Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 195
Filtrar
1.
Front Immunol ; 15: 1411872, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39034997

RESUMO

During the foreign body response (FBR), macrophages fuse to form foreign body giant cells (FBGCs). Modulation of FBGC formation can prevent biomaterial degradation and loss of therapeutic efficacy. However, the microenvironmental cues that dictate FBGC formation are poorly understood with conflicting reports. Here, we identified molecular and cellular factors involved in driving FBGC formation in vitro. Macrophages demonstrated distinct fusion competencies dependent on monocyte differentiation. The transition from a proinflammatory to a reparative microenvironment, characterised by specific cytokine and growth factor programmes, accompanied FBGC formation. Toll-like receptor signalling licensed the formation of FBGCs containing more than 10 nuclei but was not essential for cell-cell fusion to occur. Moreover, the fibroblast-macrophage crosstalk influenced FBGC development, with the fibroblast secretome inducing macrophages to secrete more PDGF, which enhanced large FBGC formation. These findings advance our understanding as to how a specific and timely combination of cellular and microenvironmental factors is required for an effective FBR, with monocyte differentiation and fibroblasts being key players.


Assuntos
Diferenciação Celular , Fusão Celular , Microambiente Celular , Fibroblastos , Reação a Corpo Estranho , Células Gigantes de Corpo Estranho , Macrófagos , Macrófagos/metabolismo , Macrófagos/imunologia , Reação a Corpo Estranho/imunologia , Fibroblastos/metabolismo , Humanos , Células Gigantes de Corpo Estranho/metabolismo , Células Gigantes de Corpo Estranho/patologia , Animais , Monócitos/imunologia , Monócitos/metabolismo , Camundongos , Citocinas/metabolismo , Transdução de Sinais , Células Cultivadas
2.
ACS Biomater Sci Eng ; 10(5): 3006-3016, 2024 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-38640484

RESUMO

Soft implantable devices are crucial to optimizing form and function for many patients. However, periprosthetic capsule fibrosis is one of the major challenges limiting the use of implants. Currently, little is understood about how spatial and temporal factors influence capsule physiology and how the local capsule environment affects the implant structure. In this work, we analyzed breast implant capsule specimens with staining, immunohistochemistry, and real-time polymerase chain reaction to investigate spatiotemporal differences in inflammation and fibrosis. We demonstrated that in comparison to the anterior capsule against the convex surface of breast implants, the posterior capsule against the flat surface of the breast implant displays several features of a dysregulated foreign body reaction including increased capsule thickness, abnormal extracellular remodeling, and infiltration of macrophages. Furthermore, the expression of pro-inflammatory cytokines increased in the posterior capsule across the lifespan of the device, but not in the anterior capsule. We also analyzed the surface oxidation of breast explant samples with XPS analysis. No significant differences in surface oxidation were identified either spatially or temporally. Collectively, our results support spatiotemporal heterogeneity in inflammation and fibrosis within the breast implant capsule. These findings presented here provide a more detailed picture of the complexity of the foreign body reaction surrounding implants destined for human use and could lead to key research avenues and clinical applications to treat periprosthetic fibrosis and improve device longevity.


Assuntos
Implantes de Mama , Fibrose , Reação a Corpo Estranho , Propriedades de Superfície , Implantes de Mama/efeitos adversos , Humanos , Reação a Corpo Estranho/patologia , Reação a Corpo Estranho/metabolismo , Reação a Corpo Estranho/imunologia , Feminino , Silicones/química , Géis de Silicone/efeitos adversos , Citocinas/metabolismo , Inflamação/patologia , Inflamação/metabolismo , Macrófagos/metabolismo , Macrófagos/imunologia
3.
Nat Commun ; 12(1): 5327, 2021 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-34493717

RESUMO

Implantation-caused foreign-body response (FBR) is a commonly encountered issue and can result in failure of implants. The high L-serine content in low immunogenic silk sericin, and the high D-serine content as a neurotransmitter together inspire us to prepare poly-DL-serine (PSer) materials in mitigating the FBR. Here we report highly water soluble, biocompatible and easily accessible PSer hydrogels that cause negligible inflammatory response after subcutaneous implantation in mice for 1 week and 2 weeks. No obvious collagen capsulation is found surrounding the PSer hydrogels after 4 weeks, 3 months and 7 months post implantation. Histological analysis on inflammatory cytokines and RNA-seq assay both indicate that PSer hydrogels show low FBR, comparable to the Mock group. The anti-FBR performance of PSer hydrogels at all time points surpass the poly(ethyleneglycol) hydrogels that is widely utilized as bio-inert materials, implying the potent and wide application of PSer materials in implantable biomaterials and biomedical devices.


Assuntos
Materiais Biocompatíveis/farmacologia , Reação a Corpo Estranho/prevenção & controle , Peptídeos/farmacologia , Próteses e Implantes , Animais , Materiais Biocompatíveis/síntese química , Citocinas/antagonistas & inibidores , Citocinas/biossíntese , Citocinas/imunologia , Reação a Corpo Estranho/imunologia , Hidrogéis , Infusões Subcutâneas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Peptídeos/síntese química , Polietilenoglicóis/farmacologia , Solubilidade , Água/química
4.
Nat Rev Urol ; 18(12): 725-738, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34545239

RESUMO

Polypropylene (PPL) mesh is widely used in pelvic floor reconstructive surgery for prolapse and stress urinary incontinence. However, some women, particularly those treated using transvaginal PPL mesh placement for prolapse, experience intractable pain and mesh exposure or extrusion. Explanted tissue from patients with complications following transvaginal implantation of mesh is typified by a dense fibrous capsule with an immune cell-rich infiltrate, suggesting that the host immune response has a role in transvaginal PPL mesh complications through the separate contributions of the host (patient), the biological niche within which the material is implanted and biomaterial properties of the mesh. This immune response might be strongly influenced by both the baseline inflammatory status of the patient, surgical technique and experience, and the unique hormonal, immune and microbial tissue niche of the vagina. Mesh porosity, surface area and stiffness also might have an effect on the immune and tissue response to transvaginal mesh placement. Thus, a regulatory pathway is needed for mesh development that recognizes the roles of host and biological factors in driving the immune response to mesh, as well as mandatory mesh registries and the longitudinal surveillance of patients.


Assuntos
Materiais Biocompatíveis/efeitos adversos , Reação a Corpo Estranho/etiologia , Prolapso de Órgão Pélvico/cirurgia , Polipropilenos/efeitos adversos , Complicações Pós-Operatórias/etiologia , Telas Cirúrgicas/efeitos adversos , Incontinência Urinária por Estresse/cirurgia , Feminino , Reação a Corpo Estranho/imunologia , Reação a Corpo Estranho/prevenção & controle , Procedimentos Cirúrgicos em Ginecologia/efeitos adversos , Procedimentos Cirúrgicos em Ginecologia/instrumentação , Humanos , Complicações Pós-Operatórias/imunologia , Complicações Pós-Operatórias/prevenção & controle , Fatores de Risco , Procedimentos Cirúrgicos Urológicos/efeitos adversos , Procedimentos Cirúrgicos Urológicos/instrumentação
5.
Cells ; 10(7)2021 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-34359963

RESUMO

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.


Assuntos
Fibroblastos/transplante , Reação a Corpo Estranho/imunologia , Miofibroblastos/citologia , Próteses e Implantes , Reação a Corpo Estranho/metabolismo , Humanos , Macrófagos/metabolismo , Mecanotransdução Celular/imunologia , Miofibroblastos/imunologia
6.
Nat Commun ; 12(1): 3256, 2021 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-34059671

RESUMO

Macrophages perform diverse functions within tissues during immune responses to pathogens and injury, but molecular mechanisms by which physical properties of the tissue regulate macrophage behavior are less well understood. Here, we examine the role of the mechanically activated cation channel Piezo1 in macrophage polarization and sensing of microenvironmental stiffness. We show that macrophages lacking Piezo1 exhibit reduced inflammation and enhanced wound healing responses. Additionally, macrophages expressing the transgenic Ca2+ reporter, Salsa6f, reveal that Ca2+ influx is dependent on Piezo1, modulated by soluble signals, and enhanced on stiff substrates. Furthermore, stiffness-dependent changes in macrophage function, both in vitro and in response to subcutaneous implantation of biomaterials in vivo, require Piezo1. Finally, we show that positive feedback between Piezo1 and actin drives macrophage activation. Together, our studies reveal that Piezo1 is a mechanosensor of stiffness in macrophages, and that its activity modulates polarization responses.


Assuntos
Materiais Biocompatíveis/efeitos adversos , Reação a Corpo Estranho/imunologia , Canais Iônicos/metabolismo , Macrófagos/imunologia , Cicatrização/imunologia , Actinas/metabolismo , Animais , Células Cultivadas , Microambiente Celular/imunologia , Modelos Animais de Doenças , Retroalimentação Fisiológica , Feminino , Humanos , Canais Iônicos/genética , Ativação de Macrófagos , Macrófagos/metabolismo , Masculino , Mecanotransdução Celular/imunologia , Camundongos , Cultura Primária de Células , Tela Subcutânea/cirurgia
7.
Histol Histopathol ; 36(9): 899-906, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-33834451

RESUMO

BACKGROUND: Peri-miniscrew implant is a temporary assistant armamentarium for the treatment of severe malocclusion and complex tooth movement, the inflammation around it is the main reason for the failure of orthodontic treatment due to the implant loosening and falling out. Inflammation around the peri-miniscrew implant is associated with the release of pro-inflammatory cytokines. These pro-inflammatory cytokines, in turn, recruit immune cells (such as macrophages, dendritic cells, T cells, and B cells), which can produce and release inflammatory biomarkers, regulate the interaction between immune cells, periodontal ligament cells, osteoblasts, and so on. However, there is currently no effective clinical treatment plan to prevent inflammation around implants. PURPOSE: To investigate the potentially essential factors in the inflammatory response around the peri-miniscrew implant and explore the signaling pathways involved. METHODS: Here, we review the studies focused on inflammatory biomarkers (Interleukins, tumor necrosis factor-α (TNF-α), receptor activator of NF-κB ligand (RANKL), matrix metalloproteinases (MMPs), and cellular adhesion molecules (CAMs)) in peri-miniscrew implant crevicular fluid (PMICF), as well as inflammatory signaling pathways (Wnt5a, JNK, Erk1/2, NF-κBp65 and TAB/TAK) in periodontal cells from 1998 to 2020. RESULTS: A literature search revealed TLR-2, TLR-4, LOX-1, and BMPs are involved in regulating ILs (IL-1ß, IL-6, IL-8, and IL-17), TNF-α, RANKL, MMP-2, MMP-9 expression via JNK, Erk1/2, Wnt5a, NF-κBp65, OPN, and TAB/TAK signaling pathways. Among them, IL-1ß and IL-6 are the critical inflammation factors in the signaling pathways inducing the inflammatory reaction surrounding implants. Besides, CAM-1 was also regulated by MMP-9 and IL-17. CONCLUSION: There are considerable potential factors involving regulating inflammatory biomarkers on downstream signaling pathways in peri-minisrew implant crevicular fluid. CLINICAL SIGNIFICANCE: This review provides the substantiation of these cell factors and signaling pathways around peri-miniscrew implants, proposes more practical clinical therapeutic ideas and schemes for improving the stability and clinical efficacy of peri-miniscrew implants.


Assuntos
Parafusos Ósseos/efeitos adversos , Reação a Corpo Estranho/metabolismo , Líquido do Sulco Gengival/metabolismo , Mediadores da Inflamação/metabolismo , Inflamação/metabolismo , Aparelhos Ortodônticos/efeitos adversos , Peri-Implantite/metabolismo , Técnicas de Movimentação Dentária/instrumentação , Animais , Reação a Corpo Estranho/imunologia , Reação a Corpo Estranho/patologia , Líquido do Sulco Gengival/imunologia , Humanos , Inflamação/imunologia , Inflamação/patologia , Peri-Implantite/imunologia , Peri-Implantite/patologia , Transdução de Sinais , Resultado do Tratamento
8.
J Tissue Eng Regen Med ; 15(1): 24-36, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33217150

RESUMO

Implanted porous precision templated scaffolds (PTS) with 40-µm spherical pores reduce inflammation and foreign body reaction (FBR) while increasing vascular density upon implantation. Larger or smaller pores, however, promote chronic inflammation and FBR. While macrophage (MØ) recruitment and polarization participates in perpetuating this pore-size-mediated phenomenon, the driving mechanism of this unique pro-healing response is poorly characterized. We hypothesized that the primarily myeloid PTS resident cells release small extracellular vesicles (sEVs) that induce pore-size-dependent pro-healing effects in surrounding T cells. Upon profiling resident immune cells and their sEVs from explanted 40-µm- (pro-healing) and 100-µm-pore diameter (inflammatory) PTS, we found that PTS pore size did not affect PTS resident immune cell population ratios or the proportion of myeloid sEVs generated from explanted PTS. However, quantitative transcriptomic assessment indicated cell and sEV phenotype were pore size dependent. In vitro experiments demonstrated the ability of PTS cell-derived sEVs to stimulate T cells transcriptionally and proliferatively. Specifically, sEVs isolated from cells inhabiting explanted 100 µm PTS significantly upregulated Th1 inflammatory gene expression in immortalized T cells. sEVs isolated from cell inhabiting both 40- and 100-µm PTS upregulated essential Treg transcriptional markers in both primary and immortalized T cells. Finally, we investigated the effects of Treg depletion on explanted PTS resident cells. FoxP3+ cell depletion suggests Tregs play a unique role in balancing T cell subset ratios, thus driving host response in 40-µm PTS. These results indicate that predominantly 40-µm PTS myeloid cell-derived sEVs affect T cells through a distinct, pore-size-mediated modality.


Assuntos
Comunicação Celular/imunologia , Vesículas Extracelulares/imunologia , Macrófagos/imunologia , Linfócitos T Reguladores/imunologia , Células Th1/imunologia , Alicerces Teciduais/química , Cicatrização/imunologia , Animais , Reação a Corpo Estranho/imunologia , Reação a Corpo Estranho/prevenção & controle , Camundongos , Camundongos Transgênicos , Porosidade
9.
Acta Orthop ; 91(6): 782-788, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32691656

RESUMO

Background and purpose - Biological patches can be used to augment rotator cuff tendon repair in an attempt to improve healing and reduce rates of re-rupture. However, little is known about the in vivo tissue response to these patches. We assessed native rotator cuff tissue response after surgical repair and augmentation with 2 commercially available extracellular matrix (ECM) patches. Patients and methods - Patients underwent a rotator cuff repair augmented with either GraftJacket (Wright Medical), Permacol (Zimmer Biomet), or no patch (Control), applied using an onlay technique. A sample of supraspinatus tendon was collected intraoperatively and 4 weeks post-surgery, using ultrasound-guided biopsy. Histology and immunohistochemistry were performed on all samples. Results - The Permacol group (n = 3) and GraftJacket group (n = 4) demonstrated some changes in native tendon ECM compared with the control group (n = 3). Significant disruption of the extracellular matrix of the repaired native supraspinatus, underlying both patches, was observed. The patches did not generally increase cellularity, foreign body giant cell count, or vascularity compared to the control group. 1 patient in the Permacol group had an adverse tissue immune response characterized by extensive infiltration of IRF5+, CD68+, and CD206+ cells, suggesting involvement of macrophages with a pro-inflammatory phenotype. No significant differences in protein expression of CD4, CD45, CD68, CD206, BMP7, IRF5, TGFß, and PDPN were observed among the groups. Interpretation - Histological and immunohistochemical analysis of native tendon tissue after patch augmentation in rotator cuff repair raises some concerns about a lack of benefit and potential for harm from these materials.


Assuntos
Artroplastia , Colágeno , Biópsia Guiada por Imagem/métodos , Inflamação , Teste de Materiais/métodos , Lesões do Manguito Rotador/cirurgia , Manguito Rotador , Artroplastia/efeitos adversos , Artroplastia/instrumentação , Artroplastia/métodos , Materiais Biocompatíveis/efeitos adversos , Materiais Biocompatíveis/uso terapêutico , Colágeno/efeitos adversos , Colágeno/uso terapêutico , Feminino , Reação a Corpo Estranho/etiologia , Reação a Corpo Estranho/imunologia , Reação a Corpo Estranho/patologia , Humanos , Inflamação/etiologia , Inflamação/imunologia , Inflamação/patologia , Imageamento por Ressonância Magnética/métodos , Masculino , Pessoa de Meia-Idade , Avaliação de Processos e Resultados em Cuidados de Saúde , Manguito Rotador/irrigação sanguínea , Manguito Rotador/imunologia , Ultrassonografia/métodos , Reino Unido , Cicatrização/imunologia
10.
Sci Rep ; 10(1): 10348, 2020 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-32587271

RESUMO

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.


Assuntos
Materiais Biocompatíveis/efeitos adversos , Adesão Celular/imunologia , Reação a Corpo Estranho/imunologia , Macrófagos/imunologia , Alicerces Teciduais/efeitos adversos , Animais , Materiais Biocompatíveis/administração & dosagem , Colágeno/administração & dosagem , Colágeno/efeitos adversos , Modelos Animais de Doenças , Humanos , Masculino , Teste de Materiais/métodos , Camundongos , Cultura Primária de Células/métodos , Células RAW 264.7 , Ratos
11.
Tissue Eng Part B Rev ; 25(6): 492-499, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31436142

RESUMO

Mesenchymal stem cells (MSCs) have great therapeutic potential for tissue engineering and regenerative medicine due to their multipotency and paracrine functions. However, shortly after in vivo implantation, MSCs tend to migrate to the lungs and undergo apoptosis, which impairs their clinical efficacy. In addition, the ex vivo two-dimensional expansion of MSCs results in changes in their immunophenotype and functional activities compared to those in vivo. The use of biomaterials to culture and deliver MSCs has the potential to overcome these limitations. MSC-biomaterial constructs retain MSCs in situ and prolong their survival, while the MSCs ameliorate the foreign body reaction and fibrosis caused by the biomaterial. Biomaterial scaffolds can both preserve the tissue architecture and provide a three-dimensional biomimetic milieu for embedded MSCs, which enhance their paracrine functions, including their immunomodulatory potential. The dimensionality, physical characteristics, topographical cues, biochemistry, and microstructure can enhance the immunomodulatory potential of MSCs. Here, we review the link between the properties of biomaterial and the immunomodulatory potential of MSCs. Impact Statement Regeneration of cells, tissues, and whole organs is challenging. Mesenchymal stem cells (MSCs) have therapeutic potential in tissue engineering and regenerative medicine due to their paracrine functions, including immunomodulatory activity. The dimensionality, physical characteristics, topographical cues, biochemistry, and microstructure of biomaterial can be harnessed to enhance the immunomodulatory potential of MSCs for tissue engineering, which will increase their clinical efficacy, particularly for immune-related diseases.


Assuntos
Materiais Biocompatíveis/farmacologia , Reação a Corpo Estranho/imunologia , Imunomodulação/efeitos dos fármacos , Células-Tronco Mesenquimais/citologia , Engenharia Tecidual/métodos , Alicerces Teciduais , Animais , Reação a Corpo Estranho/induzido quimicamente , Humanos , Células-Tronco Mesenquimais/efeitos dos fármacos
12.
BMC Musculoskelet Disord ; 20(1): 195, 2019 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-31054584

RESUMO

BACKGROUND: Adverse Reaction to Metal Debris (ARMD) is a major reason for revision surgeries in patients with metal-on-metal (MoM) hip replacements. Most failures are related to excessively wearing implant producing harmful metal debris (extrinsic factor). As ARMD may also occur in patients with low-wearing implants, it has been suggested that there are differences in host-specific intrinsic factors contributing to the development of ARMD. However, there are no studies that have directly assessed whether the development of ARMD is actually affected by these intrinsic factors. METHODS: We included all 29 patients (out of 33 patients) with sufficient data who had undergone bilateral revision of ASR MoM hips (58 hips) at our institution. Samples of the inflamed synovia and/or pseudotumour were obtained perioperatively and sent to histopathological analysis. Total wear volumes of the implants were assessed. Patients underwent MARS-MRI imaging of the hips preoperatively. Histological findings, imaging findings and total wear volumes between the hips of each patient were compared. RESULTS: The difference in wear volume between the hips was clinically and statistically significant (median difference 15.35 mm3, range 1 to 39 mm3, IQR 6 to 23 mm3) (p < 0.001). The median ratio of total wear volume between the hips was 2.0 (range 1.09 to 10.0, IQR 1.67 to 3.72). In majority of the histological features and in presence of pseudotumour, there were no differences between the left and right hip of each patient (p > 0.05 for all comparisons). These features included macrophage sheet thickness, perivascular lymphocyte cuff thickness, presence of plasma cells, presence of diffuse lymphocytic infiltration and presence of germinal centers. CONCLUSIONS: Despite the significantly differing amounts of wear (extrinsic factor) seen between the sides, majority of the histological findings were similar in both hips and the presence of pseudotumour was symmetrical in most hips. As a direct consequence, it follows that there must be intrinsic factors which contribute to the symmetry of the findings, ie. the pathogenesis of ARMD, on individual level. This has been hypothesized in the literature but no studies have been conducted to confirm the hypothesis. Further, as the threshold of metal debris needed to develop ARMD appears to be largely variable based on the previous literature, it is likely that there are between-patient differences in these intrinsic factors, ie. the host response to metal debris is individual.


Assuntos
Artroplastia de Quadril/instrumentação , Reação a Corpo Estranho/imunologia , Prótese de Quadril/efeitos adversos , Próteses Articulares Metal-Metal/efeitos adversos , Metais/imunologia , Falha de Prótese/etiologia , Idoso , Artroplastia de Quadril/efeitos adversos , Feminino , Reação a Corpo Estranho/induzido quimicamente , Reação a Corpo Estranho/patologia , Reação a Corpo Estranho/cirurgia , Articulação do Quadril/diagnóstico por imagem , Articulação do Quadril/patologia , Articulação do Quadril/cirurgia , Humanos , Imageamento por Ressonância Magnética , Masculino , Metais/efeitos adversos , Pessoa de Meia-Idade , Reoperação/estatística & dados numéricos
13.
Int J Mol Sci ; 20(3)2019 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-30717232

RESUMO

The perspectives of regenerative medicine are still severely hampered by the host response to biomaterial implantation, despite the robustness of technologies that hold the promise to recover the functionality of damaged organs and tissues. In this scenario, the cellular and molecular events that decide on implant success and tissue regeneration are played at the interface between the foreign body and the host inflammation, determined by innate and adaptive immune responses. To avoid adverse events, rather than the use of inert scaffolds, current state of the art points to the use of immunomodulatory biomaterials and their knowledge-based use to reduce neutrophil activation, and optimize M1 to M2 macrophage polarization, Th1 to Th2 lymphocyte switch, and Treg induction. Despite the fact that the field is still evolving and much remains to be accomplished, recent research breakthroughs have provided a broader insight on the correct choice of biomaterial physicochemical modifications to tune the reaction of the host immune system to implanted biomaterial and to favor integration and healing.


Assuntos
Materiais Biocompatíveis/farmacologia , Reação a Corpo Estranho/prevenção & controle , Fatores Imunológicos/farmacologia , Macrófagos/efeitos dos fármacos , Neutrófilos/efeitos dos fármacos , Próteses e Implantes , Imunidade Adaptativa/efeitos dos fármacos , Materiais Biocompatíveis/química , Diferenciação Celular/efeitos dos fármacos , Reação a Corpo Estranho/imunologia , Humanos , Imunidade Inata/efeitos dos fármacos , Fatores Imunológicos/química , Macrófagos/citologia , Macrófagos/imunologia , Ativação de Neutrófilo/efeitos dos fármacos , Neutrófilos/citologia , Neutrófilos/imunologia , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Células Th1/citologia , Células Th1/efeitos dos fármacos , Células Th1/imunologia , Equilíbrio Th1-Th2/efeitos dos fármacos , Células Th2/citologia , Células Th2/efeitos dos fármacos , Células Th2/imunologia , Alicerces Teciduais
14.
Am J Obstet Gynecol ; 220(2): 187.e1-187.e8, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30419195

RESUMO

BACKGROUND: Polypropylene mesh is used widely for surgical treatment of pelvic organ prolapse and stress urinary incontinence. Although these surgeries demonstrate favorable functional and anatomic outcomes, their use has been limited by complications, the 2 most common being exposure and pain. Growing evidence suggests that T lymphocytes play a critical role in the regulation of the host response to biomaterials. OBJECTIVE: The purpose of this study was to define and characterize the T-cell response and to correlate the response to collagen deposition in fibrotic capsules in mesh tissue complexes that are removed for the complications of pain vs exposure. STUDY DESIGN: Patients who were scheduled to undergo a surgical excision of mesh for pain or exposure at Magee-Women's Hospital were offered enrollment. Forty-two mesh-vagina tissue complexes were removed for the primary complaint of exposure (n=24) vs pain (n=18). Twenty-one patients agreed to have an additional vaginal biopsy away from the site of mesh that served as control tissue. T cells were examined via immunofluorescent labeling for cell surface markers CD4+ (Th), CD8+ (cytotoxic) and foxp3 (T-regulatory cell). Frozen sections were stained with hematoxylin-eosin for gross morphologic condition and picrosirius red for collagen fiber analysis. Interrupted sodium-dodecyl sulfate gel electrophoresis was used to quantify the content of collagens type I and III, and the collagen III/I ratio. Transforming growth factor-ß and connective tissue growth factor, which are implicated in the development of fibrosis, were measured via enzyme-linked immunosorbent assays. Data were analyzed with the Student's t tests, mixed effects linear regression, and Spearman's correlation coefficients. RESULTS: Demographic data were not different between groups, except for body mass index, which was 31.7 kg/m2 for the exposure group and 28.2 kg/m2 for pain (P=.04). Tissue complexes demonstrated a marked, but highly localized, foreign body response. We consistently observed a teardrop-shaped fibroma that encapsulated mesh fibers in both pain and exposure groups, with the T cells localized within the tip of this configuration away from the mesh-tissue interface. All 3 T-cell populations were significantly increased relative to control: CD4+ T helper (P<.001), foxp3+ T regulatory (P<.001), and CD8+ cytotoxic T cell (P=.034) in the exposure group. In the pain group, only T-helper (P<.001) and T-regulatory cells (P<.001) were increased, with cytotoxic T cells (P=.520) not different from control. Picrosirius red staining showed a greater area of green (thin) fibers in the exposure group (P=.025) and red (thick) fibers in the pain group (P<.001). The ratio of area green/(yellow + orange + red) that represented thin vs thick fibers was significantly greater in the exposure group (P=.005). Analysis of collagen showed that collagen type I was increased by 35% in samples with mesh complications (exposure and pain) when compared with control samples (P=.043). Strong correlations between the profibrosis cytokine transforming growth factor-ß and collagen type I and III were found in patients with pain (r≥0.833; P=.01) but not exposure (P>.7). CONCLUSION: T cells appear to play a critical role in the long-term host response to mesh and may be a central pathway that leads to complications. The complexity of this response warrants further investigation and has the potential to broaden our understanding of mesh biology and clinical outcomes.


Assuntos
Colágeno/metabolismo , Reação a Corpo Estranho/imunologia , Polipropilenos/efeitos adversos , Telas Cirúrgicas/efeitos adversos , Linfócitos T/metabolismo , Adulto , Idoso , Biomarcadores/metabolismo , Remoção de Dispositivo , Feminino , Migração de Corpo Estranho/imunologia , Migração de Corpo Estranho/metabolismo , Migração de Corpo Estranho/patologia , Migração de Corpo Estranho/cirurgia , Reação a Corpo Estranho/diagnóstico , Reação a Corpo Estranho/metabolismo , Reação a Corpo Estranho/patologia , Humanos , Modelos Lineares , Pessoa de Meia-Idade , Dor Pós-Operatória/diagnóstico , Dor Pós-Operatória/etiologia , Dor Pós-Operatória/cirurgia
15.
Int J Mol Sci ; 19(11)2018 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-30373272

RESUMO

Various synthetic biomaterials are used to replace lost or damaged bone tissue that, more or less successfully, osseointegrate into the bone environment. Almost all biomaterials used in orthopedic medicine activate the host-immune system to a certain degree. The complement system, which is a crucial arm of innate immunity, is rapidly activated by an implanted foreign material into the human body, and it is intensely studied regarding blood-contacting medical devices. In contrast, much less is known regarding the role of the complement system in response to implanted bone biomaterials. However, given the increasing knowledge of the complement regulation of bone homeostasis, regeneration, and inflammation, complement involvement in the immune response following biomaterial implantation into bone appears very likely. Moreover, bone cells can produce complement factors and are target cells of activated complement. Therefore, new bone formation or bone resorption around the implant area might be greatly influenced by the complement system. This review aims to summarize the current knowledge on biomaterial-mediated complement activation, with a focus on materials primarily used in orthopedic medicine. In addition, methods to modify the interactions between the complement system and bone biomaterials are discussed, which might favor osseointegration and improve the functionality of the device.


Assuntos
Substitutos Ósseos/efeitos adversos , Proteínas do Sistema Complemento/imunologia , Reação a Corpo Estranho/imunologia , Procedimentos Ortopédicos/efeitos adversos , Animais , Regeneração Óssea/imunologia , Substitutos Ósseos/química , Ativação do Complemento , Humanos , Procedimentos Ortopédicos/métodos
16.
Ann Plast Surg ; 80(6S Suppl 6): S410-S417, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29746273

RESUMO

BACKGROUND: Biomedical devices are implanted into mammalian soft tissues to improve, monitor, or restore form or function. The utility of these implants is limited by the subsequent foreign body response (FBR), beginning with inflammation and terminating in a collagen envelope around the device, known as the capsule. This capsule then can contract and distort the shape of the device or limit its effectiveness in interacting with the surrounding host tissues. In the current study, we investigated the effect of therapeutic collagen-coated silicone discs in a rat model of the FBR. METHODS: A 3-dimensional printed mold was used to fabricate collagen-coated silicone discs incorporating 3 therapeutic agents: colchicine, a function-blocking antibody against interleukin 8 (IL-8) receptor B, and a powerful anti-inflammatory steroid, dexamethasone. Discs were implanted submuscularly into a well-characterized rat model of the FBR and evaluated for inflammatory response, fibrotic development, and cytokine release. RESULTS: Coated silicone discs exhibited reduced collagen deposition and little to no foreign body giant cells at the host-silicone interface when compared with the silicone-only group. Therapeutic hydrogels demonstrate a significant decrease in cellular infiltration into the coatings over the 2-week time point in contrast to therapeutic-free hydrogel coatings. Cytokine analysis revealed significant differences between therapeutic-free and therapeutic-containing coatings when compared with silicone-only controls. Levels of IL-1ß, IL-6, monocyte chemotactic protein 1, and macrophage inflammatory protein 3α were affected 48 hours after implantation, while differences in IL-18, growth-regulated oncogene/keratinocyte chemoattractant, and macrophage inflammatory protein 3α were observed 1 week after implantation. CONCLUSIONS: By utilizing the host's innate immune response, our engineered hydrogel coatings delivered therapeutic moieties directly to the implant microenvironment, thus delaying the FBR up to 2 weeks.


Assuntos
Anti-Inflamatórios/uso terapêutico , Colágeno/uso terapêutico , Reação a Corpo Estranho/prevenção & controle , Hidrogéis/uso terapêutico , Próteses e Implantes/efeitos adversos , Animais , Biomarcadores/metabolismo , Citocinas/metabolismo , Reação a Corpo Estranho/diagnóstico , Reação a Corpo Estranho/imunologia , Reação a Corpo Estranho/patologia , Ratos , Ratos Sprague-Dawley , Silicones/efeitos adversos , Resultado do Tratamento
17.
Int J Mol Sci ; 19(4)2018 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-29649133

RESUMO

Capsular contracture, which is the pathologic development of fibrous capsules around implants, is a major complication of reconstructive and aesthetic breast surgeries. Capsular contracture can cause implant failure with breast hardening, deformity, and severe pain. The exact mechanisms underlying this complication remain unclear. In addition, anaplastic large cell lymphoma is now widely recognized as a very rare disease associated with breast implants. Foreign body reactions are an inevitable common denominator of capsular contracture. A number of studies have focused on the associated immune responses and their regulation. The present article provides an overview of the currently available techniques, including novel nano/microtechniques, to reduce silicone implant-induced contracture and associated foreign body responses.


Assuntos
Implantes de Mama/efeitos adversos , Contratura Capsular em Implantes/prevenção & controle , Linfoma Anaplásico de Células Grandes/prevenção & controle , Géis de Silicone/efeitos adversos , Animais , Materiais Biomiméticos/uso terapêutico , Feminino , Reação a Corpo Estranho/induzido quimicamente , Reação a Corpo Estranho/imunologia , Reação a Corpo Estranho/prevenção & controle , Humanos , Contratura Capsular em Implantes/induzido quimicamente , Contratura Capsular em Implantes/imunologia , Linfoma Anaplásico de Células Grandes/induzido quimicamente , Linfoma Anaplásico de Células Grandes/imunologia , Nanotecnologia
19.
Adv Drug Deliv Rev ; 114: 184-192, 2017 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-28712923

RESUMO

The compatibility of biomaterials is critical to their structural and biological function in medical applications. The immune system is the first responder to tissue trauma and to a biomaterial implant. The innate immune effector cells, most notably macrophages, play a significant role in the defense against foreign bodies and the formation of a fibrous capsule around synthetic implants. Alternatively, macrophages participate in the pro-regenerative capacity of tissue-derived biological scaffolds. Research is now elucidating the role of the adaptive immune system, and T cells in particular, in directing macrophage response to synthetic and biological materials. Here, we review basic immune cell types and discuss recent research on the role of the immune system in tissue repair and its potential relevance to scaffold design. We will also discuss new emerging immune cell types relevant to biomaterial responses and tissue repair. Finally, prospects for specifically targeting and modulating the immune response to biomaterial scaffolds for enhancing tissue repair and regeneration will be presented.


Assuntos
Materiais Biocompatíveis/efeitos adversos , Reação a Corpo Estranho/imunologia , Macrófagos/imunologia , Medicina Regenerativa , Linfócitos T/imunologia , Alicerces Teciduais/efeitos adversos , Animais , Humanos , Imunidade Inata , Regeneração/imunologia , Medicina Regenerativa/métodos
20.
Clin Exp Pharmacol Physiol ; 44(2): 313-323, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27864839

RESUMO

Implantation of electrodes or cannulae into the brain is accompanied by a tissue response referred to as foreign body response. Adenosine triphosphate (ATP) is one of the signalling molecules released by injured cells which mediate the chemoattraction of microglial cells. The constitutive release of pro-inflammatory and cytotoxic substances by microglial cells in chronic implants exacerbates neuronal cell death and the immune response. This study aimed to interfere with the initial events of the foreign body response in order to mitigate neurotoxicity and inflammation. For this purpose, the ATP-hydrolysing enzyme apyrase and the antibiotic minocycline with a broad range of anti-inflammatory, anti-apoptotic and glutamate-antagonist properties were locally infused during cannula implantation in the caudal forelimb area of the motor cortex in Lister Hooded rats. The rats' motor performance was assessed in a skilled reaching task and the distribution of neurons and glial cells in the vicinity of the implant was examined 2 and 6 weeks post-implantation. Apyrase as well as minocycline increased the number of surviving neurons and reduced microglial activation. Moreover, minocycline improved the motor performance and, additionally, caused a temporary reduction in astrogliosis, suggesting it as a possible therapeutic candidate to improve the biocompatibility of chronic brain implants.


Assuntos
Apirase/farmacologia , Encéfalo/efeitos dos fármacos , Cânula/efeitos adversos , Reação a Corpo Estranho/prevenção & controle , Minociclina/farmacologia , Fármacos Neuroprotetores/farmacologia , Animais , Encéfalo/imunologia , Encéfalo/patologia , Reação a Corpo Estranho/imunologia , Reação a Corpo Estranho/patologia , Membro Anterior/efeitos dos fármacos , Membro Anterior/imunologia , Membro Anterior/patologia , Masculino , Microglia/efeitos dos fármacos , Microglia/imunologia , Microglia/patologia , Atividade Motora/efeitos dos fármacos , Córtex Motor/efeitos dos fármacos , Córtex Motor/imunologia , Córtex Motor/patologia , Neurônios/efeitos dos fármacos , Neurônios/imunologia , Neurônios/patologia , Ratos Endogâmicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA