Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2023 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-37961399

RESUMO

Adipose tissue is an active endocrine organ that can signal bidirectionally to many tissues and organ systems in the body. With obesity, adipose tissue is a source of low-level inflammation that contributes to various co-morbidities and damage to downstream effector tissues. The ability to synthesize genetically engineered adipose tissue could have critical applications in studying adipokine signaling and the use of adipose tissue for novel therapeutic strategies. This study aimed to develop a method for non-viral adipogenic differentiation of genome-edited murine induced pluripotent stem cells (iPSCs) and to test the ability of such cells to engraft in mice in vivo . Designer adipocytes were created from iPSCs, which can be readily genetically engineered using CRISPR-Cas9 to knock out or insert individual genes of interest. As a model system for adipocyte-based drug delivery, an existing iPSC cell line that transcribes interleukin 1 receptor antagonist under the endogenous macrophage chemoattractant protein-1 promoter was tested for adipogenic capabilities under these same differentiation conditions. To understand the role of various adipocyte subtypes and their impact on health and disease, an efficient method was devised for inducing browning and whitening of IPSC-derived adipocytes in culture. Finally, to study the downstream effects of designer adipocytes in vivo , we transplanted the designer adipocytes into fat-free lipodystrophic mice as a model system for studying adipose signaling in different models of disease or repair. This novel translational tissue engineering and regenerative medicine platform provides an innovative approach to studying the role of adipose interorgan communication in various conditions.

3.
Eur Cell Mater ; 41: 40-51, 2021 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-33411938

RESUMO

The repair of focal cartilage defects remains one of the foremost issues in the field of orthopaedics. Chondral defects may arise from a variety of joint pathologies and left untreated, will likely progress to osteoarthritis. Current repair techniques, such as microfracture, result in short-term clinical improvements but have poor long-term outcomes. Emerging scaffold-based repair strategies have reported superior outcomes compared to microfracture and motivate the development of new biomaterials for this purpose. In this study, unique composite implants consisting of a base porous reinforcing component (woven poly(ε-caprolactone)) infiltrated with 1 of 2 hydrogels (self-assembling peptide or thermo-gelling hyaluronan) or bone marrow aspirate were evaluated. The objective was to evaluate cartilage repair with composite scaffold treatment compared to the current standard of care (microfracture) in a translationally relevant large animal model, the Yucatan minipig. While many cartilage-repair studies have shown some success in vivo, most are short term and not clinically relevant. Informed by promising 6-week findings, a 12-month study was carried out and those results are presented here. To aid in comparisons across platforms, several structural and functionally relevant outcome measures were performed. Despite positive early findings, the long-term results indicated less than optimal structural and mechanical results with respect to cartilage repair, with all treatment groups performing worse than the standard of care. This study is important in that it brings much needed attention to the importance of performing translationally relevant long-term studies in an appropriate animal model when developing new clinical cartilage repair approaches.


Assuntos
Cartilagem Articular , Animais , Materiais Biocompatíveis , Cartilagem Articular/cirurgia , Modelos Animais de Doenças , Ácido Hialurônico , Suínos , Porco Miniatura
4.
Osteoarthritis Cartilage ; 29(2): 248-256, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33246158

RESUMO

OBJECTIVE: This study aimed to examine the temporal activation of NF-κB and its relationship to the development of pain-related sensitivity and behavioral changes in a non-invasive murine knee loading model of PTOA. METHOD: Following knee injury NF-κB activity was assessed longitudinally via in vivo imaging in FVB. Cg-Tg (HIV-EGFP,luc)8Tsb/J mice. Measures of pain-related sensitivity and behavior were also assessed longitudinally for 16 weeks. Additionally, we antagonized NF-κB signaling via intra-articular delivery of an IκB kinase two antagonist to understand how local NF-κB inhibition might alter disease progression. RESULTS: Following joint injury NF-κB signaling within the knee joint was transiently increased and peaked on day 3 with an estimated 1.35 p/s/cm2/sr (95% CI 0.913.1.792 p/s/cm2/sr) fold increase in signaling when compared to control joints. Furthermore, injury resulted in the long-term development of hindpaw allodynia. Hyperalgesia withdrawal thresholds were reduced at injured knee joints, with the largest reduction occurring 2 days following injury (estimate of between group difference 129.1 g with 95% CI 60.9,197.4 g), static weight bearing on injured limbs was also reduced. Local delivery of an NF-κB inhibitor following joint injury reduced chondrocyte death and influenced the development of pain-related sensitivity but did not reduce long-term cartilage degeneration. CONCLUSION: These findings underscore the development of behavioral changes in this non-invasive loading model of PTOA and their relationships to NF-κB activation and pathology. They also highlight the potential chondroprotective effects of NF-κB inhibition shortly following joint injury despite limitations in preventing the long-term development of joint degeneration in this model of PTOA.


Assuntos
Cartilagem Articular/metabolismo , NF-kappa B/metabolismo , Osteoartrite/metabolismo , Joelho de Quadrúpedes/metabolismo , Suporte de Carga , Animais , Comportamento Animal , Fenômenos Biomecânicos , Cartilagem Articular/efeitos dos fármacos , Cartilagem Articular/patologia , Modelos Animais de Doenças , Hiperalgesia , Quinase I-kappa B/antagonistas & inibidores , Indazóis/farmacologia , Ácidos Isonicotínicos/farmacologia , Traumatismos do Joelho/complicações , Medições Luminescentes , Camundongos , Camundongos Transgênicos , NF-kappa B/efeitos dos fármacos , Osteoartrite/etiologia , Osteoartrite do Joelho/etiologia , Osteoartrite do Joelho/metabolismo , Joelho de Quadrúpedes/efeitos dos fármacos , Joelho de Quadrúpedes/lesões
5.
Osteoarthritis Cartilage ; 28(5): 626-638, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32044353

RESUMO

OBJECTIVE: Acute synovial inflammation following joint trauma is associated with posttraumatic arthritis. Synovial macrophages have been implicated in degenerative changes. In this study, we sought to elucidate the role of intra-articular macrophages in the acute inflammatory response to fracture in the mouse knee. METHOD: A closed articular fracture was induced in two models of synovial macrophage depletion: genetically-modified MaFIA mice administered AP20187 to induce programmed macrophage apoptosis, and wild-type C57BL/6 mice administered clodronate liposomes, both via intra-articular injection. Synovial inflammation, bone morphology, and levels of F4/80+ macrophages, NOS2+ M1 macrophages, and CD206+ M2 macrophages were quantified 7 days after fracture using histology and micro-computed tomography. RESULTS: Intra-articular macrophage depletion with joint injury did not reduce acute synovitis or the number of synovial macrophages 7 days after fracture in either macrophage-depleted MaFIA mice or in clodronate-treated C57BL/6 mice. In macrophage-depleted MaFIA mice, macrophage polarity shifted to a dominance of M1 macrophages and a reduction of M2 macrophages in the synovial stroma, indicating a shift in M1/M2 macrophage ratio in the joint following injury. Interestingly, MaFIA mice depleted 2 days prior to fracture demonstrated increased synovitis (P = 0.003), reduced bone mineral density (P = 0.0004), higher levels of M1 macrophages (P = 0.013), and lower levels of M2 macrophages (not statistically significant, P=0.084) compared to control-treated MaFIA mice. CONCLUSION: Our findings indicate that macrophages play a critical immunomodulatory role in the acute inflammatory response surrounding joint injury and suggest that inhibition of macrophage function can have prominent effects on joint inflammation and bone homeostasis after joint trauma.


Assuntos
Fraturas Intra-Articulares/imunologia , Traumatismos do Joelho/imunologia , Macrófagos/imunologia , Osteoartrite do Joelho/imunologia , Sinovite/imunologia , Animais , Apoptose , Proteínas de Ligação ao Cálcio/metabolismo , Ácido Clodrônico , Genes Transgênicos Suicidas , Injeções Intra-Articulares , Fraturas Intra-Articulares/diagnóstico por imagem , Fraturas Intra-Articulares/patologia , Traumatismos do Joelho/diagnóstico por imagem , Traumatismos do Joelho/patologia , Lectinas Tipo C/metabolismo , Lipossomos , Macrófagos/metabolismo , Masculino , Receptor de Manose , Lectinas de Ligação a Manose/metabolismo , Camundongos , Camundongos Transgênicos , Óxido Nítrico Sintase Tipo II/metabolismo , Receptores de Superfície Celular/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Sinovite/diagnóstico por imagem , Sinovite/patologia , Tacrolimo/análogos & derivados , Microtomografia por Raio-X
6.
Osteoarthritis Cartilage ; 28(5): 544-554, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31926267

RESUMO

Osteoarthritis (OA) is a family of degenerative diseases affecting multiple joint tissues. Despite the diverse etiology and pathogenesis of OA, increasing evidence suggests that macrophages can play a significant role in modulating joint inflammation, and thus OA severity, via various secreted mediators. Recent advances in next-generation sequencing technologies coupled with proteomic and epigenetic tools have greatly facilitated research to elucidate the embryonic origin of macrophages in various tissues including joint synovium. Furthermore, scientists have now begun to appreciate that macrophage polarization can span beyond the conventionally recognized binary states (i.e., pro-inflammatory M1-like vs anti-inflammatory M2-like) and may encompass a broad spectrum of phenotypes. Although the presence of these cells has been shown in multiple joint tissues, additional mechanistic studies are required to provide a comprehensive understanding of the precise role of these diverse macrophage populations in OA onset and progression. New approaches that can modulate macrophages into desired functional phenotypes may provide novel therapeutic strategies for preventing OA or enhancing cartilage repair and regeneration.


Assuntos
Cartilagem Articular/imunologia , Inflamação/imunologia , Macrófagos/imunologia , Osteoartrite/imunologia , Regeneração/imunologia , Tecido Adiposo/imunologia , Remodelação Óssea/imunologia , Cartilagem Articular/fisiologia , Humanos , Membrana Sinovial/imunologia
7.
Osteoarthritis Cartilage ; 27(12): 1778-1789, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31430535

RESUMO

OBJECTIVE: Emerging evidence suggests that injury to the anterior cruciate ligament (ACL) typically initiates biological changes that contribute to the development of osteoarthritis (OA). The molecular biomarkers or mediators of these biological events remain unknown. The goal of this exploratory study was to identify novel synovial fluid biomarkers associated with early biological changes following ACL injury distinct from findings in end-stage OA. METHODS: Synovial fluid was aspirated from patients with acute (≤30 days) and subacute (31-90 days) ACL tears and from patients with advanced OA and probed via tandem mass spectrometry for biomarkers to distinguish OA from ACL injury. Periostin (POSTN) was identified as a potential candidate. Further analyses of POSTN were performed in synovial fluid, OA cartilage, torn ACL remnants, and cultured cells and media by Western blot, PCR, immunostaining and ELISA. RESULTS: Synovial fluid analysis revealed that POSTN exhibited higher expression in subacute ACL injury than OA. POSTN expression was relatively low in cartilage/chondrocytes suggesting it is also produced by other intra-articular tissues. Conversely, high and time-dependent expression of POSTN in ACL tear remnants and isolated cells was consistent with the synovial fluid results. CONCLUSIONS: Elevated POSTN may provide a synovial fluid biomarker of subacute ACL injury setting separate from OA. Increased expression of POSTN in ACL suggests that the injured ACL may play a pivotal role in POSTN production, which is sensitive to time from injury. Previous studies have shown potential catabolic effects of POSTN, raising the possibility that POSTN contributes to the initiation of joint degeneration and may offer a window of opportunity to intervene in the early stages of post-traumatic OA.


Assuntos
Lesões do Ligamento Cruzado Anterior/metabolismo , Moléculas de Adesão Celular/metabolismo , Líquido Sinovial/metabolismo , Adolescente , Adulto , Ligamento Cruzado Anterior/metabolismo , Lesões do Ligamento Cruzado Anterior/genética , Western Blotting , Cartilagem Articular/metabolismo , Estudos de Casos e Controles , Moléculas de Adesão Celular/genética , Condrócitos/metabolismo , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Osteoartrite do Joelho/metabolismo , Reação em Cadeia da Polimerase , Proteômica , Espectrometria de Massas em Tandem , Adulto Jovem
8.
Curr Top Membr ; 79: 263-273, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28728820

RESUMO

Cartilage is an intrinsically mechanically sensitive tissue composed of chondrocytes as the only cell type. Chondrocyte mechanotransduction is not well understood, but recently we identified critical components of the mechanotransduction machinery demonstrating how mechanical stimulation of these cells can be converted into cellular calcium signals. Physiologic mechanical cues induce anabolic responses of (post-mitotic) chondrocytes via transient receptor potential vanilloid 4 ion channels, whereas injurious mechanical stress is transduced by Piezo1 jointly with Piezo2 ion channels. This chapter sheds light on the latter discovery and provides a rationale for follow-up questions, such as the nature of interaction between Piezo1 and Piezo2, and their tethering to the cytoskeleton. These recent insights can be leveraged toward translational medical progress to benefit diagnosis and treatment of osteoarthritis, representing a large and growing unmet medical need in the United States and large parts of the world.


Assuntos
Saúde , Canais Iônicos/metabolismo , Articulações/lesões , Animais , Condrócitos/citologia , Condrócitos/metabolismo , Humanos , Articulações/citologia , Mecanotransdução Celular , Estresse Mecânico
9.
Osteoarthritis Cartilage ; 24(8): 1330-9, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27063441

RESUMO

Meniscal lesions are common problems in orthopaedic surgery and sports medicine, and injury or loss of the meniscus accelerates the onset of knee osteoarthritis (OA). Despite a variety of therapeutic options in the clinics, there is a critical need for improved treatments to enhance meniscal repair. In this regard, combining gene-, cell-, and tissue engineering-based approaches is an attractive strategy to generate novel, effective therapies to treat meniscal lesions. In the present work, we provide an overview of the tools currently available to improve meniscal repair and discuss the progress and remaining challenges for potential future translation in patients.


Assuntos
Terapia Genética , Engenharia Tecidual , Humanos , Meniscos Tibiais , Menisco , Lesões do Menisco Tibial , Cicatrização
10.
Osteoarthritis Cartilage ; 24(9): 1528-34, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27090577

RESUMO

OBJECTIVE: To investigate the effects of pain coping skills training (PCST) and a lifestyle behavioral weight management (BWM) program on inflammatory markers and biomarker associations with pain and function in the OA LIFE study. METHOD: Serum samples were available from a subset (N = 169) of the overweight or obese knee OA participants in the OA LIFE study that evaluated: PCST, BWM, combined PCST + BWM, or standard care (SC). Inflammatory markers (hsCRP, IL-1ra, IL-1ß, IL-6, IL-8, TNF-α, TNFRI, TNFRII, and hyaluronic acid (HA)), and adipokines (leptin and adiponectin) were measured before and after the 24-week treatment period. Biomarkers were assessed for effects of treatment and for associations with change in weight, pain and disability (unadjusted and adjusted for age, race, sex, baseline body mass index (BMI), and baseline biomarker concentration). RESULTS: PCST + BWM was associated with significant reductions in hsCRP (P = 0.0014), IL-6 (P = 0.0075), and leptin (P = 0.0001). After adjustment, there was a significant effect of PCST + BWM on changes in leptin (b = -0.19, P = 0.01) and IL-6 (b = -0.25, P = 0.02) relative to SC. Reductions in leptin and IL-6 were significantly correlated with reductions in weight, BMI and Western Ontario and McMaster Universities Arthritis Index (WOMAC) pain; reductions in IL-6 were correlated with improvements in WOMAC and Arthritis Impact Measurement Scales (AIMS) physical function. By mediation analyses, weight loss was responsible for 54% of the change in IL-6 and all of the change in leptin. CONCLUSIONS: OA-related inflammatory markers were reduced by a 24-week combined PCST + BWM intervention. This suggests that the inflammatory state can be successfully modified in the context of a readily instituted clinical intervention with a positive clinical outcome.


Assuntos
Osteoartrite do Joelho , Adipócitos , Biomarcadores , Cognição , Humanos , Inflamação , Ontário
11.
Osteoarthritis Cartilage ; 23(10): 1627-38, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26003950

RESUMO

Animal models of osteoarthritis (OA) are essential tools for investigating the development of the disease on a more rapid timeline than human OA. Mice are particularly useful due to the plethora of genetically modified or inbred mouse strains available. The majority of available mouse models of OA use a joint injury or other acute insult to initiate joint degeneration, representing post-traumatic osteoarthritis (PTOA). However, no consensus exists on which injury methods are most translatable to human OA. Currently, surgical injury methods are most commonly used for studies of OA in mice; however, these methods may have confounding effects due to the surgical/invasive injury procedure itself, rather than the targeted joint injury. Non-invasive injury methods avoid this complication by mechanically inducing a joint injury externally, without breaking the skin or disrupting the joint. In this regard, non-invasive injury models may be crucial for investigating early adaptive processes initiated at the time of injury, and may be more representative of human OA in which injury is induced mechanically. A small number of non-invasive mouse models of PTOA have been described within the last few years, including intra-articular fracture of tibial subchondral bone, cyclic tibial compression loading of articular cartilage, and anterior cruciate ligament (ACL) rupture via tibial compression overload. This review describes the methods used to induce joint injury in each of these non-invasive models, and presents the findings of studies utilizing these models. Altogether, these non-invasive mouse models represent a unique and important spectrum of animal models for studying different aspects of PTOA.


Assuntos
Lesões do Ligamento Cruzado Anterior , Cartilagem Articular/lesões , Modelos Animais de Doenças , Traumatismos do Joelho/complicações , Camundongos , Osteoartrite do Joelho/etiologia , Tíbia/lesões , Animais , Fraturas Intra-Articulares , Fraturas da Tíbia
12.
Eur Cell Mater ; 29: 124-39; discussion 139-40, 2015 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-25636786

RESUMO

Post-traumatic arthritis (PTA) is a rapidly progressive form of arthritis that develops due to joint injury, including articular fracture. Current treatments are limited to surgical restoration and stabilization of the joint; however, evidence suggests that PTA progression is mediated by the upregulation of pro-inflammatory cytokines, such as interleukin-1 (IL-1) or tumor necrosis factor-α (TNF-α). Although these cytokines provide potential therapeutic targets for PTA, intra-articular injections of anti-cytokine therapies have proven difficult due to rapid clearance from the joint space. In this study, we examined the ability of a cross-linked elastin-like polypeptide (xELP) drug depot to provide sustained intra-articular delivery of IL-1 and TNF-α inhibitors as a beneficial therapy. Mice sustained a closed intra-articular tibial plateau fracture; treatment groups received a single intra-articular injection of drug encapsulated in xELP. Arthritic changes were assessed 4 and 8 weeks after fracture. Inhibition of IL-1 significantly reduced the severity of cartilage degeneration and synovitis. Inhibition of TNF-α alone or with IL-1 led to deleterious effects in bone morphology, articular cartilage degeneration, and synovitis. These findings suggest that IL-1 plays a critical role in the pathogenesis of PTA following articular fracture, and sustained intra-articular cytokine inhibition may provide a therapeutic approach for reducing or preventing joint degeneration following trauma.


Assuntos
Artrite Experimental/tratamento farmacológico , Cartilagem Articular/efeitos dos fármacos , Preparações de Ação Retardada/farmacologia , Proteína Antagonista do Receptor de Interleucina 1/farmacologia , Peptídeos/farmacologia , Sequência de Aminoácidos , Animais , Artrite Experimental/etiologia , Artrite Experimental/metabolismo , Proteína de Matriz Oligomérica de Cartilagem/metabolismo , Cartilagem Articular/diagnóstico por imagem , Cartilagem Articular/lesões , Preparações de Ação Retardada/administração & dosagem , Preparações de Ação Retardada/química , Sistemas de Liberação de Medicamentos/métodos , Elastina/química , Injeções Intra-Articulares , Proteína Antagonista do Receptor de Interleucina 1/administração & dosagem , Proteína Antagonista do Receptor de Interleucina 1/química , Interleucina-1/antagonistas & inibidores , Interleucina-1/metabolismo , Masculino , Metaloproteinase 3 da Matriz/sangue , Metaloproteinase 3 da Matriz/metabolismo , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Peptídeos/administração & dosagem , Peptídeos/química , Líquido Sinovial/efeitos dos fármacos , Líquido Sinovial/metabolismo , Temperatura , Fatores de Tempo , Resultado do Tratamento , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/metabolismo , Ferimentos e Lesões/complicações , Microtomografia por Raio-X
13.
Eur Cell Mater ; 27: 12-6, 2014 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-24802612

RESUMO

An AO Foundation (Davos, Switzerland) sponsored workshop "Cell Therapy in Cartilage Repair" from the Symposium "Where Science meets Clinics" (September 5-7, 2013, Davos) gathered leaders from medicine, science, industry, and regulatory organisations to debate the vision of cell therapy in articular cartilage repair and the measures that could be taken to narrow the gap between vision and current practice. Cell-based therapy is already in clinical use to enhance the repair of cartilage lesions, with procedures such as microfracture and articular chondrocyte implantation. However, even though long term follow up is good from a clinical perspective and some of the most rigorous randomised controlled trials in the regenerative medicine/orthopaedics field show beneficial effect, none of these options have proved successful in restoring the original articular cartilage structure and functionality in patients so far. With the remarkable recent advances in experimental research in cell biology (new sources for chondrocytes, stem cells), molecular biology (growth factors, genes), biomaterials, biomechanics, and translational science, a combined effort between scientists and clinicians with broad expertise may allow development of an improved cell therapy for cartilage repair. This position paper describes the current state of the art in the field to help define a procedure adapted to the clinical situation for upcoming translation in the patient.


Assuntos
Cartilagem Articular/fisiologia , Regeneração Tecidual Guiada/tendências , Regeneração , Animais , Cartilagem Articular/cirurgia , Regeneração Tecidual Guiada/métodos , Humanos
14.
Osteoarthritis Cartilage ; 22(6): 747-55, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24752039

RESUMO

OBJECTIVE: Pathological gaits have been shown to limit transfer between potential (PE) and kinetic (KE) energy during walking, which can increase locomotor costs. The purpose of this study was to examine whether energy exchange would be limited in people with knee osteoarthritis (OA). METHODS: Ground reaction forces during walking were collected from 93 subjects with symptomatic knee OA (self-selected and fast speeds) and 13 healthy controls (self-selected speed) and used to calculate their center of mass (COM) movements, PE and KE relationships, and energy recovery during a stride. Correlations and linear regressions examined the impact of energy fluctuation phase and amplitude, walking velocity, body mass, self-reported pain, and radiographic severity on recovery. Paired t-tests were run to compare energy recovery between cohorts. RESULTS: Symptomatic knee OA subjects displayed lower energetic recovery during self-selected walking speeds than healthy controls (P = 0.0018). PE and KE phase relationships explained the majority (66%) of variance in recovery. Recovery had a complex relationship with velocity and its change across speeds was significantly influenced by the self-selected walking speed of each subject. Neither radiographic OA scores nor subject self-reported measures demonstrated any relationship with energy recovery. CONCLUSIONS: Knee OA reduces effective exchange of PE and KE, potentially increasing the muscular work required to control movements of the COM. Gait retraining may return subjects to more normal patterns of energy exchange and allow them to reduce fatigue.


Assuntos
Aceleração , Metabolismo Energético/fisiologia , Limitação da Mobilidade , Osteoartrite do Joelho/diagnóstico , Caminhada/fisiologia , Idoso , Antropometria , Fenômenos Biomecânicos , Índice de Massa Corporal , Estudos de Casos e Controles , Avaliação da Deficiência , Feminino , Marcha , Humanos , Modelos Lineares , Masculino , Pessoa de Meia-Idade , Osteoartrite do Joelho/reabilitação , Medição da Dor , Prognóstico , Valores de Referência , Índice de Gravidade de Doença , Fatores de Tempo
15.
Physiol Meas ; 34(11): 1467-82, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24149574

RESUMO

We introduce an implementation of a novel spline framework for parametrically representing electrocardiogram (ECG) waveforms. This implementation enables a flexible means to study ECG structure in large databases. Our algorithm allows researchers to identify key points in the waveform and optimally locate them in long-term recordings with minimal manual effort, thereby permitting analysis of trends in the points themselves or in metrics derived from their locations. In the work described here we estimate the location of a number of commonly-used characteristic points of the ECG signal, defined as the onsets, peaks, and offsets of the P, QRS, T, and R' waves. The algorithm applies Bayesian optimization to a linear spline representation of the ECG waveform. The location of the knots-which are the endpoints of the piecewise linear segments used in the spline representation of the signal-serve as the estimate of the waveform's characteristic points. We obtained prior information of knot times, amplitudes, and curvature from a large manually-annotated training dataset and used the priors to optimize a Bayesian figure of merit based on estimated knot locations. In cases where morphologies vary or are subject to noise, the algorithm relies more heavily on the estimated priors for its estimate of knot locations. We compared optimized knot locations from our algorithm to two sets of manual annotations on a prospective test data set comprising 200 beats from 20 subjects not in the training set. Mean errors of characteristic point locations were less than four milliseconds, and standard deviations of errors compared favorably against reference values. This framework can easily be adapted to include additional points of interest in the ECG signal or for other biomedical detection problems on quasi-periodic signals.


Assuntos
Eletrocardiografia/métodos , Processamento de Sinais Assistido por Computador , Adulto , Algoritmos , Teorema de Bayes , Humanos , Pessoa de Meia-Idade , Probabilidade , Adulto Jovem
16.
Osteoarthritis Cartilage ; 21(12): 1895-903, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24025318

RESUMO

OBJECTIVE: Osteoarthritis (OA) is a degenerative joint disease characterized by the progressive loss of articular cartilage. While macroscale degradation of the cartilage extracellular matrix (ECM) has been extensively studied, microscale changes in the chondrocyte pericellular matrix (PCM) and immediate microenvironment with OA are not fully understood. The objective of this study was to quantify osteoarthritic changes in the micromechanical properties of the ECM and PCM of human articular cartilage in situ using atomic force microscopy (AFM). METHOD: AFM elastic mapping was performed on cryosections of human cartilage harvested from both condyles of macroscopically normal and osteoarthritic knee joints. This method was used to test the hypotheses that both ECM and PCM regions exhibit a loss of mechanical properties with OA and that the size of the PCM is enlarged in OA cartilage as compared to normal tissue. RESULTS: Significant decreases were observed in both ECM and PCM moduli of 45% and 30%, respectively, on the medial condyle of OA knee joints as compared to cartilage from macroscopically normal joints. Enlargement of the PCM, as measured biomechanically, was also observed in medial condyle OA cartilage, reflecting the underlying distribution of type VI collagen in the region. No significant differences were observed in elastic moduli or their spatial distribution on the lateral condyle between normal and OA joints. CONCLUSION: Our findings provide new evidence of significant site-specific degenerative changes in the chondrocyte micromechanical environment with OA.


Assuntos
Cartilagem Articular/ultraestrutura , Condrócitos/ultraestrutura , Matriz Extracelular/ultraestrutura , Osteoartrite do Joelho/patologia , Idoso , Idoso de 80 Anos ou mais , Cartilagem Articular/metabolismo , Estudos de Casos e Controles , Colágeno Tipo VI/metabolismo , Módulo de Elasticidade , Matriz Extracelular/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Masculino , Microscopia de Força Atômica , Microscopia de Varredura por Sonda , Pessoa de Meia-Idade , Osteoartrite do Joelho/metabolismo
17.
Int J Obes (Lond) ; 37(8): 1079-87, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23164698

RESUMO

INTRODUCTION: Obesity is a major risk factor for several musculoskeletal conditions that are characterized by an imbalance of tissue remodeling. Adult stem cells are closely associated with the remodeling and potential repair of several mesodermally derived tissues such as fat, bone and cartilage. We hypothesized that obesity would alter the frequency, proliferation, multipotency and immunophenotype of adult stem cells from a variety of tissues. MATERIALS AND METHODS: Bone marrow-derived mesenchymal stem cells (MSCs), subcutaneous adipose-derived stem cells (sqASCs) and infrapatellar fat pad-derived stem cells (IFP cells) were isolated from lean and high-fat diet-induced obese mice, and their cellular properties were examined. To test the hypothesis that changes in stem cell properties were due to the increased systemic levels of free fatty acids (FFAs), we further investigated the effects of FFAs on lean stem cells in vitro. RESULTS: Obese mice showed a trend toward increased prevalence of MSCs and sqASCs in the stromal tissues. While no significant differences in cell proliferation were observed in vitro, the differentiation potential of all types of stem cells was altered by obesity. MSCs from obese mice demonstrated decreased adipogenic, osteogenic and chondrogenic potential. Obese sqASCs and IFP cells showed increased adipogenic and osteogenic differentiation, but decreased chondrogenic ability. Obese MSCs also showed decreased CD105 and increased platelet-derived growth factor receptor α expression, consistent with decreased chondrogenic potential. FFA treatment of lean stem cells significantly altered their multipotency but did not completely recapitulate the properties of obese stem cells. CONCLUSIONS: These findings support the hypothesis that obesity alters the properties of adult stem cells in a manner that depends on the cell source. These effects may be regulated in part by increased levels of FFAs, but may involve other obesity-associated cytokines. These findings contribute to our understanding of mesenchymal tissue remodeling with obesity, as well as the development of autologous stem cell therapies for obese patients.


Assuntos
Tecido Adiposo/patologia , Diferenciação Celular , Ácidos Graxos não Esterificados , Células-Tronco Mesenquimais/patologia , Obesidade/patologia , Adipogenia , Animais , Técnicas de Cultura de Células , Proliferação de Células , Células Cultivadas , Dieta Hiperlipídica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Patela , Fatores de Risco , Gordura Subcutânea/patologia
18.
Ann Biomed Eng ; 40(11): 2456-74, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22648577

RESUMO

Articular cartilage experiences significant mechanical loads during daily activities. Healthy cartilage provides the capacity for load bearing and regulates the mechanobiological processes for tissue development, maintenance, and repair. Experimental studies at multiple scales have provided a fundamental understanding of macroscopic mechanical function, evaluation of the micromechanical environment of chondrocytes, and the foundations for mechanobiological response. In addition, computational models of cartilage have offered a concise description of experimental data at many spatial levels under healthy and diseased conditions, and have served to generate hypotheses for the mechanical and biological function. Further, modeling and simulation provides a platform for predictive risk assessment, management of dysfunction, as well as a means to relate multiple spatial scales. Simulation-based investigation of cartilage comes with many challenges including both the computational burden and often insufficient availability of data for model development and validation. This review outlines recent modeling and simulation approaches to understand cartilage function from a mechanical systems perspective, and illustrates pathways to associate mechanics with biological function. Computational representations at single scales are provided from the body down to the microstructure, along with attempts to explore multiscale mechanisms of load sharing that dictate the mechanical environment of the cartilage and chondrocytes.


Assuntos
Cartilagem/fisiologia , Modelos Biológicos , Animais , Fenômenos Biomecânicos , Humanos , Articulações/fisiologia , Músculo Esquelético/fisiologia
19.
Osteoarthritis Cartilage ; 19(7): 864-73, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21619936

RESUMO

OBJECTIVE: Post-traumatic arthritis is a frequent cause of disability and occurs most commonly and predictably after articular fracture. The objective of this investigation was to examine the effect of fracture severity on acute joint pathology in a novel murine model of intra-articular fracture. DESIGN: Low and high energy articular fractures (n=25 per group) of the tibial plateau were created in adult male C57BL/6 mice. The acute effect of articular fracture severity on synovial inflammation, bone morphology, liberated fracture area, cartilage pathology, chondrocyte viability, and systemic cytokines and biomarkers levels was assessed at 0, 1, 3, 5, and 7 days post-fracture. RESULTS: Increasing intra-articular fracture severity was associated with greater acute pathology in the synovium and bone compared to control limbs, including increased global synovitis and reduced periarticular bone density and thickness. Applied fracture energy was significantly correlated with degree of liberated cortical bone surface area, indicating greater comminution. Serum concentrations of hyaluronic acid (HA) were significantly increased 1 day post-fracture. While articular fracture significantly reduced chondrocyte viability, there was no relationship between fracture severity and chondrocyte viability, cartilage degeneration, or systemic levels of cytokines and biomarkers. CONCLUSIONS: This study demonstrates that articular fracture is associated with a loss of chondrocyte viability and increased levels of systemic biomarkers, and that increased intra-articular fracture severity is associated with increased acute joint pathology in a variety of joint tissues, including synovial inflammation, cortical comminution, and bone morphology. Further characterization of the early events following articular fracture could aid in the treatment of post-traumatic arthritis.


Assuntos
Fraturas Intra-Articulares/patologia , Articulação do Joelho/patologia , Membrana Sinovial/patologia , Análise de Variância , Animais , Biomarcadores/metabolismo , Condrócitos/patologia , Citocinas/metabolismo , Modelos Animais de Doenças , Inflamação/metabolismo , Inflamação/patologia , Fraturas Intra-Articulares/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Membrana Sinovial/metabolismo
20.
J Biomech Eng ; 132(2): 024501, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20370247

RESUMO

One of the functions of the meniscus is to distribute contact forces over the articular surfaces by increasing the joint contact areas. It is widely accepted that total/partial loss of the meniscus increases the risk of joint degeneration. A short-term method for evaluating whether degenerative arthritis can be prevented or not would be to determine if the peak pressure and contact area coverage of the tibial plateau (TP) in the knee are restored at the time of implantation. Although several published studies already utilized TP contact pressure measurements as an indicator for biomechanical performance of allograft menisci, there is a paucity of a quantitative method for evaluation of these parameters in situ with a single effective parameter. In the present study, we developed such a method and used it to assess the load distribution ability of various meniscal implant configurations in human cadaveric knees (n=3). Contact pressures under the intact meniscus were measured under compression (1200 N, 0 deg flexion). Next, total meniscectomy was performed and the protocol was repeated with meniscal implants. Resultant pressure maps were evaluated for the peak pressure value, total contact area, and its distribution pattern, all with respect to the natural meniscus output. Two other measures--implant-dislocation and implant-impingement on the ligaments--were also considered. If any of these occurred, the score was zeroed. The total implant score was based on an adjusted calculation of the aforementioned measures, where the natural meniscus score was always 100. Laboratory experiments demonstrated a good correlation between qualitative and quantitative evaluations of the same pressure map outputs, especially in cases where there were contradicting indications between different parameters. Overall, the proposed approach provides a novel, validated method for quantitative assessment of the biomechanical performance of meniscal implants, which can be used in various applications ranging from bench testing of design (geometry and material of an implant) to correct implant sizing.


Assuntos
Articulação do Joelho/cirurgia , Humanos , Mecânica , Osteoartrite/cirurgia , Pressão , Pesquisa , Tíbia/cirurgia , Tíbia/transplante
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...