Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
NPJ Regen Med ; 8(1): 22, 2023 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-37117221

RESUMO

Engineered whole lungs may one day expand therapeutic options for patients with end-stage lung disease. However, the feasibility of ex vivo lung regeneration remains limited by the inability to recapitulate mature, functional alveolar epithelium. Here, we modulate multimodal components of the alveolar epithelial type 2 cell (AEC2) niche in decellularized lung scaffolds in order to guide AEC2 behavior for epithelial regeneration. First, endothelial cells coordinate with fibroblasts, in the presence of soluble growth and maturation factors, to promote alveolar scaffold population with surfactant-secreting AEC2s. Subsequent withdrawal of Wnt and FGF agonism synergizes with tidal-magnitude mechanical strain to induce the differentiation of AEC2s to squamous type 1 AECs (AEC1s) in cultured alveoli, in situ. These results outline a rational strategy to engineer an epithelium of AEC2s and AEC1s contained within epithelial-mesenchymal-endothelial alveolar-like units, and highlight the critical interplay amongst cellular, biochemical, and mechanical niche cues within the reconstituting alveolus.

2.
Anesth Analg ; 134(5): 1094-1105, 2022 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-34928890

RESUMO

The coronavirus disease 2019 (COVID-19) pandemic has revealed that even the best-resourced hospitals may lack sufficient ventilators to support patients under surge conditions. During a pandemic or mass trauma, an affordable, low-maintenance, off-the-shelf device that would allow health care teams to rapidly expand their ventilator capacity could prove lifesaving, but only if it can be safely integrated into a complex and rapidly changing clinical environment. Here, we define an approach to safe ventilator sharing that prioritizes predictable and independent care of patients sharing a ventilator. Subsequently, we detail the design and testing of a ventilator-splitting circuit that follows this approach and describe our clinical experience with this circuit during the COVID-19 pandemic. This circuit was able to provide individualized and titratable ventilatory support with individualized positive end-expiratory pressure (PEEP) to 2 critically ill patients at the same time, while insulating each patient from changes in the other's condition. We share insights from our experience using this technology in the intensive care unit and outline recommendations for future clinical applications.


Assuntos
COVID-19 , Pandemias , COVID-19/terapia , Humanos , Respiração com Pressão Positiva , Respiração Artificial , Ventiladores Mecânicos
3.
Front Bioeng Biotechnol ; 9: 760309, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34869270

RESUMO

The development of an in vitro system for the study of lung vascular disease is critical to understanding human pathologies. Conventional culture systems fail to fully recapitulate native microenvironmental conditions and are typically limited in their ability to represent human pathophysiology for the study of disease and drug mechanisms. Whole organ decellularization provides a means to developing a construct that recapitulates structural, mechanical, and biological features of a complete vascular structure. Here, we developed a culture protocol to improve endothelial cell coverage in whole lung scaffolds and used single-cell RNA-sequencing analysis to explore the impact of decellularized whole lung scaffolds on endothelial phenotypes and functions in a biomimetic bioreactor system. Intriguingly, we found that the phenotype and functional signals of primary pulmonary microvascular revert back-at least partially-toward native lung endothelium. Additionally, human induced pluripotent stem cell-derived endothelium cultured in decellularized lung systems start to gain various native human endothelial phenotypes. Vascular barrier function was partially restored, while small capillaries remained patent in endothelial cell-repopulated lungs. To evaluate the ability of the engineered endothelium to modulate permeability in response to exogenous stimuli, lipopolysaccharide (LPS) was introduced into repopulated lungs to simulate acute lung injury. After LPS treatment, proinflammatory signals were significantly increased and the vascular barrier was impaired. Taken together, these results demonstrate a novel platform that recapitulates some pulmonary microvascular functions and phenotypes at a whole organ level. This development may help pave the way for using the whole organ engineering approach to model vascular diseases.

4.
Biomaterials ; 217: 119313, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31280072

RESUMO

Microvascular leak is a phenomenon witnessed in multiple disease states. In organ engineering, regaining a functional barrier is the most crucial step towards creating an implantable organ. All previous methods of measuring microvascular permeability were either invasive, lengthy, introduced exogenous macromolecules, or relied on extrapolations from cultured cells. We present here a system that enables real-time measurement of microvascular permeability in intact rat lungs. Our unique system design allows direct, non-invasive measurement of average alveolar and capillary pressures, tracks flow paths within the organ, and enables calculation of lumped internal resistances including microvascular barrier. We first describe the physiology of native and decellularized lungs and the inherent properties of the extracellular matrix as functions of perfusion rate. We next track changing internal resistances and flows in injured native rat lungs, resolving the onset of microvascular leak, quantifying changing vascular resistances, and identifying distinct phases of organ failure. Finally, we measure changes in permeability within engineered lungs seeded with microvascular endothelial cells, quantifying cellular effects on internal vascular and barrier resistances over time. This system marks considerable progress in bioreactor design for intact organs and may be used to monitor and garner physiological insights into native, decellularized, and engineered tissues.


Assuntos
Sistemas Computacionais , Pulmão/irrigação sanguínea , Microvasos/patologia , Animais , Reatores Biológicos , Endotélio/patologia , Pulmão/ultraestrutura , Lesão Pulmonar/patologia , Masculino , Ratos Sprague-Dawley , Técnicas de Cultura de Tecidos , Engenharia Tecidual
5.
Biomaterials ; 200: 25-34, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30754017

RESUMO

Whole organ engineering paradigms typically involve repopulating acellular organ scaffolds with recipient-compatible cells, to generate a neo-organ that may provide key physiological functions. In the case of whole lung engineering, functionally endothelialized pulmonary vasculature is critical for establishing a fluid-tight barrier at the level of the alveolus, so that oxygen and carbon dioxide can be exchanged in the organ. We have previously developed a protocol to efficiently seed endothelial cells into the microvascular channels of decellularized lung scaffolds, but fully functional endothelial coverage, in terms of barrier function and resistance to thrombosis, was not achieved. In this study, we investigated whether various small molecules could favorably impact endothelial functionality after seeding into decellularized lung scaffolds. We demonstrated that the Epac-selective cAMP analog 8CPT-2Me-cAMP improves endothelial barrier function in repopulated lung scaffolds. When treated with the Epac agonist, barrier function of human umbilical vein endothelial cells (HUVECs) improved, and was maintained for at least three days, whereas the effect of other tested molecules lasted for only 5 h. Treatment with the Epac agonist re-organized actin structure, and appeared to increase the continuity of junction proteins such as VE-cadherin and ZO1. Blockade of actin polymerization abolished the effect of the Epac agonist on barrier function and actin reorganization, confirming a strong actin-mediated effect. Similarly, after treatment with Epac agonist, the barrier function in iPSC-derived endothelial colony forming cells (ECFCs) was increased and the enhanced barrier was maintained for at least 60 h. After culture in lung scaffolds for 5 days, iPSC-ECFCs maintained their phenotype by expressing CD31, eNOS, vWF, and VE-Cadherin. Treatment with the Epac agonist significantly improved the barrier function of iPSC-ECFC-repopulated lung for at least 6 h. Taken together, these findings demonstrated that Epac-selective 8CPT-2Me-cAMP activation enhanced vascular barrier in iPSC-ECFC-engineered lungs, and may be useful to improve endothelial functionality for whole organ tissue engineering.


Assuntos
Fatores de Troca do Nucleotídeo Guanina/agonistas , Células Endoteliais da Veia Umbilical Humana/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Engenharia Tecidual/métodos , Citoesqueleto de Actina/metabolismo , Animais , Ensaio de Unidades Formadoras de Colônias , AMP Cíclico/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Pulmão/metabolismo , Ratos , Alicerces Teciduais/química
6.
J Tissue Eng Regen Med ; 12(3): e1623-e1635, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29024475

RESUMO

The development of an alternative source for donor lungs would change the paradigm of lung transplantation. Recent studies have demonstrated the potential feasibility of using decellularized lungs as scaffolds for lung tissue regeneration and subsequent implantation. However, finding a reliable cell source and the ability to scale up for recellularization of the lung scaffold still remain significant challenges. To explore the possibility of regeneration of human lung tissue from stem cells in vitro, populations of lung progenitor cells were generated from human iPSCs. To explore the feasibility of producing engineered lungs from stem cells, we repopulated decellularized human lung and rat lungs with iPSC-derived epithelial progenitor cells. The iPSCs-derived epithelial progenitor cells lined the decellularized human lung and expressed most of the epithelial markers when were cultured in a lung bioreactor system. In decellularized rat lungs, these human-derived cells attach and proliferate in a manner similar to what was observed in the decellularized human lung. Our results suggest that repopulation of lung matrix with iPSC-derived lung epithelial cells may be a viable strategy for human lung regeneration and represents an important early step toward translation of this technology.


Assuntos
Bioengenharia/métodos , Células Epiteliais/citologia , Matriz Extracelular/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Pulmão/fisiologia , Animais , Biomarcadores/metabolismo , Linhagem Celular , Proliferação de Células , Células Cultivadas , Endoderma/citologia , Células Endoteliais/citologia , Células Epiteliais/metabolismo , Regulação da Expressão Gênica , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Microvasos/citologia , Ratos Sprague-Dawley , Alicerces Teciduais/química
7.
J Tissue Eng Regen Med ; 12(1): e119-e129, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28083925

RESUMO

In cellular, tissue-level or whole organ bioreactors, the level of dissolved oxygen is one of the most important factors requiring control. Hypoxic environments may lead to cellular apoptosis, while hyperoxic environments may lead to cellular damage or dedifferentiation, both resulting in loss of overall tissue function. This manuscript describes the creation, characterization and validation of a bioreactor system that can control oxygen delivery based on real-time metabolic demand of cultured whole lung tissue. A mathematical model describing and predicting gas exchange within the tunable bioreactor system is developed. In addition, the inherent gas exchange properties of the bioreactor and the inherent oxygen consumption rates of native rat lungs are determined, thereby providing a quantitative relationship between system parameters and levels of dissolved oxygen. Finally, the mathematical model is validated during whole lung culture under a range of system parameters. The system presented here provides a quantitative relationship between the concentration of dissolved oxygen, tissue oxygen consumption rates, and controllable system parameters that introduce gasses into the bioreactor. This relationship not only enables the maintenance of constant levels of dissolved oxygen throughout a culture period during which cells are replicating, but also provides noninvasive and real-time estimation of the metabolic and proliferative states of native or engineered lung tissue simply through dissolved oxygen measurements. Copyright © 2017 John Wiley & Sons, Ltd.


Assuntos
Reatores Biológicos , Gases/metabolismo , Pulmão/fisiologia , Animais , Contagem de Células , DNA/metabolismo , Desenho de Equipamento , Processamento de Imagem Assistida por Computador , Modelos Biológicos , Consumo de Oxigênio , Ratos Sprague-Dawley , Reprodutibilidade dos Testes
8.
ACS Biomater Sci Eng ; 3(9): 2000-2010, 2017 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-33440555

RESUMO

To date, efforts to generate engineered lung tissue capable of long-term function have been limited by incomplete barrier formation between air and blood and by thrombosis of the microvasculature upon exposure of blood to the collagens within the decellularized scaffold. Improved barrier function and resistance to thrombosis both depend upon the recapitulation of a confluent monolayer of functional endothelium throughout the pulmonary vasculature. This manuscript describes novel strategies to increase cell coverage of the vascular surface area, compared to previous reports in our lab and others, and reports robust production of multiple anticoagulant substances that will be key to long-term function in vivo once additional strides are made in improving barrier function. Rat lung microvascular endothelial cells were seeded into decellularized rat lungs by both the pulmonary artery and veins with the use of low-concentration cell suspensions, pulsatile, gravity-driven flow, and supraphysiological vascular pressures. Together, these strategies yielded 72.44 ± 10.52% endothelial cell nuclear coverage of the acellular matrix after 3-4 d of biomimetic bioreactor culture compared to that of the native rat lung. Immunofluorescence, Western blot, and PCR analysis of these lungs indicated robust expression of phenotypic markers such as CD31 and VE-Cadherin after time in culture. Endothelial-seeded lungs had CD31 gene expression of 0.074 ± 0.015 vs 0.021 ± 0.0023 for native lungs, p = 0.025, and VE-Cadherin gene expression of 0.93 ± 0.22 compared to that of the native lung at 0.13 ± 0.02, p = 0.023. Precursors to antithrombotic substances such as tissue plasminogen activator, prostacyclin synthase, and endothelial nitric oxide synthase were expressed at levels equal to or greater than those of the native lung. Engineered lungs reseeded with endothelial cells were implanted orthotopically and contained patent microvascular networks that had gas exchange function during mechanical ventilation on 100% O2 greater than that of decelluarized lungs. Taken together, these data suggest that these engineered constructs could be compatible with long-term function in vivo when utilized in future studies in tandem with improved barrier function.

9.
Biomaterials ; 102: 220-30, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27344365

RESUMO

Lung engineering is a promising technology, relying on re-seeding of either human or xenographic decellularized matrices with patient-derived pulmonary cells. Little is known about the species-specificity of decellularization in various models of lung regeneration, or if species dependent cell-matrix interactions exist within these systems. Therefore decellularized scaffolds were produced from rat, pig, primate and human lungs, and assessed by measuring residual DNA, mechanical properties, and key matrix proteins (collagen, elastin, glycosaminoglycans). To study intrinsic matrix biologic cues, human endothelial cells were seeded onto acellular slices and analyzed for markers of cell health and inflammation. Despite similar levels of collagen after decellularization, human and primate lungs were stiffer, contained more elastin, and retained fewer glycosaminoglycans than pig or rat lung scaffolds. Human endothelial cells seeded onto human and primate lung tissue demonstrated less expression of vascular cell adhesion molecule and activation of nuclear factor-κB compared to those seeded onto rodent or porcine tissue. Adhesion of endothelial cells was markedly enhanced on human and primate tissues. Our work suggests that species-dependent biologic cues intrinsic to lung extracellular matrix could have profound effects on attempts at lung regeneration.


Assuntos
Células Endoteliais/citologia , Matriz Extracelular/química , Pulmão/química , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Animais , Linhagem Celular , Colágeno/análise , Elastina/análise , Glicosaminoglicanos/análise , Humanos , Pulmão/citologia , Pulmão/fisiologia , Pulmão/ultraestrutura , Ratos , Regeneração , Medicina Regenerativa , Suínos , Resistência à Tração
10.
Proc Natl Acad Sci U S A ; 105(7): 2463-8, 2008 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-18263733

RESUMO

During the bloodstage of malaria infection, the parasite internalizes and degrades massive amounts of hemoglobin from the host red blood cell. Using serial thin-section electron microscopy and three-dimensional reconstruction, we demonstrate four independent, but partially overlapping, hemoglobin-uptake processes distinguishable temporally, morphologically, and pharmacologically. Early ring-stage parasites undergo a profound morphological transformation in which they fold, like a cup, onto themselves and in so doing take a large first gulp of host cell cytoplasm. This event, which we term the "Big Gulp," appears to be independent of actin polymerization and marks the first step in biogenesis of the parasite's lysosomal compartment-the food vacuole. A second, previously identified uptake process, uses the cytostome, a well characterized and morphologically distinct structure at the surface of the parasite. This process is more akin to classical endocytosis, giving rise to small (<0.004 fl) vesicles that are marked by the early endosomal regulatory protein Rab5a. A third process, also arising from cytostomes, creates long thin tubes previously termed cytostomal tubes in an actin-dependent manner. The fourth pathway, which we term phagotrophy, is similar to the Big Gulp in that it more closely resembles phagocytosis, except that phagotrophy does not require actin polymerization. Each of these four processes has aspects that are unique to Plasmodium, thus opening avenues to antimalarial therapy.


Assuntos
Hemoglobinas/metabolismo , Malária/parasitologia , Plasmodium falciparum/metabolismo , Actinas/metabolismo , Animais , Hemoglobinas/ultraestrutura , Microscopia Imunoeletrônica , Plasmodium falciparum/ultraestrutura , Esquizontes/ultraestrutura , Trofozoítos/ultraestrutura , Vacúolos/ultraestrutura , Proteínas rab5 de Ligação ao GTP/metabolismo
11.
J Biol Chem ; 282(15): 11499-508, 2007 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-17289673

RESUMO

Phosphatidylinositol 3-phosphate (PI3P) is a key ligand for recruitment of endosomal regulatory proteins in higher eukaryotes. Subsets of these endosomal proteins possess a highly selective PI3P binding zinc finger motif belonging to the FYVE domain family. We have identified a single FYVE domain-containing protein in Plasmodium falciparum which we term FCP. Expression and mutagenesis studies demonstrate that key residues are involved in specific binding to PI3P. In contrast to FYVE proteins in other organisms, endogenous FCP localizes to a lysosomal compartment, the malaria parasite food vacuole (FV), rather than to cytoplasmic endocytic organelles. Transfections of deletion mutants further indicate that FCP is essential for trophozoite and FV maturation and that it traffics to the FV via a novel constitutive cytoplasmic to vacuole targeting pathway. This newly discovered pathway excludes the secretory pathway and is directed by a C-terminal 44-amino acid peptide domain. We conclude that an FYVE protein that might be expected to participate in vesicle targeting in the parasite cytosol instead has a vital and functional role in the malaria parasite FV.


Assuntos
Fosfatos de Fosfatidilinositol/metabolismo , Plasmodium falciparum/citologia , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/metabolismo , Vacúolos/metabolismo , Sequência de Aminoácidos , Animais , Transporte Biológico , Sequência Conservada , Microscopia Crioeletrônica , Deleção de Genes , Humanos , Dados de Sequência Molecular , Mutação/genética , Proteínas de Protozoários/química , Proteínas de Protozoários/genética , Alinhamento de Sequência
12.
J Cell Sci ; 117(Pt 17): 3831-8, 2004 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-15252121

RESUMO

Members of the apicomplexan family of parasites contain morphologically unique secretory organelles termed rhoptries that are essential for host cell invasion. Rhoptries contain internal membranes, and thus resemble multivesicular bodies. To determine whether multivesicular body endosomal intermediates are formed in Apicomplexa, we used the Plasmodium falciparum homolog of the class E gene, Vps4, as a probe. Endogenous P. falciparum Vps4 (PfVps4) localized to the cytoplasm of P. falciparum trophozoites, and transgenic PfVps4 localized to the cytosol in P. falciparum, in the related parasite Toxoplasma gondii and in COS cells. When mutated to block ATP hydrolysis, transiently expressed PfVps4 localized instead to large vesicular structures in P. falciparum. The same construct, and another mutant blocked in ATP binding, generated large cholesterol-enriched multivesicular bodies in both COS cells and T. gondii. Mutant PfVps4 structures in T. gondii co-localized with markers for early endosomes. These results demonstrate a conservation of Vps4 function across wide phylogenetic boundaries, and indicate that endosomal multivesicular bodies form in both P. falciparum and T. gondii.


Assuntos
Adenosina Trifosfatases/metabolismo , Adenosina Trifosfatases/fisiologia , Plasmodium falciparum/fisiologia , Trifosfato de Adenosina/metabolismo , Animais , Células COS , Proliferação de Células , Colesterol/metabolismo , Clonagem Molecular , Citoplasma/metabolismo , Citosol/metabolismo , DNA/metabolismo , Eletroporação , Complexos Endossomais de Distribuição Requeridos para Transporte , Endossomos/metabolismo , Fibroblastos/parasitologia , Proteínas de Fluorescência Verde/metabolismo , Humanos , Hidrólise , Immunoblotting , Lipoproteínas LDL/metabolismo , Microscopia Eletrônica , Mutação , Filogenia , Plasmodium falciparum/metabolismo , Estrutura Terciária de Proteína , Proteínas de Protozoários , Frações Subcelulares , Toxoplasma , Transfecção , Transgenes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...