Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
PLoS One ; 16(5): e0251885, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34003868

RESUMEN

It is widely recognized that innate macrophage immune reactions to implant debris are central to the inflammatory responses that drive biologic implant failure over the long term. Less common, adaptive lymphocyte immune reactions to implant debris, such as delayed type hypersensitivity (DTH), can also affect implant performance. It is unknown which key patient factors, if any, mediate these adaptive immune responses that potentiate particle/macrophage mediated osteolysis. The objective of this investigation was to determine to what degree known adaptive immune responses to metal implant debris can affect particle-induced osteolysis (PIO); and if this pathomechanism is dependent on: 1) innate immune danger signaling, i.e., NLRP3 inflammasome activity, 2) sex, and/or 3) age. We used an established murine calvaria model of PIO using male and female wild-type C57BL/6 vs. Caspase-1 deficient mice as well as young (12-16 weeks old) vs. aged (18-24 months old) female and male C57BL/6 mice. After induction of metal-DTH, and Cobalt-alloy particle (ASTM F-75, 0.4um median diameter) calvaria challenge, bone resorption was assessed using quantitative micro-computed tomography (micro-CT) analysis and immune responses were assessed by measuring paw inflammation, lymphocyte transformation test (LTT) reactivity and adaptive immune cytokines IFN-gamma and IL-17 (ELISA). Younger aged C57BL/6 female mice exhibited the highest rate and severity of metal sensitivity lymphocyte responses that also translated into higher PIO compared to any other experimental group. The absence of inflammasome/caspase-1 activity significantly suppressed DTH metal-reactivity and osteolysis in both male and female Caspase-1 deficient mice. These murine model results indicate that young female mice are more predisposed to metal-DTH augmented inflammatory responses to wear debris, which is highly influenced by active NLRP3 inflammasome/caspase-1 danger signaling. If these results are clinically meaningful for orthopedic patients, then younger female individuals should be appropriately assessed and followed for DTH derived peri-implant complications.


Asunto(s)
Caspasa 1/genética , Metales/efectos adversos , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Osteólisis/genética , Prótesis e Implantes/efectos adversos , Factores de Edad , Animales , Resorción Ósea/etiología , Resorción Ósea/genética , Resorción Ósea/patología , Femenino , Humanos , Hipersensibilidad/etiología , Hipersensibilidad/genética , Hipersensibilidad/fisiopatología , Inmunidad Innata/efectos de los fármacos , Inmunidad Innata/genética , Inflamasomas/efectos de los fármacos , Inflamasomas/genética , Interferón gamma/genética , Interleucina-17/genética , Macrófagos/efectos de los fármacos , Masculino , Metales/uso terapéutico , Ratones , Osteólisis/inducido químicamente , Osteólisis/patología , Caracteres Sexuales , Cráneo/efectos de los fármacos , Cráneo/crecimiento & desarrollo , Cráneo/fisiopatología , Microtomografía por Rayos X
2.
Aesthet Surg J ; 41(7): NP782-NP793, 2021 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-33564817

RESUMEN

BACKGROUND: Although breast implants (BIs) have never been safer, factors such as implant debris may influence complications such as chronic inflammation and illness such as ALCL (anaplastic large cell lymphoma). Do different types of BIs produce differential particulate debris? OBJECTIVES: The aim of this study was to quantify, investigate, and characterize the size, amount, and material type of both loosely bound and adherent surface particles on 5 different surface types of commercial BIs. METHODS: Surface particles from BIs of 5 surface types (n = 5/group), Biocell, Microcell, Siltex, Smooth, SmoothSilk, and Traditional-Smooth, were: (1) removed by a rinsing procedure and (2) removed with ultrapure adhesive carbon tabs. Particles were characterized (ASTM 1877-16) by scanning electron microscopy and energy-dispersive X-ray chemical analysis. RESULTS: Particles rinsed from Biocell, Microcell and Siltex were <1 µm in diameter whereas SmoothSilk and Traditional-Smooth surfaces had median sizes >1 µm (range, 0.4-2.7 µm). The total mass of particles rinsed from the surfaces indicated Biocell had >5-fold more particulate compared with all other implants, and >30-fold more than SmoothSilk or Traditional-Smooth implants (>100-fold more for post-rinse adhesion analysis). Energy-dispersive X-ray analysis indicated that the particulate material for Biocell, Microcell, and Siltex was silicone (>50%), whereas particulates from SmoothSilk and Traditional-Smooth implants were predominantly carbon-based polymers, eg, polycarbonate-urethane, consistent with packaging (and were detected on all implant types). Generally, SmoothSilk and Traditional-Smooth implant groups released >10-fold fewer particles than Biocell, Microcell, and Siltex surfaces. Pilot ex vivo tissue analysis supported these findings. CONCLUSIONS: Particulate debris released from BIs are highly dependent on the type of implant surface and are a likely key determinant of in vivo performance.


Asunto(s)
Implantación de Mama , Implantes de Mama , Linfoma Anaplásico de Células Grandes , Implantes de Mama/efectos adversos , Humanos , Siliconas
3.
Aesthet Surg J ; 39(Suppl_1): S36-S48, 2019 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-30715176

RESUMEN

Currently, there is a dearth of information regarding the degree of particle shedding from breast implants (BIs) and what are the general biological consequences of BI debris. Thus, it is unclear to what degree BI debris compromises the long-term biological performance of BIs. For orthopedic implants, it is well established that the severity of biological reactivity to implant debris governs long-term clinical performance. Orthopedic implant particulate debris is generally in the range of 0.01 to 100 µm in diameter. Implant debris-induced bioreactivity/inflammation is mostly a peri-implant phenomenon caused by local innate immune cells (eg, macrophages) that produce proinflammatory cytokines such as tumor necrosis factor-α, interleukin-1ß, interleukin-6, and prostaglandin 2 (PGE2). In orthopedics, there have been few systemic concerns associated with polymeric implant debris (like silicone) other than documented dissemination to remote organs (eg, liver, spleen, etc.) with no known associated pathogenicity. This is not true of metal implant debris where normal (well-functioning) implants can induce systemic reactions such as delayed type hypersensitivity. Diagnostic analysis of orthopedic tissues has focused on innate (macrophage mediated) and adaptive (lymphocyte-mediated hypersensitivity) immune responses. Orthopedic implant debris-associated lymphocyte cancers have not been reported in over 40 years of orthopedic literature. Adaptive immune responses such as hypersensitivity reactions to orthopedic implant debris have been dominated by certain implant types that produce specific kinds of debris (eg, metal-on-metal total joint prostheses). Orthopedic hypersensitivity responses and atypical BI bioreactivity such as BI-associated anaplastic large cell lymphoma share crossover markers for diagnosis. Differentiating normal innate immune reactivity to particles from anaplastic large cell lymphoma reactions from delayed type hypersensitivity reactions to BI-associated implant debris remains unclear but vital to patients and surgeons.


Asunto(s)
Implantes de Mama/efectos adversos , Inflamación/etiología , Prótesis e Implantes/efectos adversos , Citocinas/inmunología , Humanos , Inmunidad Innata/inmunología , Inflamación/diagnóstico , Inflamación/inmunología , Linfoma Anaplásico de Células Grandes/diagnóstico , Linfoma Anaplásico de Células Grandes/etiología , Linfoma Anaplásico de Células Grandes/inmunología , Procedimientos Ortopédicos/efectos adversos , Procedimientos Ortopédicos/instrumentación , Diseño de Prótesis , Falla de Prótesis
4.
PLoS One ; 14(1): e0210336, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30653583

RESUMEN

Metal hypersensitivity has been recognized as an adverse biologic reaction that can compromise total joint arthroplasty (TJA) performance. However, the etiology of metal hypersensitivity responses in TJAs remains unclear. Metal implant debris is known to act as a danger signal that drives NLRP3 inflammasome activation. It remains unknown if implant debris induced inflammasome activation regulates T cell lineage in TJA metal hypersensitivity responses. In this study, we show both in vivo and in vitro that the pathogenesis of metal hypersensitivity responses to implant debris are largely dependent on activation of the inflammasome/caspase-1 pathway and subsequent production of IL-17A/F by CD4+ T cells. Inhibiting either the inflammasome pathway or IL-17A bioactivity in vivo and in vitro (in vivo using NLRP3 and Caspase-1 deficient mice or in vitro using blocking agents such as Capase-1 inhibitor, IL-1Ra and anti-IL-17A), significantly (p<0.05) mitigated metal-DTH paw inflammation as well as lymphocyte cytokine (IFN-γ and IL-17) and proliferation responses in metal-sensitized mice and primary human PBMCs. This study provides mechanistic insight into how in vivo exposure to orthopedic implant debris, and metals in general, elicits NLRP3 inflammasome activation that mediates the generation of IL-17A/F producing CD4+ T cells, leading to metal-delayed type hypersensitivity reactions.


Asunto(s)
Hipersensibilidad Tardía/etiología , Inflamasomas/inmunología , Prótesis Articulares/efectos adversos , Metales/efectos adversos , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Células Th17/inmunología , Animales , Caspasa 1/deficiencia , Caspasa 1/genética , Caspasa 1/inmunología , Células Cultivadas , Citocinas/biosíntesis , Femenino , Humanos , Hipersensibilidad Tardía/inmunología , Técnicas In Vitro , Interleucina-17/antagonistas & inhibidores , Interleucina-17/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Inmunológicos , Proteína con Dominio Pirina 3 de la Familia NLR/deficiencia , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Níquel/efectos adversos , Falla de Prótesis/etiología , Transducción de Señal
5.
Open Orthop J ; 12: 115-124, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29785221

RESUMEN

BACKGROUND: The rate of revision for some designs of total hip replacements due to idiopathic aseptic loosening has been reported as higher for women. However, whether this is environmental or inherently sex-related is not clear. OBJECTIVE: Can particle induced osteolysis be sex dependent? And if so, is this dependent on the type of implant debris (e.g. metal vs polymer)? The objective of this study was to test for material dependent inflammatory osteolysis that may be linked to sex using CoCrMo and implant grade conventional polyethylene (UHMWPE), using an in vivo murine calvaria model. METHODS: Healthy 12 week old female and male C57BL/6J mice were treated with UHMWPE (1.0um ECD) or CoCrMo particles (0.9um ECD) or received sham surgery. Bone resorption was assessed by micro-computed tomography, histology and histomorphometry on day 12 post challenge. RESULTS: Female mice that received CoCrMo particles showed significantly more inflammatory osteolysis and bone destruction compared to the females who received UHMWPE implant debris. Moreover, females challenged with CoCrMo particles exhibited 120% more inflammatory bone loss compared to males (p<0.01) challenged with CoCrMo implant debris (but this was not the case for UHMWPE particles). CONCLUSION: We demonstrated sex-specific differences in the amount of osteolysis resulting from CoCrMo particle challenge. This suggests osteo-immune responses to metal debris are preferentially higher in female compared to male mice, and supports the contention that there may be inherent sex related susceptibility to some types of implant debris.

6.
PLoS One ; 11(7): e0160141, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27467577

RESUMEN

Cobalt alloy debris has been implicated as causative in the early failure of some designs of current total joint implants. The ability of implant debris to cause excessive inflammation via danger signaling (NLRP3 inflammasome) vs. pathogen associated pattern recognition receptors (e.g. Toll-like receptors; TLRs) remains controversial. Recently, specific non-conserved histidines on human TLR4 have been shown activated by cobalt and nickel ions in solution. However, whether this TLR activation is directly or indirectly an effect of metals or secondary endogenous alarmins (danger-associated molecular patterns, DAMPs) elicited by danger signaling, remains unknown and contentious. Our study indicates that in both a human macrophage cell line (THP-1) and primary human macrophages, as well as an in vivo murine model of inflammatory osteolysis, that Cobalt-alloy particle induced NLRP3 inflammasome danger signaling inflammatory responses were highly dominant relative to TLR4 activation, as measured respectively by IL-1ß or TNF-α, IL-6, IL-10, tissue histology and quantitative bone loss measurement. Despite the lack of metal binding histidines H456 and H458 in murine TLR4, murine calvaria challenge with Cobalt alloy particles induced significant macrophage driven in vivo inflammation and bone loss inflammatory osteolysis, whereas LPS calvaria challenge alone did not. Additionally, no significant increase (p<0.05) in inflammation and inflammatory bone loss by LPS co-challenge with Cobalt vs. Cobalt alone was evident, even at high levels of LPS (i.e. levels commiserate with hematogenous levels in fatal sepsis, >500pg/mL). Therefore, not only do the results of this investigation support Cobalt alloy danger signaling induced inflammation, but under normal homeostasis low levels of hematogenous PAMPs (<2pg/mL) from Gram-negative bacteria, seem to have negligible contribution to the danger signaling responses elicited by Cobalt alloy metal implant debris. This suggests the unique nature of Cobalt alloy particle bioreactivity is strong enough to illicit danger signaling that secondarily activate concomitant TLR activation, and may in part explain Cobalt particulate associated inflammatory and toxicity-like reactions of specific orthopedic implants.


Asunto(s)
Aleaciones/efectos adversos , Cobalto/efectos adversos , Inflamación/inducido químicamente , Osteoporosis/inducido químicamente , Prótesis e Implantes , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 4/metabolismo , Animales , Humanos , Interleucina-1beta/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Receptor Toll-Like 4/agonistas , Factor de Necrosis Tumoral alfa/metabolismo
8.
Mediators Inflamm ; 2014: 185150, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24891761

RESUMEN

All of the over 1 million total joint replacements implanted in the US each year are expected to eventually fail after 15-25 years of use, due to slow progressive subtle inflammation at the bone implant interface. This inflammatory disease state is caused by implant debris acting, primarily, on innate immune cells, that is, macrophages. This slow progressive pathological bone loss or "aseptic loosening" is a potentially life-threatening condition due to the serious complications in older people (>75 yrs) of total joint replacement revision surgery. In some people implant debris (particles and ions from metals) can influence the adaptive immune system as well, giving rise to the concept of metal sensitivity. However, a consensus of studies agrees that the dominant form of this response is due to innate reactivity by macrophages to implant debris where both danger (DAMP) and pathogen (PAMP) signalling elicit cytokine-based inflammatory responses. This paper discusses implant debris induced release of the cytokines and chemokines due to activation of the innate (and the adaptive) immune system and the subsequent formation of osteolysis. Different mechanisms of implant-debris reactivity related to the innate immune system are detailed, for example, danger signalling (e.g., IL-1ß, IL-18, IL-33, etc.), toll-like receptor activation (e.g., IL-6, TNF-α, etc.), apoptosis (e.g., caspases 3-9), bone catabolism (e.g., TRAP5b), and hypoxia responses (Hif1-α). Cytokine-based clinical and basic science studies are in progress to provide diagnosis and therapeutic intervention strategies.


Asunto(s)
Regulación de la Expresión Génica , Inmunidad Innata , Falla de Prótesis , Inmunidad Adaptativa , Anciano , Huesos/metabolismo , Quimiocinas/metabolismo , Citocinas/metabolismo , Humanos , Inflamasomas , Inflamación , Linfocitos/inmunología , Macrófagos/inmunología , Ortopedia , Osteoblastos/inmunología , Osteólisis , Transducción de Señal , Estados Unidos
9.
J Long Term Eff Med Implants ; 24(1): 37-44, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24941404

RESUMEN

Metal sensitivity testing is generally the diagnosis method of last resort for aseptic painful implants with elevated inflammatory responses. However, the relationship between implant-related pain and implant-debris-related metal sensitization remains incompletely understood. Although a sensitivity to nickel alone has been used as a general measure of metal allergy, it may lack the specificity to correlate sensitivity to specific implant metals and thus to select a biologically appropriate implant material. In this retrospective study, we report the incidence of pain and nickel sensitivity in patients with total joint arthroplasties (TJAs) referred for metal sensitivity testing (n=2018). We also correlated the degree of nickel hypersensitivity to implant pain levels (none, mild, moderate, and high, using a scale of 0-10) and the incidence of sensitivity to alternative implant metals in highly nickel-reactive subjects. Most patients (>79%) reported pain levels that were moderate to high regardless of implant age, whereas patients with severely painful TJAs had a statistically greater incidence of nickel sensitivity over the short-term post-operative period (≤4 years). Patients with moderate pain scores (4-7) and high pain scores (≥8) also exhibited significantly higher sensitivity to nickel compared to patients with no pain and no implant (controls) (p<0.05). Highly nickel-sensitive subjects (SI>8) also showed incidences of sensitization to alternative materials such as cobalt, chromium, or molybdenum (57%) or aluminum or vanadium alloy (52%). These data suggest that painful TJAs caused by metal sensitivity more likely occur relatively early in the post-operative period (≤4 years). The incidences of sensitivity to alternative implant metals in only a subset of nickel-reactive patients highlights the importance of testing for sensitization to all potential revision implant materials.


Asunto(s)
Artralgia/etiología , Artroplastia de Reemplazo/instrumentación , Hipersensibilidad Tardía/inmunología , Prótesis Articulares de Metal sobre Metal/efectos adversos , Metales/efectos adversos , Níquel/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Aluminio/inmunología , Artroplastia de Reemplazo/efectos adversos , Cromo/inmunología , Cobalto/inmunología , Femenino , Prótesis de Cadera/efectos adversos , Humanos , Hipersensibilidad Tardía/etiología , Prótesis de la Rodilla/efectos adversos , Masculino , Metales/inmunología , Persona de Mediana Edad , Molibdeno/inmunología , Dimensión del Dolor , Estudios Retrospectivos , Factores de Tiempo , Vanadio/inmunología , Adulto Joven
10.
J Long Term Eff Med Implants ; 24(1): 65-76, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24941407

RESUMEN

Adverse local tissue reactions to wear debris and corrosion products have lead to a sharp decline in the use of metal-on-metal (MOM) total hip athroplasties (THAs) clinically. Today, approximately 1 million patients are still carrying such a device. To gain a better understanding of the effect of wear and corrosion products on cells within the joint environment, it is important to generate conditions in vitro that resemble the in vivo system as closely as possible. In this paper, we present a novel tribocorrosion bioreactor that enables the simultaneous conduction of tribocorrosion and cell-culture experiments. In this setup, macrophage cell cultures are located in direct proximity to a tribological interface mimicking the sliding conditions of THA and are exposed to wear and corrosion products as they are generated. These products may include meta-stable species and metallo-organic complexes that have not been considered in earlier studies. The combination of standard tribological, electrochemical, and biological techniques is associated with several challenges that are described here in detail.


Asunto(s)
Reactores Biológicos , Técnicas de Cultivo de Célula , Células Cultivadas , Corrosión , Electroquímica , Reacción a Cuerpo Extraño/inmunología , Prótesis de Cadera/efectos adversos , Humanos , Macrófagos , Ensayo de Materiales , Prótesis Articulares de Metal sobre Metal/efectos adversos , Propiedades de Superficie
11.
J Long Term Eff Med Implants ; 24(1): 77-88, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24941408

RESUMEN

Biologic reactivity to orthopedic implant debris mediates long-term clinical performance of total joint arthroplasty implants. However, the reasons that some facets of implant debris (e.g., particle size, shape, base material, etc.) are more pro-inflammatory remain controversial. This precludes accurate prediction and optimal design of modern total joint replacements. We hypothesized that debris particle size can influence adsorbed protein film composition and affect subsequent bioreactivity. We measured size-dependent proteinfilm adsorption, and adsorbed protein-film-dependent cytokine release using equal surface areas of different sized cobalt-chromium alloy (CoCr-alloy) particles and in vitro challenge of human macrophages (THP-1 and human primary). Smaller (5 µm diameter) versus larger (70 µm diameter) particles preferentially adsorbed more serum protein in general (p<0.03), where higher molecular weight serum proteins consistent with IgG were identified. Additionally, 5-µm CoCr-alloy particles pre-coated with different protein biofilms (IgG vs. albumin) resulted in a difference in cytokine expression in which albumin-coated particles induced more TNF-α release and IgG-coated particles induced more IL-1ß release from human monocytes/macrophages. In these preliminary in vitro studies, we have demonstrated the capability of equal surface areas of different particle sizes to influence adsorbed protein composition and that adsorbed protein differences on identical particles can translate into complex differences in bioreactivity. Together, these findings suggest that adsorbed protein differences on different-sized particles of the same material may be a contributing mechanism by which certain particles induce different reactivities.


Asunto(s)
Adsorción , Fibronectinas/metabolismo , Inmunoglobulina G/metabolismo , Tamaño de la Partícula , Albúmina Sérica/metabolismo , Adulto , Supervivencia Celular , Células Cultivadas , Aleaciones de Cromo , Femenino , Fibronectinas/farmacología , Voluntarios Sanos , Humanos , Inmunoglobulina G/farmacología , Interleucina-1beta/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Persona de Mediana Edad , Peso Molecular , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Albúmina Sérica/farmacología , Factor de Necrosis Tumoral alfa/metabolismo , Adulto Joven
12.
Open Orthop J ; 7: 605-13, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24198853

RESUMEN

Long-term aseptic failures of joint replacements are generally attributed to implant debris-induced inflammation and osteolysis. This response is largely mediated by immune and bone cells (monocytes/macrophages and osteoclasts, respectively), that in the presence of implant debris (e.g. metal particles and ions), release pro-inflammatory cytokines such as IL-1ß, TNF-α, and IL-6. The relative degree to which implant debris can illicit inflammatory response(s) from osteoclasts vs monocytes/macrophages is unknown, i.e. are osteoclasts a viable target for anti-inflammatory therapy for implant debris? We investigated relative monocyte versus osteoclast inflammatory responses in a side-by-side comparison using implant debris from the perspective of both danger signaling (IL-1ß) and pathogenic recognition (TNF-α) reactivity (Challenge Agents: Cobalt-alloy, Titanium-alloy, and PMMA particles, 0.9-1.8um-dia ECD and Cobalt, and Nickel-ions 0.01-0.1mM, all with and without LPS priming). Human monocytes/macrophages reacted to implant debris with >100 fold greater production of cytokines compared to osteoclast-like cells. Particulate Co-alloy challenge induced >1000 pg/ml of IL-1ß and TNF-α, in monocytes and <50pg/mL IL-1ß and TNF-α in osteoclasts. Cobalt ions induced >3000pg/mL IL-1ß and TNF-α in monocytes/macrophages and <50pg/mL IL-1ß and TNF-α in osteoclasts. The paracrine effect of supernatants from debris-treated monocytes/macrophages was capable of inducing greater osteoclastogenesis (TRAP+, p<0.06) and inflammation than direct debris challenge on osteoclasts. Our results indicate that as monocytes/macrophages differentiate into osteoclasts, they largely lose their innate immune reactivity to implant debris and thus may not be as relevant a therapeutic target as monocytes/macrophages for mitigating debris-induced inflammation.

13.
Eur Spine J ; 21(12): 2649-58, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22407269

RESUMEN

PURPOSE: We evaluated the consequences of cobalt-chromium alloy (CoCr) wear debris challenge in the peri-spine region to determine the inflammation and toxicity associated with submicron particulates of CoCr-alloy and nickel on the peri-spine. METHODS: The lumbar epidural spaces of (n = 50) New Zealand white rabbits were challenged with: 2.5 mg CoCr, 5.0 mg CoCr, 10.0 mg CoCr, a positive control (20.0 mg of nickel) and a negative control (ISOVUE-M-300). The CoCr-alloy and Ni particles had a mean diameter of 0.2 and 0.6 µm, respectively. Five rabbits per dose group were studied at 12 and 24 weeks. Local and distant tissues were analyzed histologically and quantitatively analyzed immunohistochemically (TNF-α and IL-6). RESULTS: Histologically, wear particles were observed in all animals. There was no evidence of toxicity or local irritation noted during macroscopic observations in any CoCr-dosed animals. However, Ni-treated control animals experienced bilateral hind leg paralysis and were euthanized at Day 2. Histopathology of the Ni particle-treated group revealed severe neuropathy. Quantitative immunohistochemistry demonstrated a CoCr-alloy dose-dependent increase in cytokines (IL-6, TNF-α, p < 0.05) at 12 and 24 weeks. CONCLUSIONS: Subtle peri-spine inflammation associated with CoCr-alloy implant particles was dose dependent and persistent. Neuropathy can be induced by highly reactive Ni particles. This suggests peri-spine challenge with CoCr-alloy implant debris (e.g., TDA) is consistent with past reports using titanium alloy particles, i.e., mild persistent inflammation.


Asunto(s)
Aleaciones de Cromo/efectos adversos , Inflamación/inducido químicamente , Enfermedades de la Columna Vertebral/inducido químicamente , Animales , Citocinas/análisis , Espacio Epidural/química , Espacio Epidural/inmunología , Espacio Epidural/patología , Inmunohistoquímica , Inflamación/inmunología , Inflamación/patología , Región Lumbosacra , Masculino , Conejos , Enfermedades de la Columna Vertebral/inmunología , Enfermedades de la Columna Vertebral/patología
14.
J Biomed Mater Res B Appl Biomater ; 100(2): 480-92, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22102421

RESUMEN

Biologic reactivity to orthopedic implant debris is generally the main determinant of long-term clinical performance where released polymeric particles of Ultra-high molecular weight polyethylene (UHMWPE) remain the most prevalent debris generated from metal-on-polymer bearing total joint arthroplasties. Polymeric alternatives to UHMWPE such as polyetherether-ketone (PEEK) may have increased wear resistance but the bioreactivity of PEEK-OPTIMA particles on peri-implant inflammation remains largely uncharacterized. We evaluated human monocyte/macrophage responses (THP-1s and primary human) when challenged by PEEK-OPTIMA, UHMWPE, and X-UHMWPE particles of three particle sizes (0.7 um, 2 um, and 10 um) at a dose of 20 particles-per-cell at 24- and 48-h time points. Macrophage responses were measured using cytotoxicity assays, viability assays, proliferation assays and cytokine analysis (IL-1b, IL-6, IL-8, MCP-1, and TNF-α). In general, there were no significant differences between PEEK-OPTIMA, UHMWPE, and X-UHMWPE particles on macrophage viability or proliferation. However, macrophages demonstrated greater cytotoxicity responses to UHMWPE and X-UHMWPE than to PEEK-OPTIMA at 24 and 48 h, where 0.7 µm-UHMWPE particles produced the highest amount of cytotoxicity. Particles of X-UHMWPE more than PEEK-OPTIMA and UHMWPE induced IL-1ß, IL-6, MCP-1, and TNF-α at 24 h, p < 0.05 (no significant differences at 48 h). On average, cytokine production was more adversely affected by larger 10 µm particles than by 0.7 and 2 µm sized particles. While limitations of in vitro analysis apply to this study, PEEK-OPTIMA particles were more biocompatible than UHMWPE particles, in that they induced less inflammatory cytokine responses and thus, in part, demonstrates that PEEK-OPTIMA implant debris does not represent an increased inflammatory risk over that of UHMWPE.


Asunto(s)
Materiales Biocompatibles Revestidos , Citocinas/metabolismo , Cetonas , Macrófagos/metabolismo , Nanopartículas/química , Polietilenglicoles , Polietilenos , Artroplastia de Reemplazo , Benzofenonas , Línea Celular Tumoral , Materiales Biocompatibles Revestidos/química , Materiales Biocompatibles Revestidos/farmacología , Humanos , Cetonas/química , Cetonas/farmacología , Macrófagos/patología , Ensayo de Materiales , Tamaño de la Partícula , Polietilenglicoles/química , Polietilenglicoles/farmacología , Polietilenos/química , Polietilenos/farmacología , Polímeros
15.
Spine (Phila Pa 1976) ; 36(26): E1675-85, 2011 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-21494194

RESUMEN

STUDY DESIGN: In vitro biotribological wear particulate investigation. OBJECTIVE: To characterize poly-ether-ether-ketone (PEEK)-OPTIMA wear particulate generated from a series of wear tests used to evaluate the wear resistance and long-term biodurability of NUBAC, a PEEK-on-PEEK articulating nucleus replacement device, and compare with wear particulate associated with hip and knee total joint arthroplasties. SUMMARY OF BACKGROUND DATA: The use of PEEK in spinal arthroplasty represents a unique application of this material. Clinically, osteolysis, osteolytic changes, and adverse reactions to metal ions have been documented in spinal arthroplasty. Therefore, it is critically important to analyze the PEEK wear particulate to evaluate its resultant biologic reactivity. Historically, scanning electron microscopy (SEM) has been used for wear debris characterization. Light scattering, specifically laser diffraction, has also been successfully used. The combined use of both techniques may provide a more comprehensive analysis than either method alone. METHODS: Proteinacious serum containing PEEK wear debris generated from four groups of devices from separate wear tests underwent enzymatic and acid digestion. The particulate was analyzed using laser diffraction in duplicate, followed by SEM analysis. RESULTS: Laser diffraction analysis demonstrated a relatively large mean particle diameter on the basis of particle volume (16.5-40.0 µm) as compared with particle number (0.9-2.2 µm). For all groups, more than 50% of debris was larger than 5.0 µm. SEM analysis revealed a mean particle size consistent with the number-based laser diffraction results. The morphology of the wear particulate appeared to be similar for all the groups analyzed. CONCLUSION: The analysis of the particles generated from an articulating PEEK-on-PEEK nucleus replacement device shows debris within size ranges typical of other total joint arthroplasty implants, with relatively round morphology, along with the results suggesting a reduced particle load. These attributes tend to diminish the potential of these PEEK particles to elicit an inflammatory response.


Asunto(s)
Artroplastia de Reemplazo/métodos , Análisis de Falla de Equipo/métodos , Disco Intervertebral/cirugía , Prótesis e Implantes , Benzofenonas , Materiales Biocompatibles/uso terapéutico , Fenómenos Biomecánicos , Humanos , Disco Intervertebral/fisiopatología , Desplazamiento del Disco Intervertebral/fisiopatología , Desplazamiento del Disco Intervertebral/cirugía , Prótesis Articulares , Cetonas/uso terapéutico , Rayos Láser , Vértebras Lumbares/cirugía , Ensayo de Materiales , Microscopía Electrónica de Rastreo , Evaluación de Resultado en la Atención de Salud , Tamaño de la Partícula , Polietilenglicoles/uso terapéutico , Polímeros , Rango del Movimiento Articular
16.
J Biomed Mater Res A ; 93(1): 227-34, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19557792

RESUMEN

This study was undertaken to examine macrophage response to polycarbonate-urethane, a proposed alternative material to polyethylene in acetabular components of total hip arthroplasty. Polyethylene wear debris from total joint replacements has been linked to osteolysis and implant lifespan. It has been shown in vitro, that polyethylene particles cleaned of endotoxin generate less of an inflammatory cytokine response than endotoxin bound particles. Comparative particle induced effects on implant fixation were tested using endotoxin free cross-linked ultra-high molecular weight polyethylene (x-UHMWPE) and polycarbonate-urethane (PCU) particles with and without intraperitoneal injection (IP) of lipopolysaccharide (LPS) using a Ti-alloy femoral intramedullary nail rat model. MicroCT and mechanical testing assessment of peri-implant bone indicated significantly less bone and lower fixation strength, respectively, when the implant was surrounded by xUHMWPE particles compared to PCU particles (with and without LPS IP). This indicates particles of PCU may be less disruptive to bone-implant fixation than x-UHMWPE in vivo, under both LPS free and challenged conditions.


Asunto(s)
Reactivos de Enlaces Cruzados/farmacología , Cemento de Policarboxilato/farmacología , Polietileno/farmacología , Uretano/farmacología , Animales , Huesos/efectos de los fármacos , Fuerza Compresiva/efectos de los fármacos , Femenino , Implantes Experimentales , Ratas , Ratas Sprague-Dawley , Programas Informáticos , Titanio/farmacología , Microtomografía por Rayos X
17.
J Biomed Mater Res A ; 93(1): 347-55, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19569215

RESUMEN

This study was undertaken to compare macrophage response to polycarbonate-urethane (PCU), a proposed alternative material to polyethylene in acetabular components of total hip arthroplasty to cross-linked ultra-high molecular weight polyethylene (xUHMWPE) in the presence or absence of endotoxin. Polyethylene wear debris that is generated by total hip and knee replacements has been linked to osteolysis and limiting the lifespan of the implant. We added both lipopolysaccharide (LPS)-free and endotoxin-associated xUHMWPE and PCU particles to a human monocyte cell line (TH1) in culture and measured cell viability and tumor necrosis factor (TNF)alpha, interleukin (IL)-1beta, and prostaglandin E(2) (PGE(2)) in the medium after 24 h. Results indicate that particles (both xUHMWPE and PCU) free of endotoxin did not significantly induce secretion of TNFalpha, IL-1beta, or PGE(2) above basal levels. However, endotoxin-exposed PCU particles induced significantly less TNFalpha and IL-1beta than endotoxin-exposed xUHMWPE particles. This indicates that if endotoxin is available for binding to particles in vivo, then xUHMWPE may be more inflammatory to periprosthetic tissue and bone in part because of its affinity/reactivity with endotoxin when compared with PCU.


Asunto(s)
Macrófagos/citología , Macrófagos/efectos de los fármacos , Cemento de Policarboxilato/farmacología , Polietileno/farmacología , Uretano/farmacología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Dinoprostona/metabolismo , Humanos , Interleucina-1beta/metabolismo , Luz , Microscopía Electrónica de Rastreo , Dispersión de Radiación , Factor de Necrosis Tumoral alfa/metabolismo
18.
J Biomed Mater Res A ; 92(2): 667-82, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19235773

RESUMEN

Hypersensitivity to metallic implants remains relatively unpredictable and poorly understood. We initially hypothesized that metal-induced lymphocyte proliferation responses to soluble metal challenge (ions) are mediated exclusively by early T-cell activation (not B-cells), typical of a delayed-type-hypersensitivity response. We tested this by comparing proliferation (6 days) of primary lymphocytes with early T-cell and B-cell activation (48 h) in three groups of subjects likely to demonstrate elevated metal reactivity: group 1 (n = 12) history of metal sensitivity with no implant; group 2a (n = 6) well performing metal-on-metal THRs, and group 2b (n = 20) subjects with poorly performing metal-on-polymer total joint arthroplasties (TJA). Group 1 showed 100% (12/12) metal reactivity (stimulation index > 2) to Ni. Groups 2a and 2b were 83% (5/6) and 75% (15/22) metal reactive (to Co, Cr, or Ni), respectively. Of the n = 32 metal-reactive subjects to Co, Cr, or Ni (SI > 2), n = 22/32 demonstrated >2-fold elevations in % of T-cell or B-cell activation (CD25+, CD69+) to metal challenge when compared with untreated control. 18/22 metal-activated subjects demonstrated an exclusively T-cell or B-cell activation response to metal challenge, where 6/18 demonstrated exclusively B-cell activation and 12/18 demonstrated a T-cell only response, as measured by surface activation markers CD25+ and CD69+. However, there was no direct correlation (R(2) < 0.1) between lymphocyte proliferation and % T-cell or B-cell activation (CD25+:CD69+). Proliferation assays (LTT) showed greater ability to detect metal reactivity than did subject-dependent results of flow-cytometry analysis of T-cell or B-cell activation. The high incidence of lymphocyte reactivity and activation indicate that more complex than initially hypothesized immune responses may contribute to the etiology of debris-induced osteolysis in metal-sensitive individuals.


Asunto(s)
Linfocitos B/fisiología , Materiales Biocompatibles/farmacología , Activación de Linfocitos/efectos de los fármacos , Metales/farmacología , Prótesis e Implantes , Linfocitos T/fisiología , Líquidos Corporales/química , Proliferación Celular/efectos de los fármacos , Citometría de Flujo , Humanos , Hipersensibilidad/inmunología , Prótesis Articulares , Metales/química , Microscopía Confocal
19.
SAS J ; 3(4): 143-60, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-25802640

RESUMEN

BACKGROUND: Biologic-reactivity to implant-debris is the primary determinant of long-term clinical performance. The following reviews: 1) the physical aspects of spinal-implant debris and 2) the local and systemic biologic responses to implant debris. METHODS: Methods included are: 1) gravimetric wear analysis; 2) SEM and LALLS; 3) metal-ion analysis; 4) ELISA, toxicity testing, patch testing; and 5) metal-lymphocyte transformation testing (metal-LTT). RESULTS: Wear and corrosion of spine-implants produce particles and ions. Particles (0.01-1000 µm) are generally submicron ( <1 µm). Wear rates of metal-on-polymer and metal-on-metal disc arthroplasties are approximately 2-20 and 1 mm(3)/yr, respectively. Metal-on-metal total disc replacement components have significant increases in circulating metal (less than 10-fold that of controls at 4 ppb-Co and 3 ppb-Cr or ng/mL). Debris reactivity is local and systemic. Local inflammation is caused primarily by ingestion of debris by local macrophages, which produce pro-inflammatory cytokines TNFα, IL-1ß, IL-6, and PGE2. Systemic responses associated with implant-debris have been limited to hypersensitivity reactions. Elevated amounts of in the liver, spleen, etc of patients with failed TJA have not been associated with remote toxicological or carcinogenic pathology to date. Implant debris are differentially bioreactive. Greater numbers are pro-inflammatory; the smaller-sized debris are more bioreactive by virtue of their greater numbers (dose) for a given amount of implant mass loss (one 100-µm-diameter particle is equivalent in mass to 1 million 1-µm-diameter particles). Elongated particles are pro-inflammatory (ie, aspect ratio of greater than 3). Metal particles are more proinflammatory than polymers, ceteris paribus. CONCLUSION: Spinal arthroplasty designs have been in use for more than 20 years internationally; therefore, concerns about neuropathology, toxicity, and carcinogenicity are mitigated. Debris-induced inflammation still depends on the individual and the type of debris. The consequence of debris-induced inflammation is continued; vigilance by physicians is recommended monitoring of spinal implants using physical exams and testing of metal content and bioreactivity, as is planning for the likelihood of revision in younger individuals.

20.
J Orthop Surg Res ; 3: 6, 2008 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-18271968

RESUMEN

BACKGROUND: All prostheses with metallic components release metal debris that can potentially activate the immune system. However, implant-related metal hyper-reactivity has not been well characterized. In this study, we hypothesized that adaptive immunity reaction(s), particularly T-helper type 1 (Th1) responses, will be dominant in any metal-reactivity responses of patients with total joint replacements (TJAs). We tested this hypothesis by evaluating lymphocyte reactivity to metal "ions" in subjects with and without total hip replacements, using proliferation assays and cytokine analysis. METHODS: Lymphocytes from young healthy individuals without an implant or a history of metal allergy (Group 1: n = 8) were used to assess lymphocyte responses to metal challenge agents. In addition, individuals (Group 2: n = 15) with well functioning total hip arthroplasties (average Harris Hip Score = 91, average time in-situ 158 months) were studied. Age matched controls with no implants were also used for comparison (Group 3, n = 8, 4 male, 4 female average age 70, range 49-80). Group 1 subjects' lymphocyte proliferation response to Aluminum+3, Cobalt+2, Chromium+3, Copper+2, Iron+3, Molybdenum+5, Manganeese+2, Nickel+2, Vanadium+3 and Sodium+2 chloride solutions at a variety of concentrations (0.0, 0.05, 0.1, 0.5, 1.0 and 10.0 mM) was studied to establish toxicity thresholds. Mononuclear cells from Group 2 and 3 subjects were challenged with 0.1 mM CrCl3, 0.1 mM NiCl2, 0.1 mM CoCl2 and approx. 0.001 mM titanium and the reactions measured with proliferation assays and cytokine analysis to determine T-cell subtype prominence. RESULTS: Primary lymphocytes from patients with well functioning total hip replacements demonstrated a higher incidence and greater magnitude of reactivity to chromium than young healthy controls (p < 0.03). Of the 15 metal ion-challenged subjects with well functioning total hip arthroplasties, 7 demonstrated a proliferative response to Chromium, Nickel, Cobalt and/or Titanium (as defined by a statistically significant >2 fold stimulation index response, p < 0.05) and were designated as metal-reactive. Metals such as Cobalt, Copper, Manganese, and Vanadium were toxic at concentrations as low as 0.5 mM while other metals, such as Aluminum, Chromium, Iron, Molybdenum, and Nickel, became toxic at much higher concentrations (>10 mM). The differential secretion of signature T-cell subsets' cytokines (Th1 and Th2 lymphocytes releasing IFN-gamma and IL-4, respectively) between those total hip arthroplasty subjects which demonstrated metal-reactivity and those that did not, indicated a Th1 type (IFN-gamma) pro-inflammatory response. CONCLUSION: Elevated proliferation and production of IFN-gamma to metals in hip arthroplasty subjects' lymphocytes indicates that a Th1 (vs. Th2) type response is likely associated with any metal induced reactivity. The involvement of an elevated and specific lymphocyte response suggests an adaptive (macrophage recruiting) immunity response to metallic implant debris rather than an innate (nonspecific) immune response.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...