Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Small ; 18(17): e2106097, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35344274

RESUMO

Circulating tumor cell (CTC) clusters are associated with increased metastatic potential and worse patient prognosis, but are rare, difficult to count, and poorly characterized biophysically. The PillarX device described here is a bimodular microfluidic device (Pillar-device and an X-magnetic device) to profile single CTCs and clusters from whole blood based on their size, deformability, and epithelial marker expression. Larger, less deformable clusters and large single cells are captured in the Pillar-device and sorted according to pillar gap sizes. Smaller, deformable clusters and single cells are subsequently captured in the X-device and separated based on epithelial marker expression using functionalized magnetic nanoparticles. Clusters of established and primary breast cancer cells with variable degrees of cohesion driven by different cell-cell adhesion protein expression are profiled in the device. Cohesive clusters exhibit a lower deformability as they travel through the pillar array, relative to less cohesive clusters, and have greater collective invasive behavior. The ability of the PillarX device to capture clusters is validated in mouse models and patients of metastatic breast cancer. Thus, this device effectively enumerates and profiles CTC clusters based on their unique geometrical, physical, and biochemical properties, and could form the basis of a novel prognostic clinical tool.


Assuntos
Neoplasias da Mama , Células Neoplásicas Circulantes , Animais , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Separação Celular , Feminino , Humanos , Dispositivos Lab-On-A-Chip , Camundongos , Células Neoplásicas Circulantes/patologia , Prognóstico
3.
EBioMedicine ; 61: 103031, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33045471

RESUMO

BACKGROUND: Malignant pleural mesothelioma (MPM) is an aggressive cancer related to asbestos exposure. Early diagnosis is challenging due to generic symptoms and a lack of biomarkers. We previously demonstrated that mesothelial precursor cells (MPC) characterized by mesothelin (MSLN)+CD90+CD34+ could be implicated in the development of mesothelioma after asbestos exposure. Here, we aimed to determine the clinical significance of detecting MPC in blood for early-stage diagnosis and prognosis of mesothelioma. METHODS: Due to the rarity of MPC in blood, it is challenging to identify this cell population using conventional techniques. Hence, we have developed a microfluidic liquid biopsy platform called MesoFind that utilizes an immunomagnetic, mesothelin capture strategy coupled with immunofluorescence to identify rare populations of cells at high sensitivity and precision. To validate our technique, we compared this approach to flow cytometry for the detection of MPC in murine blood and lavage samples. Upon successful validation of the murine samples, we then proceeded to examine circulating MPC in 23 patients with MPM, 23 asbestos-exposed individuals (ASB), and 10 healthy donors (HD) to evaluate their prognostic and diagnostic value. FINDING: MPC were successfully detected in the blood of murine samples using MesoFind but were undetectable with flow cytometry. Circulating MPC were significantly higher in patients with epithelioid MPM compared to HD and ASB. The MPC subpopulation, MSLN+ and CD90+, were upregulated in ASB compared to HD suggesting an early role in pleural damage from asbestos. The MPC subpopulation, MSLN+ and CD34+, in contrast, were detected in advanced MPM and associated with markers of poor prognosis, suggesting a predominant role during cancer progression. INTERPRETATION: The identification of circulating MPC presents an attractive solution for screening and early diagnosis of epithelioid mesothelioma. The presence of different subtypes of MPC have a prognostic value that could be of assistance with clinical decisions in patients with MPM. FUNDING: Princess Margaret Hospital Foundation Mesothelioma Research Fund, Toronto General & Western Hospital Foundation.


Assuntos
Biomarcadores Tumorais , Biópsia Líquida , Mesotelioma/diagnóstico , Células Neoplásicas Circulantes/patologia , Idoso , Idoso de 80 Anos ou mais , Animais , Amianto/efeitos adversos , Linhagem Celular , Modelos Animais de Doenças , Feminino , Perfilação da Expressão Gênica/métodos , Humanos , Biópsia Líquida/métodos , Biópsia Líquida/normas , Masculino , Mesotelina , Mesotelioma/etiologia , Camundongos , Pessoa de Meia-Idade , Gradação de Tumores , Metástase Neoplásica , Estadiamento de Neoplasias , Exposição Ocupacional/efeitos adversos , Prognóstico , Reprodutibilidade dos Testes , Transcriptoma
4.
ACS Appl Mater Interfaces ; 11(44): 41030-41037, 2019 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-31600052

RESUMO

Microenvironmental factors play critical roles in regulating stem cell fate, providing a rationale to engineer biomimetic microenvironments that facilitate rapid and effective stem cell differentiation. Three-dimensional (3D) hierarchical microarchitectures have been developed to enable rapid neural differentiation of multipotent human mesenchymal stromal cells (HMSCs) via mechanotransduction. However, low cell viability during long-term culture and poor cell recovery efficiency from the architectures were also observed. Such problems hinder further applications of the architectures in stem cell differentiation. Here, we present improved 3D nanostructured microarchitectures functionalized with cell-adhesion-promoting arginylglycylaspartic acid (RGD) peptides. These RGD-functionalized architectures significantly upregulated long-term cell viability and facilitated effective recovery of differentiated cells from the architectures while maintaining high differentiation efficiency. Efficient recovery of highly viable differentiated cells enabled the downstream analysis of morphology and protein expression to be performed. Remarkably, even after the removal of the mechanical stimulus provided by the 3D microarchitectures, the recovered HMSCs showed a neuron-like elongated morphology for 10 days and consistently expressed microtubule-associated protein 2, a mature neural marker. RGD-functionalized nanostructured microarchitectures hold great potential to guide effective differentiation of highly viable stem cells.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Mecanotransdução Celular/efeitos dos fármacos , Nanoestruturas/química , Oligopeptídeos/farmacologia , Técnicas de Cultura de Células/métodos , Sobrevivência Celular/efeitos dos fármacos , Ouro/química , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Nanoestruturas/toxicidade , Oligopeptídeos/química , Impressão Tridimensional
5.
Anal Chem ; 91(15): 9348-9355, 2019 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-31264404

RESUMO

The analysis of circulating tumor cells (CTCs) provides a means to collect information about the evolving properties of a tumor during cancer progression and treatment. For patients with metastatic prostate cancer, noninvasive serial measurements of bloodborne cells may provide a means to tailor therapeutic decisions based on an individual patient's response. Here, we used a high-sensitivity profiling approach to monitor CTCs in patients with metastatic castrate-resistant prostate cancer (mCRPC) undergoing treatment with abiraterone and enzalutamide, two drugs used to treat advanced prostate cancer. The capture and profiling approach uses antibody-functionalized magnetic nanoparticles to sort cells according to protein expression levels. CTCs are tagged with magnetic nanoparticles conjugated to an antibody specific for the epithelial cell adhesion molecule (EpCAM) and sorted into four zones of a microfluidic device based on EpCAM expression levels. Our approach was compared to the FDA-cleared CellSearch method, and we demonstrate significantly higher capture efficiency of low-EpCAM cells compared to the commercial method. The nanoparticle-based approach detected CTCs from 86% of patients at baseline, compared to CellSearch which only detected CTCs from 60% of patients. Patients were stratified as prostate specific antigen (PSA) progressive versus responsive based on clinically acceptable definitions, and it was observed that patients with a limited response to therapy had elevated levels of androgen receptor variant 7 (ARV7) and the mesenchymal marker, N-cadherin, expressed on their CTCs. In addition, these CTCs exhibited lower EpCAM expression. The results highlight features of CTCs associated with disease progression on abiraterone or enzalutamide, including mesenchymal phenotypes and increased expression levels of ARV7. The use of a high-sensitivity method to capture and profile CTCs provides more informative data concerning the phenotypic properties of these cells as patients undergo treatment relative to an FDA-cleared method.


Assuntos
Nanopartículas de Magnetita/uso terapêutico , Células Neoplásicas Circulantes/patologia , Neoplasias da Próstata/patologia , Androstenos/uso terapêutico , Antineoplásicos Imunológicos/uso terapêutico , Benzamidas , Caderinas/análise , Caderinas/imunologia , Progressão da Doença , Molécula de Adesão da Célula Epitelial/imunologia , Humanos , Nanopartículas de Magnetita/química , Masculino , Nitrilas , Fenótipo , Feniltioidantoína/análogos & derivados , Feniltioidantoína/uso terapêutico , Neoplasias da Próstata/tratamento farmacológico , Receptores Androgênicos/análise , Receptores Androgênicos/imunologia
6.
Nano Lett ; 18(3): 2140-2147, 2018 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-29480726

RESUMO

Invasion of dense tissues by cancer cells involves the interplay between the penetration resistance offered by interstitial pores and the deformability of cells. Metastatic cancer cells find optimal paths of minimal resistance through an adaptive path-finding process, which leads to successful dissemination. The physical limit of nuclear deformation is related to the minimal cross section of pores that can be successfully penetrated. However, this single biophysical parameter does not fully describe the architectural complexity of tissues featuring pores of variable area and shape. Here, employing laser nanolithography, we fabricate pore microenvironment models with well-controlled pore shapes, through which human breast cells (MCF10A) and their metastatic offspring (MCF10CA1a.cl1) could pervade. In these experimental settings, we demonstrate that the actual pore shape, and not only the cross section, is a major and independent determinant of cancer penetration efficiency. In complex architectures containing pores demanding large deformations from invading cells, tall and narrow rectangular openings facilitate cancer migration. In addition, we highlight the characteristic traits of the explorative behavior enabling metastatic cells to identify and select such pore shapes in a complex multishape pore environment, pinpointing paths of least resistance to invasion.


Assuntos
Neoplasias da Mama/patologia , Movimento Celular , Metástase Neoplásica/patologia , Linhagem Celular Tumoral , Núcleo Celular/patologia , Feminino , Complexo de Golgi/patologia , Humanos , Nanoestruturas/ultraestrutura , Nanotecnologia , Porosidade
7.
ACS Appl Mater Interfaces ; 9(24): 20435-20443, 2017 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-28548481

RESUMO

Isolating subpopulations of heterogeneous cancer cells is an important capability for the meaningful characterization of circulating tumor cells at different stages of tumor progression and during the epithelial-to-mesenchymal transition. Here, we present a microfluidic device that can separate phenotypically distinct subpopulations of cancer cells. Magnetic nanoparticles coated with antibodies against the epithelial cell adhesion molecule (EpCAM) are used to separate breast cancer cells in the microfluidic platform. Cells are sorted into different zones on the basis of the levels of EpCAM expression, which enables the detection of cells that are losing epithelial character and becoming more mesenchymal. The phenotypic properties of the isolated cells with low and high EpCAM are then assessed using matrix-coated surfaces for collagen uptake analysis, and an NAD(P)H assay that assesses metabolic activity. We show that low-EpCAM expressing cells have higher collagen uptake and higher folate-induced NAD(P)H responses compared to those of high-EpCAM expressing cells. In addition, we tested SKBR3 cancer cells undergoing chemically induced hypoxia. The induced cells have reduced expression of EpCAM, and we find that these cells have higher collagen uptake and NAD(P)H metabolism relative to noninduced cells. This work demonstrates that nanoparticle-mediated binning facilitates the isolation of functionally distinct cell subpopulations and allows surface marker expression to be associated with invasiveness, including collagen uptake and metabolic activity.


Assuntos
Nanopartículas , Antígenos de Neoplasias , Moléculas de Adesão Celular , Linhagem Celular Tumoral , Molécula de Adesão da Célula Epitelial , Humanos , Células Neoplásicas Circulantes
8.
Nat Nanotechnol ; 12(3): 274-281, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27870841

RESUMO

Profiling the heterogeneous phenotypes of rare circulating tumour cells (CTCs) in whole blood is critical to unravelling the complex and dynamic properties of these potential clinical markers. This task is challenging because these cells are present at parts per billion levels among normal blood cells. Here we report a new nanoparticle-enabled method for CTC characterization, called magnetic ranking cytometry, which profiles CTCs on the basis of their surface expression phenotype. We achieve this using a microfluidic chip that successfully processes whole blood samples. The approach classifies CTCs with single-cell resolution in accordance with their expression of phenotypic surface markers, which is read out using magnetic nanoparticles. We deploy this new technique to reveal the dynamic phenotypes of CTCs in unprocessed blood from mice as a function of tumour growth and aggressiveness. We also test magnetic ranking cytometry using blood samples collected from cancer patients.


Assuntos
Biomarcadores Tumorais/sangue , Neoplasias da Mama , Separação Celular , Dispositivos Lab-On-A-Chip , Nanopartículas de Magnetita/química , Células Neoplásicas Circulantes , Neoplasias da Mama/sangue , Neoplasias da Mama/patologia , Separação Celular/instrumentação , Separação Celular/métodos , Feminino , Humanos , Células MCF-7 , Células Neoplásicas Circulantes/metabolismo , Células Neoplásicas Circulantes/patologia
9.
Angew Chem Int Ed Engl ; 55(4): 1252-65, 2016 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-26643151

RESUMO

Over the last decade, significant progress has been made towards the development of approaches that enable the capture of rare circulating tumor cells (CTCs) from the blood of cancer patients, a critical capability for noninvasive tumor profiling. These advances have leveraged new insights in materials chemistry and microfluidics and allowed the capture and enumeration of CTCs with unprecedented sensitivity. However, it has become increasingly clear that simply capturing and counting tumor cells launched into the bloodstream may not provide the information needed to advance our understanding of the biology of these rare cells, or to allow us to better exploit them in medicine. A variety of advances have now emerged demonstrating that more information can be extracted from CTCs with next-generation devices and materials featuring tailored physical and chemical properties. In this Minireview, the last ten years of work in this area will be discussed, with an emphasis on the groundbreaking work of the last five years, during which the focus has moved beyond the simple capture of CTCs and gravitated towards approaches that enable in-depth analysis.


Assuntos
Separação Celular/instrumentação , Células Neoplásicas Circulantes/patologia , Humanos
10.
Lab Chip ; 13(22): 4374-84, 2013 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-24056576

RESUMO

Pancreatic islets are heavily vascularized in vivo with fenestrated endothelial cells (ECs) to facilitate blood glucose-sensing and endocrine hormone secretion. The close proximity of insulin secreting beta cells and ECs also plays a critical role in modulating the proliferation and survival of both cell types with the mechanisms governing this interaction poorly understood. Isolated islets lose EC morphology and mass over a period of days in culture prohibiting study of this interaction in vitro. The loss of ECs also limits the efficacy of islet transplant revascularization in the treatment of Type 1 diabetes. We previously showed that microfluidically driven flow positively affects beta-cell function and EC survival in culture due to enhanced transport of media into the tissue. However, holding islets stationary in media flow using a dam-wall design also resulted in reduced glucose-stimulated metabolic and Ca(2+) responses at the periphery of the tissue consistent with shear-induced damage. We have now created a device that traps islets into sequential cup-shaped nozzles. This hydrodynamic trap design limits flow velocity around the perimeter of the islet while enhancing media flow through the tissue. We demonstrate the feasibility of this device to dynamically treat and collect effluent from islets. We further show that treating islets in this device enhances EC morphology without reducing glucose-stimulate Ca(2+) responses. These data reveal a microfluidic device to study EC and endocrine cell interaction that can be further leveraged to prime islets prior to transplantation.


Assuntos
Ilhotas Pancreáticas/citologia , Técnicas Analíticas Microfluídicas/instrumentação , Compostos de Anilina/química , Animais , Cálcio/metabolismo , Células Cultivadas , Células Endoteliais/citologia , Glucose/farmacologia , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Resistência ao Cisalhamento , Xantenos/química
11.
Biophys J ; 103(11): 2379-88, 2012 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-23283237

RESUMO

Fibroblast growth factor-21 (FGF21) has therapeutic potential for metabolic syndrome due to positive effects on fatty acid metabolism in liver and white adipose tissue. FGF21 also improves pancreatic islet survival in excess palmitate; however, much less is known about FGF21-induced metabolism in this tissue. We first confirmed FGF21-dependent activity in islets by identifying expression of the cognate coreceptor Klothoß, and by measuring a ligand-stimulated decrease in acetyl-CoA carboxylase expression. To further reveal the effect of FGF21 on metabolism, we employed a unique combination of two-photon and confocal autofluorescence imaging of the NAD(P)H and mitochondrial NADH responses while holding living islets stationary in a microfluidic device. These responses were further correlated to mitochondrial membrane potential and insulin secretion. Glucose-stimulated responses were relatively unchanged by FGF21. In contrast, responses to glucose in the presence of palmitate were significantly reduced compared to controls showing diminished NAD(P)H, mitochondrial NADH, mitochondrial membrane potential, and insulin secretion. Consistent with the glucose-stimulated responses being smaller due to continued fatty acid oxidation, mitochondrial membrane potential was increased in FGF21-treated islets by using the fatty acid transport inhibitor etomoxir. Citrate-stimulated NADPH responses were also significantly larger in FGF21-treated islets suggesting preference for citrate cycling rather than acetyl-CoA carboxylase-dependent fatty acid synthesis. Overall, these data show a reduction in palmitate-induced potentiation of glucose-stimulated metabolism and insulin secretion in FGF21-treated islets, and establish the use of autofluorescence imaging and microfluidic devices to investigate cell metabolism in a limited amount of living tissue.


Assuntos
Fatores de Crescimento de Fibroblastos/metabolismo , Fatores de Crescimento de Fibroblastos/farmacologia , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/metabolismo , NADP/metabolismo , NAD/metabolismo , Imagem Óptica/métodos , Animais , Células Cultivadas , Ilhotas Pancreáticas/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL
12.
PLoS One ; 6(9): e24904, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21961048

RESUMO

Pancreatic islets are heavily vascularized in vivo with each insulin secreting beta-cell associated with at least one endothelial cell (EC). This structure is maintained immediately post-isolation; however, in culture the ECs slowly deteriorate, losing density and branched morphology. We postulate that this deterioration occurs in the absence of blood flow due to limited diffusion of media inside the tissue. To improve exchange of media inside the tissue, we created a microfluidic device to culture islets in a range of flow-rates. Culturing the islets from C57BL6 mice in this device with media flowing between 1 and 7 ml/24 hr resulted in twice the EC-density and -connected length compared to classically cultured islets. Media containing fluorescent dextran reached the center of islets in the device in a flow-rate-dependant manner consistent with improved penetration. We also observed deterioration of EC morphology using serum free media that was rescued by addition of bovine serum albumin, a known anti-apoptotic signal with limited diffusion in tissue. We further examined the effect of flow on beta-cells showing dampened glucose-stimulated Ca(2+)-response from cells at the periphery of the islet where fluid shear-stress is greatest. However, we observed normal two-photon NAD(P)H response and insulin secretion from the remainder of the islet. These data reveal the deterioration of islet EC-morphology is in part due to restricted diffusion of serum albumin within the tissue. These data further reveal microfluidic devices as unique platforms to optimize islet culture by introducing intercellular flow to overcome the restricted diffusion of media components.


Assuntos
Células Endoteliais/metabolismo , Ilhotas Pancreáticas/metabolismo , Técnicas Analíticas Microfluídicas/métodos , Animais , Cálcio/metabolismo , Tamanho Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Células Endoteliais/efeitos dos fármacos , Glucose/farmacologia , História do Século XX , Insulina/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/irrigação sanguínea , Ilhotas Pancreáticas/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Técnicas Analíticas Microfluídicas/instrumentação , Microscopia de Fluorescência por Excitação Multifotônica/métodos , NADP/metabolismo , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Soroalbumina Bovina/farmacologia , Técnicas de Cultura de Tecidos
13.
J Biol Chem ; 282(41): 30120-30, 2007 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-17716977

RESUMO

Formins are multidomain proteins that regulate numerous cytoskeleton-dependent cellular processes. These effects are mediated by the presence of two regions of homology, formin homology 1 and FH2. The diaphanous-related formins (DRFs) are distinguished by the presence of interacting N- and C-terminal regulatory domains. The GTPase binding domain and diaphanous inhibitory domain (DID) are found in the N terminus and bind to the diaphanous autoregulatory domain (DAD) found in the C terminus. Adjacent to the DID is an N-terminal dimerization motif (DD) and coiled-coil region (CC). The N terminus of Dia1 is also proposed to contain a Rho-independent membrane-targeting motif. We undertook an extensive structure/function analysis of the mDia1 N terminus to further our understanding of its role in vivo. We show here that both DID and DD are required for efficient autoinhibition in the context of full-length mDia1 and that the DD of mDia1 and mDia2, like formin homology 2, mediates homo- but not heterodimerization with other DRF family members. In contrast, our results suggest that the DID/DAD interaction mediates heterodimerization of full-length mDia1 and mDia2 and that the auto-inhibited conformation of DRFs is oligomeric. In addition, we also show that the DD/CC region is required for the Rho-independent membrane targeting of the isolated N terminus.


Assuntos
Proteínas de Transporte/química , NADPH Desidrogenase/química , Actinas/química , Motivos de Aminoácidos , Animais , Linhagem Celular , Citoplasma/metabolismo , Dimerização , Forminas , Regulação da Expressão Gênica , Imuno-Histoquímica/métodos , Camundongos , Proteínas Associadas aos Microtúbulos , Modelos Biológicos , Células NIH 3T3 , Ligação Proteica , Estrutura Terciária de Proteína
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...