Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 85
Filtrar
1.
Endocrinology ; 162(6)2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33693651

RESUMO

The molecular interactions between the maternal environment and the developing embryo are key for early pregnancy success and are influenced by factors such as maternal metabolic status. Our understanding of the mechanism(s) through which these individual nutritional stressors alter endometrial function and the in utero environment for early pregnancy success is, however, limited. Here we report, for the first time, the use of an endometrium-on-a-chip microfluidics approach to produce a multicellular endometrium in vitro. Isolated endometrial cells (epithelial and stromal) from the uteri of nonpregnant cows in the early luteal phase (Days 4-7) were seeded in the upper chamber of the device (epithelial cells; 4-6 × 104 cells/mL) and stromal cells seeded in the lower chamber (1.5-2 × 104 cells/mL). Exposure of cells to different concentrations of glucose (0.5, 5.0, or 50 mM) or insulin (Vehicle, 1 or 10 ng/mL) was performed at a flow rate of 1 µL/minute for 72 hours. Quantitative differences in the cellular transcriptome and the secreted proteome of in vitro-derived uterine luminal fluid were determined by RNA-sequencing and tandem mass tagging mass spectrometry, respectively. High glucose concentrations altered 21 and 191 protein-coding genes in epithelial and stromal cells, respectively (P < .05), with a dose-dependent quantitative change in the protein secretome (1 and 23 proteins). Altering insulin concentrations resulted in limited transcriptional changes including transcripts for insulin-like binding proteins that were cell specific but altered the quantitative secretion of 196 proteins. These findings highlight 1 potential mechanism by which changes to maternal glucose and insulin alter uterine function.


Assuntos
Endométrio/efeitos dos fármacos , Glucose/farmacologia , Insulina/farmacologia , Dispositivos Lab-On-A-Chip , Animais , Bovinos , Técnicas de Cultura de Células/instrumentação , Técnicas de Cultura de Células/métodos , Células Cultivadas , Embrião de Mamíferos , Desenvolvimento Embrionário/efeitos dos fármacos , Desenvolvimento Embrionário/genética , Endométrio/citologia , Endométrio/metabolismo , Feminino , Perfilação da Expressão Gênica/instrumentação , Perfilação da Expressão Gênica/métodos , Gravidez , Cultura Primária de Células/instrumentação , Cultura Primária de Células/métodos , Proteoma/efeitos dos fármacos , Proteoma/metabolismo , Proteômica/instrumentação , Proteômica/métodos , Via Secretória/efeitos dos fármacos , Transcriptoma/efeitos dos fármacos
2.
Commun Biol ; 4(1): 168, 2021 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-33547387

RESUMO

The air-blood barrier with its complex architecture and dynamic environment is difficult to mimic in vitro. Lung-on-a-chips enable mimicking the breathing movements using a thin, stretchable PDMS membrane. However, they fail to reproduce the characteristic alveoli network as well as the biochemical and physical properties of the alveolar basal membrane. Here, we present a lung-on-a-chip, based on a biological, stretchable and biodegradable membrane made of collagen and elastin, that emulates an array of tiny alveoli with in vivo-like dimensions. This membrane outperforms PDMS in many ways: it does not absorb rhodamine-B, is biodegradable, is created by a simple method, and can easily be tuned to modify its thickness, composition and stiffness. The air-blood barrier is reconstituted using primary lung alveolar epithelial cells from patients and primary lung endothelial cells. Typical alveolar epithelial cell markers are expressed, while the barrier properties are preserved for up to 3 weeks.


Assuntos
Elasticidade/fisiologia , Dispositivos Lab-On-A-Chip , Pulmão/citologia , Membranas Artificiais , Alvéolos Pulmonares/fisiologia , Células Epiteliais Alveolares/citologia , Células Epiteliais Alveolares/fisiologia , Barreira Alveolocapilar/citologia , Barreira Alveolocapilar/fisiologia , Comunicação Celular/fisiologia , Permeabilidade da Membrana Celular/fisiologia , Técnicas de Cocultura/instrumentação , Técnicas de Cocultura/métodos , Humanos , Pulmão/fisiologia , Microtecnologia , Cultura Primária de Células/instrumentação , Cultura Primária de Células/métodos , Alvéolos Pulmonares/citologia , Estresse Mecânico , Engenharia Tecidual/instrumentação , Engenharia Tecidual/métodos , Alicerces Teciduais/química
3.
Methods Mol Biol ; 2286: 167-178, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33381855

RESUMO

Human induced pluripotent stem cells (hiPSCs) have the potential to be used in a variety of biomedical applications, including drug discovery and Regenerative Medicine. The success of these approaches is, however, limited by the difficulty of generating the large quantities of cells required in a reproducible and controlled system. Bioreactors, widely used for industrial manufacture of biological products, constitute a viable strategy for large-scale production of stem cell derivatives. In this chapter, we describe the expansion of hiPSCs using the Vertical-Wheel™ bioreactor, a novel bioreactor configuration specifically designed for the culture of shear-sensitive cells. We provide protocols for the expansion of hiPSCs in suspension, both as floating aggregates and using microcarriers for cell adhesion. These methods may be important for the establishment of a scalable culture of hiPSCs, allowing the manufacturing of industrial- or clinical-scale cell numbers.


Assuntos
Tecnologia Biomédica/métodos , Reatores Biológicos/normas , Células-Tronco Pluripotentes Induzidas/citologia , Cultura Primária de Células/métodos , Tecnologia Biomédica/instrumentação , Tecnologia Biomédica/normas , Células Cultivadas , Humanos , Células-Tronco Pluripotentes Induzidas/fisiologia , Guias de Prática Clínica como Assunto , Cultura Primária de Células/instrumentação , Cultura Primária de Células/normas
4.
Clin Transl Sci ; 14(3): 1049-1061, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33382907

RESUMO

Liver microphysiological systems (MPSs) are promising models for predicting hepatic drug effects. Yet, after a decade since their introduction, MPSs are not routinely used in drug development due to lack of criteria for ensuring reproducibility of results. We characterized the feasibility of a liver MPS to yield reproducible outcomes of experiments assaying drug toxicity, metabolism, and intracellular accumulation. The ability of the liver MPS to reproduce hepatotoxic effects was assessed using trovafloxacin, which increased lactate dehydrogenase (LDH) release and reduced cytochrome P450 3A4 (CYP3A4) activity. These observations were made in two test sites and with different batches of Kupffer cells. Upon culturing equivalent hepatocytes in the MPS, spheroids, and sandwich cultures, differences between culture formats were detected in CYP3A4 activity and albumin production. Cells in all culture formats exhibited different sensitivities to hepatotoxicant exposure. Hepatocytes in the MPS were more functionally stable than those of other culture platforms, as CYP3A4 activity and albumin secretion remained prominent for greater than 18 days in culture, whereas functional decline occurred earlier in spheroids (12 days) and sandwich cultures (7 days). The MPS was also demonstrated to be suitable for metabolism studies, where CYP3A4 activity, troglitazone metabolites, diclofenac clearance, and intracellular accumulation of chloroquine were quantified. To ensure reproducibility between studies with the MPS, the combined use of LDH and CYP3A4 assays were implemented as quality control metrics. Overall results indicated that the liver MPS can be used reproducibly in general drug evaluation applications. Study outcomes led to general considerations and recommendations for using liver MPSs. Study Highlights WHAT IS THE CURRENT KNOWLEDGE ON THE TOPIC? Microphysiological systems (MPSs) have been designed to recreate organ- or tissue-specific characteristics of extracellular microenvironments that enhance the physiological relevance of cells in culture. Liver MPSs enable long-lasting and stable culture of hepatic cells by culturing them in three-dimensions and exposing them to fluid flow. WHAT QUESTION DID THIS STUDY ADDRESS? What is the functional performance relative to other cell culture platforms and the reproducibility of a liver MPS for assessing drug development and evaluation questions, such as toxicity, metabolism, and pharmacokinetics? WHAT DOES THIS STUDY ADD TO OUR KNOWLEDGE? The liver MPS systematically detected the toxicity of trovafloxacin. When compared with spheroids and sandwich cultures, this system had a more stable function and different sensitivity to troglitazone, tamoxifen, and digoxin. Quantifying phase II metabolism of troglitazone and intracellular accumulation of chloroquine demonstrated the potential use of the liver MPS for studying drug metabolism and pharmacokinetics. Quality control criteria for assessing chip function were key for reliably using the liver MPS. HOW MIGHT THIS CHANGE CLINICAL PHARMACOLOGY OR TRANSLATIONAL SCIENCE? Due to its functional robustness and physiological relevance (3D culture, cells expose to fluid flow and co-culture of different cell types), the liver MPS can, in a reproducible manner: (i) detect inflammatory-induced drug toxicity, as demonstrated with trovafloxacin, (ii) detect the toxicity of other drugs, such as troglitazone, tamoxifen, and digoxin, with different effects than those detected in spheroids and sandwich cultures, (iii) enable studies of hepatic function that rely on prolonged cellular activity, and (iv) detect phase II metabolites and drug accumulation to potentially support the interpretation of clinical data. The integration of MPSs in drug development will be facilitated by careful evaluation of performance and reproducibility as performed in this study.


Assuntos
Fígado/efeitos dos fármacos , Cultura Primária de Células/métodos , Testes de Toxicidade/métodos , Células Cultivadas , Citocromo P-450 CYP3A/metabolismo , Avaliação Pré-Clínica de Medicamentos , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Humanos , Dispositivos Lab-On-A-Chip , Fígado/citologia , Fígado/metabolismo , Modelos Biológicos , Cultura Primária de Células/instrumentação , Reprodutibilidade dos Testes , Esferoides Celulares , Testes de Toxicidade/instrumentação
5.
Nat Commun ; 11(1): 6313, 2020 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-33298939

RESUMO

Cultured neuronal networks with a controlled structure have been widely studied as an in vitro model system to investigate the relationship between network structure and function. However, most cell culture techniques lack the ability to control network structures during cell cultivation, making it difficult to assess functional changes induced by specific structural changes. In this study, we present an in situ manipulation platform based on gold-nanorod-mediated thermoplasmonics to interrogate an in vitro network model. We find that it is possible to induce new neurite outgrowths, eliminate interconnecting neurites, and estimate functional relationships in matured neuronal networks. This method is expected to be useful for studying functional dynamics of neural networks under controlled structural changes.


Assuntos
Nanotecnologia/instrumentação , Rede Nervosa/fisiologia , Neuritos/fisiologia , Cultura Primária de Células/instrumentação , Animais , Células Cultivadas , Embrião de Mamíferos/citologia , Estudos de Viabilidade , Ouro/química , Hipocampo/citologia , Hidrogéis/química , Nanopartículas Metálicas/química , Nanotecnologia/métodos , Nanotubos/química , Rede Nervosa/citologia , Ratos
6.
Drug Resist Updat ; 53: 100730, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33096284

RESUMO

New treatment options of acute myeloid leukemia (AML) are rapidly emerging. Pre-clinical models such as ex vivo cultures are extensively used towards the development of novel drugs and to study synergistic drug combinations, as well as to discover biomarkers for both drug response and anti-cancer drug resistance. Although these approaches empower efficient investigation of multiple drugs in a multitude of primary AML samples, their translational value and reproducibility are hampered by the lack of standardized methodologies and by culture system-specific behavior of AML cells and chemotherapeutic drugs. Moreover, distinct research questions require specific methods which rely on specific technical knowledge and skills. To address these aspects, we herein review commonly used culture techniques in light of diverse research questions. In addition, culture-dependent effects on drug resistance towards commonly used drugs in the treatment of AML are summarized including several pitfalls that may arise because of culture technique artifacts. The primary aim of the current review is to provide practical guidelines for ex vivo primary AML culture experimental design.


Assuntos
Antineoplásicos/farmacologia , Leucemia Mieloide Aguda/tratamento farmacológico , Cultura Primária de Células/métodos , Projetos de Pesquisa/normas , Antineoplásicos/uso terapêutico , Medula Óssea/patologia , Criopreservação , Meios de Cultura/química , Meios de Cultura/normas , Resistencia a Medicamentos Antineoplásicos , Ensaios de Seleção de Medicamentos Antitumorais/instrumentação , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Ensaios de Seleção de Medicamentos Antitumorais/normas , Guias como Assunto , Humanos , Leucemia Mieloide Aguda/sangue , Leucemia Mieloide Aguda/patologia , Cultura Primária de Células/instrumentação , Cultura Primária de Células/normas , Reprodutibilidade dos Testes , Células Tumorais Cultivadas
7.
Clin Transl Sci ; 13(6): 1115-1126, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33030809

RESUMO

Ex vivo (EV)-derived megakaryocytes (MKs) have shown great promise as a substitute for platelets in transfusion medicine to alleviate a severe shortage of donor-platelets. Challenges remain that include poor efficiency, a limited scale of production, and undefined short-term storage conditions of EV-derived MKs. This study aims to develop a high-efficiency system for large-scale production of Good Manufacturing Practice (GMP)-grade MKs and determine the short-term storage condition for the MKs. A roller-bottle culture system was introduced to produce GMP-grade MKs from small-molecule/cytokine cocktail expanded hematopoietic stem cells. Various buffer systems and temperatures for the short-term storage of MKs were assessed by cell viability, biomarker expression, and DNA ploidy levels. MKs stored for 24 hours were transplanted into sublethally irradiated nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice to confirm their platelet-releasing and tissue-homing ability in vivo. A yield of ~ 2.5 × 104 CD41a+ /CD42b+ MKs with purity of ~ 80% was achieved from one original cord blood CD34+ cell. Compared with the static culture, the roller-bottle culture system significantly enhanced megakaryopoiesis, as shown by the cell size, DNA ploidy, and megakaryopoiesis-related gene expression. The optimal storage condition for the MKs was defined as normal saline with 10% human serum albumin at 22℃. Stored MKs were capable of rapidly producing functional platelets and largely distributing in the lungs of NOD/SCID mice. The novel development of efficient production and storage system for GMP-grade MKs represents a significant step toward application of these MKs in the clinic.


Assuntos
Plaquetas/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Megacariócitos/fisiologia , Transfusão de Plaquetas/métodos , Cultura Primária de Células/métodos , Animais , Diferenciação Celular/fisiologia , Células Cultivadas , Sangue Fetal/citologia , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Modelos Animais , Cultura Primária de Células/instrumentação , Trombopoese
8.
Methods Mol Biol ; 2203: 119-134, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32833209

RESUMO

Well-differentiated primary airway epithelial cell (AEC) cultures have been widely used for the characterization of several human respiratory viruses including coronaviruses. In recent years, there has been an increase in interest toward animal AEC cultures and their application to characterize veterinary viruses with zoonotic potential, as well as studying host-pathogen interactions in animal reservoir host species. In this chapter, we provide a revised and improved protocol for the isolation and establishment of well-differentiated AEC cultures from diverse mammalian species and the use of the cultures for the characterization of veterinary coronavirus. We also describe immunohistochemistry protocols with validated antibodies for the visualization and identification of viral cell tropism in well-differentiated AEC cultures from human, swine, bovine, and feline origin.


Assuntos
Brônquios/citologia , Coronavirus/fisiologia , Células Epiteliais/virologia , Cultura Primária de Células/métodos , Traqueia/citologia , Animais , Gatos , Bovinos , Diferenciação Celular , Células Epiteliais/citologia , Imunofluorescência , Humanos , Imuno-Histoquímica/métodos , Cultura Primária de Células/instrumentação , Suínos , Tropismo Viral
9.
Drug Metab Dispos ; 48(7): 594-602, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32350061

RESUMO

Despite the availability of liquid chromatography (LC)-mass spectrometry (MS) methods for quantifying cytochrome P450 (P450) proteins, incorporation of P450 protein quantification into induction study workflows has not been widely adopted. To more readily enable P450 protein quantification in induction study workflows, DMPK research groups need a simple, robust, cost-effective, high-throughput method compatible with 96-well-plated human hepatocyte formats. Here, we provide such a methodology. Our method bypasses both microsomal enrichment and antibody-based enrichment to go directly from the plate to LC-MS/MS analysis. We use this "plate-to-peaks" approach for quantifying CYP3A4, CYP2B6, and CYP1A2, the major inducible hepatic P450s representative of pregnane X receptor-, constitutive androstane receptor-, and aryl hydrocarbon receptor-mediated induction, respectively. We leveraged our induction study-aligned assay format to assess induction across mRNA, protein, and enzyme activity using known induction control compounds. As expected, results from the three methods using model inducers were broadly concordant, but the magnitude of the induction response differed. Induction of CYP3A4 using 10 µM rifampicin was 12-fold for RNA, eightfold for protein, and threefold for activity; for CYP1A2 with 50 µM omeprazole, induction was 30-fold for RNA, 13-fold for protein, and 17-fold for activity; for CYP2B6 with 50 µM phenytoin, induction was 23-fold for RNA, twofold for protein, and fivefold for activity. Most importantly, we anticipate the relative ease of this method will enable researchers to routinely adopt P450 protein quantification as part of nonclinical evaluation of P450 induction. SIGNIFICANCE STATEMENT: Current methodologies for quantifying P450 proteins by liquid chromatography (LC)-tandem mass spectrometry are either cumbersome, too costly, or both to be widely adopted into induction study workflows by the ADME research community. We present a simplified LC-MS/MS methodology for quantifying P450 proteins directly from human hepatocytes, without any form of enrichment, in 96-well induction assay plate format that should be readily adoptable by any ADME laboratory with LC-multiple-reaction monitoring capabilities.


Assuntos
Indutores das Enzimas do Citocromo P-450/farmacologia , Sistema Enzimático do Citocromo P-450/análise , Ensaios Enzimáticos/métodos , Hepatócitos/enzimologia , Células Cultivadas , Cromatografia Líquida de Alta Pressão/métodos , Sistema Enzimático do Citocromo P-450/biossíntese , Indução Enzimática/efeitos dos fármacos , Humanos , Masculino , Cultura Primária de Células/instrumentação , Cultura Primária de Células/métodos , Espectrometria de Massas em Tandem/métodos
10.
Surgery ; 167(1): 197-203, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31543319

RESUMO

BACKGROUND: As patient-derived xenografts and other preclinical models of neuroendocrine tumors for testing personalized therapeutics are lacking, we have developed a perfused, 3D bioreactor model to culture tumor surrogates from patient-derived neuroendocrine tumors. This work evaluates the duration of surrogate culture and surrogate response to a novel antibody-drug conjugate. METHODS: Twenty-seven patient-derived neuroendocrine tumors were cultured. Histologic sections of a pancreatic neuroendocrine tumor xenograft (BON-1) tumor were assessed for SSTR2 expression before tumor implantation into 2 bioreactors. One surrogate was treated with an antibody-drug conjugate composed of an anti-mitotic Monomethyl auristatin-E linked to a somatostatin receptor 2 antibody. Viability and therapeutic response were assessed by pre-imaging incubation with IR-783 and the RealTime-Glo AnnexinV Apoptosis and Necrosis Assay (Promega Corporation, Madison, WI) over 6 days. A primary human pancreatic neuroendocrine tumor was evaluated similarly. RESULTS: Mean surrogate growth duration was 34.8 days. Treated BON-1 surrogates exhibited less proliferation (1.2 vs 1.9-fold) and greater apoptosis (1.5 vs 1.1-fold) than controls, whereas treated patient-derived neuroendocrine tumor bioreactors exhibited greater degrees of apoptosis (13- vs 9-fold) and necrosis (2.5- vs 1.6-fold). CONCLUSION: Patient-derived neuroendocrine tumor surrogates can be cultured reliably within the bioreactor. This model can be used to evaluate the efficacy of antibody-guided chemotherapy ex vivo and may be useful for predicting clinical responses.


Assuntos
Reatores Biológicos , Ensaios de Seleção de Medicamentos Antitumorais/instrumentação , Imunoconjugados/farmacologia , Tumores Neuroendócrinos/tratamento farmacológico , Neoplasias Pancreáticas/tratamento farmacológico , Cultura Primária de Células/instrumentação , Animais , Antineoplásicos Imunológicos/farmacologia , Antineoplásicos Imunológicos/uso terapêutico , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Humanos , Imunoconjugados/uso terapêutico , Masculino , Camundongos , Terapia de Alvo Molecular/métodos , Tumores Neuroendócrinos/patologia , Oligopeptídeos/farmacologia , Oligopeptídeos/uso terapêutico , Neoplasias Pancreáticas/patologia , Cultura Primária de Células/métodos , Receptores de Somatostatina/antagonistas & inibidores , Reprodutibilidade dos Testes , Células Tumorais Cultivadas
12.
Exp Cell Res ; 386(1): 111706, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31697927

RESUMO

Krüppel-like factor 15 (KLF15) is a well-known transcription factor associated with podocyte injury and fibrosis. Recently, hypertensive nephropathy was discovered to be closely related to podocyte injury and fibrosis. However, methods to stimulate hypertension in vitro are lacking. Here, we constructed an in vitro model mimicking hypertension using a rotational force device to identify the role of KLF15 in fibrosis due to mechanically induced hypertensive injury. First, we found that KLF15 expression was decreased in patients with hypertensive nephropathy. Then, an in vitro study of hypertension due to rotational force was conducted, and an increase in fibrosis markers and decrease in KLF15 levels were determined after application of 4 mmHg pressure in primary cultured human podocytes. KLF15 and tight junction protein levels increased with retinoic acid treatment. siRNA-mediated inhibition of KLF15 exacerbated pressure-induced fibrosis injury, and KLF15 expression after treatment with angiotensin II was similar to that observed after treatment with the blood pressure modeling device. Furthermore, the reduced KLF15 levels after mechanical pressure application were restored after the administration of an antihypertensive drug. KLF15 expression was also low in vivo. We confirmed the protective role of KLF15 in fibrosis using a mechanically induced in vitro model of hypertensive injury.


Assuntos
Hipertensão Renal/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Nefrite/metabolismo , Podócitos/metabolismo , Adulto , Angiotensina II/farmacologia , Animais , Células Cultivadas , Feminino , Fibrose , Humanos , Hipertensão Renal/genética , Hipertensão Renal/patologia , Fatores de Transcrição Kruppel-Like/genética , Masculino , Camundongos , Nefrite/genética , Nefrite/patologia , Podócitos/efeitos dos fármacos , Podócitos/patologia , Pressão , Cultura Primária de Células/instrumentação , Rotação , Proteínas de Junções Íntimas/genética , Proteínas de Junções Íntimas/metabolismo
13.
Int J Mol Sci ; 21(1)2019 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-31877727

RESUMO

Industrialization of stem-cell based therapies requires innovative solutions to close the gap between research and commercialization. Scalable cell production platforms are needed to reliably deliver the cell quantities needed during the various stages of development and commercial supply. Human pluripotent stem cells (hPSCs) are a key source material for generating therapeutic cell types. We have developed a closed, automated and scalable stirred tank bioreactor platform, capable of sustaining high fold expansion of hPSCs. Such a platform could facilitate the in-process monitoring and integration of online monitoring systems, leading to significantly reduced labor requirements and contamination risk. hPSCs are expanded in a controlled bioreactor using perfused xeno-free media. Cell harvest and concentration are performed in closed steps. The hPSCs can be cryopreserved to generate a bank of cells, or further processed as needed. Cryopreserved cells can be thawed into a two-dimensional (2D) tissue culture platform or a three-dimensional (3D) bioreactor to initiate a new expansion phase, or be differentiated to the clinically relevant cell type. The expanded hPSCs express hPSC-specific markers, have a normal karyotype and the ability to differentiate to the cells of the three germ layers. This end-to-end platform allows a large scale expansion of high quality hPSCs that can support the required cell demand for various clinical indications.


Assuntos
Automação Laboratorial/métodos , Técnicas de Reprogramação Celular/métodos , Células-Tronco Pluripotentes Induzidas/citologia , Cultura Primária de Células/métodos , Automação Laboratorial/instrumentação , Reatores Biológicos , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/fisiologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/fisiologia , Cultura Primária de Células/instrumentação
14.
Cells ; 8(11)2019 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-31726669

RESUMO

Multiple sclerosis (MS) is a chronic inflammatory demyelinating and neurodegenerative disease of the central nervous system. Neurological deficits are attributed to inflammatory demyelination, which compromises axonal function and survival. These are mitigated in experimental models by rapid and often complete remyelination of affected axons, but in MS this endogenous repair mechanism frequently fails, leaving axons increasingly vulnerable to the detrimental effects of inflammatory and metabolic stress. Understanding the molecular basis of remyelination and remyelination failure is essential to develop improved therapies for this devastating disease. However, recent studies suggest that this is not due to a single dominant mechanism, but rather represents the biological outcome of multiple changes in the lesion microenvironment that combine to disrupt oligodendrocyte differentiation. This identifies a pressing need to develop technical platforms to investigate combinatory and/or synergistic effects of factors differentially expressed in MS lesions on oligodendrocyte proliferation and differentiation. Here we describe protocols using primary oligodendrocyte cultures from Bl6 mice on 384-well nanofiber plates to model changes affecting oligodendrogenesis and differentiation in the complex signaling environment associated with multiple sclerosis lesions. Using platelet-derived growth factor (PDGF-AA), fibroblast growth factor 2 (FGF2), bone morphogenetic protein 2 (BMP2) and bone morphogenetic protein 4 (BMP4) as representative targets, we demonstrate that we can assess their combinatory effects across a wide range of concentrations in a single experiment. This in vitro model is ideal for assessing the combinatory effects of changes in availability of multiple factors, thus more closely modelling the situation in vivo and furthering high-throughput screening possibilities.


Assuntos
Esclerose Múltipla/metabolismo , Bainha de Mielina/metabolismo , Oligodendroglia/citologia , Cultura Primária de Células/instrumentação , Animais , Proteína Morfogenética Óssea 2/farmacologia , Proteína Morfogenética Óssea 4/farmacologia , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Fator 2 de Crescimento de Fibroblastos/farmacologia , Humanos , Camundongos , Modelos Teóricos , Esclerose Múltipla/terapia , Nanofibras , Oligodendroglia/efeitos dos fármacos , Oligodendroglia/metabolismo , Fator de Crescimento Derivado de Plaquetas/farmacologia , Cultura Primária de Células/métodos
15.
Methods Cell Biol ; 153: 1-23, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31395374

RESUMO

Autosomal dominant polycystic kidney (ADPKD) is a common genetic disorder characterized by the presence of numerous fluid-filled cysts that lead to a progressive decline in renal function. Cystic tissues and primary cyst epithelial cells obtained from discarded human ADPKD kidneys provide unique biomaterials for the investigation of cellular mechanisms involved in cyst growth and changes in the microenvironment adjacent to the cysts. ADPKD cells have been used to develop straightforward in vitro cell model assays to study events down-stream of the mutant proteins in carefully controlled experimental conditions, test specific hypotheses, and evaluate the cellular response to potential therapeutic drugs. Normal cadaver kidneys deemed unsuitable for transplantation and "non-involved" portions of nephrectomy specimens removed for the treatment of kidney cancer provide important control tissues and the source of primary normal human kidney (NHK) cells for comparison to ADPKD specimens. This chapter describes the methods used in the collection of cystic and non-cystic tissues from ADPKD and normal kidneys and the generation of primary cell cultures. We also highlight strengths and weaknesses of using immortalized isogenic normal and PKD mutant cell lines.


Assuntos
Rim/citologia , Rim Policístico Autossômico Dominante/patologia , Cultura Primária de Células/métodos , Linhagem Celular , Células Epiteliais , Humanos , Rim/patologia , Mutação , Nefrectomia , Rim Policístico Autossômico Dominante/genética , Rim Policístico Autossômico Dominante/cirurgia , Cultura Primária de Células/instrumentação , Canais de Cátion TRPP/genética
16.
Methods Cell Biol ; 153: 93-111, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31395386

RESUMO

Autosomal dominant polycystic kidney disease (ADPKD) is a common genetic disorder characterized by the relentless growth of numerous fluid-filled cysts in the kidneys. Mutations in PKD1 and PKD2, genes that encode polycystin 1 and 2, respectively, are responsible for most cases of ADPKD. Currently, the cellular mechanisms responsible for cyst formation remain poorly understood. In vitro models have been used by researchers to investigate cellular processes for cyst formation in carefully controlled experimental conditions. Madin-Darby canine kidney (MDCK) cells, a distal tubule epithelial cell line, were first used to form 3-dimensional (3-D) cysts within a hydrated collagen gel. This method was applied to epithelial cells cultured from cysts of human ADPKD kidneys, allowing investigators to study cellular mechanisms for cyst growth using cells that harbor the genetic mutations responsible for ADPKD in humans. Studies using ADPKD in vitro cysts have provided insight into cellular processes regulating cell proliferation, fluid secretion, and cell polarity. These assays were used to demonstrate the central role of cAMP agonists, such as arginine vasopressin, on cyst growth; and to test the effectiveness of potential therapeutic agents, including tolvaptan. Results obtained from in vitro cyst experiments demonstrate the translational value of cell model systems for investigating the mechanisms for cyst formation in human ADPKD. In this chapter, we describe protocols for growing ADPKD cells in a 3-D in vitro cyst assay and measuring total cyst volume by microscopy and image analysis.


Assuntos
Células Epiteliais/patologia , Microscopia Intravital/métodos , Rim Policístico Autossômico Dominante/etiologia , Cultura Primária de Células/métodos , Animais , Colágeno/metabolismo , Meios de Cultura/metabolismo , AMP Cíclico/agonistas , AMP Cíclico/metabolismo , Cães , Células Epiteliais/metabolismo , Humanos , Microscopia Intravital/instrumentação , Células Madin Darby de Rim Canino , Neurofisinas/metabolismo , Rim Policístico Autossômico Dominante/patologia , Cultura Primária de Células/instrumentação , Precursores de Proteínas/metabolismo , Vasopressinas/metabolismo
17.
Methods Cell Biol ; 153: 69-92, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31395385

RESUMO

Autosomal dominant polycystic kidney disease (ADPKD) is a common genetic disorder characterized by bilateral fluid-filled cysts, renal inflammation and extensive fibrosis, leading to the progressive decline in kidney function. Renal cyst formation begins in utero from aberrant proliferation of tubule epithelial cells; however, the mechanisms for cystogenesis remain unclear. Cell proliferation and Cl--dependent fluid secretion, which drives the accumulation of cyst fluid, are responsible for inexorable growth of cysts and the remarkable appearance of massively enlarged ADPKD kidneys. Investigators have used in vitro assays to explore cellular and molecular mechanisms involved in ADPKD cyst epithelial cell proliferation and Cl--dependent fluid secretion in experimentally controlled environments. These assays have been used to evaluate potential therapeutic approaches to inhibit cellular pathways involved in cyst growth. This chapter discusses methods for measuring ADPKD cell proliferation, transepithelial Cl- secretion, and net fluid transport across cyst epithelial cell monolayers.


Assuntos
Proliferação de Células , Cloretos/metabolismo , Células Epiteliais/fisiologia , Rim Policístico Autossômico Dominante/patologia , Cultura Primária de Células/métodos , Transporte Biológico/fisiologia , Contagem de Células/instrumentação , Contagem de Células/métodos , Humanos , Rim/citologia , Rim/metabolismo , Rim/patologia , Cultura Primária de Células/instrumentação
18.
Methods Mol Biol ; 2048: 121-130, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31396936

RESUMO

Invariant natural killer T (iNKT) cells are a subset of T lymphocytes that play a crucial role in the tumor surveillance. The activation of iNKT cells by their specific ligand α-galactosylceramide (α-GalCer) induces the activation of dendritic cells (DCs) via reciprocal interaction, which results in the generation of cellular immunity against cancer. Here we describe a method to detect DC-mediated cellular adjuvant properties of human iNKT cells in vitro.


Assuntos
Células Dendríticas/imunologia , Vigilância Imunológica , Células T Matadoras Naturais/imunologia , Cultura Primária de Células/métodos , Animais , Linfócitos T CD8-Positivos , Comunicação Celular/imunologia , Diferenciação Celular , Linhagem Celular Tumoral , Meios de Cultivo Condicionados/metabolismo , Citocinas/metabolismo , Citometria de Fluxo/instrumentação , Citometria de Fluxo/métodos , Técnica Direta de Fluorescência para Anticorpo , Galactosilceramidas/metabolismo , Voluntários Saudáveis , Humanos , Separação Imunomagnética/instrumentação , Separação Imunomagnética/métodos , Células-Tronco Pluripotentes Induzidas , Células-Tronco Mesenquimais , Camundongos , Neoplasias/imunologia , Cultura Primária de Células/instrumentação , Proteínas Recombinantes/metabolismo
19.
Methods Mol Biol ; 2048: 143-153, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31396938

RESUMO

The thymus plays an essential role in the development and selection of T cells by providing a unique microenvironment that is mainly composed of thymic epithelial cells (TECs). We previously identified stem cells of medullary TECs (mTECs) that are crucial for central tolerance induction using a novel clonogenic culture system. We also found that medullary thymic epithelial stem cells (mTESCs) maintain life-long mTECs regeneration and central T cell self-tolerance in mouse models. The clonogenic efficiency of TECs in vitro is highly correlated to the TEC reconstitution activity in vivo. Here, we describe the clonogenic culture system to evaluate the self-renewing activity of TESCs. The colonies are derived from TESCs, are visualized and quantified by rhodamine-B staining on a feeder layer, and can be passaged in vitro. Thus, our system enables quantitative evaluation of TESC activity and is useful for dissecting the mechanisms that regulate TESC activity in physiological aging as well as in various clinical settings.


Assuntos
Ensaio de Unidades Formadoras de Colônias/métodos , Cultura Primária de Células/métodos , Timo/citologia , Envelhecimento/fisiologia , Animais , Diferenciação Celular/fisiologia , Linhagem Celular , Técnicas de Cocultura/instrumentação , Técnicas de Cocultura/métodos , Ensaio de Unidades Formadoras de Colônias/instrumentação , Proteínas de Ligação a DNA/genética , Células Epiteliais/fisiologia , Citometria de Fluxo/instrumentação , Citometria de Fluxo/métodos , Técnica Direta de Fluorescência para Anticorpo/instrumentação , Técnica Direta de Fluorescência para Anticorpo/métodos , Corantes Fluorescentes/química , Separação Imunomagnética/instrumentação , Separação Imunomagnética/métodos , Camundongos , Camundongos Knockout , Cultura Primária de Células/instrumentação , Rodaminas/química , Tolerância a Antígenos Próprios/fisiologia , Coloração e Rotulagem/métodos , Células-Tronco/fisiologia
20.
Methods Mol Biol ; 2048: 245-257, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31396942

RESUMO

Human pluripotent stem cells (PSCs) have the potential to provide a virtually unlimited supply of cells for transplantation therapy. When combined with recent advances in genome editing technologies, human PSCs could offer various approaches that enable gene therapy, drug discovery, disease modeling, and in vitro modeling of human development. De novo generation of hematopoietic stem cells (HSCs) from human PSCs is an important focus in the field, since it enables autologous HSC transplantation to treat many blood disorders and malignancies. Although culture conditions have been established to generate a broad spectrum of hematopoietic progenitors from human PSCs, it remains a significant challenge to generate bona fide HSCs that possess sustained self-renewal and multilineage differentiation capacities upon transplantation. In this review, recent promising advances in the efforts to generate HSCs and hematopoietic progenitors from human PSCs in vitro and in vivo or from somatic cells are discussed.


Assuntos
Diferenciação Celular , Células-Tronco Hematopoéticas/fisiologia , Células-Tronco Pluripotentes/fisiologia , Cultura Primária de Células/métodos , Animais , Autorrenovação Celular , Células Cultivadas , Técnicas de Cocultura/instrumentação , Técnicas de Cocultura/métodos , Meios de Cultura/metabolismo , Corpos Embrioides/fisiologia , Transplante de Células-Tronco Hematopoéticas/métodos , Humanos , Células-Tronco Mesenquimais , Camundongos , Camundongos SCID , Cultura Primária de Células/instrumentação , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Quimeras de Transplante , Transplante Autólogo/métodos , Transplante Heterólogo/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...