Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Artigo em Inglês | MEDLINE | ID: mdl-38772903

RESUMO

Repair and regeneration of a diseased lung using stem cells or bioengineered tissues is an exciting therapeutic approach for a variety of lung diseases and critical illnesses. Over the past decade increasing evidence from preclinical models suggests that cells, which are not normally resident in the lung can be utilized to modulate immune responses after injury, but there have been challenges in translating these promising findings to the clinic. In parallel, there has been a surge in bioengineering studies investigating the use of artificial and acellular lung matrices as scaffolds for three-dimensional lung or airway regeneration, with some recent attempts of transplantation in large animal models. The combination of these studies with those involving stem cells, induced pluripotent stem cell derivatives, and/or cell therapies is a promising and rapidly developing research area. These studies have been further paralleled by significant increases in our understanding of the molecular and cellular events by which endogenous lung stem and/or progenitor cells arise during lung development and participate in normal and pathologic remodeling after lung injury. For the 2023 Stem Cells, Cell Therapies, and Bioengineering in Lung Biology and Diseases Conference, scientific symposia were chosen to reflect the most cutting-edge advances in these fields. Sessions focused on the integration of "-omics" technologies with function, the influence of immune cells on regeneration, and the role of the extracellular matrix in regeneration. The necessity for basic science studies to enhance fundamental understanding of lung regeneration and to design innovative translational studies was reinforced throughout the conference.

2.
PLoS Comput Biol ; 20(2): e1011113, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38386693

RESUMO

A variety of pulmonary insults can prompt the need for life-saving mechanical ventilation; however, misuse, prolonged use, or an excessive inflammatory response, can result in ventilator-induced lung injury. Past research has observed an increased instance of respiratory distress in older patients and differences in the inflammatory response. To address this, we performed high pressure ventilation on young (2-3 months) and old (20-25 months) mice for 2 hours and collected data for macrophage phenotypes and lung tissue integrity. Large differences in macrophage activation at baseline and airspace enlargement after ventilation were observed in the old mice. The experimental data was used to determine plausible trajectories for a mathematical model of the inflammatory response to lung injury which includes variables for the innate inflammatory cells and mediators, epithelial cells in varying states, and repair mediators. Classification methods were used to identify influential parameters separating the parameter sets associated with the young or old data and separating the response to ventilation, which was measured by changes in the epithelial state variables. Classification methods ranked parameters involved in repair and damage to the epithelial cells and those associated with classically activated macrophages to be influential. Sensitivity results were used to determine candidate in-silico interventions and these interventions were most impact for transients associated with the old data, specifically those with poorer lung health prior to ventilation. Model results identified dynamics involved in M1 macrophages as a focus for further research, potentially driving the age-dependent differences in all macrophage phenotypes. The model also supported the pro-inflammatory response as a potential indicator of age-dependent differences in response to ventilation. This mathematical model can serve as a baseline model for incorporating other pulmonary injuries.


Assuntos
Pulmão , Lesão Pulmonar Induzida por Ventilação Mecânica , Humanos , Camundongos , Animais , Idoso , Respiração Artificial/efeitos adversos , Macrófagos , Modelos Teóricos
3.
PLoS One ; 19(1): e0270779, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38271449

RESUMO

Macrophages show high plasticity and result in heterogenic subpopulations or polarized states identified by specific cellular markers. These immune cells are typically characterized as pro-inflammatory, or classically activated M1, and anti-inflammatory, or alternatively activated M2. However, a more precise definition places them along a spectrum of activation where they may exhibit a number of pro- or anti-inflammatory roles. To understand M1-M2 dynamics in the context of a localized response and explore the results of different mathematical modeling approaches based on the same biology, we utilized two different modeling techniques, ordinary differential equation (ODE) modeling and agent-based modeling (ABM), to simulate the spectrum of macrophage activation to general pro- and anti-inflammatory stimuli on an individual and multi-cell level. The ODE model includes two hallmark pro- and anti-inflammatory signaling pathways and the ABM incorporates similar M1-M2 dynamics but in a spatio-temporal platform. Both models link molecular signaling with cellular-level dynamics. We then performed simulations with various initial conditions to replicate different experimental setups. Similar results were observed in both models after tuning to a common calibrating experiment. Comparing the two models' results sheds light on the important features of each modeling approach. When more data is available these features can be considered when choosing techniques to best fit the needs of the modeler and application.


Assuntos
Ativação de Macrófagos , Macrófagos , Ativação de Macrófagos/fisiologia , Macrófagos/metabolismo , Anti-Inflamatórios/metabolismo , Transdução de Sinais
4.
J Aerosol Med Pulm Drug Deliv ; 33(6): 314-322, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32453638

RESUMO

Background: In neonatal respiratory distress syndrome, breathing support and surfactant therapy are commonly used to enable the alveoli to expand. Surfactants are typically delivered through liquid instillation. However, liquid instillation does not specifically target the small airways. We have developed an excipient enhanced growth (EEG) powder aerosol formulation using Survanta®. Methods: EEG Survanta powder aerosol was delivered using a novel dry powder inhaler via tracheal insufflation to surfactant depleted rats at nominal doses of 3, 5, 10, and 20 mg of powder containing 0.61, 0.97, 1.73, and 3.46 mg of phospholipids (PL), whereas liquid Survanta was delivered via syringe instillation at doses of 2 and 4 mL/kg containing 18.6 and 34 mg of PL. Ventilation mechanics were measured before and after depletion, and after treatment. We hypothesized that EEG Survanta powder aerosol would improve lung mechanics compared with instilled liquid Survanta in surfactant depleted rats. Results and Conclusion: EEG Survanta powder aerosol at a dose of 0.61 mg PL significantly improved lung compliance and elastance compared with the liquid Survanta at a dose of 18.6 mg, which represents improved primary efficacy of the aerosol at a 30-fold lower dose of PL. There was no significant difference in white blood cell count of the lavage from the EEG Survanta group compared with liquid Survanta. These results provide an in vivo proof-of-concept for EEG Survanta powder aerosol as a promising method of surfactant replacement therapy.


Assuntos
Lesão Pulmonar/tratamento farmacológico , Surfactantes Pulmonares/administração & dosagem , Aerossóis , Animais , Modelos Animais de Doenças , Excipientes , Tamanho da Partícula , Pós , Ratos , Ratos Sprague-Dawley
5.
Acta Biomater ; 100: 223-234, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31593773

RESUMO

Decellularized tissues offer a unique tool for developing regenerative biomaterials or in vitro platforms for the study of cell-extracellular matrix (ECM) interactions. One main challenge associated with decellularized lung tissue is that ECM components can be stripped away or altered by the detergents used to remove cellular debris. Without characterizing the composition of lung decellularized ECM (dECM) and the cellular response caused by the altered composition, it is difficult to utilize dECM for regeneration and specifically, engineering the complexities of the alveolar-capillary barrier. This study takes steps towards uncovering if dECM must be enhanced with lost ECM proteins to achieve proper epithelial barrier formation. To achieve this, the epithelial barrier function was assessed on dECM coatings with and without the systematic addition of several key basement membrane proteins. After comparing barrier function on collagen I, fibronectin, laminin, and dECM in varying combinations as an in vitro coating, the alveolar epithelium exhibited superior barrier function when dECM was supplemented with laminin as evidenced by trans-epithelial electrical resistance (TEER) and permeability assays. Increased barrier resistance with laminin addition was associated with upregulation of Claudin-18, E-cadherin, and junction adhesion molecule (JAM)-A, and stabilization of zonula occludens (ZO)-1 at junction complexes. The Epac/Rap1 pathway was observed to play a role in the ECM-mediated barrier function determined by protein expression and Epac inhibition. These findings revealed potential ECM coatings and molecular therapeutic targets for improved regeneration with decellularized scaffolds. STATEMENT OF SIGNIFICANCE: Efforts to produce a transplantable organ-scale biomaterial for lung regeneration has not been entirely successful to date, due to incomplete cell-cell junction formation, ultimately leading to severe edema in vivo. To fully understand the process of alveolar junction formation on ECM-derived biomaterials, this research has characterized and tailored decellularized ECM (dECM) to mitigate reductions in barrier strength or cell attachment caused by abnormal ECM compositions or detergent damage to dECM. These results indicate that laminin-driven Epac signaling plays a vital role in the stabilization of the alveolar barrier. Addition of laminin or Epac agonists during alveolar regeneration can reduce epithelial permeability within bioengineered lungs.


Assuntos
Células Epiteliais/metabolismo , Matriz Extracelular/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Laminina/farmacologia , Junções Aderentes/efeitos dos fármacos , Junções Aderentes/metabolismo , Animais , Bovinos , Adesão Celular/efeitos dos fármacos , Linhagem Celular , Impedância Elétrica , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Matriz Extracelular/efeitos dos fármacos , Feminino , Masculino , Camundongos , Alvéolos Pulmonares/citologia , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Suínos , Junções Íntimas/efeitos dos fármacos , Junções Íntimas/metabolismo
6.
Biophys J ; 117(1): 170-183, 2019 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-31200935

RESUMO

Cell migration, a fundamental physiological process in which cells sense and move through their surrounding physical environment, plays a critical role in development and tissue formation, as well as pathological processes, such as cancer metastasis and wound healing. During cell migration, dynamics are governed by the bidirectional interplay between cell-generated mechanical forces and the activity of Rho GTPases, a family of small GTP-binding proteins that regulate actin cytoskeleton assembly and cellular contractility. These interactions are inherently more complex during the collective migration of mechanically coupled cells because of the additional regulation of cell-cell junctional forces. In this study, we adapted a recent minimal modeling framework to simulate the interactions between mechanochemical signaling in individual cells and interactions with cell-cell junctional forces during collective cell migration. We find that migration of individual cells depends on the feedback between mechanical tension and Rho GTPase activity in a biphasic manner. During collective cell migration, waves of Rho GTPase activity mediate mechanical contraction/extension and thus synchronization throughout the tissue. Further, cell-cell junctional forces exhibit distinct spatial patterns during collective cell migration, with larger forces near the leading edge. Larger junctional force magnitudes are associated with faster collective cell migration and larger tissue size. Simulations of heterogeneous tissue migration exhibit a complex dependence on the properties of both leading and trailing cells. Computational predictions demonstrate that collective cell migration depends on both the emergent dynamics and interactions between cellular-level Rho GTPase activity and contractility and multicellular-level junctional forces.


Assuntos
Movimento Celular , Junções Intercelulares/metabolismo , Mecanotransdução Celular , Modelos Teóricos , Citoesqueleto de Actina/metabolismo , Animais , Retroalimentação Fisiológica , Junções Intercelulares/química , Estresse Mecânico , Proteínas rho de Ligação ao GTP/metabolismo
7.
J Tissue Eng Regen Med ; 12(12): 2331-2336, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30367566

RESUMO

Decellularized extracellular matrix (ECM) is an effective tissue repair scaffold. Additionally, ECM has recently been shown to be protective to the lungs. However, current processing is inadequate for effective delivery of ECM to the lungs. Processing methods, such as milling, produce large variability in particle sizes. The size variation produced is ineffective at treating the lung because only a small range of sizes reach the distal regions of the alveoli. The aim of this work is to formulate decellularized ECM to reach the distal lung while retaining the pro-regenerative effects of ECM. We first digested the protein in acid and then electrosprayed the solution into nanoparticles. The average size of the nanoparticles was 225 (±67) nm, within size requirements to reach the alveoli. After characterizing the particles, we measured cytotoxicity of the nanoparticles. Adding 0.125 mg/ml of nanoparticles to the media increased cellular proliferation in A549 alveolar epithelial cells and caused no cytotoxicity in BEAS-2B cells. We added the formed nanoparticles to macrophages derived from murine bone marrow-derived monocytes. The macrophages exposed to the formed nanoparticles expressed cell surface marker CD206 (mannose receptor C type 1), commonly attributed to a pro-regeneration phenotype. Electrosprayed ECM formed nanoparticles may improve bronchoalveolar deposition while maintaining the pro-regenerative benefits shown by other decellularized ECM materials.


Assuntos
Células Epiteliais Alveolares/metabolismo , Matriz Extracelular/química , Macrófagos/metabolismo , Teste de Materiais , Monócitos/metabolismo , Nanopartículas/química , Regeneração , Células A549 , Células Epiteliais Alveolares/citologia , Animais , Linhagem Celular , Humanos , Macrófagos/citologia , Camundongos , Monócitos/citologia , Suínos
8.
Front Pediatr ; 4: 80, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27583245

RESUMO

The field of stem cell biology, cell therapy, and regenerative medicine has expanded almost exponentially, in the last decade. Clinical trials are evaluating the potential therapeutic use of stem cells in many adult and pediatric lung diseases with vascular component, such as bronchopulmonary dysplasia (BPD), chronic obstructive pulmonary disease (COPD), idiopathic pulmonary fibrosis (IPF), or pulmonary arterial hypertension (PAH). Extensive research activity is exploring the lung resident and circulating progenitor cells and their contribution to vascular complications of chronic lung diseases, and researchers hope to use resident or circulating stem/progenitor cells to treat chronic lung diseases and their vascular complications. It is becoming more and more clear that progress in mechanobiology will help to understand the various influences of physical forces and extracellular matrix composition on the phenotype and features of the progenitor cells and stem cells. The current review provides an overview of current concepts in the field.

9.
J Biomed Mater Res A ; 104(8): 1922-35, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27012815

RESUMO

The complexity and rapid clearance mechanisms of lung tissue make it difficult to develop effective treatments for many chronic pathologies. We are investigating lung derived extracellular matrix (ECM) hydrogels as a novel approach for delivery of cellular therapies to the pulmonary system. The main objectives of this study include effective decellularization of porcine lung tissue, development of a hydrogel from the porcine ECM, and characterization of the material's composition, mechanical properties, and ability to support cellular growth. Our evaluation of the decellularized tissue indicated successful removal of cellular material and immunogenic remnants in the ECM. The self-assembly of the lung ECM hydrogel was rapid, reaching maximum modulus values within 3 min at 37°C. Rheological characterization showed the lung ECM hydrogel to have a concentration dependent storage modulus between 15 and 60 Pa. The purpose of this study was to evaluate our novel ECM derived hydrogel and measure its ability to support 3D culture of MSCs in vitro and in vivo delivery of MSCs. Our in vitro experiments using human mesenchymal stem cells demonstrated our novel ECM hydrogel's ability to enhance cellular attachment and viability. Our in vivo experiments demonstrated that rat MSC delivery in pre-gel solution significantly increased cell retention in the lung over 24 h in an emphysema rat model. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 104A: 1922-1935, 2016.


Assuntos
Matriz Extracelular/metabolismo , Hidrogel de Polietilenoglicol-Dimetacrilato/farmacologia , Pulmão/metabolismo , Animais , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Eletroforese em Gel de Poliacrilamida , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Inflamação/patologia , Cinética , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Elastase Pancreática , Ratos Sprague-Dawley , Reologia , Sus scrofa
10.
Am J Physiol Lung Cell Mol Physiol ; 306(11): L1045-55, 2014 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-24748604

RESUMO

Epithelial injury is often detected in lung allografts, however, its relation to rejection pathogenesis is unknown. We hypothesized that sterile epithelial injury can lead to alloimmune activation in the lung. We performed adoptive transfer of mismatched splenocytes into recombinant activating gene 1 (Rag1)-deficient mice to induce an alloimmune status and then exposed these mice to naphthalene to induce sterile epithelial injury. We evaluated lungs for presence of alloimmune lung injury, endoplasmic reticulum (ER) stress, and hyaluronan expression, examined the effect of ER stress induction on hyaluronan expression and lymphocyte trapping by bronchial epithelia in vitro, and examined airways from patients with bronchiolitis obliterans syndrome and normal controls histologically. We found that Rag1-deficient mice that received mismatched splenocytes and naphthalene injection displayed bronchial epithelial ER stress, peribronchial hyaluronan expression, and lymphocytic bronchitis. Bronchial epithelial ER stress led to the expression of lymphocyte-trapping hyaluronan cables in vitro. Blockade of hyaluronan binding ameliorated naphthalene-induced lymphocytic bronchitis. ER stress was present histologically in >40% of bronchial epithelia of BOS patients and associated with subepithelial hyaluronan deposition. We conclude that sterile bronchial epithelial injury in the context of alloimmunity can lead to sustained ER stress and promote allograft rejection through hyaluronan expression.


Assuntos
Bronquiolite Obliterante/metabolismo , Células Epiteliais/imunologia , Ácido Hialurônico/metabolismo , Linfócitos/imunologia , Aloenxertos/imunologia , Animais , Brônquios/patologia , Bronquiolite Obliterante/imunologia , Células Cultivadas , Técnicas de Cocultura , Estresse do Retículo Endoplasmático , Glucuronosiltransferase/genética , Glucuronosiltransferase/metabolismo , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/metabolismo , Humanos , Hialuronan Sintases , Transplante de Pulmão , Linfócitos/patologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Mucosa Respiratória/patologia , Tenascina/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Regulação para Cima
11.
J Biol Chem ; 286(20): 17435-44, 2011 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-21398522

RESUMO

Epithelial injury is a central event in the pathogenesis of many inflammatory and fibrotic lung diseases like acute respiratory distress syndrome, pulmonary fibrosis, and iatrogenic lung injury. Mechanical stress is an often underappreciated contributor to lung epithelial injury. Following injury, differentiated epithelia can assume a myofibroblast phenotype in a process termed epithelial to mesenchymal transition (EMT), which contributes to aberrant wound healing and fibrosis. We demonstrate that cyclic mechanical stretch induces EMT in alveolar type II epithelial cells, associated with increased expression of low molecular mass hyaluronan (sHA). We show that sHA is sufficient for induction of EMT in statically cultured alveolar type II epithelial cells and necessary for EMT during cell stretch. Furthermore, stretch-induced EMT requires the innate immune adaptor molecule MyD88. We examined the Wnt/ß-catenin pathway, which is known to mediate EMT. The Wnt target gene Wnt-inducible signaling protein 1 (wisp-1) is significantly up-regulated in stretched cells in hyaluronan- and MyD88-dependent fashion, and blockade of WISP-1 prevents EMT in stretched cells. In conclusion, we show for the first time that innate immunity transduces mechanical stress responses through the matrix component hyaluronan, and activation of the Wnt/ß-catenin pathway.


Assuntos
Transição Epitelial-Mesenquimal/fisiologia , Ácido Hialurônico/farmacologia , Imunidade Inata/fisiologia , Alvéolos Pulmonares/metabolismo , Mucosa Respiratória/metabolismo , Estresse Fisiológico/fisiologia , Animais , Proteínas de Sinalização Intercelular CCN , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Ácido Hialurônico/imunologia , Ácido Hialurônico/metabolismo , Imunidade Inata/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/imunologia , Fator 88 de Diferenciação Mieloide/metabolismo , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/imunologia , Proteínas Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas , Alvéolos Pulmonares/citologia , Alvéolos Pulmonares/imunologia , Mucosa Respiratória/citologia , Estresse Fisiológico/efeitos dos fármacos , Proteínas Wnt/genética , Proteínas Wnt/imunologia , Proteínas Wnt/metabolismo , beta Catenina/genética , beta Catenina/imunologia , beta Catenina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA