Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Adv Healthc Mater ; 13(13): e2304371, 2024 05.
Artigo em Inglês | MEDLINE | ID: mdl-38320209

RESUMO

Leukemia circulates in the bloodstream and induces various symptoms and complications. Occasionally, these cells accumulate in non-marrow tissues, forming a tumor-like myeloid sarcoma (MS). When the blast-stage leukemia cells invade the brain parenchyma, intracranial MS occurs, leading to a challenging prognosis owing to the limited penetration of cytostatic drugs into the brain and the development of drug resistance. The scarcity of tissue samples from MS makes understanding the phenotypic changes occurring in leukemia cells within the brain environment challenging, thereby hindering development of effective treatment strategies for intracranial MS. This study presents a novel 3D in vitro model mimicking intracranial MS, employing a hydrogel scaffold derived from the brain-decellularized extracellular matrix in which suspended leukemia cells are embedded, simulating the formation of tumor masses in the brain parenchyma. This model reveals marked phenotypic changes in leukemia cells, including altered survival, proliferation, differentiation, and cell cycle regulation. Notably, proportion of dormant leukemia stem cells increases and expression of multidrug resistance genes is upregulated, leading to imatinib resistance, mirroring the pathological features of in vivo MS tissue. Furthermore, suppression of ferroptosis is identified as an important characteristic of intracranial MS, providing valuable insights for the development of targeted therapeutic strategies.


Assuntos
Encéfalo , Matriz Extracelular , Sarcoma Mieloide , Humanos , Encéfalo/patologia , Encéfalo/metabolismo , Linhagem Celular Tumoral , Sarcoma Mieloide/metabolismo , Sarcoma Mieloide/patologia , Matriz Extracelular/metabolismo , Resistencia a Medicamentos Antineoplásicos , Proliferação de Células/efeitos dos fármacos , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/metabolismo , Fenótipo , Hidrogéis/química , Alicerces Teciduais/química , Diferenciação Celular/efeitos dos fármacos , Animais , Ferroptose/efeitos dos fármacos
2.
Small Methods ; 8(5): e2301428, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38161256

RESUMO

Mitigating sepsis-induced severe organ dysfunction with magnetic nanoparticles has shown remarkable advances in extracorporeal blood treatment. Nevertheless, treating large septic animals remains challenging due to insufficient magnetic separation at rapid blood flow rates (>6 L h-1) and limited incubation time in an extracorporeal circuit. Herein, superparamagnetic nanoclusters (SPNCs) coated with red blood cell (RBC) membranes are developed, which promptly capture and magnetically separate a wide range of pathogens at high blood flow rates in a swine sepsis model. The SPNCs exhibited an ultranarrow size distribution of clustered iron oxide nanocrystals and exceptionally high saturation magnetization (≈ 90 emu g-1) close to that of bulk magnetite. It is also revealed that CD47 on the RBCs allows the RBC-SPNCs to remain at a consistent concentration in the blood by evading innate immunity. The uniform size distribution of the RBC-SPNCs greatly enhances their effectiveness in eradicating various pathogenic materials in extracorporeal blood. The use of RBC-SPNCs for extracorporeal treatment of swine infected with multidrug-resistant E. coli is validated and found that severe bacteremic sepsis-induced organ dysfunction is significantly mitigated after 12 h. The findings highlight the potential application of RBC-SPNCs for extracorporeal therapy of severe sepsis in large animal models and potentially humans.


Assuntos
Nanopartículas de Magnetita , Sepse , Animais , Sepse/terapia , Suínos , Nanopartículas de Magnetita/química , Eritrócitos , Insuficiência de Múltiplos Órgãos/terapia , Insuficiência de Múltiplos Órgãos/prevenção & controle , Modelos Animais de Doenças , Infecções por Escherichia coli/terapia , Nanopartículas Magnéticas de Óxido de Ferro/química , Escherichia coli
3.
Adv Sci (Weinh) ; 10(27): e2300164, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37525340

RESUMO

Several stomach diseases are attributed to the dysregulation of physiological function of gastric mucosal barrier by pathogens. Gastric organoids are a promising tool to develop treatment strategies for gastric infections. However, their functional features of in vivo gastric mucosal barrier and host-microbe interactions are limited due to the lack of physiological stimuli. Herein, a human stomach micro-physiological system (hsMPS) with physiologically relevant gastric mucosal defense system is described based on the combination of organoid and MPS technology. A fluid flow enhanced epithelial-mesenchymal interaction in the hsMPS enables functional maturation of gastric epithelial cells, which allows for the recreation of mesh-like mucus layer containing high level of mucus protective peptides and well-developed epithelial junctional complexes. Furthermore, gastroprotection mechanisms against Helicobacter pylori (H. pylori) are successfully demonstrated in this system. Therefore, hsMPS represents a new in vitro tool for research where gastric mucosal defense mechanism is pivotal for developing therapeutic strategies.


Assuntos
Mucosa , Estômago , Humanos , Células Epiteliais , Organoides , Mecanismos de Defesa
4.
Adv Mater ; 35(25): e2211149, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37052392

RESUMO

Autologous implantable scaffolds that induce vasculogenesis have shown great potential in tissue regeneration; however, previous attempts mainly relied on cell-laden hydrogel patches using fat tissues or platelet-rich plasma, which are insufficient for generating a uniform vasculature in a scalable manner. Here, implantable vascularized engineered thrombi (IVETs) are presented using autologous whole blood, which potentiate effective skin wound healing by constructing robust microcapillary vessel networks at the wound site. Microfluidic shear stresses enable the alignment of bundled fibrin fibers along the direction of the blood flow streamlines and the activation of platelets, both of which offer moderate stiffness of the microenvironment optimal for facilitating endothelial cell maturation and vascularization. Rodent dorsal skin wounds patched with IVET present superior wound closure rates (96.08 ± 1.58%), epidermis thickness, collagen deposition, hair follicle numbers, and neutrophil infiltration, which are permitted by enhanced microvascular circulation. Moreover, IVET treatment accelerates wound healing by recruiting M2 phenotype macrophages.


Assuntos
Fibrina , Trombose , Humanos , Cicatrização , Colágeno , Hidrogéis , Alicerces Teciduais , Pele
5.
Tissue Eng Regen Med ; 20(3): 341-353, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37079198

RESUMO

BACKGOUND: Considering the important role of the Peyer's patches (PPs) in gut immune balance, understanding of the detailed mechanisms that control and regulate the antigens in PPs can facilitate the development of immune therapeutic strategies against the gut inflammatory diseases. METHODS: In this review, we summarize the unique structure and function of intestinal PPs and current technologies to establish in vitro intestinal PP system focusing on M cell within the follicle-associated epithelium and IgA+ B cell models for studying mucosal immune networks. Furthermore, multidisciplinary approaches to establish more physiologically relevant PP model were proposed. RESULTS: PPs are surrounded by follicle-associated epithelium containing microfold (M) cells, which serve as special gateways for luminal antigen transport across the gut epithelium. The transported antigens are processed by immune cells within PPs and then, antigen-specific mucosal immune response or mucosal tolerance is initiated, depending on the response of underlying mucosal immune cells. So far, there is no high fidelity (patho)physiological model of PPs; however, there have been several efforts to recapitulate the key steps of mucosal immunity in PPs such as antigen transport through M cells and mucosal IgA responses. CONCLUSION: Current in vitro PP models are not sufficient to recapitulate how mucosal immune system works in PPs. Advanced three-dimensional cell culture technologies would enable to recapitulate the function of PPs, and bridge the gap between animal models and human.


Assuntos
Antígenos , Nódulos Linfáticos Agregados , Animais , Humanos , Imunidade nas Mucosas , Imunoglobulina A
6.
ACS Nano ; 17(9): 8153-8166, 2023 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-37068137

RESUMO

Blood-brain barrier (BBB) remains one of the critical challenges in developing neurological therapeutics. Short single-stranded DNA/RNA nucleotides forming a three-dimensional structure, called aptamers, have received increasing attention as BBB shuttles for efficient brain drug delivery owing to their practical advantages over Trojan horse antibodies or peptides. Aptamers are typically obtained by combinatorial chemical technology, termed Systemic Evolution of Ligands by EXponential Enrichment (SELEX), against purified targets, living cells, or animal models. However, identifying reliable BBB-penetrating aptamers that perform efficiently under human physiological conditions has been challenging because of the poor physiological relevance in the conventional SELEX process. Here, we report a human BBB shuttle aptamer (hBS) identified using a human microphysiological system (MPS)-based SELEX (MPS-SELEX) method. A two-channel MPS lined with human brain microvascular endothelial cells (BMECs) interfaced with astrocytes and pericytes, recapitulating high-level barrier function of in vivo BBB, was exploited as a screening platform. The MPS-SELEX procedure enabled robust function-based screening of the hBS candidates, which was not achievable in traditional in vitro BBB models. The identified aptamer (hBS01) through five-round of MPS-SELEX exhibited high capability to transport protein cargoes across the human BBB via clathrin-mediated endocytosis and enhanced uptake efficiency in BMECs and brain cells. The enhanced targeting specificity of hBS01 was further validated both in vitro and in vivo, confirming its powerful brain accumulation efficiency. These findings demonstrate that MPS-SELEX has potential in the discovery of aptamers with high target specificity that can be widely utilized to boost the development of drug delivery strategies.


Assuntos
Aptâmeros de Nucleotídeos , Animais , Humanos , Aptâmeros de Nucleotídeos/química , Células Endoteliais/metabolismo , Barreira Hematoencefálica/metabolismo , Sistemas Microfisiológicos , Técnica de Seleção de Aptâmeros/métodos , Ligantes
7.
Small ; 18(40): e2203746, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36070419

RESUMO

Bloodstream infection caused by antimicrobial resistance pathogens is a global concern because it is difficult to treat with conventional therapy. Here, scavenger magnetic nanoparticles enveloped by nanovesicles derived from blood cells (MNVs) are reported, which magnetically eradicate an extreme range of pathogens in an extracorporeal circuit. It is quantitatively revealed that glycophorin A and complement receptor (CR) 1 on red blood cell (RBC)-MNVs predominantly capture human fecal bacteria, carbapenem-resistant (CR) Escherichia  coli, and extended-spectrum beta-lactamases-positive (ESBL-positive) E. coli, vancomycin-intermediate Staphylococcus aureus (VISA), endotoxins, and proinflammatory cytokines in human blood. Additionally, CR3 and CR1 on white blood cell-MNVs mainly contribute to depleting the virus envelope proteins of Zika, SARS-CoV-2, and their variants in human blood. Supplementing opsonins into the blood significantly augments the pathogen removal efficiency due to its combinatorial interactions between pathogens and CR1 and CR3 on MNVs. The extracorporeal blood cleansing enables full recovery of lethally infected rodent animals within 7 days by treating them twice in series. It is also validated that parameters reflecting immune homeostasis, such as blood cell counts, cytokine levels, and transcriptomics changes, are restored in blood of the fatally infected rats after treatment.


Assuntos
Bacteriemia , Tratamento Farmacológico da COVID-19 , Infecções por Escherichia coli , Animais , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Bacteriemia/tratamento farmacológico , Bacteriemia/microbiologia , Carbapenêmicos/metabolismo , Citocinas/metabolismo , Endotoxinas/metabolismo , Escherichia coli/metabolismo , Infecções por Escherichia coli/tratamento farmacológico , Infecções por Escherichia coli/microbiologia , Glicoforinas/metabolismo , Homeostase , Humanos , Testes de Sensibilidade Microbiana , Proteínas Opsonizantes/metabolismo , Ratos , Receptores de Complemento/metabolismo , Roedores/metabolismo , SARS-CoV-2 , Proteínas do Envelope Viral/metabolismo , beta-Lactamases/metabolismo
8.
Microbiol Spectr ; 10(4): e0086422, 2022 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-35862959

RESUMO

Proper selection of susceptible antibiotics in drug-resistant bacteria is critical to treat bloodstream infection. Although biomarkers that guide antibiotic therapy have been extensively evaluated, little is known about host biomarkers targeting in vivo antibiotic susceptibility. Therefore, we aimed to evaluate the trends of hemodynamics and biomarkers in a porcine bacteremia model treated with insusceptible antibiotics compared to those in susceptible models. Extended-spectrum ß-lactamase (ESBL)-producing Escherichia coli (E. coli, 5.0 * 10^9 CFU) was intravenously administered to 11 male pigs. One hour after bacterial infusion, pigs were assigned to two groups of antibiotics, ceftriaxone (n = 6) or ertapenem (n = 5). Pigs were monitored up to 7 h after bacterial injection with fluid and vasopressor support to maintain the mean arterial blood pressure over 65 mmHg. Blood sampling for blood culture and plasma acquisition was performed before and every predefined hour after E. coli injection. Cytokine (tumor necrosis factor-α, interleukin [IL]-1ß, IL-6, IL-8, IL-10, C-reactive protein, procalcitonin, presepsin, heparan sulfate, syndecan, and soluble triggering receptor expressed on myeloid cells-1 [sTREM-1]) levels in plasma were analyzed using enzyme-linked immunosorbent assays. Bacteremia developed after intravenous injection of E. coli, and negative conversion was confirmed only in the ertapenem group. While trends of other biomarkers failed to show differences, the trend of sTREM-1 was significantly different between the two groups (P = 0.0001, two-way repeated measures analysis of variance). Among hemodynamics and biomarkers, the sTREM-1 level at post 2 h after antibiotics administration represented a significant difference depending on susceptibility, which can be suggested as a biomarker candidate of in vivo antibiotics susceptibility. Further clinical studies are warranted for validation. IMPORTANCE Early and appropriate antibiotic treatment is a keystone in treating patients with sepsis. Despite its importance, blood culture which requires a few days remains as a pillar of diagnostic method for microorganisms and their antibiotic susceptibility. Whether changes in biomarkers and hemodynamics indicate treatment response of susceptible antibiotic compared to resistant one is not well understood to date. In this study using extended-spectrum ß-lactamase -producing E. coli bacteremia porcine model, we have demonstrated the comprehensive cardiovascular hemodynamics and trends of plasma biomarkers in sepsis and compared them between two groups with susceptible and resistant antibiotics. While other hemodynamics and biomarkers have failed to differ, we have identified that levels of soluble triggering receptor expressed on myeloid cells-1 (sTREM-1) significantly differed between the two groups over time. Based on the data in this study, trends of sTREM-1 obtained before the antibiotics and 2~4 h after the antibiotics could be a novel host biomarker that triggers the step-up choice of antibiotics.


Assuntos
Bacteriemia , Sepse , Animais , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Bacteriemia/tratamento farmacológico , Bacteriemia/microbiologia , Biomarcadores , Ertapenem/uso terapêutico , Escherichia coli , Hemodinâmica , Masculino , Sepse/tratamento farmacológico , Suínos , Receptor Gatilho 1 Expresso em Células Mieloides , beta-Lactamases
9.
Small Methods ; 6(3): e2101239, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35112812

RESUMO

The current diagnosis of bacteremia mainly relies on blood culture, which is inadequate for the rapid and quantitative determination of most bacteria in blood. Here, a quantitative, multiplex, microfluidic fluorescence in situ hybridization method (µFISH) is developed, which enables early and rapid (3-h) diagnosis of bacteremia without the need for prior blood culture. This novel technology employs mannose-binding lectin-coated magnetic nanoparticles, which effectively opsonize a broad range of pathogens, magnetically sequestering them in a microfluidic device. Therein, µFISH probes, based on unique 16S rRNA sequences, enable the identification and quantification of sequestered pathogens both in saline and whole blood, which is more sensitive than conventional polymerase chain reaction. Using µFISH, Escherichia coli (E. coli) is detected in whole blood collected from a porcine disease model and from E. coli-infected patients. Moreover, the proportion of E. coli, relative to other bacterial levels in the blood, is accurately and rapidly determined, which is not possible using conventional diagnostic methods. Blood from E. coli-infected patients is differentiated from healthy donors' blood using cutoff values with a 0.05 significance level. Thus, µFISH is a versatile method that can be used to rapidly identify pathogens and determine their levels relative to other culturable and nonculturable bacteria in biological samples.


Assuntos
Bacteriemia , Infecções por Escherichia coli , Animais , Bacteriemia/diagnóstico , Bactérias , Escherichia coli/genética , Infecções por Escherichia coli/diagnóstico , Humanos , Hibridização in Situ Fluorescente/métodos , RNA Ribossômico 16S/genética , Suínos
10.
FASEB J ; 35(10): e21906, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34490940

RESUMO

Glioblastoma (GBM) is a refractory disease that has a highly infiltrative characteristic. Over the past decade, GBM perivascular niche (PVN) has been described as a route of dissemination. Here, we investigated that trailed membrane structures, namely retraction fibers (RFs), are formed by perivascular extracellular matrix (ECM) proteins. By using the anatomical GBM database, we validated that the ECM-related genes were highly expressed in the cells within the PVN where fibronectin (FN) induced RF formation. By disrupting candidates of FN-binding integrins, integrin α5ß1 was identified as the main regulator of RF formation. De novo RFs were produced at the trailing edge, and focal adhesions were actively localized in RFs, indicating that adhesive force makes RFs remain at the bottom surface. Furthermore, we observed that GBM cells more frequently migrated along the residual RFs formed by preceding cells in microfluidic channels in comparison to those in the channels without RFs, suggesting that the infiltrative characteristics GBM could be attributed to RFs formed by the preceding cells in concert with chemoattractant cues. Altogether, we demonstrated that shedding membrane structures of GBM cells are maintained by FN-integrin α5ß1 interaction and promoted their motility .


Assuntos
Neoplasias Encefálicas/metabolismo , Movimento Celular , Fibronectinas/metabolismo , Glioblastoma/metabolismo , Proteínas de Neoplasias/metabolismo , Receptores de Vitronectina/metabolismo , Animais , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Matriz Extracelular/metabolismo , Matriz Extracelular/patologia , Feminino , Glioblastoma/patologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus
11.
Biofabrication ; 13(4)2021 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-34479224

RESUMO

The properties of a semipermeable porous membrane, including pore size, pore density, and thickness, play a crucial role in creating a tissue interface in a microphysiological system (MPS) because it dictates multicellular interactions between different compartments. The small pore-sized membrane has been preferentially used in an MPS for stable cell adhesion and the formation of tissue barriers on the membrane. However, it limited the applicability of the MPS because of the hindered cell transmigration via sparse through-holes and the optical translucence caused by light scattering through pores. Thus, there remain unmet challenges to construct a compartmentalized MPS without those drawbacks. Here we report a submicrometer-thickness (∼500 nm) fibrous extracellular matrix (ECM) film selectively condensed on a large pore-sized track-etched (TE) membrane (10µm-pores) in an MPS device, which enables the generation of functional tissue barriers simultaneously achieving optical transparency, intercellular interactions, and transmigration of cells across the membrane. The condensed ECM fibers uniformly covering the surface and 10µm-pores of the TE membrane permitted sufficient surface areas where a monolayer of the human induced pluripotent stem cell-derived brain endothelial cells is formed in the MPS device. The functional maturation of the blood-brain barrier (BBB) was proficiently achieved due to astrocytic endfeet sheathing the brain endothelial cells through 10µm pores of the condensed-ECM-coated TE (cECMTE) membrane. We also demonstrated the extravasation of human metastatic breast tumor cells through the human BBB on the cECMTE membrane. Thus, the cECMTE membrane integrated with an MPS can be used as a versatile platform for studying various intercellular communications and migration, mimicking the physiological barriers of an organ compartment.


Assuntos
Comunicação Celular , Células Endoteliais , Matriz Extracelular , Humanos , Células-Tronco Pluripotentes Induzidas , Tomografia por Emissão de Pósitrons
12.
Front Cell Infect Microbiol ; 11: 676650, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34527598

RESUMO

Recent studies have suggested the existence of a blood microbiome in the healthy host. However, changes in the blood microbiome upon bloodstream infection are not known. Here, we analyzed the dynamics of the blood microbiome in a porcine model of polymicrobial bacteremia induced by fecal peritonitis. Surprisingly, we detected bacterial populations in the bloodstream even before the infection, and these populations were maintained over time. The native blood microbiome was notably taxonomically different from the fecal microbiome that was used to induce peritonitis, reflecting microbial tropism for the blood. Although the population composition after the infection was similar to that of the native blood microbiome, new bacterial strains entered the bloodstream upon peritonitis induction as clinical symptoms relevant to sepsis developed. This indicates that the bacteria detected in the blood before peritonitis induction were derived from the blood rather than a contamination. Comparison of the functional pathways enriched in the blood and fecal microbiomes revealed that communication and stress management pathways are essential for the survival of the blood microbiome.


Assuntos
Microbioma Gastrointestinal , Microbiota , Peritonite , Animais , Fezes , Suínos , Tropismo
13.
Small ; 17(23): e2100797, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33978996

RESUMO

A hemolysis-free and highly efficient plasma separation platform enabled by enhanced diamagnetic repulsion of blood cells in undiluted whole blood is reported. Complete removal of blood cells from blood plasma is achieved by supplementing blood with superparamagnetic iron oxide nanoparticles (SPIONs), which turns the blood plasma into a paramagnetic condition, and thus, all blood cells are repelled by magnets. The blood plasma is successfully collected from 4 mL of blood at flow rates up to 100 µL min-1 without losing plasma proteins, platelets, or exosomes with 83.3±1.64% of plasma volume recovery, which is superior over the conventional microfluidic methods. The theoretical model elucidates the diamagnetic repulsion of blood cells considering hematocrit-dependent viscosity, which allows to determine a range of optimal flow rates to harvest platelet-rich plasma and platelet-free plasma. For clinical validations, it is demonstrated that the method enables the greater recovery of bacterial DNA from the infected blood than centrifugation and the immunoassay in whole blood without prior plasma separation.


Assuntos
Células Sanguíneas , Plasma , Biomarcadores , Separação Celular , Hemólise , Humanos , Microfluídica
14.
Nat Commun ; 11(1): 5221, 2020 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-33060604

RESUMO

Hydrodynamic interactions play a role in synchronized motions of coupled oscillators in fluids, and understanding the mechanism will facilitate development of applications in fluid mechanics. For example, synchronization phenomenon in two-phase flow will benefit the design of future microfluidic devices, allowing spatiotemporal control of microdroplet generation without additional integration of control elements. In this work, utilizing a characteristic oscillation of adjacent interfaces between two immiscible fluids in a microfluidic platform, we discover that the system can act as a coupled oscillator, notably showing spontaneous in-phase synchronization of droplet breakup. With this observation of in-phase synchronization, the coupled droplet generator exhibits a complete set of modes of coupled oscillators, including out-of-phase synchronization and nonsynchronous modes. We present a theoretical model to elucidate how a negative feedback mechanism, tied to the distance between the interfaces, induces the in-phase synchronization. We also identify the criterion for the transition from in-phase to out-of-phase oscillations.

15.
Biosens Bioelectron ; 168: 112558, 2020 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-32911451

RESUMO

Recruitment of circulating leukocytes to sites of infection is of utmost importance in the development, propagation, and outcome of sepsis. These multi-step processes are mediated by interactions between adhesion receptors of leukocytes and cell adhesion molecules (CAMs) of endothelial cells, such as P-selectin, E-selectin and ICAM-1. However, the potential utility of the CAMs-facilitated leukocyte capture has not been thoroughly investigated as an index of the host response to infection for diagnostic purposes. Here, we report that the systemic infection affects the expression of CAMs ligands on leukocytes, upregulating the expression of P-selectin ligand-1 (PSGL-1) and increasing the number of PSGL-1- and E-selectin ligand-1 (ESL-1)-expressing leukocyte levels in septic blood. We leveraged this finding to determine infection by measuring the increased adhesion of leukocytes to an inflammatory vascular endothelium-mimicking microchannel coated with CAMs. We successfully validated that the proposed method can significantly differentiate infection in bacteremia and endotoxemia models in rats as early as an hour post-infection using a finger-prick volume of blood (50 µL), which were unachievable with the conventional diagnostic methods.


Assuntos
Técnicas Biossensoriais , Migração e Rolagem de Leucócitos , Animais , Adesão Celular , Células Endoteliais , Endotélio Vascular , Dispositivos Lab-On-A-Chip , Leucócitos , Selectina-P , Ratos
16.
Biochip J ; 14(1): 63-71, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32218896

RESUMO

Infectious diseases and their pandemics periodically attract public interests due to difficulty in treating the patients and the consequent high mortality. Sepsis caused by an imbalanced systemic inflammatory response to infection often leads to organ failure and death. The current therapeutic intervention mainly includes "the sepsis bundles," antibiotics (antibacterial, antiviral, and antifungal), intravenous fluids for resuscitation, and surgery, which have significantly improved the clinical outcomes in past decades; however, the patients with fulminant sepsis are still in desperate need of alternative therapeutic approaches. One of the potential supportive therapies, extracorporeal blood treatment, has emerged and been developed for improving the current therapeutic efficacy. Here, I overview how the treatment of infectious diseases has been assisted with the extracorporeal adjuvant therapy and the potential utility of various nanobiotechnology and microfluidic approaches for developing new auxiliary therapeutic methods.

17.
Lab Chip ; 19(21): 3706-3713, 2019 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-31577312

RESUMO

Here, we report a simple yet reliable method for bonding poly(methyl methacrylate) (PMMA) to polyethylene terephthalate (PETE) track-etched membranes using (3-glycidyloxypropyl)trimethoxysilane (GLYMO), which enables reliable cytotoxicity tests in a microfluidic device impermeable to small molecules, such as anti-cancer drugs. The porous PETE membranes treated with 5% GLYMO were assembled with microfluidic channel-engraved PMMA substrates after air plasma treatment for 1 minute, followed by heating at 100 °C for 2 minutes, which permits irreversible and complete bonding to be achieved within 1 h. The bonding strength between the two substrates (1.97 × 107 kg m-2) was robust enough to flow culture medium through the device without leakage even at a gauge pressure of above 135 kPa. For validation of its utility in drugs testing, we successfully demonstrated that human lung adenocarcinoma cells cultured in the PMMA devices show more reliable cytotoxicity results for vincristine in comparison to conventional polydimethylsiloxane (PDMS) devices due to the inherent property of PMMA of it being impervious to small molecules. Given that the current organ-on-a-chip fabrication methods mostly rely on PDMS, this bonding strategy will expand simple fabrication capability using various thermoplastics and porous track-etched membranes, and allow us to create 3D-micro-constructs that more precisely mimic organ-level physiological conditions.


Assuntos
Células Endoteliais da Veia Umbilical Humana/metabolismo , Dispositivos Lab-On-A-Chip , Membranas Artificiais , Polietilenotereftalatos/química , Polimetil Metacrilato/química , Células A549 , Avaliação Pré-Clínica de Medicamentos , Células Endoteliais da Veia Umbilical Humana/citologia , Humanos , Porosidade
18.
Lab Chip ; 19(14): 2356-2361, 2019 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-31173624

RESUMO

Herein, we described a microfluidic device that enabled the measurement of magnetic susceptibility of a subtle paramagnetic solution that could not be determined by conventional magnetic susceptibility measurement methods such as a superconducting quantum interference device (SQUID). We measured the diamagnetic repelling velocity of polystyrene microparticles suspended in a weak paramagnetic solution including 50-150 mM of gadolinium-diethylenetriamine penta-acetic acid (GD-DTPA) and superparamagnetic iron oxide nanoparticles (SPION) (10 nm in diameter) dispersed in distilled water at various concentrations. The measured diamagnetic repelling velocities correlated with our prediction and also with the magnetic susceptibility of the SPION solution measured by a SQUID magnetometer. Via this approach, we could determine the magnetic susceptibility of the subtle paramagnetic solution of GD-DTPA that could not be measured via the conventional method. This study provides us with the new capability of analyzing weakly paramagnetic solutions such as paramagnetic components or metal contaminants present in environmental liquid samples at low concentrations.

19.
RSC Adv ; 9(41): 23791-23796, 2019 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-35530621

RESUMO

We present that enhanced simultaneous incubation of multiple antibodies (Abs) can be achieved by exploiting microfluidic laminar flows and difference in diffusivity between primary Ab (pAb) and secondary Ab (sAb). We demonstrate that injecting Ab of larger and smaller diffusivity (D Ab) in an upper and lower laminar flow over the analyte-coated bottom surface, respectively, would result in enhanced signal intensity in the given reaction time. To prove this, we simultaneously infused anti-prostate specific antigen (PSA) pAb (upper laminar flow) and quantum dot (QD) labeled secondary Ab (QD-sAb) (lower laminar flow) to generate two Ab laminar flows vertically sheathing each other in the microfluidic device in which PSA was immobilized on the glass bottom surface. Because of the larger D Ab of pAb than that of QD-sAb due to the heavy metal components of QD, anti-PSA pAb diffuses more rapidly toward the bottom surface where the immune reaction between PSA, pAb, and QD-sAb instantaneously occurs. We corroborated our principle by switching the position of the two Ab laminar flows (QD-sAb in upper and pAb in lower laminar flows) in the channel, which resulted in significantly lower intensity of QD signals than the previous method. Moreover, when we adjusted the interface of pAb and QD-sAb in upper and lower laminar flows, respectively, closer toward the bottom surface, the fluorescence signal was even more intensified. This is attributed to the increased flux of anti-PSA pAb more adjacent to the reaction site, which, in turn, enhances the binding efficiency of pAb to PSA on the surface.

20.
Small ; 14(34): e1801731, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30044534

RESUMO

A major challenge to scale up a microfluidic magnetic separator for extracorporeal blood cleansing applications is to overcome low magnetic drag velocity caused by viscous blood components interfering with magnetophoresis. Therefore, there is an unmet need to develop an effective method to position magnetic particles to the area of augmented magnetic flux density gradients while retaining clinically applicable throughput. Here, a magnetophoretic cell separation device, integrated with slanted ridge-arrays in a microfluidic channel, is reported. The slanted ridges patterned in the microfluidic channels generate spiral flows along the microfluidic channel. The cells bound with magnetic particles follow trajectories of the spiral streamlines and are repeatedly transferred in a transverse direction toward the area adjacent to a ferromagnetic nickel structure, where they are exposed to a highly augmented magnetic force of 7.68 µN that is much greater than the force (0.35 pN) at the side of the channel furthest from the nickel structure. With this approach, 91.68% ± 2.18% of Escherichia coli (E. coli) bound with magnetic nanoparticles are successfully separated from undiluted whole blood at a flow rate of 0.6 mL h-1 in a single microfluidic channel, whereas only 23.98% ± 6.59% of E. coli are depleted in the conventional microfluidic device.


Assuntos
Sangue/microbiologia , Escherichia coli/isolamento & purificação , Magnetismo/métodos , Reologia/métodos , Fluorescência , Humanos , Dispositivos Lab-On-A-Chip , Lectina de Ligação a Manose/metabolismo , Nanopartículas/química , Rotação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA