Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Sci Rep ; 10(1): 15018, 2020 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-32929172

RESUMO

Cancer is the second cause of death worldwide. This devastating disease requires specific, fast, and affordable solutions to mitigate and reverse this trend. A step towards cancer-fighting lies in the isolation of natural killer (NK) cells, a set of innate immune cells, that can either be used as biomarkers of tumorigenesis or, after autologous transplantation, to fight aggressive metastatic cells. In order to specifically isolate NK cells (which express the surface NKp30 receptor) from peripheral blood mononuclear cells, a ZnO immunoaffinity-based platform was developed by electrodeposition of the metal oxide on a flexible indium tin oxide (ITO)-coated polyethylene terephthalate (PET) substrate. The resulting crystalline and well-aligned ZnO nanorods (NRs) proved their efficiency in immobilizing monoclonal anti-human NKp30 antibodies (mAb), obviating the need for additional procedures for mAb immobilization. The presence of NK cells on the peripheral blood mononuclear cell (PBMCs) fraction was evaluated by the response to their natural ligand (B7-H6) using an acridine orange (AO)-based assay. The successful selection of NK cells from PBMCs by our nanoplatform was assessed by the photoluminescent properties of AO. This easy and straightforward ZnO-mAb nanoplatform paves the way for the design of biosensors for clinic diagnosis, and, due to its inherent biocompatibility, for the initial selection of NK cells for autotransplantation immunotherapies.


Assuntos
Anticorpos Monoclonais/imunologia , Separação Celular/métodos , Células Imobilizadas/citologia , Células Matadoras Naturais/citologia , Nanoconjugados/química , Óxido de Zinco/química , Técnicas Biossensoriais/métodos , Células Imobilizadas/imunologia , Humanos , Células Matadoras Naturais/imunologia , Receptor 3 Desencadeador da Citotoxicidade Natural/imunologia , Polietilenotereftalatos/química , Compostos de Estanho/química
2.
ACS Appl Mater Interfaces ; 11(37): 33716-33724, 2019 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-31454227

RESUMO

Immunotherapy has recently garnered significant research interest in the field of clinical cancer management. The potential of tumor immunotherapy has been demonstrated for targeting a variety of tumors, both in vivo and in vitro, yielding some remarkable therapeutic effects. Herein, inspired by the stem cell niche, we developed a scale-up approach to generating porous microspheres with encapsulated natural killer (NK) cells via microfluidic electrospray for in situ tumor immunotherapy. The generated microspheres contained porous microstructures with tunable morphologies because of versatile but precise fluid control in the microfluidic electrospray system. NK-92MI cells encapsulated in porous microspheres were protected from the outer complex environment, allowing for improved proliferation and functionality. As observed, perforin and granzymes were sustainably secreted from the encapsulated NK-92MI cells, which exhibited robust killing effects on tumors both in vitro and in vivo. With continual proliferation, NK-92MI cells budded from the surface of porous microspheres and migrated into the surrounding residual tumor tissues, further destroying tumor cells. More importantly, no side effects owing to the native host immune system were observed by injecting the NK-92MI cell-encapsulated microspheres into tumors in vivo. Therefore, the NK-cell-encapsulated porous microspheres show great potential for tumor immunotherapy, offering a robust and attractive treatment option for cancer patient management.


Assuntos
Células Imobilizadas , Imunidade Celular , Imunoterapia , Células Matadoras Naturais , Microesferas , Neoplasias Experimentais , Animais , Linhagem Celular Tumoral , Células Imobilizadas/imunologia , Células Imobilizadas/patologia , Células Imobilizadas/transplante , Humanos , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/patologia , Células Matadoras Naturais/transplante , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/terapia , Porosidade
3.
Methods Mol Biol ; 1953: 151-162, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30912021

RESUMO

The cross talk between tumor cells and other cells present in the tumor microenvironment such as stromal and immune cells highly influences the behavior and progression of disease. Understanding the underlying mechanisms of interaction is a prerequisite to develop new treatment strategies and to prevent or at least reduce therapy failure in the future. Specific reactivation of the patient's immune system is one of the major goals today. However, standard two-dimensional (2D) cell culture techniques lack the necessary complexity to address related questions. Novel three-dimensional (3D) in vitro models-embedded in a matrix or encapsulated in alginate-recapitulate the in vivo situation much better. Cross talk between different cell types can be studied starting from co-cultures. As cancer immune modulation is becoming a major research topic, 3D in vitro models represent an important tool to address immune regulatory/modulatory questions for T, NK, and other cells of the immune system. The 3D systems consisting of tumor cells, fibroblasts, and immune cells (3D-3) already proved as a reliable tool for us. For instance, we made use of those models to study the molecular mechanisms of the cross talk of non-small cell lung cancer (NSCLC) and fibroblasts, to unveil macrophage plasticity in the tumor microenvironment and to mirror drug responses in vivo. Generation of those 3D models and how to use them to study immune cell infiltration and activation will be described in the present book chapter.


Assuntos
Técnicas de Cocultura/métodos , Descoberta de Drogas/métodos , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Antineoplásicos/farmacologia , Reatores Biológicos , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/imunologia , Carcinoma Pulmonar de Células não Pequenas/patologia , Células Imobilizadas/efeitos dos fármacos , Células Imobilizadas/imunologia , Células Imobilizadas/patologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/imunologia , Fibroblastos/patologia , Humanos , Imunidade Celular/efeitos dos fármacos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/patologia , Esferoides Celulares/efeitos dos fármacos , Esferoides Celulares/imunologia , Esferoides Celulares/patologia , Células Tumorais Cultivadas , Microambiente Tumoral/efeitos dos fármacos
4.
Bioconjug Chem ; 30(3): 552-560, 2019 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-30779553

RESUMO

We report the generation of magnetically responsive, cord blood-derived natural killer (NK) cells using iron oxide nanoparticles (IONPs). NK cells are a promising immune cell population for cancer cell therapy as they can target and lyse target tumor cells without prior education. However, NK cells cannot home to disease sites based on antigen recognition, instead relying primarily on external stimuli and chemotactic gradients for transport. Hence, we hypothesized that conjugating IONPs onto the surface of NK cells provides an added feature of magnetic homing to the NK cells, improving their therapeutic function. We describe a robust design for conjugating the IONPs onto the surface of NK cells, which maintains their intrinsic phenotype and function. The conferred magnetic-responsiveness is utilized to improve the cytolytic function of the NK cells for target cells in 2D and 3D models. These findings demonstrate the feasibility of improving NK cell homing and therapeutic efficacy with our NK:IONP "biohybrid".


Assuntos
Sangue Fetal/citologia , Células Matadoras Naturais/citologia , Nanopartículas de Magnetita/química , Linhagem Celular Tumoral , Células Cultivadas , Células Imobilizadas/citologia , Células Imobilizadas/imunologia , Células Imobilizadas/transplante , Humanos , Imunoterapia , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/transplante , Nanopartículas de Magnetita/uso terapêutico , Nanomedicina , Neoplasias/imunologia , Neoplasias/terapia
5.
Tissue Eng Part A ; 24(5-6): 448-457, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-28683667

RESUMO

Human perivascular stem/stromal cells (PSC) are a multipotent mesodermal progenitor cell population defined by their perivascular residence. PSC are most commonly derived from subcutaneous adipose tissue, and recent studies have demonstrated the high potential for clinical translation of this fluorescence-activated cell sorting-derived cell population for bone tissue engineering. Specifically, purified PSC induce greater bone formation than unpurified stroma taken from the same patient sample. In this study, we examined the differences in early innate immune response to human PSC or unpurified stroma (stromal vascular fraction [SVF]) during the in vivo process of bone formation. Briefly, SVF or PSC from the same patient sample were implanted intramuscularly in the hindlimb of severe combined immunodeficient (SCID) mice using an osteoinductive demineralized bone matrix carrier. Histological examination of early inflammatory infiltrates was examined by hematoxylin and eosin and immunohistochemical staining (Ly-6G, F4/80). Results showed significantly greater neutrophilic and macrophage infiltrates within and around SVF in comparison to PSC-laden implants. Differences in early postoperative inflammation among SVF-laden implants were associated with reduced osteogenic differentiation and bone formation. Similar findings were recapitulated with PSC implantation in immunocompetent mice. Exaggerated postoperative inflammation was associated with increased IL-1α, IL-1ß, IFN-γ, and TNF-α gene expression among SVF samples, and conversely increased IL-6 and IL-10 expression among PSC samples. These data document a robust immunomodulatory effect of implanted PSC, and an inverse correlation between host inflammatory cell infiltration and stromal progenitor cell-mediated ossification.


Assuntos
Matriz Óssea , Células Imobilizadas , Imunomodulação , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/imunologia , Osteogênese/imunologia , Animais , Matriz Óssea/imunologia , Matriz Óssea/transplante , Células Imobilizadas/imunologia , Células Imobilizadas/transplante , Citocinas/imunologia , Xenoenxertos , Humanos , Camundongos , Camundongos SCID
6.
Anal Chem ; 89(23): 12918-12923, 2017 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-29116752

RESUMO

In this study, we report the label-free and reliable detection of allergic response using a cell-coupled gate field-effect transistor (cell-based FET). Rat basophilic leukemia (RBL-2H3) cells were cultured as a signal transduction interface to induce allergic reaction on the gate oxide surface of the FET, because IgE antibodies, which bind to Fcε receptors at the RBL-2H3 cell membrane, are specifically cross-linked by allergens, resulting in the allergic response of RBL-2H3 cells. In fact, the surface potential at the FET gate decreased owing to secretions such as histamine from the IgE-bound RBL-2H3 cells, which reacted with the allergen. This is because histamine, as one of the candidate secretions, shows basicity, resulting in a change in pH around the cell/gate interface. That is, the RBL-2H3-cell-based FET used in this study was originally from an ion-sensitive FET (ISFET), whose oxide surface (Ta2O5) with hydroxyl groups is fully responsive to pH on the basis of the equilibrium reaction. The allergic response of RBL-2H3 cells on the gate was also confirmed by estimating the amount of ß-hexosaminidase released together with histamine and was analyzed using the electrical properties based on an inflammatory response of secreted histamine with the vascular endothelial cell-based FET. Thus, the allergic responses were monitored in a nonoptical and real-time manner using the cell-based FETs with the cellular layers on the gate, which reproduced the in vivo system and were useful for the reliable detection of the allergic reaction.


Assuntos
Células Imobilizadas/metabolismo , Técnicas Eletroquímicas/métodos , Hipersensibilidade/diagnóstico , Transistores Eletrônicos , Animais , Linhagem Celular Tumoral , Células Imobilizadas/imunologia , Técnicas Eletroquímicas/instrumentação , Histamina/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Concentração de Íons de Hidrogênio , Imunoglobulina E/imunologia , Óxidos/química , Ratos , Tantálio/química
7.
Sci Rep ; 7(1): 10059, 2017 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-28855611

RESUMO

Pericapsular fibrotic overgrowth (PFO) is associated with poor survival of encapsulated islets. A strategy to combat PFO is the use of mesenchymal stem cells (MSC). MSC have anti-inflammatory properties and their potential can be enhanced by stimulation with proinflammatory cytokines. This study investigated whether co-encapsulation or co-transplantation of MSC with encapsulated islets would reduce PFO and improve graft survival. Stimulating MSC with a cytokine cocktail of IFN-γ and TNF-α enhanced their immunosuppressive potential by increasing nitric oxide production and secreting higher levels of immunomodulatory cytokines. In vitro, co-encapsulation with MSC did not affect islet viability but significantly enhanced glucose-induced insulin secretion. In vivo, normoglycemia was achieved in 100% mice receiving islets co-encapsulated with stimulated MSC as opposed to 71.4% receiving unstimulated MSC and only 9.1% receiving encapsulated islets alone. Microcapsules retrieved from both unstimulated and stimulated MSC groups had significantly less PFO with improved islet viability and function compared to encapsulated islets alone. Levels of peritoneal immunomodulatory cytokines IL-4, IL-6, IL-10 and G-CSF were significantly higher in MSC co-encapsulated groups. Similar results were obtained when encapsulated islets and MSC were co-transplanted. In summary, co-encapsulation or co-transplantation of MSC with encapsulated islets reduced PFO and improved the functional outcome of allotransplants.


Assuntos
Composição de Medicamentos/métodos , Sobrevivência de Enxerto/fisiologia , Transplante das Ilhotas Pancreáticas/métodos , Ilhotas Pancreáticas/metabolismo , Transplante de Células-Tronco Mesenquimais/métodos , Alginatos/química , Animais , Células Imobilizadas/citologia , Células Imobilizadas/efeitos dos fármacos , Células Imobilizadas/imunologia , Citocinas/genética , Citocinas/imunologia , Feminino , Fibrose/prevenção & controle , Expressão Gênica , Insulina/biossíntese , Interferon gama/farmacologia , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/imunologia , Masculino , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Transplante Homólogo , Fator de Necrose Tumoral alfa/farmacologia
8.
Methods Mol Biol ; 1634: 173-180, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28819850

RESUMO

The RareCyte CyteFinder instrument is an automated scanner that allows rapid identification of circulating tumor cells (CTCs) on microscope slides prepared by the AccuCyte process (see Chapter 13 ) and stained by immunofluorescence. Here, we present the workflow for CyteFinder scanning, analysis, and CyteMapper scan review which includes CTC confirmation and report generation.


Assuntos
Separação Celular/métodos , Células Imobilizadas/patologia , Processamento de Imagem Assistida por Computador/métodos , Neoplasias/diagnóstico , Células Neoplásicas Circulantes/patologia , Análise de Célula Única/métodos , Anticorpos Monoclonais/química , Anticorpos Monoclonais/metabolismo , Automação Laboratorial/instrumentação , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/imunologia , Biomarcadores Tumorais/metabolismo , Contagem de Células , Linhagem Celular Tumoral , Separação Celular/instrumentação , Células Imobilizadas/imunologia , Células Imobilizadas/metabolismo , Centrifugação/instrumentação , Centrifugação/métodos , Molécula de Adesão da Célula Epitelial/genética , Molécula de Adesão da Célula Epitelial/imunologia , Molécula de Adesão da Célula Epitelial/metabolismo , Desenho de Equipamento , Imunofluorescência/métodos , Corantes Fluorescentes/química , Humanos , Imunoconjugados/química , Queratinas/genética , Queratinas/imunologia , Queratinas/metabolismo , Antígenos Comuns de Leucócito/genética , Antígenos Comuns de Leucócito/imunologia , Antígenos Comuns de Leucócito/metabolismo , Neoplasias/sangue , Neoplasias/imunologia , Neoplasias/patologia , Células Neoplásicas Circulantes/imunologia , Células Neoplásicas Circulantes/metabolismo , Ligação Proteica , Análise de Célula Única/instrumentação
9.
Methods Mol Biol ; 1634: 181-192, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28819851

RESUMO

The CytePicker module built into the RareCyte CyteFinder instrument allows researchers to easily retrieve individual cells from microscope slides for genomic analyses, including array CGH, targeted sequencing, and next-generation sequencing. Here, we describe the semiautomated retrieval of CTCs from the blood processed by AccuCyte (see Chapter 13) and amplification of genomic DNA so that molecular analysis can be performed.


Assuntos
Separação Celular/métodos , Células Imobilizadas/patologia , Neoplasias/diagnóstico , Células Neoplásicas Circulantes/patologia , Técnicas de Amplificação de Ácido Nucleico/métodos , Análise de Célula Única/métodos , Automação Laboratorial/instrumentação , Contagem de Células , Linhagem Celular Tumoral , Separação Celular/instrumentação , Células Imobilizadas/imunologia , Células Imobilizadas/metabolismo , Centrifugação/instrumentação , Centrifugação/métodos , Hibridização Genômica Comparativa , Desenho de Equipamento , Genoma Humano , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Neoplasias/sangue , Neoplasias/imunologia , Neoplasias/patologia , Células Neoplásicas Circulantes/imunologia , Células Neoplásicas Circulantes/metabolismo , Análise de Célula Única/instrumentação
10.
Stem Cell Res Ther ; 8(1): 171, 2017 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-28732530

RESUMO

BACKGROUND: Mesenchymal stromal/stem cells (MSCs) have demonstrated pro-healing properties due to their anti-inflammatory, angiogenic, and even antibacterial properties. We have shown previously that minocycline enhances the wound healing phenotype of MSCs, and MSCs encapsulated in poly(ethylene glycol) and gelatin-based hydrogels with minocycline have antibacterial properties against Staphylococcus aureus (SA). Here, we investigated the signaling pathway that minocycline modulates in MSCs which results in their enhanced wound healing phenotype and determined whether preconditioning MSCs with minocycline has an effect on antimicrobial activity. We further investigated the in-vivo antimicrobial efficacy of MSC and antibiotic-loaded hydrogels in inoculated full-thickness cutaneous wounds. METHODS: Modulation of cell signaling pathways in MSCs with minocycline was analyzed via western blot, immunofluorescence, and ELISA. Antimicrobial efficacy of MSCs pretreated with minocycline was determined by direct and transwell coculture with SA. MSC viability after SA coculture was determined via a LIVE/DEAD® stain. Internalization of SA by MSCs pretreated with minocycline was determined via confocal imaging. All protein and cytokine analysis was done via ELISA. The in-vivo antimicrobial efficacy of MSC and antibiotic-loaded hydrogels was determined in Sprague-Dawley rats inoculated with SA. Two-way ANOVA for multiple comparisons was used with Bonferroni test assessment and an unpaired two-tailed Student's t test was used to determine p values for all assays with multiple or two conditions, respectively. RESULTS: Minocycline leads to the phosphorylation of transcriptional nuclear factor-κB (NFκB), but not c-Jun NH2-terminal kinase (JNK) or mitogen-activated protein kinase (ERK). Inhibition of NFκB activation prevented the minocycline-induced increase in VEGF secretion. Preconditioning of MSCs with minocycline led to a reduced production of the antimicrobial peptide LL-37, but enhanced antimicrobial activity against SA via an increased production of IL-6 and SA internalization. MSC and antibiotic-loaded hydrogels reduced SA bioburden in inoculated wounds over 3 days and accelerated reepithelialization. CONCLUSIONS: Minocycline modulates the NFκB pathway in MSCs that leads to an enhanced production of IL-6 and internalization of SA. This mechanism may have contributed to the in-vivo antibacterial efficacy of MSC and antibiotic-loaded hydrogels.


Assuntos
Células-Tronco Mesenquimais/imunologia , Minociclina/farmacologia , NF-kappa B/imunologia , Staphylococcus aureus/imunologia , Adulto , Células Imobilizadas/imunologia , Feminino , Humanos , Masculino , Fosforilação/efeitos dos fármacos , Fosforilação/imunologia
11.
Tissue Eng Part A ; 23(5-6): 177-184, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27967655

RESUMO

Gene therapy approaches have been difficult to implement due to pre-existing immunity against the virus used for delivery. To circumvent this problem, a cell-based approach was developed that avoided the use of free virus within the animal. However, even cells transduced in vitro with E1- to E3-deleted adenovirus encoding bone morphogenetic protein 2 (AdBMP2) resulted in the production of virus-neutralizing antibodies in mice. Furthermore, when mice received an intramuscular injection of nonencoding adenovirus (AdEmpty)-transduced cells, AdBMP2-transduced cells were unable to launch bone formation when an intramuscular injection of these BMP2-producing cells was delivered 1 week later. This phenomenon was not observed in NOD/SCID mice, and could be overcome in C57BL/6 mice by encapsulating the adenovirus-transduced cells in a nondegradable hydrogel poly(ethylene glycol) diacrylate (PEGDA). Data collectively suggest that PEGDA hydrogel encapsulation of AdBMP2-transduced cells prevents pre-existing immunity from suppressing BMP2-induced bone formation.


Assuntos
Adenoviridae , Proteína Morfogenética Óssea 2/imunologia , Células Imobilizadas , Fibroblastos , Hidrogéis/química , Polietilenoglicóis/química , Transdução Genética , Animais , Proteína Morfogenética Óssea 2/genética , Células Imobilizadas/imunologia , Células Imobilizadas/transplante , Fibroblastos/imunologia , Fibroblastos/transplante , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID
12.
Methods Mol Biol ; 1479: 305-333, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27738946

RESUMO

Transplantation of alginate-encapsulated islets has the potential to treat patients suffering from type I diabetes, a condition characterized by an autoimmune attack against insulin-secreting beta cells. However, there are multiple immunological challenges associated with this procedure, all of which must be adequately addressed prior to translation from trials in small animal and nonhuman primate models to human clinical trials. Principal threats to graft viability include immune-mediated destruction triggered by immunogenic alginate impurities, unfavorable polymer composition and surface characteristics, and release of membrane-permeable antigens, as well as damage associated molecular patterns (DAMPs) by the encapsulated islets themselves. The lack of standardization of significant parameters of bioencapsulation device design and manufacture (i.e., purification protocols, surface-modification grafting techniques, alginate composition modifications) between labs is yet another obstacle that must be overcome before a clinically effective and applicable protocol for encapsulating islets can be implemented. Nonetheless, substantial progress is being made, as is evident from prolonged graft survival times and improved protection from immune-mediated graft destruction reported by various research groups, but also with regard to discoveries of specific pathways involved in explaining observed outcomes. Progress in the latter is essential for a comprehensive understanding of the mechanisms responsible for the varying levels of immunogenicity of certain alginate devices. Successful translation of encapsulated islet transplantation from in vitro and animal model testing to human clinical trials hinges on application of this knowledge of the pathways and interactions which comprise immune-mediated rejection. Thus, this review not only focuses on the different factors contributing to provocation of the immune reaction by encapsulated islets, but also on the defining characteristics of the response itself.


Assuntos
Alginatos/química , Células Imobilizadas/imunologia , Rejeição de Enxerto/imunologia , Terapia de Imunossupressão/métodos , Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas/imunologia , Transplante Heterólogo , Alginatos/efeitos adversos , Animais , Células Imobilizadas/citologia , Células Imobilizadas/transplante , Ensaios Clínicos como Assunto , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/terapia , Ácido Glucurônico/efeitos adversos , Ácido Glucurônico/química , Rejeição de Enxerto/etiologia , Sobrevivência de Enxerto , Ácidos Hexurônicos/efeitos adversos , Ácidos Hexurônicos/química , Humanos , Ilhotas Pancreáticas/citologia , Transplante das Ilhotas Pancreáticas/efeitos adversos , Transplante das Ilhotas Pancreáticas/métodos , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/imunologia , Suínos , Transplante Heterólogo/efeitos adversos , Transplante Heterólogo/métodos , Resultado do Tratamento
13.
Ultramicroscopy ; 160: 168-181, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26521163

RESUMO

A method is presented for combining atomic force microscopy (AFM) force mode and fluorescence microscopy in order to (a) mechanically stimulate immune cells while recording the subsequent activation under the form of calcium pulses, and (b) observe the mechanical response of a cell upon photoactivation of a small G protein, namely Rac. Using commercial set-ups and a robust signal coupling the fluorescence excitation light and the cantilever bending, the applied force and activation signals were very easily synchronized. This approach allows to control the entire mechanical history of a single cell up to its activation and response down to a few hundreds of milliseconds, and can be extended with very minimal adaptations to other cellular systems where mechanotransduction is studied, using either purely mechanical stimuli or via a surface bound specific ligand.


Assuntos
Mecanotransdução Celular/imunologia , Microscopia de Força Atômica/métodos , Microscopia de Fluorescência/métodos , Compostos de Anilina , Animais , Sinalização do Cálcio/imunologia , Linhagem Celular , Células Imobilizadas/imunologia , Corantes Fluorescentes , Humanos , Células Jurkat , Ativação Linfocitária , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Análise de Célula Única , Linfócitos T/imunologia , Linfócitos T/metabolismo , Xantenos , Proteínas rac de Ligação ao GTP/metabolismo
14.
Sci Rep ; 5: 16884, 2015 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-26592180

RESUMO

Cellular therapies for liver diseases and in vitro models for drug testing both require functional human hepatocytes (Hum-H), which have unfortunately been limited due to the paucity of donor liver tissues. Human pluripotent stem cells (hPSCs) represent a promising and potentially unlimited cell source to derive Hum-H. However, the hepatic functions of these hPSC-derived cells to date are not fully comparable to adult Hum-H and are more similar to fetal ones. In addition, it has been challenging to obtain functional hepatic engraftment of these cells with prior studies having been done in immunocompromised animals. In this report, we demonstrated successful engraftment of human induced pluripotent stem cell (iPSC)-derived hepatocyte-like cells (iPS-H) in immunocompetent mice by pre-engineering 3D cell co-aggregates with stromal cells (SCs) followed by encapsulation in recently developed biocompatible hydrogel capsules. Notably, upon transplantation, human albumin and α1-antitrypsin (A1AT) in mouse sera secreted by encapsulated iPS-H/SCs aggregates reached a level comparable to the primary Hum-H/SCs control. Further immunohistochemistry of human albumin in retrieved cell aggregates confirmed the survival and function of iPS-H. This proof-of-concept study provides a simple yet robust approach to improve the engraftment of iPS-H, and may be applicable to many stem cell-based therapies.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/métodos , Células Imobilizadas/transplante , Sobrevivência de Enxerto , Hepatócitos/transplante , Células-Tronco Pluripotentes Induzidas/citologia , Células Estromais/transplante , Albuminas/biossíntese , Albuminas/metabolismo , Animais , Agregação Celular/fisiologia , Diferenciação Celular , Células Imobilizadas/citologia , Células Imobilizadas/imunologia , Células Imobilizadas/metabolismo , Técnicas de Cocultura , Hepatócitos/citologia , Hepatócitos/imunologia , Hepatócitos/metabolismo , Humanos , Hidrogéis/química , Imunocompetência , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Células Estromais/citologia , Células Estromais/imunologia , Células Estromais/metabolismo , Técnicas de Cultura de Tecidos , Transplante Heterólogo , alfa 1-Antitripsina/biossíntese , alfa 1-Antitripsina/metabolismo
15.
Biomaterials ; 35(36): 9563-72, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25175597

RESUMO

The erroneous and unpredictable behavior of percutaneous glucose sensors just days following implantation has limited their clinical utility for diabetes management. Recent research has implicated the presence of adherent inflammatory cells as the key mitigating factor limiting sensor functionality in this period of days post-implantation. Here we present a novel in vitro platform to mimic the cell-embedded provisional matrix that forms adjacent to the sensor immediately after implantation for the focused investigation of the effects of early stage tissue response on sensor function. This biomimetic surrogate is formed by imbibing fibrin-based gels with physiological densities of inflammatory RAW 264.7 macrophages. When surrounding functional sensors, macrophage-embedded fibrin gels contribute to sensor signal declines that are similar in both shape and magnitude to those observed in previous whole blood and small animal studies. Signal decline in the presence of gels is both metabolically-mediated and sensitive to cell type and activation. Computational modeling of the experimental setup is also presented to validate the design by showing that the cellular glucose uptake parameters necessary to achieve such experimental declines align well with literature values. Together, these data suggest this in vitro provisional matrix surrogate may serve as an effective screening tool for testing the biocompatibility of future glucose sensor designs.


Assuntos
Técnicas Biossensoriais , Glicemia/análise , Fibrina/química , Géis/química , Inflamação/etiologia , Macrófagos/imunologia , Próteses e Implantes/efeitos adversos , Células 3T3 , Animais , Materiais Biocompatíveis/química , Técnicas Biossensoriais/instrumentação , Linhagem Celular , Células Imobilizadas/citologia , Células Imobilizadas/imunologia , Inflamação/imunologia , Macrófagos/citologia , Camundongos
16.
Biosens Bioelectron ; 24(2): 228-32, 2008 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-18468880

RESUMO

A novel yeast cell-based strategy for the immobilization of antibodies using an amine-terminated self-assembly film has been proposed. A quartz crystal microbalance sensor was according fabricated by coupling with anti-prostate specific antigen (anti-PSA) for PSA immunoassay. The crystal was modified with cysteamine to deposit yeast cells, on which anti-PSA antibodies were immobilized. The surface topologies of the as-prepared crystals were characterized by use of scanning electron microscopy. In contrast to the traditional glutaraldehyde (GLU) approach, the yeast cells could allow antibody molecules bound with higher bioactivity and achieve better immunoreaction capability. Results indicate that immunoassay prepared using the developed yeast cell-binding procedure exhibits increased analytical performance compared with that produced using the GLU cross-linking procedure. A PSA serum concentration in the range of 5.0-604.0 ng ml(-1) can be determined by this new system.


Assuntos
Bioensaio/métodos , Separação Celular/métodos , Eletroquímica/métodos , Imunoensaio/métodos , Antígeno Prostático Específico/análise , Antígeno Prostático Específico/imunologia , Saccharomyces cerevisiae/imunologia , Células Imobilizadas/imunologia
17.
Artigo em Inglês | MEDLINE | ID: mdl-12906307

RESUMO

Formulation of membrane properties is important prior the successful implantation of encapsulated cells producing therapeutically relevant compounds. The purpose of our study was to specify the methods allowing preliminary evaluation of hollow fibers (HF) chosen for immunoisolation. We have selected as estimates (1) diffusive permeability for small and large solutes, and HF cut off (in vitro), (2) histological evaluation of tissue overgrowth after sc. implantation into mice. It was found that diffusive coefficients were linearly dependent on the particle diameter except that of albumin (2-3 times higher than theoretically estimated). This discrepancy imply that for certain particles the interaction with membrane material may be significant. The histological evaluation showed that siliconized HF implanted for 105 days were accepted (there was thin fibrotic layer on the external surface of the HF, no surrounding haemopoietic cells were found). It is concluded that proposed methods for preliminary evaluation of hollow fibers chosen for immunoisolation seems to be reliable and suitable for testing diffusive permeability of each relevant cell product.


Assuntos
Cápsulas/química , Células Imobilizadas/citologia , Polipropilenos/química , Transplantes , Velocidade do Fluxo Sanguíneo , Sobrevivência Celular/imunologia , Células Imobilizadas/imunologia , Difusão , Fibroblastos , Modelos Cardiovasculares , Silício
18.
Russ J Immunol ; 7(1): 34-40, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12687264

RESUMO

Alpha2-macroglobulin (a2M) secreted by tissue macrophages and fibroblasts functions in the environment of extracellular matrix macromolecules. We supposed that it may interact with these molecules and change the properties of extracellular matrix. Modified variant of ELISA was used to prove the direct binding of human a2M to collagen. Native and transformed by plasmin a2M, as well as plasmin, used as the control, were labeled by biotin. It has been found that the transformed, but not the native a2M form binds to type I collagen molecules: K(d)=(1.168 +/- 1.14) x 10(-11) M. The data obtained give a strong evidence of high power of the interaction between a2M and type I collagen: practically no reverse dissociation may be seen for such a binding. The modification of three-dimensional collagen matrix by binding to the transformed a2M resulted in the enhancement of migration of macrophages, carrying the receptors for a2M, but not splenocytes that lack for such receptors. Our results allow to suggest that a2M may be one of the components of extracellular matrix, and may change the properties of microenvironment for immunocompetent cells during the processes of inflammation, reparation and tumor invasion.


Assuntos
Movimento Celular/imunologia , Colágeno Tipo I/imunologia , Macrófagos/imunologia , alfa-Macroglobulinas/imunologia , Células Imobilizadas/imunologia , Colágeno Tipo I/metabolismo , Matriz Extracelular/imunologia , Matriz Extracelular/metabolismo , Fibrinolisina/metabolismo , Humanos , Ativação de Macrófagos/imunologia , Macrófagos/citologia , Macrófagos/fisiologia , Ligação Proteica , Baço/citologia , Baço/imunologia , alfa-Macroglobulinas/metabolismo
19.
Clin Exp Immunol ; 115(1): 78-85, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9933423

RESUMO

IL- 12 is the prominent inducer of Th1 responses in humans and in the mouse. CD40 ligand (CD40L) plays important roles in regulation of immune responses, including T cell-dependent activation of B cells and cytokine production by monocytes and dendritic cells. The present study examined the influences of IL-12 on the CD40L expression of activated human CD4+ T cells. IL-12 enhanced CD40L expression on CD4+ T cells stimulated with immobilized anti-CD3 in the complete absence of accessory cells, whereas IL-4 and IL-10 decreased it. Exogenous interferon-gamma (IFN-gamma) did not increase CD40L expression on immobilized anti-CD3 stimulated CD4+ T cells at any time up to 168 h of culture. The IL-12-induced enhancement of CD40L expression on anti-CD3 activated CD4+ T cells was not influenced in the presence of a metalloproteinase inhibitor KB8301, which up-regulated CD40L expression by preventing the processing of membrane-bound CD40L, or B cells, which down-regulated CD40L expression by receptor-mediated endocytosis. These results indicate that IL-12 enhances the CD40L expression of activated CD4+ T cells independently of the IFN-gamma production. The data thus suggest that Th1 responses induced by IL-12 might play an important role in the regulation of humoral immune responses through up-regulated CD40L expression.


Assuntos
Interleucina-12/farmacologia , Glicoproteínas de Membrana/biossíntese , Células Th1/imunologia , Adulto , Linfócitos B/imunologia , Linfócitos B/fisiologia , Complexo CD3/imunologia , Linfócitos T CD4-Positivos/imunologia , Ligante de CD40 , Células Imobilizadas/efeitos dos fármacos , Células Imobilizadas/imunologia , Humanos , Interferon gama/farmacologia , Ligantes , Ativação Linfocitária/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA