Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
bioRxiv ; 2024 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-38712200

RESUMO

The role of extracellular vesicles (EVs) in human health and disease has garnered considerable attention over the past two decades. However, while several types of EVs are known to interact dynamically with the extracellular matrix and there is great potential value in producing high-fidelity EV micropatterns, there are currently no label-free, high-resolution, and tunable platform technologies with this capability. We introduce Light-induced Extracellular Vesicle Adsorption (LEVA) as a powerful solution to rapidly advance the study of matrix- and surface-bound EVs and other particles. The versatility of LEVA is demonstrated using commercial GFP-EV standards, EVs from glioblastoma bioreactors, and E. coli outer membrane vesicles (OMVs), with the resulting patterns used for single EV characterization, single cell migration on migrasome-mimetic trails, and OMV-mediated neutrophil swarming. LEVA will enable rapid advancements in the study of matrix- and surface-bound EVs and other particles, and should encourage researchers from many disciplines to create novel diagnostic, biomimetic, immunoengineering, and therapeutic screening assays.

2.
bioRxiv ; 2024 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-38659953

RESUMO

Obesity is a global health crisis that contributes to morbidity and mortality worldwide. Obesity's comorbid association with a variety of diseases, from metabolic syndrome to neurodegenerative disease, underscores the critical need to better understand the pathobiology of obesity. Adipose tissue, once seen as an inert storage depot, is now recognized as an active endocrine organ, regulating metabolic and systemic homeostasis. Recent studies spotlight the theranostic utility of extracellular vesicles (EVs) as novel biomarkers and drivers of disease, including obesity-related complications. Adipose-derived EVs (ADEVs) have garnered increased interest for their roles in diverse diseases, however robust isolation and characterization protocols for human, cell-specific EV subsets are limited. Herein, we directly address this technical challenge by establishing a multiparametric analysis framework that leverages bulk and single EV characterization, mRNA phenotyping and proteomics of human ADEVs directly from paired visceral adipose tissue, cultured mature adipocyte conditioned media, and plasma from obese subjects undergoing bariatric surgery. Importantly, rigorous EV phenotyping at the tissue and cell-specific level identified top 'adipose liquid biopsy' candidates that were validated in circulating plasma EVs from the same patient. In summary, our study paves the way toward a tissue and cell-specific, multiparametric framework for studying tissue and circulating adipose EVs in obesity-driven disease.

3.
Adv Sci (Weinh) ; 11(11): e2306373, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38204202

RESUMO

Detecting pancreatic duct adenocarcinoma (PDAC) in its early stages and predicting late-stage patient prognosis undergoing chemotherapy is challenging. This work shows that the activation of specific oncogenes leads to elevated expression of mRNAs and their corresponding proteins in extracellular vesicles (EVs) circulating in blood. Utilizing an immune lipoplex nanoparticle (ILN) biochip assay, these findings demonstrate that glypican 1 (GPC1) mRNA expression in the exosomes-rich (Exo) EV subpopulation and GPC1 membrane protein (mProtein) expression in the microvesicles-rich (MV) EV subpopulation, particularly the tumor associated microvesicles (tMV), served as a viable biomarker for PDAC. A combined analysis effectively discriminated early-stage PDAC patients from benign pancreatic diseases and healthy donors in sizable clinical from multiple hospitals. Furthermore, among late-stage PDAC patients undergoing chemotherapy, lower GPC1 tMV-mProtein and Exo-mRNA expression before treatment correlated significantly with prolonged overall survival. These findings underscore the potential of vesicular GPC1 expression for early PDAC screenings and chemotherapy prognosis.


Assuntos
Carcinoma Ductal Pancreático , Vesículas Extracelulares , Neoplasias Pancreáticas , Humanos , Biomarcadores Tumorais/genética , Carcinoma Ductal Pancreático/diagnóstico , Carcinoma Ductal Pancreático/genética , Vesículas Extracelulares/metabolismo , Glipicanas/genética , Glipicanas/metabolismo , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/genética , Prognóstico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
4.
J Extracell Vesicles ; 12(11): e12369, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37908159

RESUMO

The molecular heterogeneity of extracellular vesicles (EVs) and the co-isolation of physically similar particles, such as lipoproteins (LPs), confounds and limits the sensitivity of EV bulk biomarker characterization. Herein, we present a single-EV and particle (siEVP) protein and RNA assay (siEVP PRA) to simultaneously detect mRNAs, miRNAs, and proteins in subpopulations of EVs and LPs. The siEVP PRA immobilizes and sorts particles via positive immunoselection onto micropatterns and focuses biomolecular signals in situ. By detecting EVPs at a single-particle resolution, the siEVP PRA outperformed the sensitivities of bulk-analysis benchmark assays for RNA and protein. To assess the specificity of RNA detection in complex biofluids, EVs from various glioma cell lines were processed with small RNA sequencing, whereby two mRNAs and two miRNAs associated with glioblastoma multiforme (GBM) were chosen for cross-validation. Despite the presence of single-EV-LP co-isolates in serum, the siEVP PRA detected GBM-associated vesicular RNA profiles in GBM patient siEVPs. The siEVP PRA effectively examines intravesicular, intervesicular, and interparticle heterogeneity with diagnostic promise.


Assuntos
Vesículas Extracelulares , Glioblastoma , MicroRNAs , Humanos , Vesículas Extracelulares/genética , Vesículas Extracelulares/metabolismo , Lipopolissacarídeos , MicroRNAs/genética , MicroRNAs/metabolismo , RNA Mensageiro , Lipoproteínas , Glioblastoma/diagnóstico , Glioblastoma/genética
5.
Adv Sci (Weinh) ; 10(33): e2302622, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37847907

RESUMO

Regenerative medicine in tissue engineering often relies on stem cells and specific growth factors at a supraphysiological dose. These approaches are costly and may cause severe side effects. Herein, therapeutic small extracellular vesicles (t-sEVs) endogenously loaded with a cocktail of human vascular endothelial growth factor A (VEGF-A) and human bone morphogenetic protein 2 (BMP-2) mRNAs within a customized injectable PEGylated poly (glycerol sebacate) acrylate (PEGS-A) hydrogel for bone regeneration in rats with challenging femur critical-size defects are introduced. Abundant t-sEVs are produced by a facile cellular nanoelectroporation system based on a commercially available track-etched membrane (TM-nanoEP) to deliver plasmid DNAs to human adipose-derived mesenchymal stem cells (hAdMSCs). Upregulated microRNAs associated with the therapeutic mRNAs are enriched in t-sEVs for enhanced angiogenic-osteogenic regeneration. Localized and controlled release of t-sEVs within the PEGS-A hydrogel leads to the retention of therapeutics in the defect site for highly efficient bone regeneration with minimal low accumulation in other organs.


Assuntos
Osteogênese , Fator A de Crescimento do Endotélio Vascular , Ratos , Humanos , Animais , RNA Mensageiro/genética , Regeneração Óssea/genética , Hidrogéis/farmacologia
6.
Nat Commun ; 14(1): 6692, 2023 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-37872156

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) tumours carry multiple gene mutations and respond poorly to treatments. There is currently an unmet need for drug carriers that can deliver multiple gene cargoes to target high solid tumour burden like PDAC. Here, we report a dual targeted extracellular vesicle (dtEV) carrying high loads of therapeutic RNA that effectively suppresses large PDAC tumours in mice. The EV surface contains a CD64 protein that has a tissue targeting peptide and a humanized monoclonal antibody. Cells sequentially transfected with plasmid DNAs encoding for the RNA and protein of interest by Transwell®-based asymmetric cell electroporation release abundant targeted EVs with high RNA loading. Together with a low dose chemotherapy drug, Gemcitabine, dtEVs suppress large orthotopic PANC-1 and patient derived xenograft tumours and metastasis in mice and extended animal survival. Our work presents a clinically accessible and scalable way to produce abundant EVs for delivering multiple gene cargoes to large solid tumours.


Assuntos
Carcinoma Ductal Pancreático , Vesículas Extracelulares , Neoplasias Pancreáticas , Humanos , Animais , Camundongos , Desoxicitidina/uso terapêutico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/terapia , Neoplasias Pancreáticas/metabolismo , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/terapia , Carcinoma Ductal Pancreático/metabolismo , RNA , Vesículas Extracelulares/metabolismo , Linhagem Celular Tumoral , Neoplasias Pancreáticas
7.
Biomater Sci ; 11(20): 6834-6847, 2023 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-37646133

RESUMO

Tumor-associated immune cells play a crucial role in cancer progression. Myeloid-derived suppressor cells (MDSCs), for example, are immature innate immune cells that infiltrate the tumor to exert immunosuppressive activity and protect cancer cells from the host's immune system and/or cancer-specific immunotherapies. While tumor-associated immune cells have emerged as a promising therapeutic target, efforts to counter immunosuppression within the tumor niche have been hampered by the lack of approaches that selectively target the immune cell compartment of the tumor, to effectively eliminate "tumor-protecting" immune cells and/or drive an "anti-tumor" phenotype. Here we report on a novel nanotechnology-based approach to target tumor-associated immune cells and promote "anti-tumor" responses in a murine model of breast cancer. Engineered extracellular vesicles (EVs) decorated with ICAM-1 ligands and loaded with miR-146a and Glut1, were biosynthesized (in vitro or in vivo) and administered to tumor-bearing mice once a week for up to 5 weeks. The impact of this treatment modality on the immune cell compartment and tumor progression was evaluated via RT-qPCR, flow cytometry, and histology. Our results indicate that weekly administration of the engineered EVs (i.e., ICAM-1-decorated and loaded with miR-146a and Glut1) hampered tumor progression compared to ICAM-1-decorated EVs with no cargo. Flow cytometry analyses of the tumors indicated a shift in the phenotype of the immune cell population toward a more pro-inflammatory state, which appeared to have facilitated the infiltration of tumor-targeting T cells, and was associated with a reduction in tumor size and decreased metastatic burden. Altogether, our results indicate that ICAM-1-decorated EVs could be a powerful platform nanotechnology for the deployment of immune cell-targeting therapies to solid tumors.

8.
Methods Mol Biol ; 2679: 67-81, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37300609

RESUMO

Selectively capturing and releasing viable circulating tumor cells (CTCs) from the peripheral blood of cancer patients is advantageous for investigating the molecular hallmarks of metastasis and developing personalized therapeutics. CTC-based liquid biopsies are flourishing in the clinical setting, offering opportunities to track the real-time responses of patients during clinical trials and lending accessibility to cancers that are traditionally difficult to diagnose. However, CTCs are rare compared to the breadth of cells that reside in the circulatory network, which has encouraged the engineering of novel microfluidic devices. Current microfluidic technologies either extensively enrich CTCs but compromise cellular viability or sort viable CTCs at low efficiencies. Herein we present a procedure to fabricate and operate a microfluidic device capable of capturing CTCs at high efficiencies while ensuring high viability. The microvortex-inducing microfluidic device functionalized with nanointerfaces positively enriches CTCs via cancer-specific immunoaffinity, while a thermally responsive surface chemistry releases the captured cells by raising the temperature to 37 °C.


Assuntos
Células Neoplásicas Circulantes , Humanos , Células Neoplásicas Circulantes/patologia , Separação Celular/métodos , Microfluídica , Linhagem Celular Tumoral
9.
bioRxiv ; 2023 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-36993759

RESUMO

Extracellular vesicles (EVs) have emerged as promising diagnostic and therapeutic candidates in many biomedical applications. However, EV research continues to rely heavily on in vitro cell cultures for EV production, where the exogenous EVs present in fetal bovine (FBS) or other required serum supplementation can be difficult to remove entirely. Despite this and other potential applications involving EV mixtures, there are currently no rapid, robust, inexpensive, and label-free methods for determining the relative concentrations of different EV subpopulations within a sample. In this study, we demonstrate that surface-enhanced Raman spectroscopy (SERS) can biochemically fingerprint fetal bovine serum-derived and bioreactor-produced EVs, and after applying a novel manifold learning technique to the acquired spectra, enables the quantitative detection of the relative amounts of different EV populations within an unknown sample. We first developed this method using known ratios of Rhodamine B to Rhodamine 6G, then using known ratios of FBS EVs to breast cancer EVs from a bioreactor culture. In addition to quantifying EV mixtures, the proposed deep learning architecture provides some knowledge discovery capabilities which we demonstrate by applying it to dynamic Raman spectra of a chemical milling process. This label-free characterization and analytical approach should translate well to other EV SERS applications, such as monitoring the integrity of semipermeable membranes within EV bioreactors, ensuring the quality or potency of diagnostic or therapeutic EVs, determining relative amounts of EVs produced in complex co-culture systems, as well as many Raman spectroscopy applications.

10.
J Extracell Vesicles ; 11(9): e12258, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-36093740

RESUMO

Conventional PD-L1 immunohistochemical tissue biopsies only predict 20%-40% of non-small cell lung cancer (NSCLC) patients that will respond positively to anti-PD-1/PD-L1 immunotherapy. Herein, we present an immunogold biochip to quantify single extracellular vesicular RNA and protein (Au SERP) as a non-invasive alternative. With only 20 µl of purified serum, PD-1/PD-L1 proteins on the surface of extracellular vesicles (EVs) and EV PD-1/PD-L1 messenger RNA (mRNA) cargo were detected at a single-vesicle resolution and exceeded the sensitivities of their bulk-analysis conventional counterparts, ELISA and qRT-PCR, by 1000 times. By testing a cohort of 27 non-responding and 27 responding NSCLC patients, Au SERP indicated that the single-EV mRNA biomarkers surpass the single-EV protein biomarkers in predicting patient responses to immunotherapy. Dual single-EV PD-1/PD-L1 mRNA detection differentiated responders from non-responders with an accuracy of 72.2% and achieved an NSCLC diagnosis accuracy of 93.2%, suggesting the potential for Au SERP to provide enhanced immunotherapy predictions and cancer diagnoses within the clinical setting.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Vesículas Extracelulares , Neoplasias Pulmonares , Antígeno B7-H1/genética , Biomarcadores , Carcinoma Pulmonar de Células não Pequenas/genética , Vesículas Extracelulares/metabolismo , Humanos , Fatores Imunológicos/uso terapêutico , Imunoterapia , Neoplasias Pulmonares/genética , RNA/uso terapêutico , RNA Mensageiro/metabolismo
11.
Lab Chip ; 22(13): 2502-2518, 2022 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-35579189

RESUMO

Investigating cellular and vesicular heterogeneity in breast cancer remains a challenge, which encourages the development of controllable in vitro systems that mimic the tumor microenvironment. Although three-dimensional cell culture better recapitulates the heterogeneity observed in tumor growth and extracellular vesicle (EV) biogenesis, the physiological relevance is often contrasted with the control offered by two-dimensional cell culture. Therefore, to challenge this misconception we developed a novel microfluidic system harboring highly tunable three-dimensional EV microbioreactors (EVµBRs) to model micrometastatic EV release in breast cancer while capitalizing on the convenient, low-volume, and sterile interface provided by microfluidics. The diameter and cellular occupancy of the EVµBRs could be precisely tailored to various configurations, supporting the formation of breast cancer tumor spheroids. To immobilize the EVµBRs within a microchannel and facilitate EV extraction, oxygen inhibition in free-radical polymerization was repurposed to rapidly generate two-layer hydrodynamic traps in situ using a digital-micromirror device (DMD)-based ultraviolet (UV) projection system. Breast cancer tumor spheroid-derived EVs were harvested with as little as 20 µL from the microfluidic system and quantified by single-EV immunofluorescence for CD63 and CD81. Despite the low-volume extraction, differences in biomarker expression and coexpression of the tetraspanins on single EVs were observed. Furthermore, the EVµBRs were capable of recapitulating heterogeneity at a cellular and vesicular degree, indicating the utility and robustness of the microfluidic system to investigate physiologically relevant EVs in breast cancer and other disease models.


Assuntos
Neoplasias da Mama , Vesículas Extracelulares , Microgéis , Neoplasias da Mama/patologia , Técnicas de Cultura de Células , Vesículas Extracelulares/metabolismo , Feminino , Humanos , Microfluídica , Microambiente Tumoral
12.
Exp Hematol ; 99: 21-31.e5, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34015390

RESUMO

The presence of iron in circulating monocytes is well known as they play an essential role in iron recycling. It has been demonstrated that the iron content of blood cells can be measured through their magnetic behavior; however, the magnetic properties of different monocyte subtypes remain unknown. In this study we report, for the first time, the magnetic behavior of classical, intermediate and non-classical monocytes, which may be related to their iron storage capacity. The magnetic properties of monocytes were compared with those of other blood cells, such as lymphocytes and red blood cells in the oxyhemoglobin and methemoglobin states, and a cancer cell type. For this analysis, we used an instrument referred to as a Cell Tracking Velocimetry (CTV), which quantitatively characterizes the magnetic behavior of biological entities. Our results revealed that significant fractions of the intermediate and non-classical monocytes (up to 59% and 65% depending on the sample, respectively) have paramagnetic properties, suggesting their higher iron storage capacities. Moreover, our findings have implications for the immunomagnetic separation industry; we propose that negative magnetic isolation techniques for recovering monocytes from blood should be used with caution, as it is possible to lose magnetic monocytes when using this technique.


Assuntos
Eritrócitos/citologia , Citometria de Fluxo , Campos Magnéticos , Monócitos/citologia , Eritrócitos/metabolismo , Humanos , Monócitos/metabolismo
13.
Sci Rep ; 11(1): 8034, 2021 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-33850163

RESUMO

Extracellular vesicles (EVs) derived from tumor cells have the potential to provide a much-needed source of non-invasive molecular biomarkers for liquid biopsies. However, current methods for EV isolation have limited specificity towards tumor-derived EVs that limit their clinical use. Here, we present an approach called immunomagnetic sequential ultrafiltration (iSUF) that consists of sequential stages of purification and enrichment of EVs in approximately 2 h. In iSUF, EVs present in different volumes of biofluids (0.5-100 mL) can be significantly enriched (up to 1000 times), with up to 99% removal of contaminating proteins (e.g., albumin). The EV recovery rate by iSUF for cell culture media (CCM), serum, and urine corresponded to 98.0% ± 3.6%, 96.0% ± 2.0% and 94.0% ± 1.9%, respectively (p > 0.05). The final step of iSUF enables the separation of tumor-specific EVs by incorporating immunomagnetic beads to target EV subpopulations. Serum from a cohort of clinical samples from metastatic breast cancer (BC) patients and healthy donors were processed by the iSUF platform and the isolated EVs from patients showed significantly higher expression levels of BC biomarkers (i.e., HER2, CD24, and miR21).


Assuntos
Vesículas Extracelulares , Ultrafiltração , Biomarcadores Tumorais , Humanos , Biópsia Líquida , MicroRNAs
14.
Biomicrofluidics ; 14(1): 014104, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31933714

RESUMO

The applications of cell patterning are widespread due to the high-throughput testing and different resolutions offered by these platforms. Cell patterning has aided in deconvoluting in vivo experiments to better characterize cellular mechanisms and increase therapeutic output. Here, we present a technique for engineering an artificial surface via surface chemistry to form large-scale arrays of cells within a microchannel by employing microstamping. By changing the approach in surface chemistry, H1568 cells were patterned hydrodynamically using immunoaffinity, and neutrophils were patterned through self-assembly via chemotaxis. The high patterning efficiencies (93% for hydrodynamic patterning and 68% for self-assembled patterning) and the lack of secondary adhesion demonstrate the reproducibility of the platform. The interaction between H1568 and neutrophils was visualized and quantified to determine the capability of the platform to encourage cell-cell interaction. With the introduction of H1568 cells into the self-assembled patterning platform, a significant hindrance in the neutrophils' ability to swarm was observed, indicating the important roles of inflammatory mediators within the nonsmall cell lung cancer tumor microenvironment.

15.
Nat Commun ; 9(1): 175, 2018 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-29330365

RESUMO

Extracellular vesicles (EVs) carry RNA, DNA, proteins, and lipids. Specifically, tumor-derived EVs have the potential to be utilized as disease-specific biomarkers. However, a lack of methods to isolate tumor-specific EVs has limited their use in clinical settings. Here we report a sensitive analytical microfluidic platform (EVHB-Chip) that enables tumor-specific EV-RNA isolation within 3 h. Using the EVHB-Chip, we achieve 94% tumor-EV specificity, a limit of detection of 100 EVs per µL, and a 10-fold increase in tumor RNA enrichment in comparison to other methods. Our approach allows for the subsequent release of captured tumor EVs, enabling downstream characterization and functional studies. Processing serum and plasma samples from glioblastoma multiforme (GBM) patients, we can detect the mutant EGFRvIII mRNA. Moreover, using next-generation RNA sequencing, we identify genes specific to GBM as well as transcripts that are hallmarks for the four genetic subtypes of the disease.


Assuntos
Neoplasias Encefálicas/metabolismo , Vesículas Extracelulares/química , Glioblastoma/metabolismo , Microfluídica/métodos , Transporte Biológico , Neoplasias Encefálicas/química , Neoplasias Encefálicas/genética , Linhagem Celular Tumoral , Receptores ErbB/genética , Receptores ErbB/metabolismo , Vesículas Extracelulares/metabolismo , Glioblastoma/química , Glioblastoma/genética , Humanos , Microfluídica/instrumentação , RNA/genética , RNA/metabolismo
16.
Lab Chip ; 17(20): 3498-3503, 2017 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-28932842

RESUMO

The interplay between platelets and tumor cells is known to play important roles in metastasis by enhancing tumor cell survival, tumor-vascular interactions, and escape from immune surveillance. However, platelet-covered circulating tumor cells (CTC) are extremely difficult to isolate due to masking or downregulation of surface epitopes. Here we describe a microfluidic platform that takes advantage of the satellite platelets on the surface of these "stealth" CTCs as a ubiquitous surface marker for isolation. Compared to conventional CTC enrichment techniques which rely on known surface markers expressed by tumor cells, platelet-targeted isolation is generally applicable to CTCs of both epithelial and mesenchymal phenotypes. Our approach first depletes unbound, free platelets by means of hydrodynamic size-based sorting, followed by immunoaffinity-based capture of platelet-covered CTCs using a herringbone micromixing device. This method enabled the reliable isolation of CTCs from 66% of lung and 60% of breast cancer (both epithelial) patient samples, as well as in 83% of melanoma (mesenchymal) samples. Interestingly, we observed special populations of CTCs that were extensively covered by platelets, as well as CTC-leukocyte clusters. Because these cloaked CTCs often escape conventional positive and negative isolation mechanisms, further characterization of these cells may uncover important yet overlooked biological information in blood-borne metastasis and cancer immunology.


Assuntos
Plaquetas/citologia , Separação Celular/métodos , Técnicas Analíticas Microfluídicas/métodos , Células Neoplásicas Circulantes/química , Biomarcadores Tumorais , Neoplasias da Mama/patologia , Transição Epitelial-Mesenquimal , Feminino , Humanos , Imunoensaio , Neoplasias Pulmonares/patologia
17.
J Am Chem Soc ; 139(7): 2741-2749, 2017 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-28133963

RESUMO

The detection of rare circulating tumor cells (CTCs) in the blood of cancer patients has the potential to be a powerful and noninvasive method for examining metastasis, evaluating prognosis, assessing tumor sensitivity to drugs, and monitoring therapeutic outcomes. In this study, we have developed an efficient strategy to isolate CTCs from the blood of breast cancer patients using a microfluidic immune-affinity approach. Additionally, to gain further access to these rare cells for downstream characterization, our strategy allows for easy detachment of the captured CTCs from the substrate without compromising cell viability or the ability to employ next generation RNA sequencing for the identification of specific breast cancer genes. To achieve this, a chemical ligand-exchange reaction was engineered to release cells attached to a gold nanoparticle coating bound to the surface of a herringbone microfluidic chip (NP-HBCTC-Chip). Compared to the use of the unmodified HBCTC-Chip, our approach provides several advantages, including enhanced capture efficiency and recovery of isolated CTCs.


Assuntos
Ouro/química , Nanopartículas Metálicas/química , Técnicas Analíticas Microfluídicas , Células Neoplásicas Circulantes/química , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Adesão Celular , Linhagem Celular Tumoral , Feminino , Imunofluorescência , Humanos , Ligantes , Propriedades de Superfície , Transcriptoma
18.
Biomaterials ; 65: 93-102, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26142780

RESUMO

Selective isolation and purification of circulating tumor cells (CTCs) from whole blood is an important capability for both clinical medicine and biological research. Current techniques to perform this task place the isolated cells under excessive stresses that reduce cell viability, and potentially induce phenotype change, therefore losing valuable information about the isolated cells. We present a biodegradable nano-film coating on the surface of a microfluidic chip, which can be used to effectively capture as well as non-invasively release cancer cell lines such as PC-3, LNCaP, DU 145, H1650 and H1975. We have applied layer-by-layer (LbL) assembly to create a library of ultrathin coatings using a broad range of materials through complementary interactions. By developing an LbL nano-film coating with an affinity-based cell-capture surface that is capable of selectively isolating cancer cells from whole blood, and that can be rapidly degraded on command, we are able to gently isolate cancer cells and recover them without compromising cell viability or proliferative potential. Our approach has the capability to overcome practical hurdles and provide viable cancer cells for downstream analyses, such as live cell imaging, single cell genomics, and in vitro cell culture of recovered cells. Furthermore, CTCs from cancer patients were also captured, identified, and successfully released using the LbL-modified microchips.


Assuntos
Alginatos/química , Materiais Biocompatíveis/química , Separação Celular/métodos , Técnicas Analíticas Microfluídicas/métodos , Nanoestruturas/química , Neoplasias/sangue , Células Neoplásicas Circulantes/patologia , Contagem de Células , Linhagem Celular Tumoral , Ácido Glucurônico/química , Ácidos Hexurônicos/química , Humanos , Nanoestruturas/ultraestrutura , Neoplasias/diagnóstico , Poliaminas/química , Propriedades de Superfície
19.
Adv Mater ; 27(9): 1593-9, 2015 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-25640006

RESUMO

A layer-by-layer gelatin nanocoating is presented for use as a tunable, dual response biomaterial for the capture and release of circulating tumor cells (CTCs) from cancer patient blood. The entire nanocoating can be dissolved from the surface of microfluidic devices through biologically compatible temperature shifts. Alternatively, individual CTCs can be released through locally applied mechanical stress.


Assuntos
Separação Celular/instrumentação , Técnicas Analíticas Microfluídicas/instrumentação , Nanoestruturas , Células Neoplásicas Circulantes , Materiais Biocompatíveis/química , Biópsia por Agulha Fina , Neoplasias da Mama/sangue , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Separação Celular/métodos , Sobrevivência Celular , Desenho de Equipamento , Gelatina/química , Humanos , Neoplasias Pulmonares/sangue , Neoplasias Pulmonares/genética , Masculino , Teste de Materiais , Técnicas Analíticas Microfluídicas/métodos , Modelos Teóricos , Nanoestruturas/química , Células Neoplásicas Circulantes/química , Neoplasias da Próstata/sangue , Análise de Célula Única/instrumentação , Análise de Célula Única/métodos , Estresse Mecânico , Temperatura
20.
J Mater Chem B ; 2(42): 7440-7448, 2014 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-32261969

RESUMO

In this study, human foreskin fibroblasts and mouse embryonic fibroblasts were encapsulated in mechanically reversible, THEOS and THEOS-PEG gels that completely immobilized them restricting their motility, growth and proliferation. The changes in the membrane integrity and metabolic activity (MA) of the immobilized cells were measured by IR spectroscopy and fluorescence microscopy. To explore the effects of surface chemistry and porosity on immobilized cell MA, different amounts of a biocompatible polymer, polyethylene glycol PEG, was incorporated into the silica gels. To explore the effects of the proliferative stress, in selected experiments, cellular proliferation was arrested prior to immobilization by exposing the cells to irradiation. Four main factors were identified that affect the long-term survival of the cells within the immobilization matrix: (1) porosity/permeability of the gel, (2) structural homogeneity of the gel, (3) specific interactions between the cell membrane and the gel surface and (4) the proliferative stress. It was shown that the immobilized cells could easily be mechanically recovered from the gel and upon incubation, proliferated normally. It is believed that the gels and the matrix developed here have very significant potential applications in tissue engineering and in personalized cancer treatment.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA