Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Nat Mater ; 22(8): 1039-1046, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37500957

RESUMO

Hydrogels are attractive materials for tissue engineering, but efforts to date have shown limited ability to produce the microstructural features necessary to promote cellular self-organization into hierarchical three-dimensional (3D) organ models. Here we develop a hydrogel ink containing prefabricated gelatin fibres to print 3D organ-level scaffolds that recapitulate the intra- and intercellular organization of the heart. The addition of prefabricated gelatin fibres to hydrogels enables the tailoring of the ink rheology, allowing for a controlled sol-gel transition to achieve precise printing of free-standing 3D structures without additional supporting materials. Shear-induced alignment of fibres during ink extrusion provides microscale geometric cues that promote the self-organization of cultured human cardiomyocytes into anisotropic muscular tissues in vitro. The resulting 3D-printed ventricle in vitro model exhibited biomimetic anisotropic electrophysiological and contractile properties.


Assuntos
Gelatina , Alicerces Teciduais , Humanos , Alicerces Teciduais/química , Gelatina/química , Miócitos Cardíacos , Engenharia Tecidual/métodos , Hidrogéis/química , Impressão Tridimensional
2.
Circulation ; 141(4): 285-300, 2020 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-31707831

RESUMO

BACKGROUND: Current differentiation protocols to produce cardiomyocytes from human induced pluripotent stem cells (iPSCs) are capable of generating highly pure cardiomyocyte populations as determined by expression of cardiac troponin T. However, these cardiomyocytes remain immature, more closely resembling the fetal state, with a lower maximum contractile force, slower upstroke velocity, and immature mitochondrial function compared with adult cardiomyocytes. Immaturity of iPSC-derived cardiomyocytes may be a significant barrier to clinical translation of cardiomyocyte cell therapies for heart disease. During development, cardiomyocytes undergo a shift from a proliferative state in the fetus to a more mature but quiescent state after birth. The mechanistic target of rapamycin (mTOR)-signaling pathway plays a key role in nutrient sensing and growth. We hypothesized that transient inhibition of the mTOR-signaling pathway could lead cardiomyocytes to a quiescent state and enhance cardiomyocyte maturation. METHODS: Cardiomyocytes were differentiated from 3 human iPSC lines using small molecules to modulate the Wnt pathway. Torin1 (0 to 200 nmol/L) was used to inhibit the mTOR pathway at various time points. We quantified contractile, metabolic, and electrophysiological properties of matured iPSC-derived cardiomyocytes. We utilized the small molecule inhibitor, pifithrin-α, to inhibit p53 signaling, and nutlin-3a, a small molecule inhibitor of MDM2 (mouse double minute 2 homolog) to upregulate and increase activation of p53. RESULTS: Torin1 (200 nmol/L) increased the percentage of quiescent cells (G0 phase) from 24% to 48% compared with vehicle control (P<0.05). Torin1 significantly increased expression of selected sarcomere proteins (including TNNI3 [troponin I, cardiac muscle]) and ion channels (including Kir2.1) in a dose-dependent manner when Torin1 was initiated after onset of cardiomyocyte beating. Torin1-treated cells had an increased relative maximum force of contraction, increased maximum oxygen consumption rate, decreased peak rise time, and increased downstroke velocity. Torin1 treatment increased protein expression of p53, and these effects were inhibited by pifithrin-α. In contrast, nutlin-3a independently upregulated p53, led to an increase in TNNI3 expression and worked synergistically with Torin1 to further increase expression of both p53 and TNNI3. CONCLUSIONS: Transient treatment of human iPSC-derived cardiomyocytes with Torin1 shifts cells to a quiescent state and enhances cardiomyocyte maturity.


Assuntos
Células-Tronco Pluripotentes Induzidas/metabolismo , Miócitos Cardíacos/metabolismo , Naftiridinas/farmacologia , Serina-Treonina Quinases TOR/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Via de Sinalização Wnt/efeitos dos fármacos , Benzotiazóis/farmacologia , Linhagem Celular , Humanos , Imidazóis/farmacologia , Células-Tronco Pluripotentes Induzidas/citologia , Miócitos Cardíacos/citologia , Piperazinas/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Tolueno/análogos & derivados , Tolueno/farmacologia , Proteína Supressora de Tumor p53/antagonistas & inibidores
3.
Stem Cell Reports ; 18(9): 1811-1826, 2023 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-37595583

RESUMO

Arrhythmogenic cardiomyopathy (ACM) is an inherited cardiac disorder that causes life-threatening arrhythmias and myocardial dysfunction. Pathogenic variants in Plakophilin-2 (PKP2), a desmosome component within specialized cardiac cell junctions, cause the majority of ACM cases. However, the molecular mechanisms by which PKP2 variants induce disease phenotypes remain unclear. Here we built bioengineered platforms using genetically modified human induced pluripotent stem cell-derived cardiomyocytes to model the early spatiotemporal process of cardiomyocyte junction assembly in vitro. Heterozygosity for truncating variant PKP2R413X reduced Wnt/ß-catenin signaling, impaired myofibrillogenesis, delayed mechanical coupling, and reduced calcium wave velocity in engineered tissues. These abnormalities were ameliorated by SB216763, which activated Wnt/ß-catenin signaling, improved cytoskeletal organization, restored cell junction integrity in cell pairs, and improved calcium wave velocity in engineered tissues. Together, these findings highlight the therapeutic potential of modulating Wnt/ß-catenin signaling in a human model of ACM.


Assuntos
Células-Tronco Pluripotentes Induzidas , Humanos , beta Catenina/genética , Sinalização do Cálcio , Junções Intercelulares , Miócitos Cardíacos , Placofilinas/genética
4.
Nat Food ; 3(6): 428-436, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-37118042

RESUMO

Food waste and food safety motivate the need for improved food packaging solutions. However, current films/coatings addressing these issues are often limited by inefficient release dynamics that require large quantities of active ingredients. Here we developed antimicrobial pullulan fibre (APF)-based packaging that is biodegradable and capable of wrapping food substrates, increasing their longevity and enhancing their safety. APFs were spun using a high-throughput system, termed focused rotary jet spinning, with water as the only solvent, allowing the incorporation of naturally derived antimicrobial agents. Using avocados as a representative example, we demonstrate that APF-coated samples had their shelf life extended by inhibited proliferation of natural microflora, and lost less weight than uncoated control samples. This work offers a promising technique to produce scalable, low-cost and environmentally friendly biodegradable antimicrobial packaging systems.

5.
Science ; 377(6602): 180-185, 2022 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-35857545

RESUMO

Helical alignments within the heart's musculature have been speculated to be important in achieving physiological pumping efficiencies. Testing this possibility is difficult, however, because it is challenging to reproduce the fine spatial features and complex structures of the heart's musculature using current techniques. Here we report focused rotary jet spinning (FRJS), an additive manufacturing approach that enables rapid fabrication of micro/nanofiber scaffolds with programmable alignments in three-dimensional geometries. Seeding these scaffolds with cardiomyocytes enabled the biofabrication of tissue-engineered ventricles, with helically aligned models displaying more uniform deformations, greater apical shortening, and increased ejection fractions compared with circumferential alignments. The ability of FRJS to control fiber arrangements in three dimensions offers a streamlined approach to fabricating tissues and organs, with this work demonstrating how helical architectures contribute to cardiac performance.


Assuntos
Ventrículos do Coração , Nanofibras , Desenho de Prótese , Engenharia Tecidual , Animais , Humanos , Miócitos Cardíacos , Nanofibras/química , Engenharia Tecidual/métodos , Alicerces Teciduais
6.
NPJ Sci Food ; 3: 20, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31646181

RESUMO

Bioprocessing applications that derive meat products from animal cell cultures require food-safe culture substrates that support volumetric expansion and maturation of adherent muscle cells. Here we demonstrate scalable production of microfibrous gelatin that supports cultured adherent muscle cells derived from cow and rabbit. As gelatin is a natural component of meat, resulting from collagen denaturation during processing and cooking, our extruded gelatin microfibers recapitulated structural and biochemical features of natural muscle tissues. Using immersion rotary jet spinning, a dry-jet wet-spinning process, we produced gelatin fibers at high rates (~ 100 g/h, dry weight) and, depending on process conditions, we tuned fiber diameters between ~ 1.3 ± 0.1 µm (mean ± SEM) and 8.7 ± 1.4 µm (mean ± SEM), which are comparable to natural collagen fibers. To inhibit fiber degradation during cell culture, we crosslinked them either chemically or by co-spinning gelatin with a microbial crosslinking enzyme. To produce meat analogs, we cultured bovine aortic smooth muscle cells and rabbit skeletal muscle myoblasts in gelatin fiber scaffolds, then used immunohistochemical staining to verify that both cell types attached to gelatin fibers and proliferated in scaffold volumes. Short-length gelatin fibers promoted cell aggregation, whereas long fibers promoted aligned muscle tissue formation. Histology, scanning electron microscopy, and mechanical testing demonstrated that cultured muscle lacked the mature contractile architecture observed in natural muscle but recapitulated some of the structural and mechanical features measured in meat products.

7.
Bioelectrochemistry ; 72(2): 141-8, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18276199

RESUMO

We report the use of dielectrophoresis (DEP) to position U-937 monocytes within a non-uniform electric field, prior to electroporation (EP) for gene delivery. DEP positioning and EP pulsing were both accomplished using a common set of inert planar electrodes, micro-fabricated on a glass substrate. A single-shell model of the cell's dielectric properties and finite-element modeling of the electric field distribution permitted us to predict the major features of cell positioning. The extent to which electric pulses increased the permeability of the cell membranes to fluorescent molecules and to pEGFPLuc DNA plasmids were found to depend on prior positioning. For a given set of pulse parameters, EP was either irreversible (resulting in cytolysis), reversible (leading to gene delivery), or not detectable, depending on where cells were positioned. Our results clearly demonstrate that position-dependent EP of cells in a non-uniform electric field can be controlled by DEP.


Assuntos
Elétrons , Eletroporação/métodos , Monócitos/química , Monócitos/metabolismo , Transgenes , Linfoma de Burkitt , Sobrevivência Celular , Eletroforese , Humanos , Modelos Biológicos , Monócitos/citologia
8.
Biomaterials ; 172: 30-40, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29715593

RESUMO

Native and engineered tissue development are regulated by the integrative effects of multiple microenvironmental stimuli. Microfabricated bioreactor array platforms can efficiently dissect cue-response networks, and have recently integrated critical 2D and 3D mechanical stimulation for greater physiological relevance. However, a limitation of these approaches is that assessment of tissue functional properties is typically limited to end-point analyses. Here we report a new deformable membrane platform with integrated strain sensors that enables mechanical stretching or compression of 3D cell-hydrogel arrays and simultaneous measurement of hydrogel construct stiffness in situ. We tested the ability of the integrated strain sensors to measure the evolution of the stiffness of cell-hydrogel constructs for two cases. First, we demonstrated in situ stiffness monitoring of degradable poly (ethylene glycol)-norbornene (PEG-NB) hydrogels embedded with mesenchymal stromal cells (MSCs) and cultured with or without cyclic tensile stimulation for up to 15 days. Whereas statically-cultured hydrogels degraded and softened throughout the culture period, mechanically-stimulated gels initially softened and then recovered their stiffness corresponding to extensive cell network and collagen production. Second, we demonstrated in situ measurement of compressive stiffening of MSC-seeded PEG-NB gels cultured statically under osteogenic conditions, corresponding to increased mineralization and cellularization. This measurement technique can be generalized to other relevant bioreactor and organ-on-a-chip platforms to facilitate online, non-invasive, and high-throughput functional analysis, and to provide insights into the dynamics of engineered tissue development that are otherwise not available.


Assuntos
Ensaios de Triagem em Larga Escala/instrumentação , Hidrogéis/química , Teste de Materiais/métodos , Alicerces Teciduais/química , Adesão Celular/efeitos dos fármacos , Células Cultivadas/efeitos dos fármacos , Força Compressiva/efeitos dos fármacos , Humanos , Membranas Artificiais , Células-Tronco Mesenquimais/metabolismo , Microtecnologia/métodos , Norbornanos/química , Polietilenoglicóis/química , Engenharia Tecidual/métodos
9.
Nat Biomed Eng ; 2(12): 930-941, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-31015723

RESUMO

Laboratory studies of the heart use cell and tissue cultures to dissect heart function yet rely on animal models to measure pressure and volume dynamics. Here, we report tissue-engineered scale models of the human left ventricle, made of nanofibrous scaffolds that promote native-like anisotropic myocardial tissue genesis and chamber-level contractile function. Incorporating neonatal rat ventricular myocytes or cardiomyocytes derived from human induced pluripotent stem cells, the tissue-engineered ventricles have a diastolic chamber volume of ~500 µl (comparable to that of the native rat ventricle and approximately 1/250 the size of the human ventricle), and ejection fractions and contractile work 50-250 times smaller and 104-108 times smaller than the corresponding values for rodent and human ventricles, respectively. We also measured tissue coverage and alignment, calcium-transient propagation and pressure-volume loops in the presence or absence of test compounds. Moreover, we describe an instrumented bioreactor with ventricular-assist capabilities, and provide a proof-of-concept disease model of structural arrhythmia. The model ventricles can be evaluated with the same assays used in animal models and in clinical settings.


Assuntos
Ventrículos do Coração/citologia , Modelos Biológicos , Engenharia Tecidual , Animais , Arritmias Cardíacas/patologia , Desenho Assistido por Computador , Matriz Extracelular/química , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Contração Miocárdica , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Nanofibras/química , Polímeros/química , Ratos , Ratos Sprague-Dawley , Alicerces Teciduais/química , Função Ventricular
11.
Cardiovasc Pathol ; 25(4): 316-324, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27174867

RESUMO

Medications based on ergoline-derived dopamine and serotonin agonists are associated with off-target toxicities that include valvular heart disease (VHD). Reports of drug-induced VHD resulted in the withdrawal of appetite suppressants containing fenfluramine and phentermine from the US market in 1997 and pergolide, a Parkinson's disease medication, in 2007. Recent evidence suggests that serotonin receptor activity affected by these medications modulates cardiac valve interstitial cell activation and subsequent valvular remodeling, which can lead to cardiac valve fibrosis and dysfunction similar to that seen in carcinoid heart disease. Failure to identify these risks prior to market and continued use of similar drugs reaffirm the need to improve preclinical evaluation of drug-induced VHD. Here, we present two complimentary assays to measure stiffness and contractile stresses generated by engineered valvular tissues in vitro. As a case study, we measured the effects of acute (24 h) pergolide exposure to engineered porcine aortic valve interstitial cell (AVIC) tissues. Pergolide exposure led to increased tissue stiffness, but it decreased both basal and active contractile tone stresses generated by AVIC tissues. Pergolide exposure also disrupted AVIC tissue organization (i.e., tissue anisotropy), suggesting that the mechanical properties and contractile functionality of these tissues are governed by their ability to maintain their structure. We expect further use of these assays to identify off-target drug effects that alter the phenotypic balance of AVICs, disrupt their ability to maintain mechanical homeostasis, and lead to VHD.


Assuntos
Valva Aórtica/efeitos dos fármacos , Agonistas de Dopamina/toxicidade , Técnicas In Vitro/métodos , Pergolida/toxicidade , Rigidez Vascular , Animais , Western Blotting , Avaliação Pré-Clínica de Medicamentos , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/patologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/patologia , Contração Muscular/efeitos dos fármacos , Suínos , Engenharia Tecidual/métodos
12.
Annu Rev Pathol ; 10: 195-262, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25621660

RESUMO

The ultimate goal of most biomedical research is to gain greater insight into mechanisms of human disease or to develop new and improved therapies or diagnostics. Although great advances have been made in terms of developing disease models in animals, such as transgenic mice, many of these models fail to faithfully recapitulate the human condition. In addition, it is difficult to identify critical cellular and molecular contributors to disease or to vary them independently in whole-animal models. This challenge has attracted the interest of engineers, who have begun to collaborate with biologists to leverage recent advances in tissue engineering and microfabrication to develop novel in vitro models of disease. As these models are synthetic systems, specific molecular factors and individual cell types, including parenchymal cells, vascular cells, and immune cells, can be varied independently while simultaneously measuring system-level responses in real time. In this article, we provide some examples of these efforts, including engineered models of diseases of the heart, lung, intestine, liver, kidney, cartilage, skin and vascular, endocrine, musculoskeletal, and nervous systems, as well as models of infectious diseases and cancer. We also describe how engineered in vitro models can be combined with human inducible pluripotent stem cells to enable new insights into a broad variety of disease mechanisms, as well as provide a test bed for screening new therapies.


Assuntos
Modelos Biológicos , Patologia/métodos , Animais , Doença , Humanos , Técnicas In Vitro
13.
J R Soc Interface ; 10(84): 20130179, 2013 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-23635493

RESUMO

Experimental control over progenitor cell lineage specification can be achieved by modulating properties of the cell's microenvironment. These include physical properties of the cell adhesion substrate, such as rigidity, topography and deformation owing to dynamic mechanical forces. Multipotent mesenchymal stem cells (MSCs) generate contractile forces to sense and remodel their extracellular microenvironments and thereby obtain information that directs broad aspects of MSC function, including lineage specification. Various physical factors are important regulators of MSC function, but improved understanding of MSC mechanobiology requires novel experimental platforms. Engineers are bridging this gap by developing tools to control mechanical factors with improved precision and throughput, thereby enabling biological investigation of mechanics-driven MSC function. In this review, we introduce MSC mechanobiology and review emerging cell culture platforms that enable new insights into mechanobiological control of MSCs. Our main goals are to provide engineers and microtechnology developers with an up-to-date description of MSC mechanobiology that is relevant to the design of experimental platforms and to introduce biologists to these emerging platforms.


Assuntos
Bioengenharia/métodos , Linhagem da Célula/fisiologia , Microambiente Celular , Células-Tronco Mesenquimais/fisiologia , Fenômenos Biomecânicos , Citoesqueleto/fisiologia , Movimento/fisiologia
14.
Lab Chip ; 12(20): 4178-84, 2012 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-22885688

RESUMO

We report a microfabricated mechanical testing platform with on-chip strain sensors for in situ mechanical characterization of arrayed materials. The device is based on deformable elastomeric membranes that are actuated by pressure that is delivered through an underlying channel network. The bulging membranes compress material samples that are confined between the membranes and a rigid top-plate. Carbon nanotube-based strain sensors that exhibit strain-dependent electrical resistivity were integrated within the membranes to provide continuous read-out of membrane deflection amplitude. We used this platform to study the cyclic compression of several different silicone samples and thereby measured their elastic moduli. The results obtained using our miniaturized platform were in excellent agreement with those obtained using a commercially available mechanical testing platform and clearly demonstrated the utility of our platform for the mechanical testing of small samples in parallel. The miniaturized platform can significantly increase mechanical testing efficiency, particularly when testing of iterative sample formulations is required.


Assuntos
Elastômeros/química , Dispositivos Lab-On-A-Chip , Teste de Materiais/instrumentação , Teste de Materiais/métodos , Membranas Artificiais , Nanotubos de Carbono , Força Compressiva , Módulo de Elasticidade , Impedância Elétrica
15.
J Biomech ; 45(16): 2797-803, 2012 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-23021592

RESUMO

The mechanical properties of mammalian cells are largely determined by their cytoskeletons (CSKs), which comprise several distinct but interacting cytoplasmic molecular networks. To examine the influence of the CSK on cell mechanical properties, we deformed several mammalian cell-types (L929, CHO, HEK293, and U937) in suspension using time-varying non-uniform electric fields. Confocal fluorescent microscopy was also used to visualize and semi-quantitatively analyze CSK dimensions. We found mechanical properties of individually deformed cells to depend on cortical actin (CA) thickness. U937 and HEK293 cells with thin CA were more easily deformed than CHO and L929 cells, which bore thicker CA. In additional experiments, we treated U937 cells with latrunculin-A (Lat-A) and acrylamide (ACR), drugs that disrupt microfilaments (MF) and intermediate-filaments (IF), respectively, in order to assess their effects on the CSK and on the cell mechanical properties. We fit strain data using either a power-law or a viscoelasticity model of compliance. Our results demonstrated that maximal strain values observed under identical loading conditions were determined by the structural integrity and thickness of CA in suspended cells. Young's modulus values of individually deformed cells that were estimated using a power-law model showed a linear dependence on cortical actin thickness.


Assuntos
Citoesqueleto de Actina/fisiologia , Animais , Células CHO , Cricetinae , Cricetulus , Elasticidade , Estimulação Elétrica , Células HEK293 , Humanos , Camundongos , Estresse Mecânico , Suspensões , Células U937 , Viscosidade
16.
Arthritis Res Ther ; 12(5): R201, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20977750

RESUMO

INTRODUCTION: Objectives were to investigate whether interactions between human osteoarthritic chondrocytes and 4-hydroxynonenal (HNE)-modified type II collagen (Col II) affect cell phenotype and functions and to determine the protective role of carnosine (CAR) treatment in preventing these effects. METHODS: Human Col II was treated with HNE at different molar ratios (MR) (1:20 to 1:200; Col II:HNE). Articular chondrocytes were seeded in HNE/Col II adduct-coated plates and incubated for 48 hours. Cell morphology was studied by phase-contrast and confocal microscopy. Adhesion molecules such as intercellular adhesion molecule-1 (ICAM-1) and α1ß1 integrin at protein and mRNA levels were quantified by Western blotting, flow cytometry and real-time reverse transcription-polymerase chain reaction. Cell death, caspases activity, prostaglandin E2 (PGE2), metalloproteinase-13 (MMP-13), mitogen-activated protein kinases (MAPKs) and nuclear factor-kappa B (NF-κB) were assessed by commercial kits. Col II, cyclooxygenase-2 (COX-2), MAPK, NF-κB-p65 levels were analyzed by Western blotting. The formation of α1ß1 integrin-focal adhesion kinase (FAK) complex was revealed by immunoprecipitation. RESULTS: Col II modification by HNE at MR approximately 1:20, strongly induced ICAM-1, α1ß1 integrin and MMP-13 expression as well as extracellular signal-regulated kinases 1 and 2 (ERK1/2) and NF-κB-p65 phosphorylation without impacting cell adhesion and viability or Col II expression. However, Col II modification with HNE at MR approximately 1:200, altered chondrocyte adhesion by evoking cell death and caspase-3 activity. It inhibited α1ß1 integrin and Col II expression as well as ERK1/2 and NF-κB-p65 phosphorylation, but, in contrast, markedly elicited PGE2 release, COX-2 expression and p38 MAPK phosphorylation. Immunoprecipitation assay revealed the involvement of FAK in cell-matrix interactions through the formation of α1ß1 integrin-FAK complex. Moreover, the modification of Col II by HNE at a 1:20 or approximately 1:200 MR affects parameters of the cell shape. All these effects were prevented by CAR, an HNE-trapping drug. CONCLUSIONS: Our novel findings indicate that HNE-binding to Col II results in multiple abnormalities of chondrocyte phenotype and function, suggesting its contribution in osteoarthritis development. CAR was shown to be an efficient HNE-snaring agent capable of counteracting these outcomes.


Assuntos
Aldeídos/metabolismo , Moléculas de Adesão Celular/metabolismo , Condrócitos/metabolismo , Colágeno Tipo II/metabolismo , Osteoartrite/metabolismo , Idoso , Western Blotting , Carnosina/farmacologia , Moléculas de Adesão Celular/efeitos dos fármacos , Separação Celular , Células Cultivadas , Condrócitos/efeitos dos fármacos , Condrócitos/patologia , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/metabolismo , Matriz Extracelular/patologia , Citometria de Fluxo , Imunofluorescência , Perfilação da Expressão Gênica , Humanos , Imunoprecipitação , Microscopia Confocal , Osteoartrite/patologia , Fenótipo , RNA Mensageiro/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa