Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
ACS Appl Mater Interfaces ; 15(35): 41299-41309, 2023 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-37616579

RESUMO

Intracellular delivery technologies that are cost-effective, non-cytotoxic, efficient, and cargo-agnostic are needed to enable the manufacturing of cell-based therapies as well as gene manipulation for research applications. Current technologies capable of delivering large cargoes, such as plasmids and CRISPR-Cas9 ribonucleoproteins (RNPs), are plagued with high costs and/or cytotoxicity and often require substantial specialized equipment and reagents, which may not be available in resource-limited settings. Here, we report an intracellular delivery technology that can be assembled from materials available in most research laboratories, thus democratizing access to intracellular delivery for researchers and clinicians in low-resource areas of the world. These filtroporation devices permeabilize cells by pulling them through the pores of a cell culture insert by the application of vacuum available in biosafety cabinets. In a format that costs less than $10 in materials per experiment, we demonstrate the delivery of fluorescently labeled dextran, expression plasmids, and RNPs for gene knockout to Jurkat cells and human CD34+ hematopoietic stem and progenitor cell populations with delivery efficiencies of up to 40% for RNP knockout and viabilities of >80%. We show that functionalizing the surfaces of the filters with fluorinated silane moieties further enhances the delivery efficiency. These devices are capable of processing 500,000 to 4 million cells per experiment, and when combined with a 3D-printed vacuum application chamber, this throughput can be straightforwardly increased 6-12-fold in parallel experiments.


Assuntos
Silanos , Células-Tronco , Humanos , Técnicas de Inativação de Genes , Técnicas de Cultura de Células , Terapia Baseada em Transplante de Células e Tecidos
2.
ACS Appl Nano Mater ; 6(8): 6454-6460, 2023 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-37152920

RESUMO

We developed an unconventional seed-mediated in situ synthetic method, whereby gold nanostars are formed directly on the internal walls of microfluidic reactors. The dense plasmonic substrate coatings were grown in microfluidic channels with different geometries to elucidate the impacts of flow rate and profile on reagent consumption, product morphology, and density. Nanostar growth was found to occur in the flow-limited regime and our results highlight the possibility of creating shape gradients or incorporating multiple morphologies in the same microreactor, which is challenging to achieve with traditional self-assembly. The plasmonic-microfluidic platforms developed herein have implications for a broad range of applications, including cell culture/sorting, catalysis, sensing, and drug/gene delivery.

4.
Adv Mater ; 35(7): e2206933, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36468617

RESUMO

Developing scalable electrical stimulating platforms for cell and tissue engineering applications is limited by external power source dependency, wetting resistance, microscale size requirements, and suitable flexibility. Here, a versatile and scalable platform is developed to enable tunable electrical stimulation for biological applications by harnessing the giant magnetoelastic effect in soft systems, converting gentle air pressure (100-400 kPa) to yield a current of up to 10.5 mA and a voltage of 9.5 mV. The platform can be easily manufactured and scaled up for integration in multiwell magnetoelastic plates via 3D printing. The authors demonstrate that the electrical stimulation generated by this platform enhances the conversion of fibroblasts into neurons up to 2-fold (104%) and subsequent neuronal maturation up to 3-fold (251%). This easily configurable electrical stimulation device has broad applications in high throughput organ-on-a-chip systems, and paves the way for future development of neural engineering, including cellular therapy via implantable self-powered electrical stimulation devices.


Assuntos
Fontes de Energia Elétrica , Neurônios , Próteses e Implantes , Fibroblastos , Engenharia Tecidual
5.
Front Bioeng Biotechnol ; 10: 973326, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36225598

RESUMO

Clustered Regularly Interspaced Short Palindromic Repeats associated protein 9 (CRISPR/Cas9) has transformed our ability to edit the human genome selectively. This technology has quickly become the most standardized and reproducible gene editing tool available. Catalyzing rapid advances in biomedical research and genetic engineering, the CRISPR/Cas9 system offers great potential to provide diagnostic and therapeutic options for the prevention and treatment of currently incurable single-gene and more complex human diseases. However, significant barriers to the clinical application of CRISPR/Cas9 remain. While in vitro, ex vivo, and in vivo gene editing has been demonstrated extensively in a laboratory setting, the translation to clinical studies is currently limited by shortfalls in the precision, scalability, and efficiency of delivering CRISPR/Cas9-associated reagents to their intended therapeutic targets. To overcome these challenges, recent advancements manipulate both the delivery cargo and vehicles used to transport CRISPR/Cas9 reagents. With the choice of cargo informing the delivery vehicle, both must be optimized for precision and efficiency. This review aims to summarize current bioengineering approaches to applying CRISPR/Cas9 gene editing tools towards the development of emerging cellular therapeutics, focusing on its two main engineerable components: the delivery vehicle and the gene editing cargo it carries. The contemporary barriers to biomedical applications are discussed within the context of key considerations to be made in the optimization of CRISPR/Cas9 for widespread clinical translation.

6.
Adv Mater ; 34(37): e2205330, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35903851

RESUMO

Precise arrangements of plasmonic nanoparticles on substrates are important for designing optoelectronics, sensors and metamaterials with rational electronic, optical and magnetic properties. Bottom-up synthesis offers unmatched control over morphology and optical response of individual plasmonic building blocks. Usually, the incorporation of nanoparticles made by bottom-up wet chemistry starts from batch synthesis of colloids, which requires time-consuming and hard-to-scale steps like ligand exchange and self-assembly. Herein, an unconventional bottom-up wet-chemical synthetic approach for producing gold nanoparticle ordered arrays is developed. Water-processable hydroxypropyl cellulose stencils facilitate the patterning of a reductant chemical ink on which nanoparticle growth selectively occurs. Arrays exhibiting lattice plasmon resonances in the visible region and near infrared (quality factors of >20) are produced following a rapid synthetic step (<10 min), all without cleanroom fabrication, specialized equipment, or self-assembly, constituting a major step forward in establishing in situ growth approaches. Further, the technical capabilities of this method through modulation of the particle size, shape, and array spacings directly on the substrate are demonstrated. Ultimately, establishing a fundamental understanding of in situ growth has the potential to inform the fabrication of plasmonic materials; opening the door for in situ growth fabrication of waveguides, lasing platforms, and plasmonic sensors.

7.
Plasma Process Polym ; 19(9): 2200012, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35574246

RESUMO

The coronavirus disease 2019 (COVID-19) pandemic has greatly stressed the global community, exposing vulnerabilities in the supply chains for disinfection materials, personal protective equipment, and medical resources worldwide. Disinfection methods based on cold atmospheric plasma (CAP) technologies offer an intriguing solution to many of these challenges because they are easily deployable and do not require resource-constrained consumables or reagents needed for conventional decontamination practices. CAP technologies have shown great promise for a wide range of medical applications from wound healing and cancer treatment to sterilization methods to mitigate airborne and fomite transfer of viruses. This review engages the broader community of scientists and engineers that wish to help the medical community with the ongoing COVID-19 pandemic by establishing methods to utilize broadly applicable CAP technologies.

8.
Adv Sci (Weinh) ; 9(14): e2105853, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35486030

RESUMO

Well-preserved molecular cargo in circulating extracellular vesicles (EVs) offers an ideal material for detecting oncogenic gene alterations in cancer patients, providing a noninvasive diagnostic solution for detection of disease status and monitoring treatment response. Therefore, technologies that conveniently isolate EVs with sufficient efficiency are desperately needed. Here, a lipid labeling and click chemistry-based EV capture platform ("Click Beads"), which is ideal for EV message ribonucleic acid (mRNA) assays due to its efficient, convenient, and rapid purification of EVs, enabling downstream molecular quantification using reverse transcription digital polymerase chain reaction (RT-dPCR) is described and demonstrated. Ewing sarcoma protein (EWS) gene rearrangements and kirsten rat sarcoma viral oncogene homolog (KRAS) gene mutation status are detected and quantified using EVs isolated by Click Beads and matched with those identified in biopsy specimens from Ewing sarcoma or pancreatic cancer patients. Moreover, the quantification of gene alterations can be used for monitoring treatment responses and disease progression.


Assuntos
Vesículas Extracelulares , Sarcoma de Ewing , Carcinogênese/genética , Química Click , Vesículas Extracelulares/genética , Vesículas Extracelulares/metabolismo , Genes ras , Humanos , Lipídeos , Proteína EWS de Ligação a RNA/genética , Proteína EWS de Ligação a RNA/metabolismo , Sarcoma de Ewing/genética , Sarcoma de Ewing/metabolismo
9.
Pediatr Dermatol ; 39(3): 443-446, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35322460

RESUMO

Severe mosquito bite allergy (SMBA) is characterized by necrotic skin lesions and systemic symptoms. Chronic active Epstein-Barr virus (EBV) infection, when superimposed with SMBA, is a key driver for catastrophic clinical consequences, such as uncontrolled lymphoproliferation. This interplay is of clinical significance due to its association with hemophagocytic lymphohistiocytosis (HLH) and/or EBV-driven malignancies. Here, we report a case of SMBA that developed in a 14-year-old Hispanic boy that led to fatal secondary HLH.


Assuntos
Infecções por Vírus Epstein-Barr , Hipersensibilidade , Mordeduras e Picadas de Insetos , Linfo-Histiocitose Hemofagocítica , Transtornos Linfoproliferativos , Adolescente , Infecções por Vírus Epstein-Barr/complicações , Infecções por Vírus Epstein-Barr/diagnóstico , Herpesvirus Humano 4 , Humanos , Mordeduras e Picadas de Insetos/complicações , Linfo-Histiocitose Hemofagocítica/complicações , Linfo-Histiocitose Hemofagocítica/diagnóstico , Transtornos Linfoproliferativos/diagnóstico , Masculino
10.
ACS Mater Lett ; 3(3): 282-289, 2021 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-34337418

RESUMO

Micro- and nanoscale patterned monolayers of plasmonic nanoparticles were fabricated by combining concepts from colloidal chemistry, self-assembly, and subtractive soft lithography. Leveraging chemical interactions between the capping ligands of pre-synthesized gold colloids and a polydimethylsiloxane stamp, we demonstrated patterning gold nanoparticles over centimeter-scale areas with a variety of micro- and nanoscale geometries, including islands, lines, and chiral structures (e.g., square spirals). By successfully achieving nanoscale manipulation over a wide range of substrates and patterns, we establish a powerful and straightforward strategy, nanoparticle chemical lift-off lithography (NP-CLL), for the economical and scalable fabrication of functional plasmonic materials with colloidal nanoparticles as building blocks, offering a transformative solution for designing next-generation plasmonic technologies.

11.
Small ; 17(28): e2100546, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34105245

RESUMO

The clustered regularly interspaced short palindromic repeats (CRISPR)-associated protein 9 (CRISPR/Cas9) is an efficient and precise gene-editing technology that offers a versatile solution for establishing treatments directed at genetic diseases. Currently, CRISPR/Cas9 delivery into cells relies primarily on viral vectors, which suffer from limitations in packaging capacity and safety concerns. These issues with a nonviral delivery strategy are addressed, where Cas9•sgRNA ribonucleoprotein (RNP) complexes can be encapsulated into supramolecular nanoparticles (SMNP) to form RNP⊂SMNPs, which can then be delivered into targeted cells via supramolecular nanosubstrate-mediated delivery. Utilizing the U87 glioblastoma cell line as a model system, a variety of parameters for cellular-uptake of the RNP-laden nanoparticles are examined. Dose- and time-dependent CRISPR/Cas9-mediated gene disruption is further examined in a green fluorescent protein (GFP)-expressing U87 cell line (GFP-U87). The utility of an optimized SMNP formulation in co-delivering Cas9 protein and two sgRNAs that target deletion of exons 45-55 (708 kb) of the dystrophin gene is demonstrated. Mutations in this region lead to Duchenne muscular dystrophy, a severe genetic muscle wasting disease. Efficient delivery of these gene deletion cargoes is observed in a human cardiomyocyte cell line (AC16), induced pluripotent stem cells, and mesenchymal stem cells.


Assuntos
Sistemas CRISPR-Cas , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Proteína 9 Associada à CRISPR , Edição de Genes , Vetores Genéticos , Humanos
12.
ACS Cent Sci ; 6(11): 2105-2116, 2020 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-33274287

RESUMO

Gold nanostars (AuNSTs) are biocompatible, have large surface areas, and are characterized by high near-infrared extinction, making them ideal for integration with technologies targeting biological applications. We have developed a robust and simple microfluidic method for the direct growth of anisotropic AuNSTs on oxide substrates including indium tin oxide and glass. The synthesis was optimized to yield AuNSTs with high anisotropy, branching, uniformity, and density in batch and microfluidic systems for optimal light-to-heat conversion upon laser irradiation. Surface-enhanced Raman scattering spectra and mesoscale temperature measurements were combined with spatially correlated scanning electron microscopy to monitor nanostar and ligand stability and microbubble formation at different laser fluences. The capability of the platform for generating controlled localized heating was used to explore hyperthermia-assisted detachment of adherent glioblastoma cells (U87-GFP) grafted to the capillary walls. Both flow and laser fluence can be tuned to induce different biological responses, such as ablation, cell deformation, release of intracellular components, and the removal of intact cells. Ultimately, this platform has potential applications in biological and chemical sensing, hyperthermia-mediated drug delivery, and microfluidic soft-release of grafted cells with single-cell specificity.

13.
Sci Adv ; 6(48)2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33246950

RESUMO

Skin-mountable microelectronics are garnering substantial interest for various promising applications including human-machine interfaces, biointegrated devices, and personalized medicine. However, it remains a critical challenge to develop e-skins to mimic the human somatosensory system in full working range. Here, we present a multifunctional e-skin system with a heterostructured configuration that couples vinyl-hybrid-silica nanoparticle (VSNP)-modified polyacrylamide (PAM) hydrogel with two-dimensional (2D) MXene through nano-bridging layers of polypyrrole nanowires (PpyNWs) at the interfaces, featuring high toughness and low hysteresis, in tandem with controlled crack generation and distribution. The multidimensional configurations endow the e-skin with an extraordinary working range (2800%), ultrafast responsiveness (90 ms) and resilience (240 ms), good linearity (800%), tunable sensing mechanisms, and excellent reproducibility. In parallel, this e-skin platform is capable of detecting, quantifying, and remotely monitoring stretching motions in multiple dimensions, tactile pressure, proximity sensing, and variations in temperature and light, establishing a promising platform for next-generation smart flexible electronics.

14.
Sci Adv ; 6(43)2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33097539

RESUMO

Leveraging the endogenous homology-directed repair (HDR) pathway, the CRISPR-Cas9 gene-editing system can be applied to knock in a therapeutic gene at a designated site in the genome, offering a general therapeutic solution for treating genetic diseases such as hemoglobinopathies. Here, a combined supramolecular nanoparticle (SMNP)/supramolecular nanosubstrate-mediated delivery (SNSMD) strategy is used to facilitate CRISPR-Cas9 knockin of the hemoglobin beta (HBB) gene into the adeno-associated virus integration site 1 (AAVS1) safe-harbor site of an engineered K562 3.21 cell line harboring the sickle cell disease mutation. Through stepwise treatments of the two SMNP vectors encapsulating a Cas9•single-guide RNA (sgRNA) complex and an HBB/green fluorescent protein (GFP)-encoding plasmid, CRISPR-Cas9 knockin was successfully achieved via HDR. Last, the HBB/GFP-knockin K562 3.21 cells were introduced into mice via intraperitoneal injection to show their in vivo proliferative potential. This proof-of-concept demonstration paves the way for general gene therapeutic solutions for treating hemoglobinopathies.


Assuntos
Sistemas CRISPR-Cas , Hemoglobinopatias , Animais , Edição de Genes , Vetores Genéticos/genética , Hemoglobinopatias/genética , Hemoglobinopatias/terapia , Hemoglobinas/genética , Camundongos
15.
ACS Appl Mater Interfaces ; 12(41): 45744-45752, 2020 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-32940030

RESUMO

Innovative technologies for intracellular delivery are ushering in a new era for gene editing, enabling the utilization of a patient's own cells for stem cell and immunotherapies. In particular, cell-squeezing platforms provide unconventional forms of intracellular delivery, deforming cells through microfluidic constrictions to generate transient pores and to enable effective diffusion of biomolecular cargo. While these devices are promising gene-editing platforms, they require frequent maintenance due to the accumulation of cellular debris, limiting their potential for reaching the throughputs necessary for scalable cellular therapies. As these cell-squeezing technologies are improved, there is a need to develop next-generation platforms with higher throughput and longer lifespan, importantly, avoiding the buildup of cell debris and thus channel clogging. Here, we report a versatile strategy to coat the channels of microfluidic devices with lipid bilayers based on noncovalent lipid bicelle technology, which led to substantial improvements in reducing cell adhesion and protein adsorption. The antifouling properties of the lipid bilayer coating were evaluated, including membrane uniformity, passivation against nonspecific protein adsorption, and inhibition of cell attachment against multiple cell types. This surface functionalization approach was applied to coat constricted microfluidic channels for the intracellular delivery of fluorescently labeled dextran and plasmid DNA, demonstrating significant reductions in the accumulation of cell debris. Taken together, our work demonstrates that lipid bicelles are a useful tool to fabricate antifouling lipid bilayer coatings in cell-squeezing devices, resulting in reduced nonspecific fouling and cell clogging to improve performance.


Assuntos
Incrustação Biológica/prevenção & controle , Dispositivos Lab-On-A-Chip , Bicamadas Lipídicas/química , Adesão Celular , Células Cultivadas , Humanos , Células Jurkat , Estrutura Molecular , Tamanho da Partícula , Propriedades de Superfície
16.
Proc Natl Acad Sci U S A ; 117(20): 10976-10982, 2020 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-32358194

RESUMO

Advances in gene editing are leading to new medical interventions where patients' own cells are used for stem cell therapies and immunotherapies. One of the key limitations to translating these treatments to the clinic is the need for scalable technologies for engineering cells efficiently and safely. Toward this goal, microfluidic strategies to induce membrane pores and permeability have emerged as promising techniques to deliver biomolecular cargo into cells. As these technologies continue to mature, there is a need to achieve efficient, safe, nontoxic, fast, and economical processing of clinically relevant cell types. We demonstrate an acoustofluidic sonoporation method to deliver plasmids to immortalized and primary human cell types, based on pore formation and permeabilization of cell membranes with acoustic waves. This acoustofluidic-mediated approach achieves fast and efficient intracellular delivery of an enhanced green fluorescent protein-expressing plasmid to cells at a scalable throughput of 200,000 cells/min in a single channel. Analyses of intracellular delivery and nuclear membrane rupture revealed mechanisms underlying acoustofluidic delivery and successful gene expression. Our studies show that acoustofluidic technologies are promising platforms for gene delivery and a useful tool for investigating membrane repair.


Assuntos
Técnicas de Transferência de Genes , Terapia Genética/métodos , Transplante de Células-Tronco Hematopoéticas/métodos , Sistema Hematopoético , Células-Tronco , Sobrevivência Celular , Citoplasma , Expressão Gênica , Técnicas de Transferência de Genes/instrumentação , Terapia Genética/instrumentação , Proteínas de Fluorescência Verde/genética , Humanos , Células Jurkat , Plasmídeos , Som
17.
ACS Appl Mater Interfaces ; 12(11): 13447-13455, 2020 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-32092250

RESUMO

Supported lipid membranes are versatile biomimetic coatings for the chemical functionalization of inorganic surfaces. Developing simple and effective fabrication strategies to form supported lipid membranes with micropatterned geometries is a long-standing challenge. Herein, we demonstrate how the combination of chemical lift-off lithography (CLL) and easily prepared lipid bicelle nanostructures can yield micropatterned, supported lipid membranes on gold surfaces with high pattern resolution, conformal character, and biofunctionality. Using CLL, we functionalized gold surfaces with patterned arrays of hydrophilic and hydrophobic self-assembled monolayers (SAMs). Time-lapse fluorescence microscopy imaging revealed that lipid bicelles adsorbed preferentially onto the hydrophilic SAM regions, while there was negligible lipid adsorption onto the hydrophobic SAM regions. Functional receptors could be embedded within the lipid bicelles, which facilitated selective detection of receptor-ligand binding interactions in a model streptavidin-biotin system. Quartz crystal microbalance-dissipation measurements further identified that lipid bicelles adsorb irreversibly and remain intact on top of the hydrophilic SAM regions. Taken together, our findings indicate that lipid bicelles are useful lipid nanostructures for reproducibly assembling micropatterned, supported lipid membranes with precise pattern fidelity.

18.
ACS Mater Lett ; 2(11): 1475-1483, 2020 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-34708213

RESUMO

Local heating using pulsed laser-induced photothermal effects on plasmonic nanostructured substrates can be used for intracellular delivery applications. However, the fabrication of plasmonic nanostructured interfaces is hampered by complex nanomanufacturing schemes. Here, we demonstrate the fabrication of large-area plasmonic gold (Au) nanodisk arrays that enable photothermal intracellular delivery of biomolecular cargo at high efficiency. The Au nanodisks (350 nm in diameter) were fabricated using chemical lift-off lithography (CLL). Nanosecond laser pulses were used to excite the plasmonic nanostructures, thereby generating transient pores at the outer membranes of targeted cells that enable the delivery of biomolecules via diffusion. Delivery efficiencies of >98% were achieved using the cell impermeable dye calcein (0.6 kDa) as a model payload, while maintaining cell viabilities at >98%. The highly efficient intracellular delivery approach demonstrated in this work will facilitate translational studies targeting molecular screening and drug testing that bridge laboratory and clinical investigations.

19.
Adv Funct Mater ; 30(49)2020 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-34220409

RESUMO

Tumor-derived extracellular vesicles (EVs) play essential roles in intercellular communication during tumor growth and metastatic evolution. Currently, little is known about the possible roles of tumor-derived EVs in sarcoma because the lack of specific surface markers makes it technically challenging to purify sarcoma-derived EVs. In this study, a specific purification system is developed for Ewing sarcoma (ES)-derived EVs by coupling covalent chemistry-mediated EV capture/ release within a nanostructure-embedded microchip. The purification platform-ES-EV Click Chip-takes advantage of specific anti-LINGO-1 recognition and sensitive click chemistry-mediated EV capture, followed by disulfide cleavage-driven EV release. Since the device is capable of specific and efficient purification of intact ES EVs with high purity, ES-EV Click Chip is ideal for conducting downstream functional studies of ES EVs. Absolute quantification of the molecular hallmark of ES (i.e., EWS rearrangements) using reverse transcription Droplet Digital PCR enables specific quantification of ES EVs. The purified ES EVs can be internalized by recipient cells and transfer their mRNA cargoes, exhibiting their biological intactness and potential role as biological shuttles in intercellular communication.

20.
Proc Natl Acad Sci U S A ; 116(51): 25870-25879, 2019 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-31796586

RESUMO

Deficiencies in either lamin B1 or lamin B2 cause both defective migration of cortical neurons in the developing brain and reduced neuronal survival. The neuronal migration abnormality is explained by a weakened nuclear lamina that interferes with nucleokinesis, a nuclear translocation process required for neuronal migration. In contrast, the explanation for impaired neuronal survival is poorly understood. We hypothesized that the forces imparted on the nucleus during neuronal migration result in nuclear membrane (NM) ruptures, causing interspersion of nuclear and cytoplasmic contents-and ultimately cell death. To test this hypothesis, we bred Lmnb1-deficient mice that express a nuclear-localized fluorescent Cre reporter. Migrating neurons within the cortical plate of E18.5 Lmnb1-deficient embryos exhibited NM ruptures, evident by the escape of the nuclear-localized reporter into the cytoplasm and NM discontinuities by electron microscopy. The NM ruptures were accompanied by DNA damage and cell death. The NM ruptures were not observed in nonmigrating cells within the ventricular zone. NM ruptures, DNA damage, and cell death were also observed in cultured Lmnb1-/- and Lmnb2-/- neurons as they migrated away from neurospheres. To test whether mechanical forces on the cell nucleus are relevant to NM ruptures in migrating neurons, we examined cultured Lmnb1-/- neurons when exposed to external constrictive forces (migration into a field of tightly spaced silicon pillars). As the cells entered the field of pillars, there were frequent NM ruptures, accompanied by DNA damage and cell death.


Assuntos
Morte Celular/fisiologia , Movimento Celular/fisiologia , Lamina Tipo B/metabolismo , Neurônios/metabolismo , Membrana Nuclear/metabolismo , Lâmina Nuclear/metabolismo , Animais , Linhagem Celular , Sobrevivência Celular , Citoplasma/metabolismo , Dano ao DNA , Regulação da Expressão Gênica , Lamina Tipo B/genética , Camundongos , Camundongos Knockout , Neurônios/citologia , Lâmina Nuclear/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...