Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Biotechnol Bioeng ; 114(2): 407-415, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27543759

RESUMEN

Microbial cells have for many years been engineered to facilitate efficient production of biologics, chemicals, and other compounds. As the "metabolic" burden of synthetic genetic components can impair cell performance, microbial consortia are being developed to piece together specialized subpopulations that collectively produce desired products. Their use, however, has been limited by the inability to control their composition and function. One approach to leverage advantages of the division of labor within consortia is to link microbial subpopulations together through quorum sensing (QS) molecules. Previously, we directed the assembly of "quantized quorums," microbial subpopulations that are parsed through QS activation, by the exogenous addition of QS signal molecules to QS synthase mutants. In this work, we develop a more facile and general platform for creating "quantized quorums." Moreover, the methodology is not restricted to QS-mutant populations. We constructed quorum quenching capsules that partition QS-mediated phenotypes into discrete subpopulations. This compartmentalization guides QS subpopulations in a dose-dependent manner, parsing cell populations into activated or deactivated groups. The capsular "devices" consist of polyelectrolyte alginate-chitosan beads that encapsulate high-efficiency (HE) "controller cells" that, in turn, provide rapid uptake of the QS signal molecule AI-2 from culture fluids. In this methodology, instead of adding AI-2 to parse QS-mutants into subpopulations, we engineered cells to encapsulate them into compartments, and they serve to deplete AI-2 from wild-type populations. These encapsulated bacteria therefore, provide orthogonal control of population composition while allowing only minimal interaction with the product-producing cell population or consortia. We envision that compartmentalized control of QS should have applications in both metabolic engineering and human disease. Biotechnol. Bioeng. 2017;114: 407-415. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Bacterias , Ingeniería Metabólica/métodos , Consorcios Microbianos/fisiología , Modelos Biológicos , Percepción de Quorum/fisiología , Bacterias/citología , Bacterias/metabolismo , Fenotipo
2.
Sci Rep ; 13(1): 5032, 2023 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-36977745

RESUMEN

Hutchinson-Gilford Progeria Syndrome (HGPS) is a rare, fatal genetic disease that accelerates atherosclerosis. With a limited pool of HGPS patients, clinical trials face unique challenges and require reliable preclinical testing. We previously reported a 3D tissue engineered blood vessel (TEBV) microphysiological system fabricated with iPSC-derived vascular cells from HGPS patients. HGPS TEBVs exhibit features of HGPS atherosclerosis including loss of smooth muscle cells, reduced vasoactivity, excess extracellular matrix (ECM) deposition, inflammatory marker expression, and calcification. We tested the effects of HGPS therapeutics Lonafarnib and Everolimus separately and together, currently in Phase I/II clinical trial, on HGPS TEBVs. Everolimus decreased reactive oxygen species levels, increased proliferation, reduced DNA damage in HGPS vascular cells, and improved vasoconstriction in HGPS TEBVs. Lonafarnib improved shear stress response of HGPS iPSC-derived endothelial cells (viECs) and reduced ECM deposition, inflammation, and calcification in HGPS TEBVs. Combination treatment with Lonafarnib and Everolimus produced additional benefits such as improved endothelial and smooth muscle marker expression and reduced apoptosis, as well as increased TEBV vasoconstriction and vasodilation. These results suggest that a combined trial of both drugs may provide cardiovascular benefits beyond Lonafarnib, if the Everolimus dose can be tolerated.


Asunto(s)
Aterosclerosis , Calcinosis , Células Madre Pluripotentes Inducidas , Progeria , Humanos , Progeria/genética , Everolimus/farmacología , Everolimus/uso terapéutico , Everolimus/metabolismo , Células Endoteliales/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Aterosclerosis/metabolismo , Calcinosis/metabolismo , Lamina Tipo A/genética
3.
Methods Mol Biol ; 2375: 77-90, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34591300

RESUMEN

Human tissue-engineered blood vessels (TEBVs) that exhibit vasoactivity can be used to test drug toxicity, modulate pro-inflammatory cytokines, and model disease states in vitro. We developed a novel device to fabricate arteriole-scale human endothelialized TEBVs in situ with smaller volumes and higher throughput than previously reported. Both primary and induced pluripotent stem cell (iPSC)-derived cells can be used. Four collagen TEBVs with 600µm inner diameter and 2.9 mm outer diameter are fabricated by pipetting a solution of collagen and medial cells into a three-layer acrylic mold. After gelation, the TEBVs are released from the mold and dehydrated. After suturing the TEBVs in place and changing the mold parts to form a perfusion chamber, the TEBVs are endothelialized in situ, and then media is perfused through the lumen. By removing 90% of the water after gelation, the TEBVs become mechanically strong enough for perfusion at the physiological shear stress of 0.4 Pa within 24 h of fabrication and maintain function for at least 5 weeks.


Asunto(s)
Ingeniería de Tejidos , Arteriolas , Vasos Sanguíneos , Colágeno , Humanos , Células Madre Pluripotentes Inducidas , Perfusión
4.
STAR Protoc ; 2(2): 100394, 2021 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-33796871

RESUMEN

Induced pluripotent stem cells (iPSCs) offer a potentially unlimited source to generate endothelial cells (ECs) for numerous applications. Here, we describe a 7-day protocol to differentiate up to 55 million vascular endothelial cells (viECs) from 3.5 million human iPSCs using small molecules to regulate specific transcription factors. We also describe a parallel-plate flow chamber system to study EC behavior under physiological shear stress. For complete details on the use and execution of this protocol, please refer to Atchison et al. (2020).


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Células Endoteliales , Células Madre Pluripotentes Inducidas , Estrés Mecánico , Estrés Fisiológico/fisiología , Diferenciación Celular/fisiología , Células Cultivadas , Células Endoteliales/citología , Células Endoteliales/fisiología , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/fisiología
5.
Front Cardiovasc Med ; 8: 625016, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33659279

RESUMEN

The vascular endothelium is present in all organs and blood vessels, facilitates the exchange of nutrients and waste throughout different organ systems in the body, and sets the tone for healthy vessel function. Mechanosensitive in nature, the endothelium responds to the magnitude and temporal waveform of shear stress in the vessels. Endothelial dysfunction can lead to atherosclerosis and other diseases. Modeling endothelial function and dysfunction in organ systems in vitro, such as the blood-brain barrier and tissue-engineered blood vessels, requires sourcing endothelial cells (ECs) for these biomedical engineering applications. It can be difficult to source primary, easily renewable ECs that possess the function or dysfunction in question. In contrast, human pluripotent stem cells (hPSCs) can be sourced from donors of interest and renewed almost indefinitely. In this review, we highlight how knowledge of vascular EC development in vivo is used to differentiate induced pluripotent stem cells (iPSC) into ECs. We then describe how iPSC-derived ECs are being used currently in in vitro models of organ function and disease and in vivo applications.

6.
Cell Stem Cell ; 26(2): 136-137, 2020 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-32032523

RESUMEN

Human induced pluripotent stem cells (hiPSCs) provide a potentially unlimited cell source for producing autologous tissue-engineered vascular grafts (TEVGs), which currently suffer from low mechanical strength. In this issue of Cell Stem Cell, Luo et al. (2020) describe optimized culture media and a mechanical stretching regimen to produce hiPSC-derived TEVGs with mechanical behavior similar to that of natural vessels.


Asunto(s)
Células Madre Pluripotentes Inducidas , Prótesis Vascular , Humanos , Ingeniería de Tejidos
7.
Stem Cell Reports ; 14(2): 325-337, 2020 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-32032552

RESUMEN

Hutchinson-Gilford progeria syndrome (HGPS) is a rare disorder caused by a point mutation in the Lamin A gene that produces the protein progerin. Progerin toxicity leads to accelerated aging and death from cardiovascular disease. To elucidate the effects of progerin on endothelial cells, we prepared tissue-engineered blood vessels (viTEBVs) using induced pluripotent stem cell-derived smooth muscle cells (viSMCs) and endothelial cells (viECs) from HGPS patients. HGPS viECs aligned with flow but exhibited reduced flow-responsive gene expression and altered NOS3 levels. Relative to viTEBVs with healthy cells, HGPS viTEBVs showed reduced function and exhibited markers of cardiovascular disease associated with endothelium. HGPS viTEBVs exhibited a reduction in both vasoconstriction and vasodilation. Preparing viTEBVs with HGPS viECs and healthy viSMCs only reduced vasodilation. Furthermore, HGPS viECs produced VCAM1 and E-selectin protein in TEBVs with healthy or HGPS viSMCs. In summary, the viTEBV model has identified a role of the endothelium in HGPS.


Asunto(s)
Prótesis Vascular , Vasos Sanguíneos/fisiopatología , Células Madre Pluripotentes Inducidas/patología , Modelos Biológicos , Progeria/patología , Ingeniería de Tejidos , Vasos Sanguíneos/patología , Células Clonales , Regulación de la Expresión Génica , Humanos , Masculino , Fenotipo , Donantes de Tejidos
8.
Biomaterials ; 221: 119416, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31419653

RESUMEN

Skeletal muscle is the largest organ of human body with several important roles in everyday movement and metabolic homeostasis. The limited ability of small animal models of muscle disease to accurately predict drug efficacy and toxicity in humans has prompted the development in vitro models of human skeletal muscle that fatefully recapitulate cell and tissue level functions and drug responses. We first review methods for development of three-dimensional engineered muscle tissues and organ-on-a-chip microphysiological systems and discuss their potential utility in drug discovery research and development of new regenerative therapies. Furthermore, we describe strategies to increase the functional maturation of engineered muscle, and motivate the importance of incorporating multiple tissue types on the same chip to model organ cross-talk and generate more predictive drug development platforms. Finally, we review the ability of available in vitro systems to model diseases such as type II diabetes, Duchenne muscular dystrophy, Pompe disease, and dysferlinopathy.


Asunto(s)
Descubrimiento de Drogas/métodos , Músculo Esquelético/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Músculo Esquelético/citología , Distrofias Musculares/metabolismo , Distrofias Musculares/terapia , Ingeniería de Tejidos/métodos
9.
Biotechnol Prog ; 35(6): e2881, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31306566

RESUMEN

The autoinducer-2 (AI-2) quorum sensing system is involved in a range of population-based bacterial behaviors and has been engineered for cell-cell communication in synthetic biology systems. Investigation into the cellular mechanisms of AI-2 processing has determined that overexpression of uptake genes increases AI-2 uptake rate, and genomic deletions of degradation genes lowers the AI-2 level required for activation of reporter genes. Here, we combine these two strategies to engineer an Escherichia coli strain with enhanced ability to detect and respond to AI-2. In an E. coli strain that does not produce AI-2, we monitored AI-2 uptake and reporter protein expression in a strain that overproduced the AI-2 uptake or phosphorylation units LsrACDB or LsrK, a strain with the deletion of AI-2 degradation units LsrF and LsrG, and an "enhanced" strain with both overproduction of AI-2 uptake and deletion of AI-2 degradation elements. By adding up to 40 µM AI-2 to growing cell cultures, we determine that this "enhanced" AI-2 sensitive strain both uptakes AI-2 more rapidly and responds with increased reporter protein expression than the others. This work expands the toolbox for manipulating AI-2 quorum sensing processes both in native environments and for synthetic biology applications.


Asunto(s)
Escherichia coli/fisiología , Homoserina/análogos & derivados , Percepción de Quorum/fisiología , Bioingeniería , Escherichia coli/genética , Homoserina/fisiología , Lactonas
10.
Nat Biomed Eng ; 2(12): 942-954, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30581652

RESUMEN

Adult skeletal muscle has a robust capacity for self-repair, owing to synergies between muscle satellite cells and the immune system. In vitro models of muscle self-repair would facilitate the basic understanding of muscle regeneration and the screening of therapies for muscle disease. Here, we show that the incorporation of macrophages into muscle tissues engineered from adult-rat myogenic cells enables near-complete structural and functional repair after cardiotoxic injury in vitro. First, we show that-in contrast with injured neonatal-derived engineered muscle-adult-derived engineered muscle fails to properly self-repair after injury, even when treated with pro-regenerative cytokines. We then show that rat bone-marrow-derived macrophages or human blood-derived macrophages resident within the in vitro engineered tissues stimulate muscle satellite cell-mediated myogenesis while significantly limiting myofibre apoptosis and degeneration. Moreover, bone-marrow-derived macrophages within engineered tissues implanted in a mouse dorsal window-chamber model augmented blood vessel ingrowth, cell survival, muscle regeneration and contractile function.

SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda