Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Nucleic Acids Res ; 52(10): e47, 2024 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-38709890

RESUMEN

Sequence verification of plasmid DNA is critical for many cloning and molecular biology workflows. To leverage high-throughput sequencing, several methods have been developed that add a unique DNA barcode to individual samples prior to pooling and sequencing. However, these methods require an individual plasmid extraction and/or in vitro barcoding reaction for each sample processed, limiting throughput and adding cost. Here, we develop an arrayed in vivo plasmid barcoding platform that enables pooled plasmid extraction and library preparation for Oxford Nanopore sequencing. This method has a high accuracy and recovery rate, and greatly increases throughput and reduces cost relative to other plasmid barcoding methods or Sanger sequencing. We use in vivo barcoding to sequence verify >45 000 plasmids and show that the method can be used to transform error-containing dispersed plasmid pools into sequence-perfect arrays or well-balanced pools. In vivo barcoding does not require any specialized equipment beyond a low-overhead Oxford Nanopore sequencer, enabling most labs to flexibly process hundreds to thousands of plasmids in parallel.


Asunto(s)
Biblioteca de Genes , Secuenciación de Nucleótidos de Alto Rendimiento , Plásmidos , Plásmidos/genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Análisis de Secuencia de ADN/métodos , ADN/genética , Código de Barras del ADN Taxonómico/métodos , Secuenciación de Nanoporos/métodos
2.
Cytotherapy ; 26(7): 714-718, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38506768

RESUMEN

BACKGROUND AIMS: In this paper, we present a review of several selected talks presented at the CTTACC conference (Cellular Therapies in Trauma and Critical Care) held in Scottsdale, AZ in May 2023. This conference review highlights the potential for cellular therapies to "reset" the dysregulated immune response and restore physiologic functions to normal. Improvements in medical care systems and technology have increasingly saved lives after major traumatic events. However, many of these patients have complicated post-traumatic sequelae, ranging from short-term multi-organ failure to chronic critical illness. METHODS/RESULTS: Patients with chronic critical illness have been found to have dysregulated immune responses. These abnormal and harmful immune responses persist for years after the initial insult and can potentially be mitigated by treatment with cellular therapies. CONCLUSIONS: The sessions emphasized the need for more research and clinical trials with cellular therapies for the treatment of a multitude of chronic illnesses: post-trauma, radiation injury, COVID-19, burns, traumatic brain injury (TBI) and other chronic infections.


Asunto(s)
Quemaduras , COVID-19 , Tratamiento Basado en Trasplante de Células y Tejidos , Humanos , Lesiones Traumáticas del Encéfalo/terapia , Lesiones Traumáticas del Encéfalo/inmunología , Lesiones Traumáticas del Encéfalo/complicaciones , Quemaduras/terapia , Quemaduras/inmunología , Quemaduras/complicaciones , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Enfermedad Crónica , COVID-19/inmunología , COVID-19/terapia , Enfermedad Crítica , Sistema Inmunológico , Infecciones/terapia , Infecciones/inmunología , Infecciones/etiología , SARS-CoV-2 , Heridas y Lesiones/terapia , Heridas y Lesiones/inmunología , Heridas y Lesiones/complicaciones , Congresos como Asunto
3.
Int J Mol Sci ; 24(11)2023 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-37298556

RESUMEN

Mesenchymal stromal cells (MSCs) are a candidate for cell immunotherapy due to potent immunomodulatory activity found in their secretome. Though studies on their secreted substances have been reported, the time dynamics of MSC potency remain unclear. Herein, we report on the dynamics of MSC secretome potency in an ex vivo hollow fiber bioreactor using a continuous perfusion cell culture system that fractionated MSC-secreted factors over time. Time-resolved fractions of MSC-conditioned media were evaluated for potency by incubation with activated immune cells. Three studies were designed to characterize MSC potency under: (1) basal conditions, (2) in situ activation, and (3) pre-licensing. Results indicate that the MSC secretome is most potent in suppressing lymphocyte proliferation during the first 24 h and is further stabilized when MSCs are prelicensed with a cocktail of pro-inflammatory cytokines, IFNγ, TNFα, and IL-1ß. The evaluation of temporal cell potency using this integrated bioreactor system can be useful in informing strategies to maximize MSC potency, minimize side effects, and allow greater control for the duration of ex vivo administration approaches.


Asunto(s)
Citocinas , Células Madre Mesenquimatosas , Inmunomodulación , Medios de Cultivo Condicionados , Perfusión , Proliferación Celular
4.
Cytotherapy ; 24(8): 774-788, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35613962

RESUMEN

The ISCT Scientific Signature Series Symposium "Advances in Cell and Gene Therapies for Lung Diseases and Critical Illnesses" was held as an independent symposium in conjunction with the biennial meeting, "Stem Cells, Cell Therapies, and Bioengineering in Lung Biology and Diseases," which took place July 12-15, 2021, at the University of Vermont. This is the third Respiratory System-based Signature Series event; the first 2, "Tracheal Bioengineering, the Next Steps" and "Cellular Therapies for Pulmonary Diseases and Critical Illnesses: State of the Art of European Science," took place in 2014 and 2015, respectively. Cell- and gene-based therapies for respiratory diseases and critical illnesses continue to be a source of great promise and opportunity. This reflects ongoing advancements in understanding of the mechanisms by which cell-based therapies, particularly those using mesenchymal stromal cells (MSCs), can mitigate different lung injuries and the increasing sophistication with which preclinical data is translated into clinical investigations. This also reflects continuing evolution in gene transfer vectors, including those designed for in situ gene editing in parallel with those targeting gene or cell replacement. Therefore, this symposium convened global thought leaders in a forum designed to catalyze communication and collaboration to bring the greatest possible innovation and value of cell- and gene-based therapies for patients with respiratory diseases and critical illnesses.


Asunto(s)
Enfermedad Crítica , Enfermedades Pulmonares , Tratamiento Basado en Trasplante de Células y Tejidos , Enfermedad Crítica/terapia , Terapia Genética , Humanos , Enfermedades Pulmonares/genética , Enfermedades Pulmonares/terapia , Células Madre
5.
J Fluoresc ; 32(2): 521-531, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34989923

RESUMEN

Tumor spheroid models have proven useful in the study of cancer cell responses to chemotherapeutic compounds by more closely mimicking the 3-dimensional nature of tumors in situ. Their advantages are often offset, however, by protocols that are long, complicated, and expensive. Efforts continue for the development of high-throughput assays that combine the advantages of 3D models with the convenience and simplicity of traditional 2D monolayer methods. Herein, we describe the development of a breast cancer spheroid image cytometry assay using T47D cells in Aggrewell™400 spheroid plates. Using the Celigo® automated imaging system, we developed a method to image and individually track thousands of spheroids within the Aggrewell™400 microwell plate over time. We demonstrate the use of calcein AM and propidium iodide staining to study the effects of known anti-cancer drugs Doxorubicin, Everolimus, Gemcitabine, Metformin, Paclitaxel and Tamoxifen. We use the image cytometry results to quantify the fluorescence of calcein AM and PI as well as spheroid size in a dose dependent manner for each of the drugs. We observe a dose-dependent reduction in spheroid size and find that it correlates well with the viability obtained from the CellTiter96® endpoint assay. The image cytometry method we demonstrate is a convenient and high-throughput drug-response assay for breast cancer spheroids under 400 µm in diameter, and may lay a foundation for investigating other three-dimensional spheroids, organoids, and tissue samples.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Ensayos Analíticos de Alto Rendimiento/métodos , Citometría de Imagen/métodos , Esferoides Celulares/efectos de los fármacos , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Fluoresceínas , Colorantes Fluorescentes , Humanos , Propidio
6.
Anal Biochem ; 625: 114213, 2021 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-33887234

RESUMEN

Cells exchange substances with their surroundings during metabolism, signaling, and other functions. These fluxes are dynamic, changing in response to external cues and internal programs. Static cultures are inadequate for measuring these dynamics because the environments of the cells change as substances accumulate or deplete from medium, unintentionally affecting cell behavior. Static cultures offer limited time resolution due to the impracticality of frequent or prolonged manual sampling, and cannot expose cells to smooth, transient changes in stimulus concentrations. In contrast, perfusion cultures constantly maintain cellular environments and continuously sample the effluent stream. Existing perfusion culture systems are either microfluidic, which are difficult to make and use, or macrofluidic devices built from custom parts that neglect solute dispersion. In this study, a multiplexed macrofluidic perfusion culture platform was developed to measure secretion and absorption rates of substances by cells in a temporally controlled environment. The modular platform handles up to 31 streams with automated fraction collection. This paper presents the assembly of this dynamic bioreactor from commercially available parts, and a method for quantitatively handling the effects of dispersion using residence time distributions. The system is then applied to monitor the secretion of a circadian clock gene-driven reporter from engineered cells.


Asunto(s)
Reactores Biológicos , Técnicas de Cultivo de Célula/métodos , Fenómenos Fisiológicos Celulares , Supervivencia Celular , Células Cultivadas , Medios de Cultivo
7.
PLoS Biol ; 16(9): e2005046, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30180168

RESUMEN

The microenvironment of lymphoid organs can aid healthy immune function through provision of both structural and molecular support. In mice, fibroblastic reticular cells (FRCs) create an essential T-cell support structure within lymph nodes, while human FRCs are largely unstudied. Here, we show that FRCs create a regulatory checkpoint in human peripheral T-cell activation through 4 mechanisms simultaneously utilised. Human tonsil and lymph node-derived FRCs constrained the proliferation of both naïve and pre-activated T cells, skewing their differentiation away from a central memory T-cell phenotype. FRCs acted unilaterally without requiring T-cell feedback, imposing suppression via indoleamine-2,3-dioxygenase, adenosine 2A Receptor, prostaglandin E2, and transforming growth factor beta receptor (TGFßR). Each mechanistic pathway was druggable, and a cocktail of inhibitors, targeting all 4 mechanisms, entirely reversed the suppressive effect of FRCs. T cells were not permanently anergised by FRCs, and studies using chimeric antigen receptor (CAR) T cells showed that immunotherapeutic T cells retained effector functions in the presence of FRCs. Since mice were not suitable as a proof-of-concept model, we instead developed a novel human tissue-based in situ assay. Human T cells stimulated using standard methods within fresh tonsil slices did not proliferate except in the presence of inhibitors described above. Collectively, we define a 4-part molecular mechanism by which FRCs regulate the T-cell response to strongly activating events in secondary lymphoid organs while permitting activated and CAR T cells to utilise effector functions. Our results define 4 feasible strategies, used alone or in combinations, to boost primary T-cell responses to infection or cancer by pharmacologically targeting FRCs.


Asunto(s)
Diferenciación Celular/inmunología , Microambiente Celular , Ganglios Linfáticos/inmunología , Activación de Linfocitos/inmunología , Linfocitos T/citología , Adulto , Proliferación Celular , Niño , Fibroblastos/citología , Humanos , Memoria Inmunológica , Fenotipo
8.
BMC Biotechnol ; 19(1): 50, 2019 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-31340783

RESUMEN

BACKGROUND: Long Adapter Single-Stranded Oligonucleotide (LASSO) probes were developed as a novel tool for massively parallel cloning of kilobase-long genomic DNA sequences. LASSO dramatically improves the capture length limit of current DNA padlock probe technology from approximately 150 bps to several kbps. High-throughput LASSO capture involves the parallel assembly of thousands of probes. However, malformed probes are indiscernible from properly formed probes using gel electrophoretic techniques. Therefore, we used next-generation sequencing (NGS) to assess the efficiency of LASSO probe assembly and how it relates to the nature of DNA capture and amplification. Additionally, we introduce a simplified single target LASSO protocol using classic molecular biology techniques for qualitative and quantitative assessment of probe specificity. RESULTS: A LASSO probe library targeting 3164 unique E. coli ORFs was assembled using two different probe assembly reaction conditions with a 40-fold difference in DNA concentration. Unique probe sequences are located within the first 50 bps of the 5' and 3' ends, therefore we used paired-end NGS to assess probe library quality. Properly mapped read pairs, representing correctly formed probes, accounted for 10.81 and 0.65% of total reads, corresponding to ~ 80% and ~ 20% coverage of the total probe library for the lower and higher DNA concentration conditions, respectively. Subsequently, we used single-end NGS to correlate probe assembly efficiency and capture quality. Significant enrichment of LASSO targets over non-targets was only observed for captures done using probes assembled with a lower DNA concentration. Additionally, semi-quantitative polyacrylamide gel electrophoresis revealed a ~ 10-fold signal-to-noise ratio of LASSO capture in a simplified system. CONCLUSIONS: These results suggest that LASSO probe coverage for target sequences is more predictive of successful capture than probe assembly depth-enrichment. Concomitantly, these results demonstrate that DNA concentration at a critical step in the probe assembly reaction significantly impacts probe formation. Additionally, we show that a simplified LASSO capture protocol coupled to PAGE (polyacrylamide gel electrophoresis) is highly specific and more amenable to small-scale LASSO approaches, such as screening novel probes and templates.


Asunto(s)
Clonación Molecular/métodos , Sondas de ADN/genética , ADN de Cadena Simple/genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Oligonucleótidos/genética , ADN/análisis , ADN/genética , Cartilla de ADN/genética , Electroforesis en Gel de Poliacrilamida/métodos , Proteínas de Escherichia coli/genética , Amplificación de Genes , Biblioteca de Genes , Sistemas de Lectura Abierta/genética , Reacción en Cadena de la Polimerasa/métodos , Reproducibilidad de los Resultados
9.
J Transl Med ; 17(1): 263, 2019 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-31409424

RESUMEN

BACKGROUND: Mesenchymal stromal cells (MSC) demonstrate innate and regulatory immunologic functions and have been widely explored for cell therapy applications. Mechanisms by which MSCs achieve therapeutic effects are theorized, though appropriate dosing and duration of these mechanisms in vivo warrant deeper investigation. One, rapid immunosuppressive function of MSCs is through ectoenzyme expression of CD73 and CD39 which cooperatively hydrolyze inflammatory, extracellular adenosine triphosphate (ATP) to anti-inflammatory adenosine. Extracellular ATP has a key role in autoimmune and inflammatory diseases, which administered MSCs have the potential to modulate in a timescale that is befitting of shorter acting therapeutic function. METHODS: In vitro experiments were performed to determine the hydrolysis rates of ATP by MSCs. Through kinetic modeling from experimental results, the rate at which a single cell can metabolize ATP was determined. Based on these rates, the ability of MSCs to downregulate inflammatory signaling pathways was prospectively validated using model system parameters with respect to two different mechanisms: extracellular ATP stimulates lymphocytes to suppress proliferation and induce apoptosis and with co-stimulation, it stimulates monocytes to release pro-inflammatory IL-1ß. MSCs were co-cultured with immune cells using transwell inserts and compared to immune cell only groups. RESULTS: Hydrolysis of ATP was efficiently modeled by first-order enzyme kinetics. For in vitro culture, the rate at which a single cell can hydrolyize ATP is 8.9 nmol/min. In the presence of extracellular ATP, cocultures of MSCs reduced cytotoxicity and allows for proliferation of lymphocytes while limiting IL-1ß secretion from monocytes. CONCLUSIONS: Such use of these models may allow for better dosing predictions for MSCs to be used therapeutically for chronic inflammatory diseases such as rheumatoid arthritis, diabetes, pancreatitis, and other systemic inflammatory response syndromes. For the first time, the effect of MSCs on ATP hydrolysis in immune cell response is quantitatively analyzed on a cell-molecule basis by modeling the hydrolysis as an enzyme-substrate reaction. The results also give insight into MSCs' dynamic response mechanisms to ameliorate effects of extracellular ATP whether it be through positive or negative regulation.


Asunto(s)
5'-Nucleotidasa/uso terapéutico , Antígenos CD/uso terapéutico , Apirasa/uso terapéutico , Inmunomodulación , Células Madre Mesenquimatosas/citología , Adenosina/metabolismo , Adenosina Trifosfato/metabolismo , Muerte Celular , Proliferación Celular , Humanos , Hidrólisis , Interleucina-1beta/metabolismo , Cinética , Linfocitos/citología , Trasplante de Células Madre Mesenquimatosas , Especificidad por Sustrato
10.
Cytotherapy ; 21(1): 76-82, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30497956

RESUMEN

BACKGROUND AIM: Translation of therapeutic cell therapies to clinical-scale products is critical to realizing widespread success. Currently, however, there are limited tools that are accessible at the research level and readily scalable to clinical-scale needs. METHODS: We herein developed and assessed a closed loop bioreactor system in which (i) a highly gas-permeable silicone material was used to fabricate cell culture bags and (ii) dynamic flow was introduced to allow for dissociation of activated T-cell aggregates. RESULTS: Using this system, we find superior T-cell proliferation compared with conventional bag materials and flasks, especially at later time points. Furthermore, intermittent dynamic flow could easily break apart T-cell clusters. CONCLUSIONS: Our novel closed loop bioreactor system is amenable to enhanced T-cell proliferation and has broader implications for being easily scaled for use in larger need settings.


Asunto(s)
Técnicas de Cultivo Celular por Lotes/métodos , Reactores Biológicos , Proliferación Celular/fisiología , Linfocitos T/fisiología , Agregación Celular , Humanos , Activación de Linfocitos , Permeabilidad , Siliconas/química
11.
Exp Cell Res ; 362(1): 102-110, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29137914

RESUMEN

Adult bone marrow mesenchymal stromal cells (MSCs) have cross-functional, intrinsic potency that is of therapeutic interest. Their ability to regenerate bone, fat, and cartilage, modulate the immune system, and nurture the growth and function of other bone marrow hematopoietic stem/progenitor cells have all been evaluated by transplant applications of MSCs. These applications require the isolation and expansion scaled cell production. To investigate biophysical properties of MSCs that can be feasibly utilized as predictors of bioactivity during biomanufacturing, we used a low-density seeding model to drive MSCs into proliferative stress and exhibit the hallmark characteristics of in vitro aging. A low-density seeding method was used to generate MSCs from passages 1-7 to simulate serial expansion of these cells to maximize yield from a single donor. MSCs were subjected to three bioactivity assays in parallel to ascertain whether patterns in MSC age, size, and shape were associated with the outcomes of the potency assays. MSC age was found to be a predictor of adipogenesis, while cell and nuclear shape was strongly associated to hematopoietic-supportive potency. Together, these data evaluate morphological changes associated with cell potency and highlight new strategies for purification or alternatives to assessing MSC quality.


Asunto(s)
Células de la Médula Ósea/citología , Células de la Médula Ósea/fisiología , Técnicas de Cultivo de Célula/métodos , Senescencia Celular/fisiología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/fisiología , Adipogénesis/fisiología , Adulto , Médula Ósea/patología , Técnicas de Cultivo de Célula/normas , Diferenciación Celular , Proliferación Celular , Forma de la Célula , Células Cultivadas , Criopreservación , Humanos , Cultivo Primario de Células/métodos , Cultivo Primario de Células/normas
12.
Biomed Microdevices ; 20(1): 13, 2018 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-29353324

RESUMEN

There is an emerging need to process, expand, and even genetically engineer hematopoietic stem and progenitor cells (HSPCs) prior to administration for blood reconstitution therapy. A closed-system and automated solution for ex vivo HSC processing can improve adoption and standardize processing techniques. Here, we report a recirculating flow bioreactor where HSCs are stabilized and enriched for short-term processing by indirect fibroblast feeder coculture. Mouse 3 T3 fibroblasts were seeded on the extraluminal membrane surface of a hollow fiber micro-bioreactor and were found to support HSPC cell number compared to unsupported BMCs. CFSE analysis indicates that 3 T3-support was essential for the enhanced intrinsic cell cycling of HSPCs. This enhanced support was specific to the HSPC population with little to no effect seen with the Lineagepositive and Lineagenegative cells. Together, these data suggest that stromal-seeded hollow fiber micro-reactors represent a platform to screening various conditions that support the expansion and bioprocessing of HSPCs ex vivo.


Asunto(s)
Reactores Biológicos , Técnicas de Cultivo de Célula/instrumentación , Técnicas de Cultivo de Célula/métodos , Células Madre Hematopoyéticas , Animales , Línea Celular , Linaje de la Célula , Separación Celular/instrumentación , Separación Celular/métodos , Técnicas de Cocultivo , Diseño de Equipo , Femenino , Fibroblastos/citología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/fisiología , Membranas Artificiales , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-kit/metabolismo , Células del Estroma/citología
13.
Cytotherapy ; 19(12): 1537-1545, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28917628

RESUMEN

BACKGROUND AIMS: Cell transplants offer a new opportunity to deliver therapies with novel and complex mechanisms of action. Understanding the pharmacology of cell transplants is important to deliver this new therapy effectively. Currently, however, there are limited techniques to easily track cells after intravenous administration due to the dispersion of the graft throughout the entire body. METHODS: We herein developed an engineered cell system that secretes a luciferase enzyme to sensitively detect cell transplants independent of their locale by a simple blood test. We specifically studied a unique feature of cell transplant pharmacology-namely, immune clearance-using mesenchymal stromal cells (MSCs) as a proof-of-concept cell therapy. MSCs are a clinically relevant cell therapy that has been explored in several disease indications due to their innate properties of altering an immune response. RESULTS: Using this engineered reporter, we observed specific sensitivity of cell therapy exposure to the preparation of cells, cytolysis of MSCs in an allogeneic setting and a NK cell-mediated destruction of MSCs in an autologous setting. CONCLUSIONS: Our cellular tracking method has broader implications at large for assessing in vivo kinetics of various other cell therapies.


Asunto(s)
Biomarcadores/análisis , Ingeniería Genética/métodos , Células Asesinas Naturales/inmunología , Trasplante de Células Madre Mesenquimatosas/métodos , Animales , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Femenino , Proteínas Fluorescentes Verdes/genética , Humanos , Luciferasas/análisis , Luciferasas/genética , Luciferasas/metabolismo , Mediciones Luminiscentes , Células Madre Mesenquimatosas/fisiología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Trasplante Homólogo/métodos
15.
Mol Ther ; 22(5): 999-1007, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24496384

RESUMEN

The secreted proteins from a cell constitute a natural biologic library that can offer significant insight into human health and disease. Discovering new secreted proteins from cells is bounded by the limitations of traditional separation and detection tools to physically fractionate and analyze samples. Here, we present a new method to systematically identify bioactive cell-secreted proteins that circumvent traditional proteomic methods by first enriching for protein candidates by differential gene expression profiling. The bone marrow stromal cell secretome was analyzed using enriched gene expression datasets in combination with potency assay testing. Four proteins expressed by stromal cells with previously unknown anti-inflammatory properties were identified, two of which provided a significant survival benefit to mice challenged with lethal endotoxic shock. Greater than 85% of secreted factors were recaptured that were otherwise undetected by proteomic methods, and remarkable hit rates of 18% in vitro and 9% in vivo were achieved.


Asunto(s)
Proteínas Contráctiles/genética , Proteínas Contráctiles/metabolismo , Encefalinas/genética , Proteínas de la Matriz Extracelular/metabolismo , Glicoproteínas/genética , Interleucina-10/metabolismo , Precursores de Proteínas/genética , Proteínas/metabolismo , Choque Séptico/terapia , Animales , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Encefalinas/metabolismo , Proteínas de la Matriz Extracelular/genética , Perfilación de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intercelular , Interleucina-10/genética , Células Madre Mesenquimatosas/inmunología , Células Madre Mesenquimatosas/metabolismo , Ratones , Biosíntesis de Proteínas/genética , Precursores de Proteínas/metabolismo , Proteínas/genética , Proteómica , Factores de Empalme de ARN , Choque Séptico/genética
16.
Proc Natl Acad Sci U S A ; 109(48): 19638-43, 2012 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-23150542

RESUMEN

The environments that harbor hematopoietic stem and progenitor cells are critical to explore for a better understanding of hematopoiesis during health and disease. These compartments often are inaccessible for controlled and rapid experimentation, thus limiting studies to the evaluation of conventional cell culture and transgenic animal models. Here we describe the manufacture and image-guided monitoring of an engineered microenvironment with user-defined properties that recruits hematopoietic progenitors into the implant. Using intravital imaging and fluorescence molecular tomography, we show in real time that the cell homing and retention process is efficient and durable for short- and long-term engraftment studies. Our results indicate that bone marrow stromal cells, precoated on the implant, accelerate the formation of new sinusoidal blood vessels with vascular integrity at the microcapillary level that enhances the recruitment hematopoietic progenitor cells to the site. This implantable construct can serve as a tool enabling the study of hematopoiesis.


Asunto(s)
Células Madre Hematopoyéticas/citología , Neoplasias/patología , Nicho de Células Madre , Andamios del Tejido , Microambiente Tumoral , Animales , Matriz Extracelular , Humanos , Hidrogeles , Ratones , Ratones Endogámicos NOD , Ratones Desnudos , Microscopía Confocal , Tomografía/métodos
17.
Biomed Microdevices ; 16(4): 609-15, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24781882

RESUMEN

Multicellular spheroids are an important 3-dimensional cell culture model that reflects many key aspects of in vivo microenvironments. This paper presents a scalable, self-assembly based approach for fabricating microcavity substrates for multicellular spheroid cell culture. Hydrophobic glass microbeads were self-assembled into a tightly packed monolayer through the combined actions of surface tension, gravity, and lateral capillary forces at the water-air interface of a polymer solution. The packed bead monolayer was subsequently embedded in the dried polymer layer. The surface was used as a template for replicating microcavity substrates with perfect spherical shapes. We demonstrated the use of the substrate in monitoring the formation process of tumor spheroids, a proof-of-concept scale-up fabrication procedure into standard microplate formats, and its application in testing cancer drug responses in the context of bone marrow stromal cells. The presented technique offers a simple and effective way of forming high-density uniformly-sized spheroids without microfabrication equipment for biological and drug screening applications.


Asunto(s)
Técnicas de Cultivo de Célula , Microesferas , Esferoides Celulares/efectos de los fármacos , Neoplasias de la Mama , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Microtecnología , Modelos Moleculares , Polímeros/química , Esferoides Celulares/metabolismo , Propiedades de Superficie
18.
Biotechnol J ; 19(1): e2300097, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37718481

RESUMEN

Mammalian cell culture is quickly becoming the go to engineering vehicle to mass produce viral vectors in a manner that is safe, convenient, reproducible, and cost and scale effective. Human embryonic kidney (HEK293) cells, in particular, have been utilized and customized (via differentiated transgene expression, modified culture parameters, addition of cytostatic culture agents) to increase vector yields. However, less attention has been made to understanding innate processes within the cells (such as, immune response, cell cycle, metabolism) themselves to better control or increase viral vector product yield. Accordingly, herein, the variation in viral production was studied from HEK cells over time using a one-way perfusion system and bioreactor to study the impact of external factors on secretion dynamics without retrotransduction. Specifically, the impact of cell density on viral titer, transduction efficiency, and LDH, was studied. Next, we look at the impact of using an inflammatory reporter cell line on viral output, and the secretion dynamics from HEK cells when we use sodium butyrate (cell cycle arrest agent). Lastly, we assess how downregulation of the PDK pathway increases viral titer. Altogether, we investigated the impact of various interventions to increase transient protein expression and viral output from HEK cells in a controlled and measurable environment to ultimately increase the efficiency of HEK cells for downstream clinical applications.


Asunto(s)
Vectores Genéticos , Lentivirus , Animales , Humanos , Lentivirus/genética , Células HEK293 , Vectores Genéticos/genética , Técnicas de Cultivo de Célula , Perfusión , Mamíferos
19.
Langmuir ; 29(34): 10611-6, 2013 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-23927769

RESUMEN

Poly(dimethylsiloxane) (PDMS) has emerged as an extremely useful polymer for various biological applications. The conjugation of PDMS with bioactive molecules to create functional surfaces is feasible yet limited to a single-molecule display with imprecise localization of the molecules on PDMS. Here we report a robust technique that can transfer and print the membrane surface of glutaraldehyde-fixed stromal cells intact onto a PDMS substrate using an intermediate polyvinylalcohol (PVA) film as a transporter system. The cell-PVA film capturing the entirety of surface molecules can be peeled off and subsequently printed onto PDMS while maintaining the spatial display of the original cell surface molecules. Proof-of-concept studies are described using human bone marrow stromal cell membranes including a demonstration of the bioactivity of transferred membranes to capture and adhere hematopoietic cells. The presented process is applicable to virtually any adherent cell and can broaden the functional display of biomolecules on PDMS for biotechnology applications.


Asunto(s)
Polímeros/química , Células del Estroma/química , Biotecnología , Membrana Celular/química , Dimetilpolisiloxanos/química , Humanos , Células Madre Mesenquimatosas/química , Nylons/química
20.
Mol Ther ; 20(1): 178-86, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21952165

RESUMEN

Mesenchymal stem cells (MSCs) are emerging as a promising immunotherapeutic, based largely on their overt suppression of T lymphocytes under inflammatory and autoimmune conditions. While paracrine cross-talk between MSCs and T cells has been well-studied, an intrinsic transcriptional switch that programs MSCs for immunomodulation has remained undefined. Here we show that bone marrow-derived MSCs require the transcriptional regulator Aire to suppress T cell-mediated pathogenesis in a mouse model of chronic colitis. Surprisingly, Aire did not control MSC suppression of T cell proliferation in vitro. Instead, Aire reduced T cell mitochondrial reductase by negatively regulating a proinflammatory cytokine, early T cell activation factor (Eta)-1. Neutralization of Eta-1 enabled Aire(-/-) MSCs to ameliorate colitis, reducing the number of infiltrating effector T cells in the colon, and normalizing T cell reductase levels. We propose that Aire represents an early molecular switch imposing a suppressive MSC phenotype via regulation of Eta-1. Monitoring Aire expression in MSCs may thus be a critical parameter for clinical use.


Asunto(s)
Enfermedad de Crohn/metabolismo , Células Madre Mesenquimatosas/metabolismo , Factores de Transcripción/metabolismo , Animales , Técnicas de Cocultivo , Enfermedad de Crohn/genética , Enfermedad de Crohn/inmunología , Femenino , Humanos , Terapia de Inmunosupresión , Inflamación/inmunología , Inflamación/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/inmunología , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Osteopontina/genética , Osteopontina/metabolismo , Oxidación-Reducción , Linfocitos T/inmunología , Factores de Transcripción/genética , Transcripción Genética , Proteína AIRE
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA