Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
1.
J Immunol ; 210(7): 895-904, 2023 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-36947817

RESUMO

Hematoimmunopoiesis takes place in the adult human bone marrow (BM), which is composed of heterogeneous niches with complex architecture that enables tight regulation of homeostatic and stress responses. There is a paucity of representative culture systems that recapitulate the heterogeneous three-dimensional (3D) human BM microenvironment and that can endogenously produce soluble factors and extracellular matrix that deliver culture fidelity for the study of both normal and abnormal hematopoiesis. Native BM lymphoid populations are also poorly represented in current in vitro and in vivo models, creating challenges for the study and treatment of BM immunopathology. BM organoid models leverage normal 3D organ structure to recreate functional niche microenvironments. Our focus herein is to review the current state of the art in the use of 3D BM organoids, focusing on their capacities to recreate critical quality attributes of the in vivo BM microenvironment for the study of human normal and abnormal hematopoiesis.


Assuntos
Células da Medula Óssea , Medula Óssea , Adulto , Humanos , Hematopoese/fisiologia , Organoides , Nicho de Células-Tronco
2.
Adv Sci (Weinh) ; 9(23): e2200244, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35644929

RESUMO

Neuroblastoma (NB) is the most common extracranial tumor in children resulting in substantial morbidity and mortality. A deeper understanding of the NB tumor microenvironment (TME) remains an area of active research but there is a lack of reliable and biomimetic experimental models. This study utilizes a 3D bioprinting approach, in combination with NB spheroids, to create an in vitro vascular model of NB for exploring the tumor function within an endothelialized microenvironment. A gelatin methacryloyl (gelMA) bioink is used to create multi-channel cubic tumor analogues with high printing fidelity and mechanical tunability. Human-derived NB spheroids and human umbilical vein endothelial cells (HUVECs) are incorporated into the biomanufactured gelMA and cocultured under static versus dynamic conditions, demonstrating high levels of survival and growth. Quantification of NB-EC integration and tumor cell migration suggested an increased aggressive behavior of NB when cultured in bioprinted endothelialized models, when cocultured with HUVECs, and also as a result of dynamic culture. This model also allowed for the assessment of metabolic, cytokine, and gene expression profiles of NB spheroids under varying TME conditions. These results establish a high throughput research enabling platform to study the TME-mediated cellular-molecular mechanisms of tumor growth, aggression, and response to therapy.


Assuntos
Células Endoteliais da Veia Umbilical Humana , Neuroblastoma , Bioimpressão , Comunicação Celular , Criança , Gelatina , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Metacrilatos , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Impressão Tridimensional , Microambiente Tumoral
3.
Tissue Eng Part A ; 28(1-2): 38-53, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34130508

RESUMO

Cellular microenvironments provide stimuli, including paracrine and autocrine growth factors and physicochemical cues, which support efficient in vivo cell production unmatched by current in vitro biomanufacturing platforms. While three-dimensional (3D) culture systems aim to recapitulate niche architecture and function of the target tissue/organ, they are limited in accessing spatiotemporal information to evaluate and optimize in situ cell/tissue process development. Herein, a mathematical modeling framework is parameterized by single-cell phenotypic imaging and multiplexed biochemical assays to simulate the nonuniform tissue distribution of nutrients/metabolites and growth factors in cell niche environments. This model is applied to a bone marrow mimicry 3D perfusion bioreactor containing dense stromal and hematopoietic tissue with limited red blood cell (RBC) egress. The model characterized an imbalance between endogenous cytokine production and nutrient starvation within the microenvironmental niches and recommended increased cell inoculum density and enhanced medium exchange, guiding the development of a miniaturized prototype bioreactor. The second-generation prototype improved the distribution of nutrients and growth factors and supported a 50-fold increase in RBC production efficiency. This image-informed bioprocess modeling framework leverages spatiotemporal niche information to enhance biochemical factor utilization and improve cell manufacturing in 3D systems. Impact statement Three-dimensional (3D) culture systems are becoming increasingly important because they recapitulate the architecture and, consequently, physiological function of the target tissue/organ. Design and optimization of these 3D biomanufacturing platforms require evaluation of in situ spatiotemporal information. We have developed an integrated experimental-computational framework that captures the spatiotemporal distribution of cells, nutrients, and cytokines within a marrow biomimicry perfusion bioreactor. The model simulated biochemical factor utilization and guided the design of an improved second-generation bioreactor that achieved 50-fold increase in RBC production with improved cost efficiency. Such a modeling framework provides an essential platform for the optimization of 3D biomanufacturing systems.


Assuntos
Reatores Biológicos , Técnicas de Cultura de Células , Medula Óssea/metabolismo , Técnicas de Cultura de Células/métodos , Microambiente Celular , Perfusão
4.
Cancer Immunol Immunother ; 70(4): 1127-1142, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33033851

RESUMO

Cytokine storm can result from cancer immunotherapy or certain infections, including COVID-19. Though short-term immune-related adverse events are routinely described, longer-term immune consequences and sequential immune monitoring are not as well defined. In 2006, six healthy volunteers received TGN1412, a CD28 superagonist antibody, in a first-in-man clinical trial and suffered from cytokine storm. After the initial cytokine release, antibody effect-specific immune monitoring started on Day + 10 and consisted mainly of evaluation of dendritic cell and T-cell subsets and 15 serum cytokines at 21 time-points over 2 years. All patients developed problems with concentration and memory; three patients were diagnosed with mild-to-moderate depression. Mild neutropenia and autoantibody production was observed intermittently. One patient suffered from peripheral dry gangrene, required amputations, and had persistent Raynaud's phenomenon. Gastrointestinal irritability was noted in three patients and coincided with elevated γδT-cells. One had pruritus associated with elevated IgE levels, also found in three other asymptomatic patients. Dendritic cells, initially undetectable, rose to normal within a month. Naïve CD8+ T-cells were maintained at high levels, whereas naïve CD4+ and memory CD4+ and CD8+ T-cells started high but declined over 2 years. T-regulatory cells cycled circannually and were normal in number. Cytokine dysregulation was especially noted in one patient with systemic symptoms. Over a 2-year follow-up, cognitive deficits were observed in all patients following TGN1412 infusion. Some also had signs or symptoms of psychological, mucosal or immune dysregulation. These observations may discern immunopathology, treatment targets, and long-term monitoring strategies for other patients undergoing immunotherapy or with cytokine storm.


Assuntos
Anticorpos Monoclonais Humanizados/efeitos adversos , Antígenos CD28/agonistas , COVID-19/imunologia , Disfunção Cognitiva/imunologia , Síndrome da Liberação de Citocina/imunologia , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/imunologia , Imunoterapia/efeitos adversos , SARS-CoV-2/fisiologia , Linfócitos T/imunologia , Adulto , Anticorpos Monoclonais Humanizados/farmacologia , Disfunção Cognitiva/etiologia , Estudos de Coortes , Síndrome da Liberação de Citocina/etiologia , Seguimentos , Humanos , Masculino , Adulto Jovem
5.
Sci Adv ; 6(30): eaba5672, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32743074

RESUMO

The biological pathways that affect drug delivery in vivo remain poorly understood. We hypothesized that altering cell metabolism with phosphatidylinositol (3,4,5)-triphosphate (PIP3), a bioactive lipid upstream of the metabolic pathway PI3K (phosphatidylinositol 3-kinase)/AKT/ mTOR (mammalian target of rapamycin) would transiently increase protein translated by nanoparticle-delivered messenger RNA (mRNA) since these pathways increase growth and proliferation. Instead, we found that PIP3 blocked delivery of clinically-relevant lipid nanoparticles (LNPs) across multiple cell types in vitro and in vivo. PIP3-driven reductions in LNP delivery were not caused by toxicity, cell uptake, or endosomal escape. Interestingly, RNA sequencing and metabolomics analyses suggested an increase in basal metabolic rate. Higher transcriptional activity and mitochondrial expansion led us to formulate two competing hypotheses that explain the reductions in LNP-mediated mRNA delivery. First, PIP3 induced consumption of limited cellular resources, "drowning out" exogenously-delivered mRNA. Second, PIP3 triggers a catabolic response that leads to protein degradation and decreased translation.


Assuntos
Lipídeos , Nanopartículas , Fosfatos de Fosfatidilinositol , Lipossomos , Nanopartículas/metabolismo , Fosfatidilinositol 3-Quinases , Fosfatos de Fosfatidilinositol/metabolismo , RNA Mensageiro/genética
6.
Biomed Mater ; 15(6): 064101, 2020 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-32629436

RESUMO

In the present study we explore the extracellular matrix (ECM) produced by human bone marrow mesenchymal stem/stromal cells (BM-MSCs) induced to undergo osteogenic differentiation within porous chitosan/gelatin (CS:Gel) scaffolds by investigating their multiple gene expression profile and mechanical behavior. Initially, the efficiency of the BM-MSCs osteogenic differentiation within the constructs was confirmed by the significant rise in the expression of the osteogenesis associated genes DLX5, RUNX2, ALP and OSC. In line with these findings, OSC and Col1A1 protein expression was also detected in BM-MSCs on the CS:Gel scaffolds at day 14 of osteogenic differentiation. We then profiled, for the first time, the expression of 84 cell adhesion and ECM molecules using PCR arrays. The arrays, which were conducted at day 14 of osteogenic differentiation, demonstrated that 49 genes including collagens, integrins, laminins, ECM proteases, catenins, thrombospondins, ECM protease inhibitors and cell-cell adhesion molecules were differentially expressed in BM-MSCs seeded on scaffolds compared to tissue culture polystyrene control. Moreover, we performed dynamic mechanical analysis of the cell-loaded scaffolds on days 0, 7 and 14 to investigate the correlation between the biological results and the mechanical behavior of the constructs. Our data demonstrate a significant increase in the stiffness of the constructs with storage modulus values of 2 MPa on day 7, compared to 0.5 MPa on day 0, following a drop of the stiffness at 0.8 MPa on day 14, that may be attributed to the significant increase of specific ECM protease gene expression such as MMP1, MMP9, MMP11 and MMP16 at this time period.


Assuntos
Materiais Biocompatíveis/química , Quitosana/química , Gelatina/química , Perfilação da Expressão Gênica , Células-Tronco Mesenquimais/citologia , Células da Medula Óssea/citologia , Adesão Celular , Diferenciação Celular , Proliferação de Células , Colágeno/metabolismo , Matriz Extracelular/metabolismo , Humanos , Imunofenotipagem , Metaloproteinases da Matriz/biossíntese , Osteogênese , Poliestirenos/química , Pressão , Alicerces Teciduais/química , Transcriptoma
7.
Biotechnol Bioeng ; 117(6): 1761-1778, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32100872

RESUMO

Mesenchymal stromal cells (MSCs) have failed to consistently demonstrate their therapeutic efficacy in clinical trials, due in part to variability in culture conditions used for their production. Of various culture conditions used for MSC production, aggregate culture has been shown to improve secretory capacity (a putative mechanism of action in vivo) compared with standard monolayer culture. The purpose of this study was to perform multiomics characterization of MSCs cultured in monolayer and as aggregates to identify aspects of cell physiology that differ between these culture conditions to begin to understand cellular-level changes that might be related to secretory capacity. Targeted secretome characterization was performed on multiple batches of MSC-conditioned media, while nontargeted proteome and metabolome characterization was performed and integrated to identify cellular processes differentially regulated between culture conditions. Secretome characterization revealed a reduction in MSC batch variability when cultured as aggregates. Proteome and metabolome characterization showed upregulation of multiple protein and lipid metabolic pathways, downregulation of several cytoskeletal processes, and differential regulation of extracellular matrix synthesis. Integration of proteome and metabolome characterization revealed individual lipid metabolites and vesicle-trafficking proteins as key features for discriminating between culture conditions. Overall, this study identifies several aspects of MSC physiology that are altered by aggregate culture. Further exploration of these processes and pathways is needed to determine their potential role in regulating cell secretory capacity.


Assuntos
Técnicas de Cultura de Células/métodos , Células-Tronco Mesenquimais/metabolismo , Metaboloma , Proteoma , Agregação Celular , Células Cultivadas , Humanos , Células-Tronco Mesenquimais/citologia , Proteoma/análise , Proteoma/metabolismo
8.
Biotechnol J ; 14(11): e1800573, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31329373

RESUMO

The monoclonal antibody (mAb) industry is witnessing unprecedented growth, with an increasing range of new molecules and biosimilars as well as disease targets approved than ever before. Competition necessitates pharmaceutical companies to reduce development/production costs and time-to-market. To this aim, mathematical modeling can aid traditional experiment-only-based process development by reducing the design space, integrating scales, and assisting in identifying optimal operating conditions in less time and with lower expense. Mathematical models have been employed by other industries for control and optimization purposes and are important decisional tools for testing scenarios, process configurations, operating conditions, etc. Herein, a predictive, experimentally validated mathematical model that captures cellular metabolism and growth with cell cycle, cell death (apoptosis), and mAb production in GS-NS0 cells is presented. The model utilizes cellular, metabolic, and gene expression data, highlighting how multiple data sources can be integrated in one tool with the aim of optimizing mammalian cell bioprocessing.


Assuntos
Anticorpos Monoclonais/biossíntese , Apoptose , Ciclo Celular , Modelos Teóricos , Mieloma Múltiplo/metabolismo , Proteínas do Mieloma/metabolismo , Animais , Anticorpos Monoclonais/genética , Medicamentos Biossimilares , Linhagem Celular Tumoral , Técnicas de Cultura/métodos , Regulação Neoplásica da Expressão Gênica , Camundongos , Mieloma Múltiplo/genética
9.
Cell Death Dis ; 10(4): 277, 2019 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-30894508

RESUMO

Variability among donors, non-standardized methods for isolation, and characterization contribute to mesenchymal stem/stromal cell (MSC) heterogeneity. Induced pluripotent stem cell (iPSCs)-derived MSCs would circumvent many of current issues and enable large-scale production of standardized cellular therapy. To explore differences between native MSCs (nMSCs) and iPSC-derived MSCs (iMSCs), we developed isogeneic lines from Wharton's jelly (WJ) from the umbilical cords of two donors (#12 and #13) under xeno-free conditions. Next, we reprogrammed them into iPSCs (iPSC12 and iPSC13) and subsequently differentiated them back into iMSCs (iMSC12 and iMSC13) using two different protocols, which we named ARG and TEX. We assessed their differentiation capability, transcriptome, immunomodulatory potential, and interferon-γ (IFNG)-induced changes in metabolome. Our data demonstrated that although both differentiation protocols yield iMSCs similar to their parental nMSCs, there are substantial differences. The ARG protocol resulted in iMSCs with a strong immunomodulatory potential and lower plasticity and proliferation rate, whereas the TEX protocol raised iMSCs with a higher proliferation rate, better differentiation potential, though weak immunomodulatory response. Our data suggest that, following a careful selection and screening of donors, nMSCs from umbilical's cord WJ can be easily reprogrammed into iPSCs, providing an unlimited source of material for differentiation into iMSCs. However, the differentiation protocol should be chosen depending on their clinical use.


Assuntos
Diferenciação Celular/fisiologia , Reprogramação Celular/fisiologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Interferon gama/farmacologia , Células-Tronco Mesenquimais/metabolismo , Metaboloma/efeitos dos fármacos , Cordão Umbilical/citologia , Plasticidade Celular , Proliferação de Células , Células Cultivadas , Técnicas de Reprogramação Celular/métodos , Feminino , Humanos , Transcriptoma/efeitos dos fármacos
10.
Acta Biomater ; 88: 224-240, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30772514

RESUMO

Oxidized alginate hydrogels are appealing alternatives to natural alginate due to their favourable biodegradability profiles and capacity to self-crosslink with amine containing molecules facilitating functionalization with extracellular matrix cues, which enable modulation of stem cell fate, achieve highly viable 3-D cultures, and promote cell growth. Stem cell metabolism is at the core of cellular fate (proliferation, differentiation, death) and metabolomics provides global metabolic signatures representative of cellular status, being able to accurately identify the quality of stem cell differentiation. Herein, umbilical cord blood mesenchymal stem cells (UCB MSCs) were encapsulated in novel oxidized alginate hydrogels functionalized with the glycine-histidine-lysine (GHK) peptide and differentiated towards the osteoblastic lineage. The ADA-GHK hydrogels significantly improved osteogenic differentiation compared to gelatin-containing control hydrogels, as demonstrated by gene expression, alkaline phosphatase activity and bone extracellular matrix deposition. Metabolomics revealed the high degree of metabolic heterogeneity in the gelatin-containing control hydrogels, captured the enhanced osteogenic differentiation in the ADA-GHK hydrogels, confirmed the similar metabolism between differentiated cells and primary osteoblasts, and elucidated the metabolic mechanism responsible for the function of GHK. Our results suggest a novel paradigm for metabolomics-guided biomaterial design and robust stem cell bioprocessing. STATEMENT OF SIGNIFICANCE: Producing high quality engineered bone grafts is important for the treatment of critical sized bone defects. Robust and sensitive techniques are required for quality assessment of tissue-engineered constructs, which result to the selection of optimal biomaterials for bone graft development. Herein, we present a new use of metabolomics signatures in guiding the development of novel oxidised alginate-based hydrogels with umbilical cord blood mesenchymal stem cells and the glycine-histidine-lysine peptide, demonstrating that GHK induces stem cell osteogenic differentiation. Metabolomics signatures captured the enhanced osteogenesis in GHK hydrogels, confirmed the metabolic similarity between differentiated cells and primary osteoblasts, and elucidated the metabolic mechanism responsible for the function of GHK. In conclusion, our results suggest a new paradigm of metabolomics-driven design of biomaterials.


Assuntos
Diferenciação Celular , Sangue Fetal/metabolismo , Hidrogéis/química , Células-Tronco Mesenquimais/metabolismo , Metaboloma , Osteogênese , Peptídeos/química , Proliferação de Células , Matriz Extracelular/química , Sangue Fetal/citologia , Humanos , Teste de Materiais , Células-Tronco Mesenquimais/citologia , Metabolômica
11.
Biomaterials ; 188: 24-37, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30317113

RESUMO

Traditional culture systems for human erythropoiesis lack microenvironmental niches, spatial marrow gradients and dense cellularity rendering them incapable of effectively translating marrow physiology ex vivo. Herein, a bio-inspired three-dimensional (3D) perfusion bioreactor was engineered and inoculated with unselected single donor umbilical cord blood mononuclear cells (CBMNCs). Functional stromal and hematopoietic environments supporting long-term erythropoiesis were generated using defined medium supplemented only with stem cell factor (SCF) and erythropoietin (EPO) at near physiological concentrations. Quantitative 3D image analyses spatiotemporally mapped 21 multi-lineal cell distributions and interactions within multiple microenvironments that secreted extracellular matrix proteins and at least 16 endogenous hematopoietic and stromal growth factors. Tissue-like culture densities (≥2∙109 cells/mL), 1000-fold above flask cultures, were attained with continuous erythropoiesis and erythrocyte harvest. We propose this physiologically-relevant system for understanding normal and abnormal erythropoiesis, as well as for drug testing and/or discovery aimed at clinical translation.


Assuntos
Reatores Biológicos , Microambiente Celular , Eritrócitos/citologia , Eritropoese , Sangue Fetal/citologia , Medula Óssea/metabolismo , Técnicas de Cultura de Células/instrumentação , Técnicas de Cultura de Células/métodos , Hipóxia Celular , Proliferação de Células , Células Cultivadas , Eritrócitos/metabolismo , Eritropoetina/metabolismo , Humanos , Fator de Células-Tronco/metabolismo
12.
J Tissue Eng Regen Med ; 13(2): 232-243, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30537385

RESUMO

Biomimetic materials are essential for the production of clinically relevant bone grafts for bone tissue engineering applications. Their ability to modulate stem cell proliferation and differentiation can be used to harness the regenerative potential of those cells and optimize the efficiency of engineered bone grafts. The arginyl-glycyl-aspartic acid (RGD) peptide has been recognized as the adhesion motif of various extracellular matrix proteins and can affect stem cell behaviour in biomaterials. Attempts to functionalize biomaterials with RGD have been limited to a maximum of 1- to 3-mm thickness scaffolds, overlooking the issue of core infiltration that represents a major hurdle in developing real thickness scaffolds. Herein, we present the cross-linking of RGD on the surface of "real thickness" (5 × 5 × 5 mm) porous polyurethane scaffolds (PU-RGD), to be used for the expansion and osteogenic differentiation of umbilical cord blood mesenchymal stem cells (UCB MSCs). RGD-functionalized scaffolds increased initial cell adhesion (1.5-fold to twofold) and achieved a 3.4-fold increase in cell numbers at the end of culture compared with a 1.5-fold increase in non-functionalized controls. Homogenous cell infiltration to the scaffold core was observed in the PU-RGD scaffolds. Importantly, PU-RGD scaffolds were able to enhance the osteogenic differentiation of UCB MSCs. Osteogenic gene and protein expression increased in scaffolds functionalized with 100 µg/ml RGD. Higher RGD concentrations (200 µg/ml) were less efficient in stimulating osteogenic differentiation. We conclude that robust RGD tethering to 3D PU "real thickness" scaffolds is possible and that it promotes core infiltration, expansion, and osteogenic differentiation of UCB MSCs for the purposes of bone regeneration.


Assuntos
Diferenciação Celular , Sangue Fetal/metabolismo , Células-Tronco Mesenquimais/metabolismo , Oligopeptídeos/química , Osteogênese , Poliuretanos/química , Alicerces Teciduais/química , Sangue Fetal/citologia , Humanos , Células-Tronco Mesenquimais/citologia
13.
Stem Cells Int ; 2018: 6230214, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29760729

RESUMO

Tissue vasculature efficiently distributes nutrients, removes metabolites, and possesses selective cellular permeability for tissue growth and function. Engineered tissue models have been limited by small volumes, low cell densities, and invasive cell extraction due to ineffective nutrient diffusion and cell-biomaterial attachment. Herein, we describe the fabrication and testing of ceramic hollow fibre membranes (HFs) able to separate red blood cells (RBCs) and mononuclear cells (MNCs) and be incorporated into 3D tissue models to improve nutrient and metabolite exchange. These HFs filtered RBCs from human umbilical cord blood (CB) suspensions of 20% RBCs to produce 90% RBC filtrate suspensions. When incorporated within 5 mL of 3D collagen-coated polyurethane porous scaffold, medium-perfused HFs maintained nontoxic glucose, lactate, pH levels, and higher cell densities over 21 days of culture in comparison to nonperfused 0.125 mL scaffolds. This hollow fibre bioreactor (HFBR) required a smaller per-cell medium requirement and operated at cell densities > 10-fold higher than current 2D methods whilst allowing for continuous cell harvest through HFs. Herein, we propose HFs to improve 3D cell culture nutrient and metabolite diffusion, increase culture volume and cell density, and continuously harvest products for translational cell therapy biomanufacturing protocols.

14.
Metab Eng ; 47: 21-30, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29501926

RESUMO

Demand for high-value biologics, a rapidly growing pipeline, and pressure from competition, time-to-market and regulators, necessitate novel biomanufacturing approaches, including Quality by Design (QbD) principles and Process Analytical Technologies (PAT), to facilitate accelerated, efficient and effective process development platforms that ensure consistent product quality and reduced lot-to-lot variability. Herein, QbD and PAT principles were incorporated within an innovative in vitro-in silico integrated framework for upstream process development (UPD). The central component of the UPD framework is a mathematical model that predicts dynamic nutrient uptake and average intracellular ATP content, based on biochemical reaction networks, to quantify and characterize energy metabolism and its adaptive response, metabolic shifts, to maintain ATP homeostasis. The accuracy and flexibility of the model depends on critical cell type/product/clone-specific parameters, which are experimentally estimated. The integrated in vitro-in silico platform and the model's predictive capacity reduced burden, time and expense of experimentation resulting in optimal medium design compared to commercially available culture media (80% amino acid reduction) and a fed-batch feeding strategy that increased productivity by 129%. The framework represents a flexible and efficient tool that transforms, improves and accelerates conventional process development in biomanufacturing with wide applications, including stem cell-based therapies.


Assuntos
Anticorpos Monoclonais Murinos/biossíntese , Técnicas de Cultura de Células/métodos , Simulação por Computador , Meios de Cultura , Modelos Biológicos , Animais , Linhagem Celular Tumoral , Meios de Cultura/química , Meios de Cultura/farmacologia , Camundongos
15.
RSC Adv ; 8(37): 20928-20940, 2018 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-35542351

RESUMO

Pancreatic ductal adenocarcinoma is an aggressive disease with an extremely low survival rate. This is due to the (i) poor prognosis and (ii) high resistance of the disease to current treatment options. The latter is partly due to the very complex and dense tissue/tumour microenvironment of pancreatic cancer, which contributes to the disease's progression and the inhibition of apoptotic pathways. Over the last years, advances in tissue engineering and the development of three-dimensional (3D) culture systems have shed more light into cancer research by enabling a more realistic recapitulation of the niches and structure of the tumour microenvironment. Herein, for the first time, 3D porous polyurethane scaffolds were fabricated and coated with fibronectin to mimic features of the structure and extracellular matrix present in the pancreatic cancer tumour microenvironment. The developed 3D scaffold could support the proliferation of the pancreatic tumour cells, which was enhanced with the presence of fibronectin, for a month, which is a significantly prolonged in vitro culturing duration. Furthermore, in situ imaging of cellular and biomarker distribution showed the formation of dense cellular masses, the production of collagen-I by the cells and the formation of environmental stress gradients (e.g. HIF-1α) with similar heterogeneity trends to the ones reported in in vivo studies. The results obtained in this study suggest that this bioinspired porous polyurethane based scaffold has great potential for in vitro high throughput studies of pancreatic cancer including drug and treatment screening.

16.
Biotechnol J ; 12(9)2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28731612

RESUMO

Various studies have presented different approaches to direct pluripotent stem cell differentiation such as applying defined sets of exogenous biochemical signals and genetic/epigenetic modifications. Although differentiation to target lineages can be successfully regulated, such conventional methods are often complicated, laborious, and not cost-effective to be employed to the large-scale production of 3D stem cell-based tissue constructs. A 3D-culture platform that could realize the large-scale production of mesoderm lineage tissue constructs from embryonic stem cells (ESCs) is developed. ESCs are cultured using our previously established 3D-bioprocess platform which is amenable to mass-production of 3D ESC-based tissue constructs. Hepatocarcinoma cell line conditioned medium is introduced to the large-scale 3D culture to provide a specific biomolecular microenvironment to mimic in vivo mesoderm formation process. After 5 days of spontaneous differentiation period, the resulting 3D tissue constructs are composed of multipotent mesodermal progenitor cells verified by gene and molecular expression profiles. Subsequently the optimal time points to trigger terminal differentiation towards cardiomyogenesis or osteogenesis from the mesodermal tissue constructs is found. A simple and affordable 3D ESC-bioprocess that can reach the scalable production of mesoderm origin tissues with significantly improved correspondent tissue properties is demonstrated.


Assuntos
Reatores Biológicos , Diferenciação Celular/fisiologia , Células-Tronco Embrionárias , Mesoderma/metabolismo , Engenharia Tecidual/métodos , Animais , Linhagem Celular , Meios de Cultivo Condicionados , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Células Hep G2 , Humanos , Mesoderma/citologia , Camundongos , Osteogênese/fisiologia
17.
Biotechnol Prog ; 33(4): 966-988, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28401675

RESUMO

Current industrial trends encourage the development of sustainable, environmentally friendly processes with minimal energy and material consumption. In particular, the increasing market demand in biopharmaceutical industry and the tight regulations in product quality necessitate efficient operating procedures that guarantee products of high purity. In this direction, process intensification via continuous operation paves the way for the development of novel, eco-friendly processes, characterized by higher productivity and lower production costs. This work focuses on the development of advanced control strategies for (i) a cell culture system in a bioreactor and (ii) a semicontinuous purification process. More specifically, we consider a fed-batch culture of GS-NS0 cells and the semicontinuous Multicolumn Countercurrent Solvent Gradient Purification (MCSGP) for the purification process. The controllers are designed following the PAROC framework/software platform and their capabilities are assessed in silico, against the process models. It is demonstrated that the proposed controllers efficiently manage to increase the system productivity, returning strategies that can lead to continuous, stable process operation. © 2017 American Institute of Chemical Engineers Biotechnol. Prog., 33:966-988, 2017.


Assuntos
Anticorpos Monoclonais/biossíntese , Simulação por Computador , Animais , Anticorpos Monoclonais/isolamento & purificação , Reatores Biológicos , Técnicas de Cultura de Células , Camundongos , Software , Células Tumorais Cultivadas
18.
Stem Cells Dev ; 26(10): 723-733, 2017 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-28418785

RESUMO

Mesenchymal stem cells (MSCs) of fetal origin, such as umbilical cord blood MSCs (UCB MSCs), have emerged as a promising cell source for musculoskeletal tissue regeneration because of their higher proliferation potential, lack of donor site morbidity, and their off-the-shelf potential. MSCs differentiated toward the osteogenic lineage exhibit a specific metabolic phenotype characterized by reliance to oxidative phosphorylation for energy production and reduced glycolytic rates. Currently, limited information exists on the metabolic transitions at different stages of the osteogenic process after osteoinduction with different agents. Herein, the osteoinduction efficiency of BMP-2 and dexamethasone on UCB MSCs was assessed using gas chromatography-mass spectrometry (GC-MS) metabolomics analysis, revealing metabolic discrepancies at 7, 14, and 21 days of induction. Whereas both agents when administered individually were able to induce collagen I, osteocalcin, and osteonectin expression, BMP-2 was less effective than dexamethasone in promoting alkaline phosphatase expression. The metabolomics analysis revealed that each agent induced distinct metabolic alterations, including changes in amino acid pools, glutaminolysis, one-carbon metabolism, glycolysis, and tricarboxylic acid cycle. Importantly, we showed that in vitro-differentiated UCB MSCs acquire a metabolic physiology similar to primary osteoblasts when induced with dexamethasone but not with BMP-2, highlighting the fact that metabolomics analysis is sensitive enough to reveal potential differences in the osteogenic efficiency and can be used as a quality control assay for evaluating the osteogenic process.


Assuntos
Proteína Morfogenética Óssea 2/farmacologia , Diferenciação Celular , Células-Tronco Mesenquimais/citologia , Metaboloma , Osteoblastos/citologia , Fosfatase Alcalina/genética , Fosfatase Alcalina/metabolismo , Células Cultivadas , Ciclo do Ácido Cítrico , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Dexametasona/farmacologia , Glicólise , Humanos , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Osteoblastos/metabolismo , Osteocalcina/genética , Osteocalcina/metabolismo , Osteonectina/genética , Osteonectina/metabolismo , Cordão Umbilical/citologia
19.
Sci Rep ; 7: 42138, 2017 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-28165055

RESUMO

Human pluripotent stem cells (hPSCs) are adhesion-dependent cells that require cultivation in colonies to maintain growth and pluripotency. Robust differentiation protocols necessitate single cell cultures that are achieved by use of ROCK (Rho kinase) inhibitors. ROCK inhibition enables maintenance of stem cell phenotype; its effects on metabolism are unknown. hPSCs were exposed to 10 µM ROCK inhibitor for varying exposure times. Pluripotency (TRA-1-81, SSEA3, OCT4, NANOG, SOX2) remained unaffected, until after prolonged exposure (96 hrs). Gas chromatography-mass spectrometry metabolomics analysis identified differences between ROCK-treated and untreated cells as early as 12 hrs. Exposure for 48 hours resulted in reduction in glycolysis, glutaminolysis, the citric acid (TCA) cycle as well as the amino acids pools, suggesting the adaptation of the cells to the new culture conditions, which was also reflected by the expression of the metabolic regulators, mTORC1 and tp53 and correlated with cellular proliferation status. While gene expression and protein levels did not reveal any changes in the physiology of the cells, metabolomics revealed the fluctuating state of the metabolism. The above highlight the usefulness of metabolomics in providing accurate and sensitive information on cellular physiological status, which could lead to the development of robust and optimal stem cell bioprocesses.


Assuntos
Amidas/farmacologia , Células-Tronco Embrionárias/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Metaboloma , Piridinas/farmacologia , Quinases Associadas a rho/genética , Antígenos de Superfície/genética , Antígenos de Superfície/metabolismo , Antígenos Glicosídicos Associados a Tumores/genética , Antígenos Glicosídicos Associados a Tumores/metabolismo , Biomarcadores/metabolismo , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Expressão Gênica , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Proteína Homeobox Nanog/genética , Proteína Homeobox Nanog/metabolismo , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Fenótipo , Espécies Reativas de Nitrogênio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Antígenos Embrionários Estágio-Específicos/genética , Antígenos Embrionários Estágio-Específicos/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Quinases Associadas a rho/antagonistas & inibidores , Quinases Associadas a rho/metabolismo
20.
Drug Discov Today ; 22(4): 690-701, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28153670

RESUMO

Pancreatic cancer is one of the most aggressive and lethal human malignancies. Drug therapies and radiotherapy are used for treatment as adjuvants to surgery, but outcomes remain disappointing. Advances in tissue engineering suggest that 3D cultures can reflect the in vivo tumor microenvironment and can guarantee a physiological distribution of oxygen, nutrients, and drugs, making them promising low-cost tools for therapy development. Here, we review crucial structural and environmental elements that should be considered for an accurate design of an ex vivo platform for studies of pancreatic cancer. Furthermore, we propose environmental stress response biomarkers as platform readouts for the efficient control and further prediction of the pancreatic cancer response to the environmental and treatment input.


Assuntos
Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Neoplasias Pancreáticas/tratamento farmacológico , Animais , Biomimética/métodos , Humanos , Engenharia Tecidual/métodos , Microambiente Tumoral/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA