Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 333
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 184(18): 4597-4611, 2021 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-34478657

RESUMO

We explore the utility of bioengineered human tissues-individually or connected into physiological units-for biological research. While much smaller and simpler than their native counterparts, these tissues are complex enough to approximate distinct tissue phenotypes: molecular, structural, and functional. Unlike organoids, which form spontaneously and recapitulate development, "organs-on-a-chip" are engineered to display some specific functions of whole organs. Looking back, we discuss the key developments of this emerging technology. Thinking forward, we focus on the challenges faced to fully establish, validate, and utilize the fidelity of these models for biological research.


Assuntos
Dispositivos Lab-On-A-Chip , Modelos Biológicos , Pesquisa , Animais , Engenharia Celular , Microambiente Celular , Humanos , Engenharia Tecidual
2.
Cell ; 176(4): 913-927.e18, 2019 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-30686581

RESUMO

Tissue engineering using cardiomyocytes derived from human pluripotent stem cells holds a promise to revolutionize drug discovery, but only if limitations related to cardiac chamber specification and platform versatility can be overcome. We describe here a scalable tissue-cultivation platform that is cell source agnostic and enables drug testing under electrical pacing. The plastic platform enabled on-line noninvasive recording of passive tension, active force, contractile dynamics, and Ca2+ transients, as well as endpoint assessments of action potentials and conduction velocity. By combining directed cell differentiation with electrical field conditioning, we engineered electrophysiologically distinct atrial and ventricular tissues with chamber-specific drug responses and gene expression. We report, for the first time, engineering of heteropolar cardiac tissues containing distinct atrial and ventricular ends, and we demonstrate their spatially confined responses to serotonin and ranolazine. Uniquely, electrical conditioning for up to 8 months enabled modeling of polygenic left ventricular hypertrophy starting from patient cells.


Assuntos
Miócitos Cardíacos/citologia , Técnicas de Cultura de Tecidos/instrumentação , Engenharia Tecidual/métodos , Potenciais de Ação , Diferenciação Celular , Células Cultivadas , Fenômenos Eletrofisiológicos , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Modelos Biológicos , Miocárdio/citologia , Miócitos Cardíacos/metabolismo , Células-Tronco Pluripotentes/citologia , Técnicas de Cultura de Tecidos/métodos
3.
Nat Methods ; 19(9): 1064-1071, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-36064773

RESUMO

Engineered cardiac tissues derived from human induced pluripotent stem cells offer unique opportunities for patient-specific disease modeling, drug discovery and cardiac repair. Since the first engineered hearts were introduced over two decades ago, human induced pluripotent stem cell-based three-dimensional cardiac organoids and heart-on-a-chip systems have now become mainstays in basic cardiovascular research as valuable platforms for investigating fundamental human pathophysiology and development. However, major obstacles remain to be addressed before the field can truly advance toward commercial and clinical translation. Here we provide a snapshot of the state-of-the-art methods in cardiac tissue engineering, with a focus on in vitro models of the human heart. Looking ahead, we discuss major challenges and opportunities in the field and suggest strategies for enabling broad acceptance of engineered cardiac tissues as models of cardiac pathophysiology and testbeds for the development of therapies.


Assuntos
Células-Tronco Pluripotentes Induzidas , Engenharia Tecidual , Descoberta de Drogas , Coração/fisiologia , Humanos , Células-Tronco Pluripotentes Induzidas/fisiologia , Miócitos Cardíacos , Organoides , Engenharia Tecidual/métodos
4.
Nature ; 572(7769): E16-E17, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31363231

RESUMO

An Amendment to this paper has been published and can be accessed via a link at the top of the paper.

5.
Nature ; 556(7700): 239-243, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29618819

RESUMO

Cardiac tissues generated from human induced pluripotent stem cells (iPSCs) can serve as platforms for patient-specific studies of physiology and disease1-6. However, the predictive power of these models is presently limited by the immature state of the cells1, 2, 5, 6. Here we show that this fundamental limitation can be overcome if cardiac tissues are formed from early-stage iPSC-derived cardiomyocytes soon after the initiation of spontaneous contractions and are subjected to physical conditioning with increasing intensity over time. After only four weeks of culture, for all iPSC lines studied, such tissues displayed adult-like gene expression profiles, remarkably organized ultrastructure, physiological sarcomere length (2.2 µm) and density of mitochondria (30%), the presence of transverse tubules, oxidative metabolism, a positive force-frequency relationship and functional calcium handling. Electromechanical properties developed more slowly and did not achieve the stage of maturity seen in adult human myocardium. Tissue maturity was necessary for achieving physiological responses to isoproterenol and recapitulating pathological hypertrophy, supporting the utility of this tissue model for studies of cardiac development and disease.


Assuntos
Diferenciação Celular , Coração/crescimento & desenvolvimento , Células-Tronco Pluripotentes Induzidas/citologia , Miocárdio/citologia , Miócitos Cardíacos/citologia , Técnicas de Cultura de Tecidos , Adulto , Cálcio/metabolismo , Diferenciação Celular/genética , Metabolismo Energético/efeitos dos fármacos , Coração/efeitos dos fármacos , Humanos , Isoproterenol/farmacologia , Mitocôndrias/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/ultraestrutura , Sarcômeros/metabolismo , Transcriptoma
6.
J Mol Cell Cardiol ; 166: 137-151, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35219725

RESUMO

Ischemic and non-ischemic cardiomyopathies have distinct etiologies and underlying disease mechanisms, which require in-depth investigation for improved therapeutic interventions. The goal of this study was to use clinically obtained myocardium from healthy and heart failure patients, and characterize the changes in extracellular matrix (ECM) in ischemic and non-ischemic failing hearts, with and without mechanical unloading. Using tissue engineering methodologies, we also investigated how diseased human ECM, in the absence of systemic factors, can influence cardiomyocyte function. Heart tissues from heart failure patients with ischemic and non-ischemic cardiomyopathy were compared to explore differential disease phenotypes and reverse remodeling potential of left ventricular assisted device (LVAD) support at transcriptomic, proteomic and structural levels. The collected data demonstrated that the differential ECM compositions recapitulated the disease microenvironment and induced cardiomyocytes to undergo disease-like functional alterations. In addition, our study also revealed molecular profiles of non-ischemic and ischemic heart failure patients and explored the underlying mechanisms of etiology-specific impact on clinical outcome of LVAD support and tendency towards reverse remodeling.


Assuntos
Insuficiência Cardíaca , Coração Auxiliar , Matriz Extracelular , Coração Auxiliar/efeitos adversos , Humanos , Miocárdio/química , Proteômica
7.
Ann Plast Surg ; 89(2): 218-224, 2022 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-35276708

RESUMO

PURPOSE: Gradual elevation of periosteum from the bone surface is known to promote the adaptation of soft tissues and the formation of hard tissues. The aim of our study was to estimate the benefit of periosteal distraction osteogenesis (PDO) on de novo bone formation in a rat model. MATERIALS AND METHODS: After device placement, animals were allowed for a latency period of 7 days. Animals in the PDO group were subjected to distraction at a rate of 0.1 mm/d for 10 days. In the periosteal pumping (PP) group, the animals were subjected to distraction at a rate of 0.1 mm/d. The direction of distraction was alternated every 2 days. The animals were euthanized at 17, 31, and 45 days after surgery, and the samples were analyzed histologically and by microcomputed tomography. RESULTS: In both groups, the new bone was characterized as primary woven bone that was located at the leading edge of bone apposition. Bone volumes significantly increased throughout the observation period both in the PP group ( P = 0.018) and in the PDO group ( P < 0.001). The new bone was denser and more mature in the PP group than in the PDO group, and the difference was significant at the 31-day time point ( P = 0.024). However, the volume of the new bone was higher in the PDO at the 45-day time point ( P < 0.001). CONCLUSIONS: We propose that the PP may be applied to enhance the osteogenic capacity of periosteum without plate elevation. Because this is only a proof-of-principle study, the alternated protocol of periosteal distraction warrants evaluation in the future studies.


Assuntos
Osteogênese por Distração , Periósteo , Animais , Estudos de Viabilidade , Osteogênese , Osteogênese por Distração/métodos , Periósteo/cirurgia , Ratos , Crânio/cirurgia , Microtomografia por Raio-X
8.
Am J Physiol Gastrointest Liver Physiol ; 320(1): G1-G11, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33174453

RESUMO

Gastrointestinal disease burden continues to rise in the United States and worldwide. The development of bioengineering strategies to model gut injury or disease and to reestablish functional gut tissue could expand therapeutic options and improve clinical outcomes. Current approaches leverage a rapidly evolving gut bioengineering toolkit aimed at 1) de novo generation of gutlike tissues at multiple scales for microtissue models or implantable grafts and 2) regeneration of functional gut in vivo. Although significant progress has been made in intestinal organoid cultures and engineered tissues, development of predictive in vitro models and effective regenerative therapies remains challenging. In this review, we survey emerging bioengineering tools and recent methodological advances to identify current challenges and future opportunities in gut bioengineering for disease modeling and regenerative medicine.


Assuntos
Microbioma Gastrointestinal/fisiologia , Regeneração/fisiologia , Medicina Regenerativa , Células-Tronco/citologia , Animais , Bioengenharia/métodos , Humanos , Organoides/metabolismo
9.
Circ Res ; 124(1): 161-169, 2019 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-30605412

RESUMO

On March 1 and 2, 2018, the National Institutes of Health 2018 Progenitor Cell Translational Consortium, Cardiovascular Bioengineering Symposium, was held at the University of Alabama at Birmingham. Convergence of life sciences and engineering to advance the understanding and treatment of heart failure was the theme of the meeting. Over 150 attendees were present, and >40 scientists presented their latest work on engineering human functional myocardium for disease modeling, drug development, and heart failure research. The scientists, engineers, and physicians in the field of cardiovascular sciences met and discussed the most recent advances in their work and proposed future strategies for overcoming the major roadblocks of cardiovascular bioengineering and therapy. Particular emphasis was given for manipulation and using of stem/progenitor cells, biomaterials, and methods to provide molecular, chemical, and mechanical cues to cells to influence their identity and fate in vitro and in vivo. Collectively, these works are profoundly impacting and progressing toward deciphering the mechanisms and developing novel treatments for left ventricular dysfunction of failing hearts. Here, we present some important perspectives that emerged from this meeting.


Assuntos
Disciplinas das Ciências Biológicas , Engenharia Biomédica , Pesquisa Biomédica , Insuficiência Cardíaca , Comunicação Interdisciplinar , Animais , Comportamento Cooperativo , Difusão de Inovações , Coração/fisiopatologia , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/fisiopatologia , Insuficiência Cardíaca/terapia , Humanos , Miocárdio/metabolismo , Miocárdio/patologia , Recuperação de Função Fisiológica , Regeneração
10.
Proc Natl Acad Sci U S A ; 115(6): 1256-1261, 2018 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-29363599

RESUMO

Eight out of 10 breast cancer patients die within 5 years after the primary tumor has spread to the bones. Tumor cells disseminated from the breast roam the vasculature, colonizing perivascular niches around blood capillaries. Slow flows support the niche maintenance by driving the oxygen, nutrients, and signaling factors from the blood into the interstitial tissue, while extracellular matrix, endothelial cells, and mesenchymal stem cells regulate metastatic homing. Here, we show the feasibility of developing a perfused bone perivascular niche-on-a-chip to investigate the progression and drug resistance of breast cancer cells colonizing the bone. The model is a functional human triculture with stable vascular networks within a 3D native bone matrix cultured on a microfluidic chip. Providing the niche-on-a-chip with controlled flow velocities, shear stresses, and oxygen gradients, we established a long-lasting, self-assembled vascular network without supplementation of angiogenic factors. We further show that human bone marrow-derived mesenchymal stem cells, which have undergone phenotypical transition toward perivascular cell lineages, support the formation of capillary-like structures lining the vascular lumen. Finally, breast cancer cells exposed to interstitial flow within the bone perivascular niche-on-a-chip persist in a slow-proliferative state associated with increased drug resistance. We propose that the bone perivascular niche-on-a-chip with interstitial flow promotes the formation of stable vasculature and mediates cancer cell colonization.


Assuntos
Neoplasias Ósseas/secundário , Neoplasias da Mama/patologia , Técnicas de Cocultura/instrumentação , Dispositivos Lab-On-A-Chip , Matriz Óssea/patologia , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/patologia , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Técnicas de Cocultura/métodos , Resistencia a Medicamentos Antineoplásicos , Feminino , Humanos , Células-Tronco Mesenquimais/citologia , Oxigênio , Perfusão , Alicerces Teciduais
11.
Adv Funct Mater ; 30(37)2020 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-33708027

RESUMO

From micro-scaled capillaries to millimeter-sized arteries and veins, human vasculature spans multiple scales and cell types. The convergence of bioengineering, materials science, and stem cell biology has enabled tissue engineers to recreate the structure and function of different hierarchical levels of the vascular tree. Engineering large-scale vessels has been pursued over the past thirty years to replace or bypass damaged arteries, arterioles, and venules, and their routine application in the clinic may become a reality in the near future. Strategies to engineer meso- and microvasculature have been extensively explored to generate models to study vascular biology, drug transport, and disease progression, as well as for vascularizing engineered tissues for regenerative medicine. However, bioengineering of large-scale tissues and whole organs for transplantation, have failed to result in clinical translation due to the lack of proper integrated vasculature for effective oxygen and nutrient delivery. The development of strategies to generate multi-scale vascular networks and their direct anastomosis to host vasculature would greatly benefit this formidable goal. In this review, we discuss design considerations and technologies for engineering millimeter-, meso-, and micro-scale vessels. We further provide examples of recent state-of-the-art strategies to engineer multi-scale vasculature. Finally, we identify key challenges limiting the translation of vascularized tissues and offer our perspective on future directions for exploration.

12.
Biotechnol Bioeng ; 117(5): 1584-1596, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31985051

RESUMO

Articular cartilage injuries are a common source of joint pain and dysfunction. We hypothesized that pulsed electromagnetic fields (PEMFs) would improve growth and healing of tissue-engineered cartilage grafts in a direction-dependent manner. PEMF stimulation of engineered cartilage constructs was first evaluated in vitro using passaged adult canine chondrocytes embedded in an agarose hydrogel scaffold. PEMF coils oriented parallel to the articular surface induced superior repair stiffness compared to both perpendicular PEMF (p = .026) and control (p = .012). This was correlated with increased glycosaminoglycan deposition in both parallel and perpendicular PEMF orientations compared to control (p = .010 and .028, respectively). Following in vitro optimization, the potential clinical translation of PEMF was evaluated in a preliminary in vivo preclinical adult canine model. Engineered osteochondral constructs (∅ 6 mm × 6 mm thick, devitalized bone base) were cultured to maturity and implanted into focal defects created in the stifle (knee) joint. To assess expedited early repair, animals were assessed after a 3-month recovery period, with microfracture repairs serving as an additional clinical control. In vivo, PEMF led to a greater likelihood of normal chondrocyte (odds ratio [OR]: 2.5, p = .051) and proteoglycan (OR: 5.0, p = .013) histological scores in engineered constructs. Interestingly, engineered constructs outperformed microfracture in clinical scoring, regardless of PEMF treatment (p < .05). Overall, the studies provided evidence that PEMF stimulation enhanced engineered cartilage growth and repair, demonstrating a potential low-cost, low-risk, noninvasive treatment modality for expediting early cartilage repair.


Assuntos
Cartilagem Articular/efeitos da radiação , Campos Eletromagnéticos , Engenharia Tecidual/métodos , Cicatrização/efeitos dos fármacos , Animais , Cartilagem Articular/lesões , Células Cultivadas , Condrócitos/efeitos da radiação , Cães , Masculino , Joelho de Quadrúpedes/lesões
13.
Circ Res ; 123(2): 244-265, 2018 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-29976691

RESUMO

Some of the most significant leaps in the history of modern civilization-the development of article in China, the steam engine, which led to the European industrial revolution, and the era of computers-have occurred when science converged with engineering. Recently, the convergence of human pluripotent stem cell technology with biomaterials and bioengineering have launched a new medical innovation: functional human engineered tissue, which promises to revolutionize the treatment of failing organs including most critically, the heart. This compendium covers recent, state-of-the-art developments in the fields of cardiovascular tissue engineering, as well as the needs and challenges associated with the clinical use of these technologies. We have not attempted to provide an exhaustive review in stem cell biology and cardiac cell therapy; many other important and influential reports are certainly merit but already been discussed in several recent reviews. Our scope is limited to the engineered tissues that have been fabricated to repair or replace components of the heart (eg, valves, vessels, contractile tissue) that have been functionally compromised by diseases or developmental abnormalities. In particular, we have focused on using an engineered myocardial tissue to mitigate deficiencies in contractile function.


Assuntos
Doenças Cardiovasculares/terapia , Medicina Regenerativa/métodos , Transplante de Células-Tronco/métodos , Engenharia Tecidual/métodos , Animais , Ensaios Clínicos como Assunto , Humanos , Transplante de Células-Tronco/efeitos adversos
14.
Proc Natl Acad Sci U S A ; 114(10): 2556-2561, 2017 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-28228529

RESUMO

Standard isotropic culture fails to recapitulate the spatiotemporal gradients present during native development. Cartilage grown from human mesenchymal stem cells (hMSCs) is poorly organized and unstable in vivo. We report that human cartilage with physiologic organization and in vivo stability can be grown in vitro from self-assembling hMSCs by implementing spatiotemporal regulation during induction. Self-assembling hMSCs formed cartilage discs in Transwell inserts following isotropic chondrogenic induction with transforming growth factor ß to set up a dual-compartment culture. Following a switch in the basal compartment to a hypertrophic regimen with thyroxine, the cartilage discs underwent progressive deep-zone hypertrophy and mineralization. Concurrent chondrogenic induction in the apical compartment enabled the maintenance of functional and hyaline cartilage. Cartilage homeostasis, chondrocyte maturation, and terminal differentiation markers were all up-regulated versus isotropic control groups. We assessed the in vivo stability of the cartilage formed under different induction regimens. Cartilage formed under spatiotemporal regulation in vitro resisted endochondral ossification, retained the expression of cartilage markers, and remained organized following s.c. implantation in immunocompromised mice. In contrast, the isotropic control groups underwent endochondral ossification. Cartilage formed from hMSCs remained stable and organized in vivo. Spatiotemporal regulation during induction in vitro recapitulated some aspects of native cartilage development, and potentiated the maturation of self-assembling hMSCs into stable and organized cartilage resembling the native articular cartilage.


Assuntos
Técnicas de Cultura de Células , Condrócitos/citologia , Condrogênese/efeitos dos fármacos , Células-Tronco Mesenquimais/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Engenharia Tecidual/métodos , Animais , Biomarcadores/metabolismo , Cartilagem Articular , Diferenciação Celular/efeitos dos fármacos , Condrócitos/imunologia , Condrócitos/transplante , Condrogênese/fisiologia , Colágeno Tipo I/genética , Colágeno Tipo I/imunologia , Cultura em Câmaras de Difusão , Feminino , Expressão Gênica , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/imunologia , Camundongos , Camundongos SCID , Osteogênese/fisiologia , Cultura Primária de Células , Tiroxina/farmacologia , Alicerces Teciduais , Fator de Crescimento Transformador beta/farmacologia , Transplante Heterólogo
15.
Proc Natl Acad Sci U S A ; 112(37): 11530-5, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-26324893

RESUMO

The ability to deliver drugs to specific sites in the lung could radically improve therapeutic outcomes of a variety of lung diseases, including cystic fibrosis, severe bronchopneumonia, chronic obstructive pulmonary disease, and lung cancer. Using conventional methods for pulmonary drug administration, precise, localized delivery of exact doses of drugs to target regions remains challenging. Here we describe a more controlled delivery of soluble reagents (e.g., drugs, enzymes, and radionuclides) in microvolume liquid plugs to targeted branches of the pulmonary airway tree: upper airways, small airways (bronchioles), or the most distal alveoli. In this approach, a soluble liquid plug of very small volume (<1 mL) is instilled into the upper airways, and with programmed air ventilation of the lungs, the plug is pushed into a specific desired (more distal) airway to achieve deposition of liquid film onto the lung epithelium. The plug volume and ventilation conditions were determined by mathematical modeling of plug transport in a tubular geometry, and targeted liquid film deposition was demonstrated in rat lungs by three different in vivo imaging modalities. The experimental and modeling data suggest that instillation of microvolumes of liquid into a ventilated pulmonary airway could be an effective strategy to deliver exact doses of drugs to targeted pathologic regions of the lung, especially those inaccessible by bronchoscopy, to increase in situ efficacy of the drug and minimize systemic side effects.


Assuntos
Sistemas de Liberação de Medicamentos , Pulmão/fisiologia , Surfactantes Pulmonares/administração & dosagem , Animais , Masculino , Microscopia de Fluorescência , Modelos Teóricos , Alvéolos Pulmonares/fisiologia , Ventilação Pulmonar , Ratos , Ratos Sprague-Dawley , Mecânica Respiratória/fisiologia , Sistema Respiratório/efeitos dos fármacos
16.
Exp Cell Res ; 347(1): 1-13, 2016 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-26500109

RESUMO

The mechanisms by which macrophages control the inflammatory response, wound healing, biomaterial-interactions, and tissue regeneration appear to be related to their activation/differentiation states. Studies of macrophage behavior in vitro can be useful for elucidating their mechanisms of action, but it is not clear to what extent the source of macrophages affects their apparent behavior, potentially affecting interpretation of results. Although comparative studies of macrophage behavior with respect to cell source have been conducted, there has been no direct comparison of the three most commonly used cell sources: murine bone marrow, human monocytes from peripheral blood (PB), and the human leukemic monocytic cell line THP-1, across multiple macrophage phenotypes. In this study, we used multivariate discriminant analysis to compare the in vitro expression of genes commonly chosen to assess macrophage phenotype across all three sources of macrophages, as well as those derived from induced pluripotent stem cells (iPSCs), that were polarized towards four distinct phenotypes using the same differentiation protocols: M(LPS,IFN) (aka M1), M(IL4,IL13) (aka M2a), M(IL10) (aka M2c), and M(-) (aka M0) used as control. Several differences in gene expression trends were found among the sources of macrophages, especially between murine bone marrow-derived and human blood-derived M(LPS,IFN) and M(IL4,IL13) macrophages with respect to commonly used phenotype markers like CCR7 and genes associated with angiogenesis and tissue regeneration like FGF2 and MMP9. We found that the genes with the most similar patterns of expression among all sources were CXCL-10 and CXCL-11 for M(LPS,IFN) and CCL17 and CCL22 for M(IL4,IL13). Human PB-derived macrophages and human iPSC-derived macrophages showed similar gene expression patterns among the groups and genes studied here, suggesting that iPSC-derived monocytes have the potential to be used as a reliable cell source of human macrophages for in vitro studies. These findings could help select appropriate markers when testing macrophage behavior in vitro and highlight those markers that may confuse interpretation of results from experiments employing macrophages from different sources.


Assuntos
Polaridade Celular/genética , Perfilação da Expressão Gênica , Macrófagos/citologia , Macrófagos/metabolismo , Animais , Materiais Biocompatíveis/farmacologia , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Polaridade Celular/efeitos dos fármacos , Análise Discriminante , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Interferon gama/farmacologia , Análise dos Mínimos Quadrados , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Camundongos Endogâmicos BALB C , Fenótipo
17.
Proc Natl Acad Sci U S A ; 111(19): 6940-5, 2014 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-24778247

RESUMO

The efforts to grow mechanically functional cartilage from human mesenchymal stem cells have not been successful. We report that clinically sized pieces of human cartilage with physiologic stratification and biomechanics can be grown in vitro by recapitulating some aspects of the developmental process of mesenchymal condensation. By exposure to transforming growth factor-ß, mesenchymal stem cells were induced to condense into cellular bodies, undergo chondrogenic differentiation, and form cartilagenous tissue, in a process designed to mimic mesenchymal condensation leading into chondrogenesis. We discovered that the condensed mesenchymal cell bodies (CMBs) formed in vitro set an outer boundary after 5 d of culture, as indicated by the expression of mesenchymal condensation genes and deposition of tenascin. Before setting of boundaries, the CMBs could be fused into homogenous cellular aggregates giving rise to well-differentiated and mechanically functional cartilage. We used the mesenchymal condensation and fusion of CMBs to grow centimeter-sized, anatomically shaped pieces of human articular cartilage over 5 wk of culture. For the first time to our knowledge biomechanical properties of cartilage derived from human mesenchymal cells were comparable to native cartilage, with the Young's modulus of >800 kPa and equilibrium friction coeffcient of <0.3. We also demonstrate that CMBs have capability to form mechanically strong cartilage-cartilage interface in an in vitro cartilage defect model. The CMBs, which acted as "lego-like" blocks of neocartilage, were capable of assembling into human cartilage with physiologic-like structure and mechanical properties.


Assuntos
Cartilagem Articular/citologia , Cartilagem Articular/fisiologia , Células-Tronco Mesenquimais/citologia , Engenharia Tecidual/métodos , Animais , Fenômenos Biomecânicos/fisiologia , Biomimética/métodos , Osso e Ossos/citologia , Osso e Ossos/fisiologia , Doenças das Cartilagens/terapia , Cartilagem Articular/crescimento & desenvolvimento , Bovinos , Diferenciação Celular/fisiologia , Células Cultivadas , Meios de Cultura/farmacologia , Módulo de Elasticidade/fisiologia , Fricção/fisiologia , Humanos , Células-Tronco Mesenquimais/fisiologia
18.
Methods ; 84: 109-14, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25828645

RESUMO

For a long time, clinically sized and mechanically functional cartilage could be engineered from young animal chondrocytes, but not from adult human mesenchymal stem cells that are of primary clinical interest. The approaches developed for primary chondrocytes were not successful when used with human mesenchymal cells. The method discussed here was designed to employ a mechanism similar to pre-cartilaginous condensation and fusion of mesenchymal stem cells at a precisely defined time. The formation of cartilage was initiated by press-molding the mesenchymal bodies onto the surface of a bone substrate. By image-guided fabrication of the bone substrate and the molds, the osteochondral constructs were engineered in anatomically precise shapes and sizes. After 5 weeks of cultivation, the cartilage layer assumed physiologically stratified histomorphology, and contained lubricin at the surface, proteoglycans and type II collagen in the bulk phase, collagen type X at the interface with the bone substrate, and collagen type I within the bone phase. For the first time, the Young's modulus and the friction coefficient of human cartilage engineered from mesenchymal stem cells reached physiological levels for adult human cartilage. We propose that this method can be effective for generating human osteochondral tissue constructs.


Assuntos
Cartilagem/fisiologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Engenharia Tecidual/métodos , Adulto , Células-Tronco Adultas/citologia , Células-Tronco Adultas/fisiologia , Animais , Fenômenos Biomecânicos , Reatores Biológicos , Cartilagem/citologia , Cartilagem/crescimento & desenvolvimento , Fusão Celular/métodos , Condrócitos/citologia , Condrócitos/fisiologia , Módulo de Elasticidade , Humanos , Regeneração , Alicerces Teciduais
20.
J Biomech Eng ; 138(2): 021010, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26720588

RESUMO

The notion that both adaptive and maladaptive cardiac remodeling occurs in response to mechanical loading has informed recent progress in cardiac tissue engineering. Today, human cardiac tissues engineered in vitro offer complementary knowledge to that currently provided by animal models, with profound implications to personalized medicine. We review here recent advances in the understanding of the roles of mechanical signals in normal and pathological cardiac function, and their application in clinical translation of tissue engineering strategies to regenerative medicine and in vitro study of disease.


Assuntos
Coração/fisiologia , Fenômenos Mecânicos , Miocárdio/citologia , Engenharia Tecidual/métodos , Animais , Fenômenos Biomecânicos , Coração/fisiopatologia , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA