Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Small ; : e2311702, 2024 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-38456371

RESUMEN

The PD1/PD-L1 immune checkpoint blocking is a promising therapy, while immunosuppressive tumor microenvironment (TME) and poor tumor penetration of therapeutic antibodies limit its efficacy. Repolarization of tumor-associated macrophages (TAMs) offers a potential method to ameliorate immunosuppression of TME and further boost T cell antitumor immunity. Herein, hybrid cell membrane biomimetic nanovesicles (hNVs) are developed by fusing M1 macrophage-derived nanovesicles (M1-NVs) and PD1-overexpressed tumor cell-derived nanovesicles (PD1-NVs) to improve cancer immunotherapy. The M1-NVs promote the transformation of M2-like TAMs to M1-like phenotype and further increase the release of pro-inflammatory cytokines, resulting in improved immunosuppressive TME. Concurrently, the PD1-NVs block PD1/PD-L1 pathway, which boosts cancer immunotherapy when combined with M1-NVs. In a breast cancer mouse model, the hNVs efficiently accumulate at the tumor site after intravenous injection and significantly inhibit the tumor growth. Mechanically, the M1 macrophages and CD8+ T lymphocytes in TME increase by twofold after the treatment, indicating effective immune activation. These results suggest the hNVs as a promising strategy to integrate TME improvement with PD1/PD-L1 blockade for cancer immunotherapy.

2.
Adv Healthc Mater ; 13(13): e2400068, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38320299

RESUMEN

Cancer nanovaccines have attracted widespread attention by inducing potent cytotoxic T cell responses to improve immune checkpoint blockade (ICB) therapy, while the lack of co-stimulatory molecules limits their clinical applications. Here, a genetically engineered cancer cytomembrane nanovaccine is reported that simultaneously overexpresses co-stimulatory molecule CD40L and immune checkpoint inhibitor PD1 to elicit robust antitumor immunity for cancer immunotherapy. The CD40L and tumor antigens inherited from cancer cytomembranes effectively stimulate dendritic cell (DC)-mediated immune activation of cytotoxic T cells, while the PD1 on cancer cytomembranes significantly blocks PD1/PD-L1 signaling pathway, synergistically stimulating antitumor immune responses. Benefiting from the targeting ability of cancer cytomembranes, this nanovaccines formula shows an enhanced lymph node trafficking and retention. Compared with original cancer cytomembranes, this genetically engineered nanovaccine induces twofold DC maturation and shows satisfactory precaution efficacy in a breast tumor mouse model. This genetically engineered cytomembrane nanovaccine offers a simple, safe, and robust strategy by incorporating cytomembrane components and co-stimulatory molecules for enhanced cancer immunotherapy.


Asunto(s)
Vacunas contra el Cáncer , Células Dendríticas , Inmunoterapia , Animales , Inmunoterapia/métodos , Ratones , Vacunas contra el Cáncer/inmunología , Células Dendríticas/inmunología , Femenino , Humanos , Receptor de Muerte Celular Programada 1/inmunología , Receptor de Muerte Celular Programada 1/metabolismo , Línea Celular Tumoral , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/genética , Ingeniería Genética/métodos , Nanopartículas/química , Ratones Endogámicos BALB C , Linfocitos T Citotóxicos/inmunología , Antígeno B7-H1/metabolismo , Antígeno B7-H1/inmunología , Neoplasias/terapia , Neoplasias/inmunología , Nanovacunas
4.
Adv Mater ; 35(12): e2207875, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36721058

RESUMEN

The high stability of antibodies and their ability to precisely bind to antigens and endogenous immune receptors, as well as their susceptibility to protein engineering, enable antibody-based therapeutics to be widely applied in cancer, inflammation, infection, and other disorders. Nevertheless, the application of traditional antibody-based therapeutics has certain limitations, such as high price, limited permeability, and protein engineering complexity. Recent breakthroughs in cell membrane nanotechnology have deepened the understanding of the critical role of membrane protein receptors in disease treatment, enabling vesicular-antibody-based therapeutics. Here, the concept of vesicular antibodies that are obtained by modifying target antibodies onto cell membranes for biomedical applications is proposed. Given that an antibody is basically a protein, as an extension of this concept, vesicles or membrane-coated nanoparticles that use surface antibodies and protein receptors on cell membranes for biomedical applications as vesicular antibodies are defined. Furthermore, several engineering strategies for vesicular antibodies are summarized and how vesicular antibodies can be used in a variety of situations is highlighted. In addition, current challenges and future prospects of vesicular antibodies are also discussed. It is anticipated this perspective will provide new insights on the development of next-generation antibodies for enhanced therapeutics.


Asunto(s)
Anticuerpos , Ingeniería de Proteínas , Anticuerpos/uso terapéutico , Antígenos , Membrana Celular , Nanotecnología
5.
J Nanobiotechnology ; 20(1): 546, 2022 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-36585678

RESUMEN

Noninvasive prenatal diagnosis (NIPD) aims to detect fetal-related genetic disorders before birth by detecting markers in the peripheral blood of pregnant women, holding the potential in reducing the risk of fetal birth defects. Fetal-nucleated red blood cells (fNRBCs) can be used as biomarkers for NIPD, given their remarkable nature of carrying the entire genetic information of the fetus. Here, we review recent advances in NIPD technologies based on the isolation and analysis of fNRBCs. Conventional cell separation methods rely primarily on physical properties and surface antigens of fNRBCs, such as density gradient centrifugation, fluorescence-activated cell sorting, and magnetic-activated cell sorting. Due to the limitations of sensitivity and purity in Conventional methods, separation techniques based on micro-/nanomaterials have been developed as novel methods for isolating and enriching fNRBCs. We also discuss emerging methods based on microfluidic chips and nanostructured substrates for static and dynamic isolation of fNRBCs. Additionally, we introduce the identification techniques of fNRBCs and address the potential clinical diagnostic values of fNRBCs. Finally, we highlight the challenges and the future directions of fNRBCs as treatment guidelines in NIPD.


Asunto(s)
Pruebas Prenatales no Invasivas , Embarazo , Femenino , Humanos , Feto/metabolismo , Eritroblastos/química , Separación Celular/métodos , Citometría de Flujo
6.
Anal Chem ; 94(29): 10479-10486, 2022 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-35834188

RESUMEN

As the key player of a new restriction modification system, DNA phosphorothioate (PT) modification, which swaps oxygen for sulfur on the DNA backbone, protects the bacterial host from foreign DNA invasion. The identification of PT sites helps us understand its physiological defense mechanisms, but accurately quantifying this dynamic modification remains a challenge. Herein, we report a simple quantitative analysis method for optical mapping of PT sites in the single bacterial genome. DNA molecules are fully stretched and immobilized in a microfluidic chip by capillary flow and electrostatic interactions, improving the labeling efficiency by maximizing exposure of PT sites on DNA while avoiding DNA loss and damage. After screening 116 candidates, we identified a bifunctional chemical compound, iodoacetyl-polyethylene glycol-biotin, that can noninvasively and selectively biotinylate PT sites, enabling further labeling with streptavidin fluorescent nanoprobes. With this method, PT sites in PT+ DNA can be easily detected by fluorescence, while almost no detectable ones were found in PT- DNA, achieving real-time visualization of PT sites on a single DNA molecule. Collectively, this facile genome-wide PT site detection method directly characterizes the distribution and frequency of DNA modification, facilitating a better understanding of its modification mechanism that can be potentially extended to label DNAs in different species.


Asunto(s)
Genoma Bacteriano , Microfluídica , ADN , ADN Bacteriano/genética , Azufre
7.
Biosens Bioelectron ; 213: 114425, 2022 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-35688024

RESUMEN

The isolation and analysis of scarce circulating tumor cells (CTCs) with immunomagnetic nanoparticles (IMNs) have shown promising outcomes in noninvasive cancer diagnosis. However, the IMNs adsorb nonspecific proteins after entering into biofluids and the formed protein coronas cover surface targeting ligands, limiting the detection efficiency of IMNs. In addition, the interaction between surface targeting ligands and white blood cells (WBCs) significantly limits the purity of CTCs isolated by IMNs. Furthermore, the interfacial collision of nanoparticles and cells has negative effects on the viability of isolated CTCs. All of these limitations synthetically restrict the isolation and analysis of rare CTCs for early diagnosis and precision medicine. Here, we proposed that surface functionalization of IMNs with neutrophil membranes can simultaneously reduce nonspecific protein adsorption, enhance the interaction with CTCs, reduce the distraction from WBCs, and improve the viability of isolated CTCs. In spiked blood samples, our neutrophil membrane-coated IMNs (Neu-IMNs) exhibited a superior separation efficiency from 41.36% to 96.82% and an improved purity from 40.25% to 90.68% when compared to bare IMNs. Additionally, we successfully isolated CTCs in 19 out of total 20 blood samples from breast cancer patients using Neu-IMNs and further confirmed the feasibility of the isolated CTCs for downstream cell sequencing. Our work provides a new perspective on engineered IMNs for efficient isolation and analysis of CTCs, paving the way for early noninvasive diagnosis of cancer.


Asunto(s)
Técnicas Biosensibles , Nanopartículas , Células Neoplásicas Circulantes , Línea Celular Tumoral , Separación Celular , Humanos , Separación Inmunomagnética , Ligandos , Células Neoplásicas Circulantes/patología , Neutrófilos/patología
8.
ACS Appl Bio Mater ; 5(6): 2768-2776, 2022 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-35537085

RESUMEN

Circulating tumor cells (CTCs) are rare, meaning that current isolation strategies can hardly satisfy efficiency and cell biocompatibility requirements, which hinders clinical applications. In addition, the selected cells require immunofluorescence identification, which is a time-consuming and expensive process. Here, we developed a method to simultaneously separate and identify CTCs by the integration of optical force and fluorescent microspheres. Our method achieved high-purity separation of CTCs without damage through light manipulation and avoided additional immunofluorescence staining procedures, thus achieving rapid identification of sorted cells. White blood cells (WBCs) and CTCs are similar in size and density, which creates difficulties in distinguishing them optically. Therefore, fluorescent PS microspheres with high refractive index (RI) are designed here to capture the CTCs (PS-CTCs) and increase the average index of refraction of PS-CTCs. In optofluidic chips, PS-CTCs were propelled to the collection channel from the sample mixture, under the radiation of light force. Cells from the collection outlet were easily identified under a fluorescence microscope due to the fluorescence signals of PS microspheres. This method provides an approach for the sorting and identification of CTCs, which holds great potential for clinical applications in early diagnosis of disease.


Asunto(s)
Células Neoplásicas Circulantes , Recuento de Células , Línea Celular Tumoral , Separación Celular/métodos , Humanos , Microesferas , Células Neoplásicas Circulantes/patología
9.
Nanoscale ; 14(9): 3504-3512, 2022 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-35171188

RESUMEN

Isolation of circulating tumor cells (CTCs) from patients is a challenge due to the rarity of CTCs. Recently, various platforms to capture and release CTCs for downstream analysis have been developed. However, most of the reported release methods provide external stimuli to release all captured cells, which lead to lack of specificity in the pool of collected cells, and the external stimuli may affect the activity of the released cells. Here, we presented a simple method for single-cell recovery to overcome the shortcomings, which combined the nanostructures with a photocurable hydrogel, chondroitin sulfate methacryloyl (CSMA). In brief, we synthesized gelatin nanoparticles (Gnps) and modified them on flat glass (Gnp substrate) for the specific capture of CTCs. A 405 nm laser was projected onto the selected cells, and then CSMA was cured to encapsulate the selected CTCs. Unselected cells were removed with MMP-9 enzyme solution, and selected CTCs were recovered using a microcapillary. Finally, the photocurable hydrogel-encapsulated cells were analyzed by nucleic acid detection. In addition, the results suggested that the isolation platform showed good biocompatibility and successfully achieved the isolation of selected cells. In summary, our light-induced hydrogel responsive platform holds certain potential for clinical applications.


Asunto(s)
Nanoestructuras , Células Neoplásicas Circulantes , Recuento de Células , Línea Celular Tumoral , Separación Celular/métodos , Gelatina , Humanos , Hidrogeles , Nanoestructuras/química , Células Neoplásicas Circulantes/patología
10.
ACS Appl Mater Interfaces ; 13(34): 40656-40663, 2021 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-34406735

RESUMEN

Nontoxic tin-based perovskite solar cells (Sn-PSCs) as a promising alternative to toxic Pb-PSCs have drawn great attention in recent years for their environmental friendliness and unique optoelectronic properties. However, both the efficiency and long-term stability of Sn-PSCs are considerably inferior to those of Pb-based ones. One of the main reasons is the difficulty in obtaining high-quality Sn-perovskite films due to the rapid crystallization of Sn-perovskites, which also results in poor device reproducibility. Here, we report a novel cation exchange strategy to prepare high-quality formamidinium tin triiodide (FASnI3) perovskite films with a better controlled crystallization process and improved reproducibility, which allows easy access to smooth and pinhole-free perovskite films with oriented crystal growth, enlarged grain size, and reduced trap-state density. The corresponding Sn-PSCs show excellent photovoltaic performance with a champion efficiency of 9.11%, comparable to the best results reported for FASnI3-PSCs, and the devices also demonstrate outstanding long-term stability without encapsulation. Our results offer a practical strategy for fabricating Sn-PSCs with superb performance and stability.

11.
J Mater Chem B ; 9(13): 3047-3054, 2021 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-33885666

RESUMEN

Prenatal diagnostics holds great significance for pregnant women desiring healthy babies. Fetal nucleated red blood cells (fNRBCs), bearing the complete genome of the fetus, have been regarded as an important biomarker for noninvasive prenatal diagnostics (NIPD). The high-performance detection and enrichment of fNRBCs from maternal blood, especially during early pregnancy, is urgently needed for NIPD, which, unfortunately, remains a big challenge for early-pregnancy fNRBC isolation. In this study, we developed an innovative platform based on silica microbeads for fNRBC isolation and release in early pregnancy. Microbeads were coated with self-assembled MnO2 nanoparticles (SiO2@MnO2) and then modified with a specific antibody. Benefiting from the three-dimensional nanostructure of the MnO2 nanoparticles, the isolation efficiency of the fNRBCs was enhanced. Subsequently, fNRBCs were released via dissolving the MnO2-nanoparticle coating using oxalic acid. We successfully isolated fNRBCs from the maternal peripheral blood samples of 20 pregnant women in the early pregnancy period, ranging from 41 to 62 gestational days. More importantly, the fetal origin of isolated cells was confirmed via fluorescent in situ hybridization and short tandem repeat analysis. This platform based on SiO2@MnO2 microbeads has verified the existence of fNRBCs in early-pregnancy maternal blood and is a promising approach for NIPD in early pregnancy.


Asunto(s)
Eritrocitos/citología , Sangre Fetal/citología , Microesferas , Nanoestructuras/química , Diagnóstico Prenatal , Femenino , Humanos , Compuestos de Manganeso/química , Óxidos/química , Embarazo , Dióxido de Silicio/química
12.
Anal Chem ; 93(2): 1033-1042, 2021 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-33296189

RESUMEN

Fetal nucleated red blood cells (fNRBCs) in maternal peripheral blood containing the whole genetic information of the fetus may serve for noninvasive pregnant diagnostics (NIPD). However, the fetal cell-based NIPD is seriously limited by the poor purity of the isolated fNRBCs. Recently, the biomimetic cell membrane-camouflaged nanoparticles containing outstanding features have been widely used to detect and isolate rare cells from the peripheral blood samples. In this work, enythrocyte (RBC) and leukocyte (WBC) membranes are fused and coated onto magnet nanoparticles and then modified with anti-CD147 to isolate fNRBCs from the maternal peripheral blood with significant efficiency (∼90%) and purity (∼87%) in simulated spiked blood samples. Further, fNRBCs were isolated and identified from a series of maternal peripheral blood samples coming from pregnant women of 11-13 gestational weeks, and different chromosomal aneuploidies were diagnosed using fNRBCs isolated from maternal blood in early pregnancy. Our strategy may offer additional opportunity to overcome the limitations of current cell-based NIPD platforms.


Asunto(s)
Aneuploidia , Membrana Celular/química , Eritrocitos/citología , Feto/citología , Leucocitos/citología , Nanopartículas de Magnetita/química , Cromosomas/genética , Femenino , Humanos , Hibridación Fluorescente in Situ , Tamaño de la Partícula , Embarazo , Propiedades de Superficie
13.
ACS Appl Bio Mater ; 4(2): 1140-1155, 2021 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-35014471

RESUMEN

Blood tests have been a powerful tool for the clinical analysis of many diseases. With the advances in microfluidic technology, two more specific indicators from the circulation system, namely, emerging "liquid biopsy" of circulating tumor cells (CTCs) and fetal nucleated red blood cells (fNRBCs), can be screened and analyzed as a simple blood test for the noninvasive diagnosis of cancers as well as fetal disorders. The unique feature of precisely manipulating a trace of fluid endows microfluidic devices with the ability to isolate CTCs or fNRBCs from numerous blood cells with high performance, which undoubtedly facilitates biomedical applications of these two kinds of rare cells. In this review, advanced developments in microfluidic technologies focusing on the detection and sorting of rare CTCs and fNRBCs from peripheral blood are summarized. The development of microfluidic devices incorporated with various multifunctional microstructures and nanomaterials for enhancing the sensitivity, purity, and viability of CTC or fNRBC detection enables CTC molecular analysis and fNRBC-based noninvasive prenatal diagnosis (NIPD). These microfluidics-based approaches provide great potential opportunities in noninvasive cancer diagnosis or NIPD applications.


Asunto(s)
Separación Celular/métodos , Eritroblastos/citología , Técnicas Analíticas Microfluídicas/métodos , Células Neoplásicas Circulantes/patología , Separación Celular/instrumentación , Recuento de Eritrocitos/instrumentación , Recuento de Eritrocitos/métodos , Humanos , Hidrodinámica , Dispositivos Laboratorio en un Chip , Técnicas Analíticas Microfluídicas/instrumentación , Nanopartículas/química
14.
Biomater Sci ; 9(3): 826-834, 2021 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-33215618

RESUMEN

Increasing number of resistant bacteria have emerged with the overuse of antibiotics, which indicates that the bacterial infection has become a global challenge. Furthermore, the pollution of antibiotics to the environment has become a serious threat to public health. It is known that toxins produced by bacteria are the main cause of bacterial infections. Photothermal therapy is an effective antibacterial approach. However, the photothermal reagents cannot eliminate bacterial toxins, and even some anti-bacterial materials are toxic. Here, we synthesized a biomimetic recycled nanoparticle, red blood cell (RBC) membrane-coated Fe3O4 nanoparticles (RBC@Fe3O4), as an antibacterial agent. The RBC@Fe3O4 nanoparticles act as nano-sponges to trap toxins and then kill them all with a photothermal effect. We can describe this process simply as a battle between two armies. Our strategy is to disarm the "enemy" so that we can easily kill the "enemy" who has no power, which results in enhancing the bactericidal efficacy. The toxin of methicillin-resistant Staphylococcus aureus (MRSA) was absorbed by RBC@Fe3O4in vitro. In addition, in vivo studies proved that the RBC@Fe3O4 nanoparticles confer obvious survival benefits against toxin-induced lethality by absorbing the toxin of MRSA. Furthermore, using a mouse model of MRSA wound infection, the RBC@Fe3O4 nanoparticles with laser irradiation were found to have a superior wound-healing effect. Simultaneously, the RBC@Fe3O4 nanoparticles could be recycled in a simple way without affecting the bactericidal efficacy. The highly biocompatible and recyclable RBC@Fe3O4 biomimetic nanoparticles based on photothermal therapy and bacterial toxin adsorption strategy are promising for treating bacterial infections.


Asunto(s)
Infecciones Bacterianas , Staphylococcus aureus Resistente a Meticilina , Nanopartículas , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Infecciones Bacterianas/tratamiento farmacológico , Biomimética , Humanos
15.
ACS Appl Mater Interfaces ; 12(46): 51563-51572, 2020 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-33146992

RESUMEN

Li metal is considered a highly desirable anode for next-generation high-energy-density rechargeable lithium batteries. However, irregular Li dendrite formation and infinite relative volume changes prevent the commercial adoption of Li-metal anodes. Here, electrophoretic deposition of black phosphorus (BP) on commercial Cu foam (BP@Cu foam) is reported to regulate Li nucleation for the first time. First-principles calculations reveal that the unique two-dimensional (2D) structure of BP is beneficial to Li intercalation and propagation. Compared with the random Li nucleation and growth on bare Cu foam, Li ions are preferably confined into the BP layers, which induces uniform Li nucleation at the early stage of the Li deposition and guides the following lateral Li growth on BP@Cu foam. In addition, the three-dimensional (3D) porous and conductive framework of Cu foams further mitigate the volume change and dissipate the current density. Attributing to these merits, the BP@Cu foam exhibits significantly enhanced Coulombic efficiency and cycling stability compared with bare Cu foam. In the full-cell configuration paired with a Li4Ti5O12 or LiFePO4 cathode, the BP@Cu foam also boosts the battery performances. This work provides new insights into the development of BP and other elaborate 2D materials for achieving dendrite-free Li-metal anodes.

16.
Biomed Microdevices ; 22(4): 75, 2020 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-33079273

RESUMEN

Being easy, safe and reliable, non-invasive prenatal diagnosis (NIPD) has been greatly pursued in recent years. Holding the complete genetic information of the fetus, fetal nucleated red blood cells (fNRBCs) are viewed as a suitable target for NIPD application. However, effective separating fNRBCs from maternal peripheral blood for clinic use still faces great challenges, given that fNRBCs are extremely rare in maternal blood circulation. Here, by combining the high-throughput inertial microfluidic chip with multifunctional microspheres as size amplification, we develop a novel method to isolate fNRBCs with high performance. To enlarge the size difference between fNRBCs and normal blood cells, we use the gelatin coated microspheres to capture fNRBCs with anti-CD147 as specific recognizer at first. The size difference between fNRBCs captured by the microspheres and normal blood cells makes it easy to purify the captured fNRBCs through the spiral microfluidic chip. Finally, the purified fNRBCs are mildly released from the microspheres by enzymatically degrading the gelatin coating. Cell capture efficiency about 81%, high purity of 83%, as well as cell release viability over 80% were achieved using spiked samples by this approach. Additionally, fNRBCs were successfully detected from peripheral blood of pregnant women with an average of 24 fNRBCs per mL, suggesting the great potential of this method for clinical non-invasive prenatal diagnosis.


Asunto(s)
Separación Celular/instrumentación , Eritroblastos/citología , Feto/citología , Dispositivos Laboratorio en un Chip , Microesferas , Femenino , Humanos , Embarazo , Diagnóstico Prenatal
17.
Nanotechnology ; 31(49): 495102, 2020 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-32990263

RESUMEN

Constructing biological affinity devices is considered as an effective strategy for isolating circulating tumor cells (CTCs), and electrospun nanofibers (ESNFs) have recently received attention. However, the current research focuses on polymer fibers, and fabricating stimuli-responsive inorganic nanofibers for cancer diagnosis and analysis is still challenging. In this work, Zn-Mn oxide nanofibers (ZnMnNFs) are used to capture and purify cancer cells after modification with specific antibodies. Then, the hierarchical nanofibers are degraded by reductive weak acid to release the captured cells efficiently without residues. Fusion of Zn and Mn, two transition metals, enhances the surface activity of oxides so that ZnMnNFs are easier to be degraded and modified. By using MCF-7 cancer cells, the cell capture efficiency of ZnMnNFs is up to 88.2%. Furthermore, by using citric acid, it is discovered that, by comparison with Mn oxide nanofibers, the cell release efficiency of ZnMnNFs is improved to 95.1% from 15.4%. In addition, the viability of released cells exceeds 90%. Lastly, the robustness of ZnMnNFs substrates is tested in peripheral blood from breast cancer patients (BCP) and colorectal cancer patients (CCP). Combined with fluorescence labeling, CTCs are confirmed to be isolated from all the clinical samples. This is the first trial of using ternary inorganic ESNFs for cancer cell capture. It is anticipated that the degradable ESNFs will provide biocompatible theranostic platforms and overcome the current limitations of cell release for high-precision gene analysis.


Asunto(s)
Separación Celular/métodos , Manganeso/química , Nanofibras/química , Células Neoplásicas Circulantes/patología , Óxidos/química , Zinc/química , Neoplasias de la Mama/sangre , Neoplasias de la Mama/patología , Supervivencia Celular , Femenino , Humanos , Células MCF-7
18.
Nanoscale ; 12(3): 1455-1463, 2020 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-31808771

RESUMEN

Circulating tumor cells (CTCs) are one type of significant biomarker in cancer patients' blood that have been attracting attention from researchers for decades, and their efficient and viable isolation is of vital importance in cancer prevention and treatment. However, the development of efficient and low-cost bio-microchips still faces significant challenges. In this paper, we construct a novel three-dimensional micro-nano bio-microchip that has dual functions of specifically capturing and non-destructively releasing cancer cells. ZnO nanowire arrays were vertically grown on the surface of a polydimethylsiloxane (PDMS) pillar substrate with a gear structure (ZnO-coated G-PDMS pillar microchips). The gear structure provides more binding sites for antibodies and target cancer cells, while ZnO nanowires provide a rough surface for CTC attachment and size-specific effects for retaining CTCs. For subsequent culture and bioanalysis, the captured CTCs can be non-destructively released with high efficiency and good viability using a mild acidic solution treatment. Furthermore, the manufacturing process of the G-PDMS pillar microchips is convenient and low-cost, and the preparation approach of the ZnO nanowire is mature and simple to operate. In particular, the bio-microchips showed high capture efficiency (91.11% ± 5.53%) and excellent cell viability (96%) using a spiked cell sample. Moreover, we successfully achieved the specific fluorescent labeling of CTCs in 9 clinical breast cancer patients' samples. The ZnO-coated G-PDMS pillar microchips not only have great potential for new target drug development for cancer stem cells but also open up new opportunities for individualized treatment.


Asunto(s)
Separación Celular , Dispositivos Laboratorio en un Chip , Nanocables/química , Células Neoplásicas Circulantes , Óxido de Zinc/química , Humanos , Células MCF-7
19.
Angew Chem Int Ed Engl ; 59(10): 4075-4081, 2020 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-31829491

RESUMEN

The role of endogenous serotonin (5-HT) in gastrointestinal motility is still highly controversial. Although electrochemical techniques allow for direct and real-time recording of biomolecules, the dynamic monitoring of 5-HT release from elastic and tubular intestine during motor reflexes remains a great challenge because of the specific peristalsis patterns and inevitable passivation of the sensing interface. A stretchable sensor with antifouling and decontamination properties was assembled from gold nanotubes, titanium dioxide nanoparticles, and carbon nanotubes. The sandwich-like structure endowed the sensor with satisfying mechanical stability and electrochemical performance, high resistance against physical adsorption, and superior efficiency in the photodegradation of biofouling molecules. Insertion of the sensor into the lumen of rat ileum (the last section of the small intestine) successfully mimics intestinal peristalsis, and simultaneous real-time monitoring of distension-evoked 5-HT release was possible for the first time. Our results unambiguously reveal that mechanical distension of the intestine induces endogenous 5-HT overflow, and 5-HT level is closely associated with the physiological or pathological states of the intestine.


Asunto(s)
Técnicas Electroquímicas , Intestinos/química , Serotonina/metabolismo , Animales , Ratas , Serotonina/química , Estrés Mecánico
20.
ACS Appl Mater Interfaces ; 11(44): 41118-41126, 2019 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-31612699

RESUMEN

The recovery of rare single circulating tumor cells (CTCs) from patients has great potential to facilitate the study of cell heterogeneity and cancer metastasis, which may promote the development of individualized cancer immunotherapy. Herein, a versatile single-cell recovery approach that utilizes an acoustic droplet-induced enzyme responsive platform for the capture and on-demand release of single CTCs is proposed. The platform combines a multifunctional enzyme-responsive gelatin nanoparticle (GNP)-decorated substrate (GNP-chip) for specific capture with an acoustic droplet positioning technique to realize on-demand release of single CTCs. The acoustic droplet dispenser is employed to generate oxidized alginate microdroplets containing the MMP-9 enzyme (OA-MMP-9) with controllable size and precise positioning upon the cell-attached GNP-chip, allowing controlled cell-surface biodegradation under enzymatic reactions followed by calcium chloride (CaCl2) solution treatment to form single-cell encapsulated calcium alginate hydrogels. Benefitting from the existence of hydrogels, the released cells could be efficiently recovered by microcapillary. Results demonstrate that the encapsulated cells maintain good cell morphology in the hydrogels, which allow further single-cell nucleic acid analysis. As a proof-of-concept platform, this approach enables reliable and efficient retrieval of single CTCs and holds the potential for versatility in single-cell analysis systems.


Asunto(s)
Separación Celular/métodos , Hidrogeles/química , Metaloproteinasa 9 de la Matriz/metabolismo , Células Neoplásicas Circulantes/química , Alginatos/química , Cloruro de Calcio/química , Gelatina/química , Humanos , Células MCF-7 , Metaloproteinasa 9 de la Matriz/química , Microscopía Fluorescente , Nanopartículas/química , Análisis de la Célula Individual
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...