Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Stem Cells ; 41(11): 1037-1046, 2023 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-37632456

RESUMO

Inherited retinal degeneration is a term used to describe heritable disorders that result from the death of light sensing photoreceptor cells. Although we and others believe that it will be possible to use gene therapy to halt disease progression early in its course, photoreceptor cell replacement will likely be required for patients who have already lost their sight. While advances in autologous photoreceptor cell manufacturing have been encouraging, development of technologies capable of efficiently delivering genome editing reagents to stem cells using current good manufacturing practices (cGMP) are needed. Gene editing reagents were delivered to induced pluripotent stem cells (iPSCs) using a Zephyr microfluidic transfection platform (CellFE). CRISPR-mediated cutting was quantified using an endonuclease assay. CRISPR correction was confirmed via digital PCR and Sanger sequencing. The resulting corrected cells were also karyotyped and differentiated into retinal organoids. We describe use of a novel microfluidic transfection platform to correct, via CRISPR-mediated homology-dependent repair (HDR), a disease-causing NR2E3 mutation in patient-derived iPSCs using cGMP compatible reagents and approaches. We show that the resulting cell lines have a corrected genotype, exhibit no off-target cutting, retain pluripotency and a normal karyotype and can be differentiated into retinal tissue suitable for transplantation. The ability to codeliver CRISPR/Cas9 and HDR templates to patient-derived iPSCs without using proprietary transfection reagents will streamline manufacturing protocols, increase the safety of resulting cell therapies, and greatly reduce the regulatory burden of clinical trials.


Assuntos
Edição de Genes , Células-Tronco Pluripotentes Induzidas , Humanos , Edição de Genes/métodos , Sistemas CRISPR-Cas/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Microfluídica , Transfecção
2.
Sci Rep ; 11(1): 21407, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34725429

RESUMO

Messenger RNA (mRNA) delivery provides gene therapy with the potential to achieve transient therapeutic efficacy without risk of insertional mutagenesis. Amongst other applications, mRNA can be employed as a platform to deliver gene editing molecules, to achieve protein expression as an alternative to enzyme replacement therapies, and to express chimeric antigen receptors (CARs) on immune cells for the treatment of cancer. We designed a novel microfluidic device that allows for efficient mRNA delivery via volume exchange for convective transfection (VECT). In the device, cells flow through a ridged channel that enforces a series of ultra-fast and large intensity deformations able to transiently open pores and induce convective transport of mRNA into the cell. Here, we describe efficient delivery of mRNA into T cells, natural killer (NK) cells and hematopoietic stem and progenitor cells (HSPCs), three human primary cell types widely used for ex vivo gene therapy applications. Results demonstrate that the device can operate at a wide range of cell and payload concentrations and that ultra-fast compressions do not have a negative impact on T cell function, making this a novel and competitive platform for the development of ex vivo mRNA-based gene therapies and other cell products engineered with mRNA.


Assuntos
Células-Tronco Hematopoéticas/citologia , Linfócitos/metabolismo , Microfluídica , Células-Tronco/citologia , Transfecção/métodos , Antígenos CD34/biossíntese , Transporte Biológico , Sobrevivência Celular , Eletroporação , Citometria de Fluxo , Terapia Genética , Humanos , Células Matadoras Naturais/citologia , Dispositivos Lab-On-A-Chip , Engenharia de Proteínas , RNA Mensageiro/metabolismo , Linfócitos T/citologia
3.
Artigo em Inglês | MEDLINE | ID: mdl-29780657

RESUMO

The mechanical properties of cells change with their differentiation, chronological age, and malignant progression. Consequently, these properties may be useful label-free biomarkers of various functional or clinically relevant cell states. Here, we demonstrate mechano-node-pore sensing (mechano-NPS), a multi-parametric single-cell-analysis method that utilizes a four-terminal measurement of the current across a microfluidic channel to quantify simultaneously cell diameter, resistance to compressive deformation, transverse deformation under constant strain, and recovery time after deformation. We define a new parameter, the whole-cell deformability index (wCDI), which provides a quantitative mechanical metric of the resistance to compressive deformation that can be used to discriminate among different cell types. The wCDI and the transverse deformation under constant strain show malignant MCF-7 and A549 cell lines are mechanically distinct from non-malignant, MCF-10A and BEAS-2B cell lines, and distinguishes between cells treated or untreated with cytoskeleton-perturbing small molecules. We categorize cell recovery time, ΔTr, as instantaneous (ΔTr ~ 0 ms), transient (ΔTr ≤ 40ms), or prolonged (ΔTr > 40ms), and show that the composition of recovery types, which is a consequence of changes in cytoskeletal organization, correlates with cellular transformation. Through the wCDI and cell-recovery time, mechano-NPS discriminates between sub-lineages of normal primary human mammary epithelial cells with accuracy comparable to flow cytometry, but without antibody labeling. Mechano-NPS identifies mechanical phenotypes that distinguishes lineage, chronological age, and stage of malignant progression in human epithelial cells.

4.
Adv Healthc Mater ; 7(12): e1800122, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29700986

RESUMO

Engineering physiologically relevant in vitro models of human organs remains a fundamental challenge. Despite significant strides made within the field, many promising organ-on-a-chip models fall short in recapitulating cellular interactions with neighboring cell types, surrounding extracellular matrix (ECM), and exposure to soluble cues due, in part, to the formation of artificial structures that obstruct >50% of the surface area of the ECM. Here, a 3D cell culture platform based upon hydrophobic patterning of hydrogels that is capable of precisely generating a 3D ECM within a microfluidic channel with an interaction area >95% is reported. In this study, for demonstrative purposes, type I collagen (COL1), Matrigel (MAT), COL1/MAT mixture, hyaluronic acid, and cell-laden MAT are formed in the device. Three potential applications are demonstrated, including creating a 3D endothelium model, studying the interstitial migration of cancer cells, and analyzing stem cell differentiation in a 3D environment. The hydrophobic patterned-based 3D cell culture device provides the ease-of-fabrication and flexibility necessary for broad potential applications in organ-on-a-chip platforms.


Assuntos
Técnicas de Cultura de Células , Dispositivos Lab-On-A-Chip , Técnicas Analíticas Microfluídicas , Técnicas de Cultura de Células/instrumentação , Técnicas de Cultura de Células/métodos , Diferenciação Celular , Movimento Celular , Humanos , Células MCF-7 , Técnicas Analíticas Microfluídicas/instrumentação , Técnicas Analíticas Microfluídicas/métodos
5.
J Control Release ; 229: 1-9, 2016 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-26956592

RESUMO

Aptamers have recently emerged as reliable and promising targeting agents in the field of biology. However, their therapeutic potential has yet to be completely assessed due to their poor pharmacokinetics for systemic administration. Here, we describe a novel aptamer-antibody complex, designated an "oligobody" (oligomer+antibody) that may overcome the therapeutic limitations of aptamers. To provide proof-of-principle study, we investigated the druggability of oligobody in vivo using cotinine conjugated t44-OMe aptamer, which is specific for the sequence of pegaptanib, and an anti-cotinine antibody. The antibody part of oligobody resulted in extended in vivo pharmacokinetics of the aptamer without influencing its binding affinity. Moreover, the aptamer of oligobody penetrated deeply into the tumor tissues whereas the anti-VEGF antibody did not. Finally, the systemic administration of this oligobody reduced the tumor burden in a xenograft mouse model. Together, these results suggested that our oligobody strategy may represent a novel platform for rapid, low-cost and high-throughput cancer therapy.


Assuntos
Anticorpos Monoclonais , Aptâmeros de Nucleotídeos , Cotinina , Neoplasias Pulmonares/tratamento farmacológico , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Células A549 , Animais , Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Aptâmeros de Nucleotídeos/farmacocinética , Aptâmeros de Nucleotídeos/uso terapêutico , Cotinina/química , Cotinina/imunologia , Sistemas de Liberação de Medicamentos , Feminino , Células Hep G2 , Células Endoteliais da Veia Umbilical Humana , Humanos , Neoplasias Pulmonares/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Nus , Terapia de Alvo Molecular
6.
Biomaterials ; 63: 177-88, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26113074

RESUMO

Paracrine signals produced from stem cells influence tissue regeneration by inducing the differentiation of endogenous stem or progenitor cells. However, many recent studies that have investigated paracrine signaling of stem cells have relied on either two-dimensional transwell systems or conditioned medium culture, neither of which provide optimal culture microenvironments for elucidating the effects of paracrine signals in vivo. In this study, we recapitulated in vivo-like paracrine signaling of human mesenchymal stem cells (hMSCs) to enhance functional neuronal differentiation of human neural stem cells (hNSCs) in three-dimensional (3D) extracellular matrices (ECMs) within a microfluidic array platform. In order to amplify paracrine signaling, hMSCs were genetically engineered using cationic polymer nanoparticles to overexpress glial cell-derived neurotrophic factor (GDNF). hNSCs were cultured in 3D ECM hydrogel used to fill central channels of the microfluidic device, while GDNF-overexpressing hMSCs (GDNF-hMSCs) were cultured in channels located on both sides of the central channel. This setup allowed for mimicking of paracrine signaling between genetically engineered hMSCs and endogenous hNSCs in the brain. Co-culture of hNSCs with GDNF-hMSCs in the 3D microfluidic system yielded reduced glial differentiation of hNSCs while significantly enhancing differentiation into neuronal cells including dopaminergic neurons. Neuronal cells produced from hNSCs differentiating in the presence of GDNF-hMSCs exhibited functional neuron-like electrophysiological features. The enhanced paracrine ability of GDNF-hMSCs was finally confirmed using an animal model of hypoxic-ischemic brain injury. This study demonstrates the presented 3D microfluidic array device can provide an efficient co-culture platform and provide an environment for paracrine signals from transplanted stem cells to control endogenous neuronal behaviors in vivo.


Assuntos
Técnicas de Cultura de Células/instrumentação , Dispositivos Lab-On-A-Chip , Células-Tronco Mesenquimais/citologia , Células-Tronco Neurais/citologia , Neurogênese , Comunicação Parácrina , Animais , Lesões Encefálicas/terapia , Desenho de Equipamento , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Humanos , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Camundongos , Células-Tronco Neurais/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Regulação para Cima
7.
Integr Biol (Camb) ; 6(7): 654-61, 2014 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-24844199

RESUMO

We present a novel in vitro breast tumor model to mimic intratumoral phenotypic heterogeneity based on a microfluidic system incorporating ECM scaffolds capable of providing a physiologically relevant tumor microenvironment. To study the regulation of invasive potentials by intratumoral subpopulation conditions, we developed heterogeneous cancer cell subpopulations by co-culturing two breast cancer cell types with distinct phenotypes, specifically, highly invasive and epithelial-like cancer cells. Our results indicate that intratumoral phenotypic heterogeneity acts as an encourager of cancer cell invasion through a 3D matrix depending on the neighboring ECM, with highly invasive cancer cells acting as the 'leader' and epithelial-like cancer cells as the 'follower', therefore enhancing the metastatic potential.


Assuntos
Adenocarcinoma/patologia , Neoplasias da Mama/patologia , Matriz Extracelular/patologia , Microambiente Tumoral/fisiologia , Linhagem Celular Tumoral , Técnicas de Cocultura , Células Epiteliais , Feminino , Humanos , Microfluídica/instrumentação , Microfluídica/métodos , Microscopia de Fluorescência , Fenótipo
8.
Adv Healthc Mater ; 2(6): 790-4, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23184641

RESUMO

Plasticity and reciprocity of breast cancer cells to various extracellular matrice (ECMs) are three-dimensionally analyzed in quantitative way in a novel and powerful microfluidic in vitro platform. This successfully demonstrates the metastatic potential of cancer cells and their effective strategies of ECM proteolytic remodeling and morphological change, while interacting with other cells and invading into heterogeneous ECMs.


Assuntos
Adenocarcinoma/patologia , Adenocarcinoma/fisiopatologia , Neoplasias da Mama/patologia , Neoplasias da Mama/fisiopatologia , Matriz Extracelular/diagnóstico por imagem , Matriz Extracelular/metabolismo , Técnicas Analíticas Microfluídicas/instrumentação , Linhagem Celular Tumoral , Separação Celular/instrumentação , Desenho de Equipamento , Análise de Falha de Equipamento , Humanos , Invasividade Neoplásica/patologia , Invasividade Neoplásica/fisiopatologia , Ultrassonografia
9.
Lab Chip ; 11(13): 2175-81, 2011 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-21617793

RESUMO

Sprouting angiogenesis requires a coordinated guidance from a variety of angiogenic factors. Here, we have developed a unique hydrogel incorporating microfluidic platform which mimics the physiological microenvironment in 3D under a precisely orchestrated gradient of soluble angiogenic factors, VEGF and ANG-1. The system enables the quantified investigation in chemotactic response of endothelial cells during the collective angiogenic sprouting process. While the presence of a VEGF gradient alone was sufficient in inducing a greater number of tip cells, addition of ANG-1 to the VEGF gradient enhanced the number of tip cells that are attached to collectively migrated stalk cells. The chemotactic response of tip cells attracted by the VEGF gradient and the stabilizing role of ANG-1 were morphologically investigated, elucidating the 3D co-operative migration of tip and stalk cells as well as their structures. We found that ANG-1 enhanced the connection of the stalk cells with the tip cells, and then the direct connection regulated the morphogenesis and/or life cycle of stalk cells.


Assuntos
Angiopoietina-1/farmacologia , Técnicas Analíticas Microfluídicas/métodos , Neovascularização Fisiológica/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/farmacologia , Angiopoietina-1/metabolismo , Linhagem Celular , Difusão , Relação Dose-Resposta a Droga , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/metabolismo , Humanos , Técnicas Analíticas Microfluídicas/instrumentação , Fator A de Crescimento do Endotélio Vascular/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA