284 research outputs found

    Th1 type lymphocyte reactivity to metals in patients with total hip arthroplasty

    Get PDF
    <p>Abstract</p> <p>Background</p> <p>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.</p> <p>Methods</p> <p>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<sup>+3</sup>, Cobalt<sup>+2</sup>, Chromium<sup>+3</sup>, Copper<sup>+2</sup>, Iron<sup>+3</sup>, Molybdenum<sup>+5</sup>, Manganeese<sup>+2</sup>, Nickel<sup>+2</sup>, Vanadium<sup>+3 </sup>and Sodium<sup>+2 </sup>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 CrCl<sub>3</sub>, 0.1 mM NiCl<sub>2</sub>, 0.1 mM CoCl<sub>2 </sub>and approx. 0.001 mM titanium and the reactions measured with proliferation assays and cytokine analysis to determine T-cell subtype prominence.</p> <p>Results</p> <p>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.</p> <p>Conclusion</p> <p>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 <it>adaptive </it>(macrophage recruiting) immunity response to metallic implant debris rather than an <it>innate </it>(nonspecific) immune response.</p

    Postoperative spinal infection mimicking systemic vasculitis with titanium-spinal implants

    Get PDF
    <p>Abstract</p> <p>Background</p> <p>Secondary systemic vasculitis after posterior spinal fusion surgery is rare. It is usually related to over-reaction of immune-system, to genetic factors, toxicity, infection or metal allergies.</p> <p>Case Description</p> <p>A 14 year-old girl with a history of extended posterior spinal fusion due to idiopathic scoliosis presented to our department with diffuse erythema and nephritis (macroscopic hemuresis and proteinuria) 5 months post surgery. The surgical trauma had no signs of inflammation or infection. The blood markers ESR and CRP were increased. Skin tests were positive for nickel allergy, which is a content of titanium alloy. The patient received corticosteroids systematically (hydrocortisone 10 mg) for 6 months, leading to total recess of skin and systemic reaction. However, a palpable mass close to the surgical wound raised the suspicion of a late infection. The patient had a second surgery consisting of surgical debridement and one stage revision of posterior spinal instrumentation. Intraoperative cultures were positive to Staphylococcus aureus. Intravenous antibiotics were administered. The patient is now free of symptoms 24 months post revision surgery without any signs of recurrence of either vasculitis or infection.</p> <p>Literature Review</p> <p>Systemic vasculitis after spinal surgery is exceptionally rare. Causative factors are broad and sometimes controversial. In general, it is associated with allergy to metal ions. This is usually addressed with metal on metal total hip bearings. In spinal surgery, titanium implants are considered to be inert and only few reports have presented cases with systemic vasculitides. Therefore, other etiologies of immune over-reaction should always be considered, such as drug toxicity, infection, or genetic predisposition.</p> <p>Purposes and Clinical Relevance</p> <p>Our purpose was to highlight the difficulties during the diagnostic work-up for systemic vasculitis and management in cases of posterior spinal surgery.</p

    Bearings in Hip Arthroplasty:Joint Registries vs Precision Medicine: Review Article

    Get PDF
    Background: Precision medicine has been adopted in a range of clinical settings where omics data have led to greater characterisation of disease and stratification of patients into subcategories of phenotypes and pathologies. However, in orthopaedics, precision medicine lags behind other disciplines such as cancer. Joint registries have now amassed a huge body of data pertaining to implant performance which can be broken down into performance statistics for different material types in different cohorts of patients. The National Joint Registry of England, Wales and Northern Ireland (NJR) is now one of the largest datasets available. Other registries such as those from Sweden and Australia however contain longer follow-up. Together, these registries can provide a wealth of informative for the orthopaedics community when considering which implant to give to any particular patient. Questions/Purposes: We aim to explore the benefits of combining multiple large data streams including joint registries, published data on osteoarthritis (OA) pathogenesis and pathology and data concerning performance of each implant material combination in terms of biocompatibility. We believe that this analysis will provide a comprehensive overview of implant performance hopefully aiding surgeons in making more informed choices about which implant should be used in which patient. Methods: Data from three joint registries were combined with established literature to highlight the heterogeneity of OA disease and the different clinical outcomes following arthroplasty with a range of material types. Results: This review confirms that joint registries are unable to consider differences in arthritis presentation or underlying drivers of pathology. OA is now recognised to present with varying pathology with differing morbidity in different patient populations. Equally, just as OA is a heterogeneous disease, there are disparate responses to wear debris from different material combinations used in joint replacement surgery. This has been highlighted by recent high-profile scrutiny of early failure of metal-on-metal total hip replacement (THR) implants. Conclusions: Bringing together data from joint registries, biomarker analysis, phenotyping of OA patients and knowledge of how different patients respond to implant debris will lead to a truly personalised approach to treating OA patients, ensuring that the correct implant is given to the correct patient at the correct time

    Fretting corrosion of hafnium in simulated body fluids.

    Get PDF
    Hafnium has been suggested as an interesting material for biomedical applications due to its good biocompatibility and osteogenesis. However, its behaviour under fretting corrosion conditions, found in applications such as dental and joint implants, has not been studied in depth. A three-electrode electrochemical cell integrated with a ball-on-flat reciprocating tribometer was used to investigate the corrosion of hafnium and commercially pure (CP) titanium in simulated body fluids. An increased susceptibility to pitting corrosion was observed when hafnium was subjected to fretting. Open circuit potential measurements showed a more severe mechanical depassivation due to fretting in the case of CP titanium in comparison to hafnium. In addition, the anodic currents measured during potentiostatic tests were also higher for CP titanium

    Exposure effects of endotoxin-free titanium-based wear particles to human osteoblasts

    Get PDF
    Titanium-based materials are widely employed by the biomedical industry in orthopedic and dental implants. However, when placed into the human body, these materials are highly susceptible to degradation processes, such as corrosion, wear, and tribocorrosion. As a consequence, metallic ions or particles (debris) may be released, and although several studies have been conducted in recent years to better understand the effects of their exposure to living cells, a consensual opinion has not yet been obtained. In this work, we produced metallic based wear particles by tribological tests carried out on Ti-6Al-4V and Ti-15Zr-15Mo alloys. They were posteriorly physicochemically characterized according to their crystal structure, size, morphology, and chemical composition and compared to Ti-6Al-4V commercially available particles. Finally, adsorbed endotoxins were removed (by applying a specific thermal treatment) and endotoxin-free particles were used in cell experiments to evaluate effects of their exposure to human osteoblasts (MG-63 and HOb), namely cell viability/metabolism, proinflammatory cytokine production (IL-6 and PGE2), and susceptibility to internalization processes. Our results indicate that tribologically-obtained wear particles exhibit fundamental differences in terms of size (smaller) and morphology (irregular shapes and rough surfaces) when compared to the commercial ones. Consequently, both Ti-6Al-4V and Ti-15Zr-15Mo particles were able to induce more pronounced effects on cell viability (decrease) and cytokine production (increase) than did Ti-6Al-4V commercial particles. Furthermore, both types of wear particles penetrated osteoblast membranes and were internalized by the cells. Influences on cytokine production by endotoxins were also demonstrated.This work was supported by Fundacao de Amparo a Pesquisa do Estado de Sao Paulo - FAPESP (2015/50280-5 and 2017/24300-4), Fundacao para a Ciencia e Tecnologia - FCT (UID/EEA/04436/2013), Coordenacao de Aperfeicoamento de Pessoal de Nivel Superior - CAPES (Finance Code 0001), FCT/CAPES Joint Research Project (99999.008666/2014-08), FCT COMPETE 2020 (POCI-01-0145-FEDER-006941 and POCI-01-0145-FEDER-007265) and M-ERA-NET (0001/2015)

    Organic electrode coatings for next-generation neural interfaces

    Get PDF
    Traditional neuronal interfaces utilize metallic electrodes which in recent years have reached a plateau in terms of the ability to provide safe stimulation at high resolution or rather with high densities of microelectrodes with improved spatial selectivity. To achieve higher resolution it has become clear that reducing the size of electrodes is required to enable higher electrode counts from the implant device. The limitations of interfacing electrodes including low charge injection limits, mechanical mismatch and foreign body response can be addressed through the use of organic electrode coatings which typically provide a softer, more roughened surface to enable both improved charge transfer and lower mechanical mismatch with neural tissue. Coating electrodes with conductive polymers or carbon nanotubes offers a substantial increase in charge transfer area compared to conventional platinum electrodes. These organic conductors provide safe electrical stimulation of tissue while avoiding undesirable chemical reactions and cell damage. However, the mechanical properties of conductive polymers are not ideal, as they are quite brittle. Hydrogel polymers present a versatile coating option for electrodes as they can be chemically modified to provide a soft and conductive scaffold. However, the in vivo chronic inflammatory response of these conductive hydrogels remains unknown. A more recent approach proposes tissue engineering the electrode interface through the use of encapsulated neurons within hydrogel coatings. This approach may provide a method for activating tissue at the cellular scale, however, several technological challenges must be addressed to demonstrate feasibility of this innovative idea. The review focuses on the various organic coatings which have been investigated to improve neural interface electrodes
    corecore