Poly(ethylene glycol) (PEG)-based hydrogels are promising for cell encapsulation and tissue engineering, but are known to elicit a foreign body response (FBR) in vivo. The impact of the FBR on encapsulated cells and their ability to synthesize tissue has not been well studied. This study utilizes thiol-ene click chemistry to create a biomimetic, enzymatically degradable hydrogel system with which to encapsulate MC3T3-E1 pre-osteoblasts. The osteogenic capabilities and differentiation of these cellswerestudied in co-culture with macrophages, known drivers of the FBR.This study demonstrates that macrophages reduce osteogenic capabilities of encapsulated cellsin vitroand suggestthat the FBR should be considered for in vivo tissue engineering.


Subject(s)
Biomimetic Materials/chemistry , Hydrogels/chemistry , Macrophages/metabolism , Osteoblasts/metabolism , Osteogenesis , Polyethylene Glycols/chemistry , Animals , Coculture Techniques , Macrophages/cytology , Mice , Osteoblasts/cytology , RAW 264.7 Cells
10.
Biomed Mater ; 13(4): 045009, 2018 05 02.
Article in English | MEDLINE | ID: mdl-29611815

ABSTRACT

This study investigated the effects of introducing hydroxyapatite nanoparticles into a matrix metalloproteinase (MMP) sensitive poly(ethylene glycol) (PEG) hydrogel containing cell adhesion peptides of RGD for bone tissue engineering. MC3T3-E1 pre-osteoblasts were encapsulated in the biomimetic PEG hydrogel, which was formed from the photoclick thiol-norbornene reaction system, cultured for up to 28 d in growth medium or osteogenic differentiation medium, and evaluated by cellular morphology and differentiation by alkaline phosphatase (ALP) activity and bone-like extracellular matrix deposition for mineral and collagen. Hydroxyapatite nanoparticles were incorporated during hydrogel formation and cell encapsulation at 0%, 0.1% or 1% (w/w). Incorporation of hydroxyapatite nanoparticles did not affect the hydrogel properties as measured by compressive modulus and equilibrium swelling. In growth medium, encapsulated MC3T3-E1 cells remained largely round regardless of hydroxyapatite concentration. ALP activity increased by 25% at day 14 and total collagen content increased by 55% at day 28 with increasing hydroxyapatite concentration from 0% to 1%. In differentiation medium, cell spreading was evident regardless of hydroxyapatite indicating that the MC3T3-E1 cells were able to degrade the hydrogel. For the 1% hydroxyapatite condition, ALP activity was 27% higher at day 14 and total collagen content was 22% higher at day 28 in differentiation medium when compared to growth medium. Mineral deposits were more abundant and spatial elaboration of collagen type I was more evident in the 1% (w/w) hydroxyapatite condition with differentiation medium when compared to all other conditions. Overall, osteogenesis was observed in the hydrogels with hydroxyapatite nanoparticles in growth medium but was enhanced in differentiation medium. In summary, a biomimetic hydrogel comprised of MMP-sensitive crosslinks, RGD cell adhesion peptides, and 1% (w/w) hydroxyapatite nanoparticles is promising for bone tissue engineering.


Subject(s)
Durapatite/chemistry , Matrix Metalloproteinases/chemistry , Metal Nanoparticles/chemistry , Osteoblasts/cytology , Polyethylene Glycols/chemistry , Tissue Engineering/methods , 3T3 Cells , Alkaline Phosphatase/metabolism , Animals , Biocompatible Materials , Bone Substitutes/chemistry , Cell Adhesion , Cell Differentiation , Cell Survival , Cells, Cultured , Collagen/chemistry , Culture Media , Extracellular Matrix/metabolism , Hydrogels/chemistry , Ligands , Mice , Norbornanes/chemistry , Oligopeptides/chemistry , Osteoblasts/metabolism , Osteogenesis , Stress, Mechanical , Sulfhydryl Compounds , Tissue Scaffolds/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL