Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
J Drugs Dermatol ; 22(5): 457-464, 2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-37133478

RESUMEN

BACKGROUND: The standard of care when treating melanoma in situ (MMIS) is an excision with at least 5 mm surgical margins.1 Some studies have suggested up to 9 mm margins to maximize local recurrence-free survival.2 This retrospective review aims to assess the efficacy of imiquimod as a topical treatment for persistently positive MMIS at the margins of prior excisions or where surgery is not an option. METHODS: Retrospective study conducted at Moffitt Cancer Center between 2019 and 2021 with patients aged > 18 years with MMIS at the margins of excision of an invasive melanoma or MMIS. Included patients were not ideal candidates for primary or additional surgical resection due to non-feasibility of surgery because of comorbidity or cosmetically sensitive location and/or the need for repeated skin grafting, or due to patient's refusal. Patients received imiquimod on protocol for 16 weeks and were monitored for treatment response and side effects. Following completion of the treatment, scouting biopsies were performed to assess histological response, and dermoscopy was used to determine the clinical disease status. RESULTS: Ten patients completed 16 weeks of imiquimod. Seven (75%) had a median of 2 surgical resections, and 3 refused surgery despite discussion that surgery was standard of care. Seven were deemed free of disease on post-imiquimod treatment scouting biopsies, while 2 were found to be clinically free of disease following confocal microscopy, indicating a tumor clearance rate of 90% with imiquimod treatment. One patient was found to have persistent residual disease following 2 rounds of imiquimod and was taken for an additional surgical excision after which they were deemed free of disease. Median follow-up duration from the onset of imiquimod therapy to the last clinic visit was 18 months, without any recurrences to date. CONCLUSION: Imiquimod appears to demonstrate an encouraging tumor clearance among patients with persistent MMIS after surgery where further surgical resection may not be feasible. Although long-term durability has not been demonstrated in this study, a 90% tumor clearance rate is promising. J Drugs Dermatol. 2023;22(5): doi:10.36849/JDD.6987.


Asunto(s)
Antineoplásicos , Melanoma , Neoplasias Cutáneas , Humanos , Imiquimod/efectos adversos , Antineoplásicos/efectos adversos , Estudios Retrospectivos , Melanoma/tratamiento farmacológico , Melanoma/cirugía , Melanoma/patología , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/cirugía , Neoplasias Cutáneas/patología , Aminoquinolinas/efectos adversos , Resultado del Tratamiento , Melanoma Cutáneo Maligno
2.
Neuromodulation ; 2023 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-37530695

RESUMEN

OBJECTIVES: Low-intensity, focused ultrasound (FUS) is an emerging noninvasive neuromodulation approach, with improved spatial and temporal resolution and penetration depth compared to other noninvasive electrical stimulation strategies. FUS has been used to modulate circuits in the brain and the peripheral nervous system, however, its potential to modulate spinal circuits is unclear. In this study, we assessed the effect of trans-spinal FUS (tsFUS) on spinal reflexes in healthy rats. MATERIALS AND METHODS: tsFUS targeting different spinal segments was delivered for 1 minute, under anesthesia. Monosynaptic H-reflex of the sciatic nerve, polysynaptic flexor reflex of the sural nerve, and withdrawal reflex tested with a hot plate were measured before, during, and after tsFUS. RESULTS: tsFUS reversibly suppresses the H-reflex in a spinal segment-, acoustic pressure- and pulse-repetition frequency (PRF)-dependent manner. tsFUS with high PRF augments the degree of homosynaptic depression of the H-reflex observed with paired stimuli. It suppresses the windup of components of the flexor reflex associated with slower, C-afferent, but not faster, A- afferent fibers. Finally, it increases the latency of the withdrawal reflex. tsFUS does not elicit neuronal loss in the spinal cord. CONCLUSIONS: Our study provides evidence that tsFUS reversibly suppresses spinal reflexes and suggests that tsFUS could be a safe and effective strategy for spinal cord neuromodulation in disorders associated with hyperreflexia, including spasticity after spinal cord injury and painful syndromes.

3.
Proc Natl Acad Sci U S A ; 116(21): 10270-10279, 2019 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-31068473

RESUMEN

Infectious diseases caused by bacterial pathogens remain one of the most common causes of morbidity and mortality worldwide. Rapid microbiological analysis is required for prompt treatment of bacterial infections and to facilitate antibiotic stewardship. This study reports an adaptable microfluidic system for rapid pathogen classification and antimicrobial susceptibility testing (AST) at the single-cell level. By incorporating tunable microfluidic valves along with real-time optical detection, bacteria can be trapped and classified according to their physical shape and size for pathogen classification. By monitoring their growth in the presence of antibiotics at the single-cell level, antimicrobial susceptibility of the bacteria can be determined in as little as 30 minutes compared with days required for standard procedures. The microfluidic system is able to detect bacterial pathogens in urine, blood cultures, and whole blood and can analyze polymicrobial samples. We pilot a study of 25 clinical urine samples to demonstrate the clinical applicability of the microfluidic system. The platform demonstrated a sensitivity of 100% and specificity of 83.33% for pathogen classification and achieved 100% concordance for AST.


Asunto(s)
Antiinfecciosos , Microfluídica , Antibacterianos , Disbiosis , Humanos , Pruebas de Sensibilidad Microbiana
4.
Anal Chem ; 92(19): 13254-13261, 2020 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-32869628

RESUMEN

Digital nucleic acid amplification testing (dNAAT) and analysis techniques, such as digital polymerase chain reaction (PCR), have become useful clinical diagnostic tools. However, nucleic acid (NA) sample preparation preceding dNAAT is generally laborious and performed manually, thus creating the need for a simple sample preparation technique and a facile coupling strategy for dNAAT. Therefore, we demonstrate a simple workflow which automates magnetic bead-based extraction of NAs with a one-step transfer to dNAAT. Specifically, we leverage droplet magnetofluidics (DM) to automate the movement of magnetic beads between small volumes of reagents commonly employed for NA extraction and purification. Importantly, the buffer typically used to elute the NAs off the magnetic beads is replaced by a carefully selected PCR solution, enabling direct transfer from sample preparation to dNAAT. Moreover, we demonstrate the potential for multiplexing using a digital high-resolution melt (dHRM) after the digital PCR (dPCR). The utility of this workflow is demonstrated with duplexed detection of bacteria in a sample imitating a coinfection. We first purify the bacterial DNA into a PCR solution using our DM-based sample preparation. We then transfer the purified bacterial DNA to our microfluidic nanoarray to amplify 16S rRNA using dPCR and then perform dHRM to identify the two bacterial species.


Asunto(s)
Automatización , Escherichia coli/genética , Técnicas de Amplificación de Ácido Nucleico , Reacción en Cadena de la Polimerasa , ARN Ribosómico 16S/genética , Staphylococcus aureus/genética , Tamaño de la Partícula , Propiedades de Superficie
5.
Nanomedicine ; 17: 246-253, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30794964

RESUMEN

A nanotube assisted microwave electroporation (NAME) technique is demonstrated for delivering molecular biosensors into viable bacteria for multiplex single cell pathogen identification to advance rapid diagnostics in clinical microbiology. Due to the small volume of a bacterial cell (~femtoliter), the intracellular concentration of the target molecule is high, which results in a strong signal for single cell detection without amplification. The NAME procedure can be completed in as little as 30 minutes and can achieve over 90% transformation efficiency. We demonstrate the feasibility of NAME for identifying clinical isolates of bloodborne and uropathogenic pathogens and detecting bacterial pathogens directly from patient's samples. In conjunction with a microfluidic single cell trapping technique, NAME allows single cell pathogen identification and antimicrobial susceptibility testing concurrently. Using this approach, the time for microbiological analysis reduces from days to hours, which will have a significant impact on the clinical management of bacterial infections.


Asunto(s)
Bacterias/aislamiento & purificación , Infecciones Bacterianas/diagnóstico , Electroporación/instrumentación , Nanotubos/química , Análisis de la Célula Individual/instrumentación , Antibacterianos/farmacología , Bacterias/efectos de los fármacos , Infecciones Bacterianas/tratamiento farmacológico , Infecciones Bacterianas/microbiología , Diseño de Equipo , Humanos , Pruebas de Sensibilidad Microbiana/instrumentación , Técnicas Analíticas Microfluídicas/instrumentación , Microondas
6.
Brain Stimul ; 16(3): 703-711, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37055009

RESUMEN

Focused ultrasound stimulation (FUS) activates mechanosensitive ion channels and is emerging as a method of noninvasive neuromodulation. In preclinical studies, FUS of the spleen (sFUS) activates an anti-inflammatory neural pathway which suppresses acute and chronic inflammation. However, the relevance of sFUS for regulating inflammatory responses in humans is unknown. Here, we used a modified diagnostic ultrasound imaging system to target the spleen of healthy human subjects with 3 min of continuously swept or stationary focused pulsed ultrasound, delivered at three different energy levels within allowable safety exposure limits. Potential anti-inflammatory effects of sFUS were assessed by measuring sFUS-elicited changes in endotoxin-induced tumor necrosis factor (TNF) production in whole blood samples from insonified subjects. We found that stimulation with either continuously swept or focused pulsed ultrasound has an anti-inflammatory effect: sFUS lowers TNF production for >2 h, with TNF returning to baseline by 24 h following sFUS. This response is independent of anatomical target (i.e., spleen hilum or parenchyma) or ultrasound energy level. No clinical, biochemical, or hematological parameters are adversely impacted. This is the first demonstration that sFUS suppresses the normal inflammatory response in humans, with potential implications for noninvasive bioelectronic therapy of inflammatory disorders.


Asunto(s)
Bazo , Terapia por Ultrasonido , Humanos , Bazo/diagnóstico por imagen , Ultrasonografía , Terapia por Ultrasonido/métodos , Vías Nerviosas , Ondas Ultrasónicas
7.
Front Immunol ; 13: 892086, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35784337

RESUMEN

Interfaces between the nervous and immune systems have been shown essential for the coordination and regulation of immune responses. Non-invasive ultrasound stimulation targeted to the spleen has recently been shown capable of activating one such interface, the splenic cholinergic anti-inflammatory pathway (CAP). Over the past decade, CAP and other neuroimmune pathways have been activated using implanted nerve stimulators and tested to prevent cytokine release and inflammation. However, CAP studies have typically been performed in models of severe, systemic (e.g., endotoxemia) or chronic inflammation (e.g., collagen-induced arthritis or DSS-induced colitis). Herein, we examined the effects of activation of the splenic CAP with ultrasound in a model of local bacterial infection by lung instillation of 105 CFU of Streptococcus pneumoniae. We demonstrate a time-dependent effect of CAP activation on the cytokine response assay during infection progression. CAP activation-induced cytokine suppression is absent at intermediate times post-infection (16 hours following inoculation), but present during the early (4 hours) and later phases (48 hours). These results indicate that cytokine inhibition associated with splenic CAP activation is not observed at all timepoints following bacterial infection and highlights the importance of further studying neuroimmune interfaces within the context of different immune system and inflammatory states.


Asunto(s)
Neumonía , Bazo , Antiinflamatorios/farmacología , Citocinas/metabolismo , Humanos , Inflamación/metabolismo , Neumonía/metabolismo , Nervio Vago/fisiología
8.
Nat Biomed Eng ; 6(6): 683-705, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35361935

RESUMEN

Peripheral neurons that sense glucose relay signals of glucose availability to integrative clusters of neurons in the brain. However, the roles of such signalling pathways in the maintenance of glucose homoeostasis and their contribution to disease are unknown. Here we show that the selective activation of the nerve plexus of the hepatic portal system via peripheral focused ultrasound stimulation (pFUS) improves glucose homoeostasis in mice and rats with insulin-resistant diabetes and in swine subject to hyperinsulinemic-euglycaemic clamps. pFUS modulated the activity of sensory projections to the hypothalamus, altered the concentrations of metabolism-regulating neurotransmitters, and enhanced glucose tolerance and utilization in the three species, whereas physical transection or chemical blocking of the liver-brain nerve pathway abolished the effect of pFUS on glucose tolerance. Longitudinal multi-omic profiling of metabolic tissues from the treated animals confirmed pFUS-induced modifications of key metabolic functions in liver, pancreas, muscle, adipose, kidney and intestinal tissues. Non-invasive ultrasound activation of afferent autonomic nerves may represent a non-pharmacologic therapy for the restoration of glucose homoeostasis in type-2 diabetes and other metabolic diseases.


Asunto(s)
Diabetes Mellitus Experimental , Glucosa , Animales , Diabetes Mellitus Experimental/terapia , Glucosa/metabolismo , Homeostasis , Hipotálamo/metabolismo , Hígado/metabolismo , Ratones , Ratas , Porcinos
9.
Biosensors (Basel) ; 11(8)2021 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-34436090

RESUMEN

Bloodstream infections are a significant cause of morbidity and mortality worldwide. The rapid initiation of effective antibiotic treatment is critical for patients with bloodstream infections. However, the diagnosis of bloodborne pathogens is largely complicated by the matrix effect of blood and the lengthy blood tube culture procedure. Here we report a culture-free workflow for the rapid isolation and enrichment of bacterial pathogens from whole blood for single-cell antimicrobial susceptibility testing (AST). A dextran sedimentation step reduces the concentration of blood cells by 4 orders of magnitude in 20-30 min while maintaining the effective concentration of bacteria in the sample. Red blood cell depletion facilitates the downstream centrifugation-based enrichment step at a sepsis-relevant bacteria concentration. The workflow is compatible with common antibiotic-resistant bacteria and does not influence the minimum inhibitory concentrations. By applying a microfluidic single-cell trapping device, we demonstrate the workflow for the rapid determination of bacterial infection and antimicrobial susceptibility testing at the single-cell level. The entire workflow from blood to categorical AST result can be completed in less than two hours.


Asunto(s)
Pruebas de Sensibilidad Microbiana , Sepsis/diagnóstico , Flujo de Trabajo , Antibacterianos , Bacterias , Infecciones Bacterianas , Humanos , Dispositivos Laboratorio en un Chip , Microfluídica
10.
Lab Chip ; 21(6): 1073-1083, 2021 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-33529300

RESUMEN

Single-molecule and single-cell analysis techniques have opened new opportunities for characterizing and analyzing heterogeneity within biological samples. These detection methods are often referred to as digital assays because the biological sample is partitioned into many small compartments and each compartment contains a discrete number of targets (e.g. cells). Using digital assays, researchers can precisely detect and quantify individual targets, and this capability has made digital techniques the basis for many modern bioanalytical tools (including digital PCR, single cell RNA sequencing, and digital ELISA). However, digital assays are dominated by optical analysis systems that typically utilize microscopy to analyze partitioned samples. The utility of digital assays may be dramatically enhanced by implementing cost-efficient and portable electrical detection capabilities. Herein, we describe a digital electrical impedance sensing platform that enables direct multiplexed measurement of single cell bacterial cells. We outline our solutions to the challenge of multiplexing impedance sensing across many culture compartments and demonstrate the potential for rapidly differentiating antimicrobial resistant versus susceptible strains of bacteria.


Asunto(s)
Antiinfecciosos , Bacterias , Bacterias/genética , Impedancia Eléctrica , Ensayo de Inmunoadsorción Enzimática , Reacción en Cadena de la Polimerasa
11.
Bioelectron Med ; 7(1): 7, 2021 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-34024277

RESUMEN

There is a broad and growing interest in Bioelectronic Medicine, a dynamic field that continues to generate new approaches in disease treatment. The fourth bioelectronic medicine summit "Technology targeting molecular mechanisms" took place on September 23 and 24, 2020. This virtual meeting was hosted by the Feinstein Institutes for Medical Research, Northwell Health. The summit called international attention to Bioelectronic Medicine as a platform for new developments in science, technology, and healthcare. The meeting was an arena for exchanging new ideas and seeding potential collaborations involving teams in academia and industry. The summit provided a forum for leaders in the field to discuss current progress, challenges, and future developments in Bioelectronic Medicine. The main topics discussed at the summit are outlined here.

12.
J Neurosci Methods ; 341: 108721, 2020 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-32387189

RESUMEN

BACKGROUND: A fundamental limit to the study of the peripheral nervous system and its effect on organ function is the lack of tools to selectively target and stimulate specific neurons. Traditional implant and electrode-based systems remain too large and invasive for use at the organ or sub-organ level (without stimulating or effecting neighboring organs and tissues). Recent progress in optical and genetic tools (such as optogenetics) has provided a new level of molecular specificity and selectivity to the neurons that are stimulated by bioelectronic devices. However, the modified neurons that result from use of these tools (that can be selectively activated based on expression of light, heat, or stimuli sensitive ion channels) often still require stimulation by implantable devices and face difficult scientific, technical, and regulatory hurdles for clinical translation. NEW METHOD: Herein, we present a new tool for selective activation of neuronal pathways using anatomical site-specific, peripheral focused ultrasound neuromodulation (pFUS). RESULTS: We utilize three experimental models to expand upon and further characterize pFUS beyond data outlined to our initial report (Cotero et al., 2019a), and further demonstrate its importance as a new investigative and translational tool. First, we utilized an interconnected microporous gel scaffold to culture isolated dorsal root ganglion (DRG) neurons in an interconnected, three-dimensional in vitro culture. (Griffin et al., 2015, Tay et al., 2018) Using this system, we directly applied ultrasound (US) stimuli and confirmed US activation of peripheral neurons at pressures consistent with recent in vivo observations. (Cotero et al., 2019a, Zachs, 2019, Gigliotti et al., 2013) Next, we tested the capability of pFUS to activate previously reported nerve pathways at multiple locations within the neural circuit, including primary sensory ganglia (i.e. inferior ganglion of the vagus nerve), peripheral ganglia (i.e. sacral ganglia), and within target end-organs. In addition, we compared selective activation of multiple anatomically overlapping neural pathways (i.e. activation of the cholinergic anti-inflammatory pathway (Tracey, 2009, Pavlov and Tracey, 2012) vs. metabolic sensory pathways (O'Hare and Zsombok, 2015, Roh et al., 2016, Pocai et al., 2005) after stimulation of each separate target site. Finally, we utilized an established model of metabolic dysfunction (the LPS-induced inflammation/hyperglycemia model) to demonstrate pFUS capability to stimulate and assess alternative therapeutic stimulation sites (i.e. liver, pancreas, and intestines) in a simple and clinically relevant manner. This is demonstrated by ultrasound induced attenuation of LPS-induced hyperglycemia by stimulation at all three anatomical targets, and mapping of the effect to a specific molecular product of excitable cell types within each stimulus site. COMPARISON WITH EXISTING METHODS: The ease-of-use and non-invasive nature of pFUS provides a solution to many of the challenges facing traditional toolsets, such as implantable electrodes and genetic/optogenetic nerve stimulation strategies. CONCLUSIONS: The pFUS tool described herein provides a fundamental technology for the future study and manipulation of the peripheral nervous and neuroendocrine systems.


Asunto(s)
Neuronas , Optogenética , Vías Aferentes , Ganglios Espinales , Vías Nerviosas
13.
SLAS Technol ; 24(4): 448-452, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31226243

RESUMEN

A new study published in Nature Communications outlines our group's results using focused ultrasound stimulation within peripheral organs to precisely activate autonomic nerve circuits. The concept is demonstrated by modulating two different (and potentially therapeutic) targets in animal models, a neuroimmune connection in the spleen (that modulates blood cytokine concentrations) and a nutrient sensory pathway within the liver (that modulates metabolism). Connected to this work is a companion Nature Communications publication that utilizes an ultrasound stimulus focused on the spleen to reduce disease severity in a serum-transferred rodent model of inflammatory arthritis. These reports highlight the growing evidence that ultrasound energy (previously shown to enable activation or modulation of central nervous system pathways) may be used to perform peripheral neuromodulation. In this commentary, we highlight the main findings and discuss their implications for new forms of ultrasound-based therapy. Though challenges remain, a new noninvasive method for precision neuromodulation could solve many of the challenges facing the nascent field of bioelectronic medicine. That is, the use of ultrasound to directly modulate neurophysiological systems therapeutically may provide alternatives to traditional pharmaceuticals. However, to alter the current pharmaceutical paradigm, the field will need to develop a new understanding of how traditional drug concepts (such as dose and pharmacokinetics-pharmacodynamics) relate to the parameters, protocols, and outcomes of this new stimulation technology.


Asunto(s)
Terapia por Ultrasonido , Vías Aferentes , Animales , Citocinas , Bazo , Ultrasonografía
14.
ACS Biomater Sci Eng ; 4(5): 1900-1907, 2018 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-33445345

RESUMEN

Microelectrode arrays of carbon nanotube (CNT)/carbon composite posts with high aspect ratio and millimeter-length were fabricated using carbon-nanotube-templated microfabrication with a sacrificial "hedge". The high aspect ratio, mechanical robustness, and electrical conductivity of these electrodes make them a potential candidate for next-generation neural interfacing. Electrochemical measurements were also demonstrated using an individual CNT post microelectrode with a diameter of 25 µm and a length of 1 mm to perform cyclic voltammetry on both methyl viologen and dopamine in a phosphate-buffered saline solution. In addition to detection of the characteristic peaks, the CNT post microelectrodes show a fast electrochemical response, which may be enabling for in vivo and/or in vitro measurements. The CNT post electrode fabrication process was also integrated with other microfabrication techniques, resulting in individually addressable electrodes.

15.
Lab Chip ; 18(23): 3703, 2018 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-30420988

RESUMEN

Correction for 'Unsupervised capture and profiling of rare immune cells using multi-directional magnetic ratcheting' by Coleman Murray et al., Lab Chip, 2018, 18, 2396-2409.

16.
Lab Chip ; 18(16): 2396-2409, 2018 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-30039125

RESUMEN

Immunotherapies (IT) require induction, expansion, and maintenance of specific changes to a patient's immune cell repertoire which yield a therapeutic benefit. Recently, mechanistic understanding of these changes at the cellular level has revealed that IT results in complex phenotypic transitions in target cells, and that therapeutic effectiveness may be predicted by monitoring these transitions during therapy. However, monitoring will require unique tools that enable capture, manipulation, and profiling of rare immune cell populations. In this study, we introduce a method of automated and unsupervised separation and processing of rare immune cells, using high-force and multidimensional magnetic ratcheting (MR). We demonstrate capture of target immune cells using samples with up to 1 : 10 000 target cell to background cell ratios from input volumes as small as 25 microliters (i.e. a low volume and low cell frequency sample sparing assay interface). Cell capture is shown to achieve up to 90% capture efficiency and purity, and captured cell analysis is shown using both on-chip culture/activity assays and off-chip ejection and nucleic acid analysis. These results demonstrate that multi-directional magnetic ratcheting offers a unique separation system for dealing with blood cell samples that contain either rare cells or significantly small volumes, and the "sample sparing" capability leads to an expanded spectrum of parameters that can be measured. These tools will be paramount to advancing techniques for immune monitoring under conditions in which both the sample volume and number of antigen-specific target cells are often exceedingly small, including during IT and treatment of allergy, asthma, autoimmunity, immunodeficiency, cell based therapy, transplantation, and infection.


Asunto(s)
Separación Celular/instrumentación , Sistema Inmunológico/citología , Campos Magnéticos , Citocinas/metabolismo , Humanos
17.
SLAS Technol ; 22(4): 425-430, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-27864340

RESUMEN

We present a mesodissection platform that retains the advantages of laser-based dissection instrumentation with the speed and ease of manual dissection. Tissue dissection in clinical laboratories is often performed by manually scraping a physician-selected region from standard glass slide mounts. In this manner, costs associated with dissection remain low, but spatial resolution is compromised. In contrast, laser microdissection methods maintain spatial resolution that matches the requirements for analysis of important tissue heterogeneity but remains costly and labor intensive. We demonstrate a microfluidic tool for rapid extraction of histological regions of interest from formalin-fixed paraffin-embedded tissue, which uses a simple and automated method that is compatible with most downstream enzymatic reactions, including protocols used for next-generation DNA sequencing.


Asunto(s)
Disección/métodos , Microfluídica/métodos , Técnicas de Diagnóstico Molecular/métodos , Neoplasias/diagnóstico , Patología Molecular/métodos , Automatización de Laboratorios , Disección/instrumentación , Humanos , Microfluídica/instrumentación , Patología Molecular/instrumentación
18.
Lab Chip ; 12(18): 3341-7, 2012 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-22842841

RESUMEN

In this article we present a novel droplet microfluidic chip enabling amplification-free detection of single pathogenic cells. The device streamlines multiple functionalities to carry out sample digitization, cell lysis, probe-target hybridization for subsequent fluorescent detection. A peptide nucleic acid fluorescence resonance energy transfer probe (PNA beacon) is used to detect 16S rRNA present in pathogenic cells. Initially the sensitivity and quantification abilities of the platform are tested using a synthetic target mimicking the actual expression level of 16S rRNA in single cells. The capability of the device to perform "sample-to-answer" pathogen detection of single cells is demonstrated using E. coli as a model pathogen.


Asunto(s)
Escherichia coli/aislamiento & purificación , Microfluídica/métodos , Escherichia coli/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Microfluídica/instrumentación , Hibridación de Ácido Nucleico , Ácidos Nucleicos de Péptidos/química , Ácidos Nucleicos de Péptidos/metabolismo , ARN Ribosómico 16S/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA