Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cureus ; 14(3): e23195, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35444866

RESUMEN

The Ad26.COV2.S vaccine, developed by Janssen (Beerse, Belgium), the pharmaceutical wing of Johnson & Johnson (JNJ), is one of the three vaccines approved for use against coronavirus disease 2019 (COVID-19) infection in the United States. We present a case of a 66-year-old female who presented to the emergency department with a one-day history of nausea, vomiting, room-spinning vertigo, and complete right facial weakness immediately after getting vaccinated with Ad26.COV2.S. Initial workup focused on uncovering a possible association between the vaccine and Bell's palsy. However, her prior history of stroke, presence of predisposing risk factors, and additional symptoms of nausea, vomiting, and vertigo prompted further neurological testing, which revealed an isolated right pontine lacunar infarct involving the right facial colliculus, mimicking Bell's palsy. Isolated dorsal pontine lesion presenting as hemifacial palsy is very rare and can be easily missed by clinicians. Relevant history and thorough neurological examination can help guide appropriate diagnostic testing and prevent potential biases. It is crucial for clinicians to know the distinguishing features between true Bell's palsy and acute brainstem infarction masquerading as Bell's.

2.
J Immunol ; 208(4): 929-940, 2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-35091434

RESUMEN

CD8+ T cell responses are the foundation of the recent clinical success of immunotherapy in oncologic indications. Although checkpoint inhibitors have enhanced the activity of existing CD8+ T cell responses, therapeutic approaches to generate Ag-specific CD8+ T cell responses have had limited success. Here, we demonstrate that cytosolic delivery of Ag through microfluidic squeezing enables MHC class I presentation to CD8+ T cells by diverse cell types. In murine dendritic cells (DCs), squeezed DCs were ∼1000-fold more potent at eliciting CD8+ T cell responses than DCs cross-presenting the same amount of protein Ag. The approach also enabled engineering of less conventional APCs, such as T cells, for effective priming of CD8+ T cells in vitro and in vivo. Mixtures of immune cells, such as murine splenocytes, also elicited CD8+ T cell responses in vivo when squeezed with Ag. We demonstrate that squeezing enables effective MHC class I presentation by human DCs, T cells, B cells, and PBMCs and that, in clinical scale formats, the system can squeeze up to 2 billion cells per minute. Using the human papillomavirus 16 (HPV16) murine model, TC-1, we demonstrate that squeezed B cells, T cells, and unfractionated splenocytes elicit antitumor immunity and correlate with an influx of HPV-specific CD8+ T cells such that >80% of CD8s in the tumor were HPV specific. Together, these findings demonstrate the potential of cytosolic Ag delivery to drive robust CD8+ T cell responses and illustrate the potential for an autologous cell-based vaccine with minimal turnaround time for patients.


Asunto(s)
Presentación de Antígeno , Células Presentadoras de Antígenos/inmunología , Linfocitos T CD8-positivos/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Microfluídica , Neoplasias/inmunología , Traslado Adoptivo , Animales , Células Presentadoras de Antígenos/metabolismo , Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/metabolismo , Técnicas de Cultivo de Célula , Femenino , Humanos , Inmunización , Inmunofenotipificación , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Ratones , Ratones Noqueados , Microfluídica/métodos , Modelos Biológicos , Neoplasias/metabolismo , Neoplasias/patología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
3.
Sci Rep ; 5: 10276, 2015 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-25999171

RESUMEN

B-cells are promising candidate autologous antigen-presenting cells (APCs) to prime antigen-specific T-cells both in vitro and in vivo. However to date, a significant barrier to utilizing B-cells as APCs is their low capacity for non-specific antigen uptake compared to "professional" APCs such as dendritic cells. Here we utilize a microfluidic device that employs many parallel channels to pass single cells through narrow constrictions in high throughput. This microscale "cell squeezing" process creates transient pores in the plasma membrane, enabling intracellular delivery of whole proteins from the surrounding medium into B-cells via mechano-poration. We demonstrate that both resting and activated B-cells process and present antigens delivered via mechano-poration exclusively to antigen-specific CD8(+)T-cells, and not CD4(+)T-cells. Squeezed B-cells primed and expanded large numbers of effector CD8(+)T-cells in vitro that produced effector cytokines critical to cytolytic function, including granzyme B and interferon-γ. Finally, antigen-loaded B-cells were also able to prime antigen-specific CD8(+)T-cells in vivo when adoptively transferred into mice. Altogether, these data demonstrate crucial proof-of-concept for mechano-poration as an enabling technology for B-cell antigen loading, priming of antigen-specific CD8(+)T-cells, and decoupling of antigen uptake from B-cell activation.


Asunto(s)
Antígenos/inmunología , Linfocitos B/inmunología , Vacunas/inmunología , Animales , Antígenos/metabolismo , Linfocitos B/citología , Linfocitos B/metabolismo , Antígeno B7-2/metabolismo , Antígenos CD40/metabolismo , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Técnicas de Cultivo de Célula , Proliferación Celular , Células Cultivadas , Citocinas/metabolismo , Interferón gamma/metabolismo , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Técnicas Analíticas Microfluídicas/instrumentación , Técnicas Analíticas Microfluídicas/métodos , Estrés Mecánico
4.
PLoS One ; 10(4): e0118803, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25875117

RESUMEN

Intracellular delivery of biomolecules, such as proteins and siRNAs, into primary immune cells, especially resting lymphocytes, is a challenge. Here we describe the design and testing of microfluidic intracellular delivery systems that cause temporary membrane disruption by rapid mechanical deformation of human and mouse immune cells. Dextran, antibody and siRNA delivery performance is measured in multiple immune cell types and the approach's potential to engineer cell function is demonstrated in HIV infection studies.


Asunto(s)
Anticuerpos/administración & dosificación , Dextranos/administración & dosificación , Sistemas de Liberación de Medicamentos/instrumentación , Dispositivos Laboratorio en un Chip , ARN Interferente Pequeño/administración & dosificación , Animales , Linfocitos B/metabolismo , Células Cultivadas , Células Dendríticas/metabolismo , VIH/genética , Infecciones por VIH/terapia , Infecciones por VIH/virología , Humanos , Ratones , Ratones Endogámicos C57BL , ARN Interferente Pequeño/genética , Tratamiento con ARN de Interferencia , Linfocitos T/metabolismo
5.
Integr Biol (Camb) ; 6(4): 470-5, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24496115

RESUMEN

Intracellular delivery of materials is a challenge in research and therapeutic applications. Physical methods of plasma membrane disruption have recently emerged as an approach to facilitate the delivery of a variety of macromolecules to a range of cell types. We use the microfluidic CellSqueeze delivery platform to examine the kinetics of plasma membrane recovery after disruption and its dependence on the calcium content of the surrounding buffer (recovery time ∼ 5 min without calcium vs. ∼ 30 s with calcium). Moreover, we illustrate that manipulation of the membrane repair kinetics can yield up to 5× improvement in delivery efficiency without significantly impacting cell viability. Membrane repair characteristics initially observed in HeLa cells are shown to translate to primary naïve murine T cells. Subsequent manipulation of membrane repair kinetics also enables the delivery of larger materials, such as antibodies, to these difficult to manipulate cells. This work provides insight into the membrane repair process in response to mechanical delivery and could potentially enable the development of improved delivery methods.


Asunto(s)
Calcio/metabolismo , Membrana Celular/metabolismo , Sustancias Macromoleculares/metabolismo , Animales , Células HeLa , Humanos , Cinética , Masculino , Ratones , Ratones Endogámicos C57BL , Microfluídica
6.
J Vis Exp ; (81): e50980, 2013 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-24300077

RESUMEN

Rapid mechanical deformation of cells has emerged as a promising, vector-free method for intracellular delivery of macromolecules and nanomaterials. This technology has shown potential in addressing previously challenging applications; including, delivery to primary immune cells, cell reprogramming, carbon nanotube, and quantum dot delivery. This vector-free microfluidic platform relies on mechanical disruption of the cell membrane to facilitate cytosolic delivery of the target material. Herein, we describe the detailed method of use for these microfluidic devices including, device assembly, cell preparation, and system operation. This delivery approach requires a brief optimization of device type and operating conditions for previously unreported applications. The provided instructions are generalizable to most cell types and delivery materials as this system does not require specialized buffers or chemical modification/conjugation steps. This work also provides recommendations on how to improve device performance and trouble-shoot potential issues related to clogging, low delivery efficiencies, and cell viability.


Asunto(s)
Técnicas Citológicas/instrumentación , Técnicas Citológicas/métodos , Técnicas Analíticas Microfluídicas/instrumentación , Técnicas Analíticas Microfluídicas/métodos , Forma de la Célula , Sistemas de Liberación de Medicamentos , Células HeLa , Humanos , Sustancias Macromoleculares/administración & dosificación
7.
Proc Natl Acad Sci U S A ; 110(6): 2082-7, 2013 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-23341631

RESUMEN

Intracellular delivery of macromolecules is a challenge in research and therapeutic applications. Existing vector-based and physical methods have limitations, including their reliance on exogenous materials or electrical fields, which can lead to toxicity or off-target effects. We describe a microfluidic approach to delivery in which cells are mechanically deformed as they pass through a constriction 30-80% smaller than the cell diameter. The resulting controlled application of compression and shear forces results in the formation of transient holes that enable the diffusion of material from the surrounding buffer into the cytosol. The method has demonstrated the ability to deliver a range of material, such as carbon nanotubes, proteins, and siRNA, to 11 cell types, including embryonic stem cells and immune cells. When used for the delivery of transcription factors, the microfluidic devices produced a 10-fold improvement in colony formation relative to electroporation and cell-penetrating peptides. Indeed, its ability to deliver structurally diverse materials and its applicability to difficult-to-transfect primary cells indicate that this method could potentially enable many research and clinical applications.


Asunto(s)
Sistemas de Liberación de Medicamentos , Técnicas Analíticas Microfluídicas , Animales , Fenómenos Biomecánicos , Permeabilidad de la Membrana Celular , Forma de la Célula , Células Cultivadas , Citosol/metabolismo , Células Dendríticas/citología , Células Dendríticas/metabolismo , Difusión , Expresión Génica , Células HeLa , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Nanotubos de Carbono , Proteínas/administración & dosificación , ARN Interferente Pequeño/administración & dosificación
8.
Anal Chem ; 84(15): 6438-43, 2012 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-22746217

RESUMEN

Mechanical properties of cells have been shown to have a significant role in disease, as in many instances cell stiffness changes when a cell is no longer healthy. We present a high-throughput microfluidics-based approach that exploits the connection between travel time of a cell through a narrow passage and cell stiffness. The system resolves both cell travel time and relative cell diameter while retaining information on the cell level. We show that stiffer cells have longer transit times than less stiff ones and that cell size significantly influences travel times. Experiments with untreated HeLa cells and cells made compliant with latrunculin A and cytochalasin B further demonstrate that travel time is influenced by cell stiffness, with the compliant cells having faster transit time.


Asunto(s)
Microfluídica , Compuestos Bicíclicos Heterocíclicos con Puentes/toxicidad , Tamaño de la Célula/efectos de los fármacos , Citocalasina B/toxicidad , Electrodos , Células HeLa , Humanos , Microfluídica/instrumentación , Tiazolidinas/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...