77 research outputs found

    Enginnering cardiac tissue using human induced pluripotent stem cell derivatives: Proteomic characterization of co-cultures of cardiomyocytes and endothelial cells

    Get PDF
    Prediction of cardiac toxicity effect is extremely relevant in the development of new drugs for different medical applications. In this way, it is important to develop more predictable human cell-based models which physiologically better mimic the human heart and allow the prediction of this toxic effect as well as establish the tools that enable the characterization of these complex cell models. To recreate engineered cardiac tissue, it is essential to reproduce the complexity of the heart by resorting to different cell types. Cardiomyocytes (CMs) are functional contractile units of the heart, and it is known that their communication with endothelial cells (ECs) is crucial for cardiac homeostasis. The aim of this study is to recreate a human pluripotent stem cells (hiPSC)-based cardiac tissue model and evaluate the impact of communication between both cell types on the phenotype of CMs. Co-cultures of hiPSC-CM and hiPSC-EC were established and maintained for 12 days as confirmed by immunofluorescence microscopy. Quantitative whole-proteome analysis was performed using SWATH Mass Spectrometry tools to compare the conditions of hiPSC-CM mono-culture and the co-culture of hiPSC-CM and hiPSC-EC. Our data showed relative increase of expression ratios of morphological maturation-related cardiac proteins in hiPSC-CM co-cultures. In particular, the expression ratios of MYH7/MYH6, MYL2/MYL7, TNNI3/TNNI1 increased 2.4-, 5.1-, and 5-fold, respectively, when compared to the mono-culture condition, indicating that in the presence of hiPSC-EC, hPSC-CM display a more adult- and ventricular- like phenotype. Changes in the extracellular matrix composition were also observed, especially related with the increased expression of ECM proteins in co-culture condition namely, collagens I and III (8.6-fold and 6-fold, respectively), fibronectin (3.5-fold) and thrombospondin-4 (2.5-fold). Other growth factors attributed to the extracellular space (e.g. CTGF, PAI1, CRTAP, IGFBP7, and NPPB) that may be responsible for the communication between both cell types have also shown to be up-regulated in the co-culture condition. The presence of a SMA+ (myofibroblast-like) population in the co-culture condition was observed by immunofluorescence microscopy images, which is in agreement with the more complex and fibrotic extracellular matrix found by whole proteome analysis. Ultrastructure characterization of CMs was carried out by transmission electron microscopy. In both conditions, hiPSC-CM displayed aligned myofibrils composed by sarcomeres with organized Z-disks, A- and I-bands, intercalated discs between adjacent cells as well as abundant mitochondria. Noteworthy, sarcomere length was higher in hiPSC-CM cultured with hiPSC-EC, suggesting structural changes associated with cardiomyocyte maturation. Calcium imaging is being performed to evaluate calcium handling of hiPSC-CMs and their response to drugs. All together our data revealed that promoting the communication of hiPSC-CM and hiPSC-EC induced structural changes in hiPSC-CM associated with maturation. This study provides important insights towards the development of more complex cardiac tissues and establishes potent analytical tools for the characterization of these models. This work was supported by Fundação para a Ciência e Tecnologia (FCT)-funded project CARDIOSTEM (MITP-TB/ECE/0013/2013); and iNOVA4Health UID/Multi/04462/2013, a program supported by FCT/Ministério da Educação e Ciência, through national funds and cofounded by FEDER under the PT2020 Partnership Agreement. BA. was supported by FCT Grant SFRH/BD/52475/201

    Dielectric spectroscopy monitoring of a bioreactor process for hiPSC expansion and differentiation

    Get PDF
    Bioprocessing strategies using 3D cell culturing approaches, such as cell aggregates, are promising solutions to achieve efficient and scalable bioprocesses for stem cell expansion and differentiation. However, tracking viable and total cell numbers in such culture systems is not straightforward. It requires cell detachment, disaggregation or disruption, which results in measurements that are laborious, biased and with high variability. In this work, we used a commercially available capacitance probe to explore the applicability of dielectric spectroscopy for in situ monitoring of a multistep process for expansion and differentiation of human induced pluripotent stem cells (hiPSC) cultivated as cell aggregates. After 5 days of cell expansion in a bioreactor, the hepatic differentiation step was integrated by addition of different levels of specific soluble factors at various stages of the process to promote growth and generate populations successively enriched for definitive endoderm, hepatoblasts, hepatocyte progenitors and mature hepatocytes. While this differentiation procedure has been previously validated for monolayer cultures, this was the first time it was carried out in a stirred tank bioreactor operated in perfusion mode. Phenotype analysis confirmed a marked increase in key hepatic differentiation markers culminating at day 21 of differentiation. Our data shows a good correlation between total volume of the cell aggregates and permittivity measured by the probe (R2 = 0.84). However, there was a delay between changes in cell concentration and the permittivity signal. This suggests that cell expansion requires a few days to result in increased volume of the cell aggregates and that each aggregate behaves as one overall inducible dipole. The β-dispersion curve shape also appears to change over culture time and could eventually be used as an indicator for differentiation progression. Dielectric spectroscopy has been used successfully to monitor viable cell concentration in different single-cell suspension cultures, but there are few published applications to 3D cultures. Our results demonstrate the potential of dielectric spectroscopy to monitor complex bioprocesses for human stem cell aggregates in stirred cultures. Acknowledgements: Funding provided by ERA-NET/E-Rare3 programme through research project ERAdicatPH (E-Rare3/0002/2015). The authors acknowledge Dr Juan Rodriguez-Madoz (University of Navarra, Spain) and Dr Anders Aspegren (Takara Bio Europe – Cellartis AB, Sweden) for helpful discussions on hepatic differentiation of hiPSC.

    Bioprocess intensification for the continuous expansion of 3D human induced pluripotent stem cell aggregates in bioreactors

    Get PDF
    Human induced pluripotent stem cells (hiPSC) are attractive tools for drug screening and disease modeling and promising candidates for cell therapy applications. However, to achieve the high numbers of cells required for these purposes, scalable and clinical-grade technologies must be established. In this study, we use environmentally controlled stirred-tank bioreactors operating in perfusion as a powerful tool for bioprocess intensification of hiPSC production. Firstly, we demonstrate the importance of controlling the dissolved oxygen concentration at low levels (4% oxygen) and perfusion at 1.3 day-1 dilution rate to improve hiPSC growth as 3D aggregates in xeno-free medium (Cellartis® DEF-CS™ 500 Xeno-Free Culture Medium). This strategy allowed for increased cell specific growth rate, maximum volumetric cell concentrations (4.7x106 cell/mL) and expansion factors (approximately 19), resulting in an overall improvement of 2.6-fold in cell yields. Extensive cell characterization, including whole proteomic analysis was performed to confirm that the pluripotent phenotype was maintained during culture. Secondly, we have tested different chemical and mechanical strategies for hiPSC aggregate dissociation, revealing similar viable cell recovery yields (approximately 50%). However, only the mechanical dissociation strategies enabled the re-aggregation of hiPSC in stirred conditions, with the mechanical dissociation using a 70 μm pore size nylon mesh allowing a higher expansion factor after dissociation. Finally, a scalable protocol for continuous expansion of hiPSC aggregates in bioreactors was implemented using the mechanical dissociation for aggregate disruption/passaging. A total expansion factor of 1100 in viable cells was obtained in 11 days of culture after 3 sequential passages in bioreactors, while cells maintained their proliferation capacity, pluripotent phenotype and potential as well as genomic stability. To our knowledge, this is the highest expansion factor reported for hiPSC for such a short culture time frame. The strategy described herein for continuous expansion of hiPSC provides important insights towards up-scaling the production of hiPSC. Integrative biomanufacturing processes using this continuous strategy are now being pursued for hiPSC expansion and differentiation towards cardiac lineages in order to recreate cardiac models for drug discovery, toxicity testing and disease modeling. Acknowledgments: This work was supported by Fundação para a Ciência e Tecnologia (FCT)-funded projects CARDIOSTEM (MITP-TB/ECE/0013/2013) and CardioRegen (HMSP-ICT/0039/2013); and iNOVA4Health UID/Multi/04462/2013, a program supported by FCT/Ministério da Educação e Ciência, through national funds and cofounded by FEDER under the PT2020 Partnership Agreement. BA. was supported by FCT Grant SFRH/BD/52475/2013. The work was also funded by a Vinnova Grant, registration number 2014-00310 (Cell therapies via large scale expansion of pluripotent stem cells)

    Expansion of 3D human induced pluripotent stem cell aggregates in bioreactors: Bioprocess intensification and scaling-up approaches

    Get PDF
    Human induced pluripotent stem cells (hiPSC) are attractive tools for drug screening and disease modeling and promising candidates for cell therapy applications. However, to achieve the high numbers of cells required for these purposes, scalable and clinical-grade technologies must be established. In this study, we use environmentally controlled stirred-tank bioreactors operating in perfusion as a powerful tool for bioprocess intensification of hiPSC production. We demonstrate the importance of controlling the dissolved oxygen concentration at low levels (4% oxygen) and perfusion at 1.3 day-1 dilution rate to improve hiPSC growth as 3D aggregates in xeno-free medium (Cellartis® DEF-CS™ 500 Xeno-Free Culture Medium). This strategy allowed for increased cell specific growth rate, maximum volumetric cell concentrations (4.7x106 cell/mL) and expansion factors (approximately 19), resulting in an overall improvement of 2.6-fold in cell yields. Extensive cell characterization, including whole proteomic analysis was performed to confirm that the pluripotent phenotype was maintained during culture. Furthermore, a scalable protocol for continuous expansion of hiPSC aggregates in bioreactors was implemented using mechanical dissociation protocols for aggregate disruption and cell passaging. A total expansion factor of 1100 in viable cells was obtained in 11 days of culture (Figure 1), while cells maintained their proliferation capacity, pluripotent phenotype and potential as well as genomic stability after 3 sequential passages in bioreactors. To our knowledge, this is the highest expansion factor reported for hiPSC for such a short culture time frame. The strategy described herein for continuous expansion of hiPSC provides important insights towards up-scale production of hiPSC. This will strengthen the utility of hiPSC in cell therapy, drug discovery, toxicity testing and disease modeling. Please click Additional Files below to see the full abstract

    Toward a Microencapsulated 3D hiPSC-Derived in vitro Cardiac Microtissue for Recapitulation of Human Heart Microenvironment Features

    Get PDF
    SAICTPAC/0047/2015 PTDC/BTMSAL/32566/ 2017 PTDC/MEC-CAR/29590/2017 UIDB/04462/2020 UIDP/04462/2020 H2020, ID:874827 SFRH/BD/52475/2013 SFRH/BPD/120595/2016The combination of cardiomyocytes (CM) and non-myocyte cardiac populations, such as endothelial cells (EC), and mesenchymal cells (MC), has been shown to be critical for recapitulation of the human heart tissue for in vitro cell-based modeling. However, most of the current engineered cardiac microtissues still rely on either (i) murine/human limited primary cell sources, (ii) animal-derived and undefined hydrogels/matrices with batch-to-batch variability, or (iii) culture systems with low compliance with pharmacological high-throughput screenings. In this work, we explored a culture platform based on alginate microencapsulation and suspension culture systems to develop three-dimensional (3D) human cardiac microtissues, which entails the co-culture of human induced pluripotent stem cell (hiPSC) cardiac derivatives including aggregates of hiPSC–CM and single cells of hiPSC–derived EC and MC (hiPSC–EC+MC). We demonstrate that the 3D human cardiac microtissues can be cultured for 15 days in dynamic conditions while maintaining the viability and phenotype of all cell populations. Noteworthy, we show that hiPSC–EC+MC survival was promoted by the co-culture with hiPSC–CM as compared to the control single-cell culture. Additionally, the presence of the hiPSC–EC+MC induced changes in the physical properties of the biomaterial, as observed by an increase in the elastic modulus of the cardiac microtissue when compared to the hiPSC–CM control culture. Detailed characterization of the 3D cardiac microtissues revealed that the crosstalk between hiPSC–CM, hiPSC–EC+MC, and extracellular matrix induced the maturation of hiPSC–CM. The cardiac microtissues displayed functional calcium signaling and respond to known cardiotoxins in a dose-dependent manner. This study is a step forward on the development of novel 3D cardiac microtissues that recapitulate features of the human cardiac microenvironment and is compliant with the larger numbers needed in preclinical research for toxicity assessment and disease modeling.publishersversionpublishe

    Human Extracellular-Matrix Functionalization of 3D hiPSC-Based Cardiac Tissues Improves Cardiomyocyte Maturation

    Get PDF
    The work here presented was funded by Fundacao para a Ciencia e Tecnologia (FCT) projects NETDIAMOND (SAICTPAC/0047/2015), financially supported by FEEI-Lisboa2020 and FCT/POCI-01-0145-FEDER-016385, and MetaCardio (PTDC/BTM-SAL/32566/2017); iNOVA4-Health -UIDB/04462/2020 and UIDP/04462/2020, a program financially supported by FCT/Ministerio da Ciencia, Tecnologia e Ensino Superior, through national funds is acknowledged; Funding from INTERFACE Programme, through the Innovation, Technology and Circular Economy Fund (FITEC), is gratefully acknowledged; and EU-funded projects BRAV3 (H2020, ID:874827) and ERAatUC (ref. 669088). HVA, AFL, and DS were financed by FCT Grants SFRH/BPD/120595/2016 and PD/BD/139078/2018 and PD/BD/106051/2015, respectively.Human induced pluripotent stem cells (hiPSC) possess significant therapeutic potential due to their high self-renewal capability and potential to differentiate into specialized cells such as cardiomyocytes. However, generated hiPSC-derived cardiomyocytes (hiPSC-CM) are still immature, with phenotypic and functional features resembling the fetal rather than their adult counterparts, which limits their application in cell-based therapies, in vitro cardiac disease modeling, and drug cardiotoxicity screening. Recent discoveries have demonstrated the potential of the extracellular matrix (ECM) as a critical regulator in development, homeostasis, and injury of the cardiac microenvironment. Within this context, this work aimed to assess the impact of human cardiac ECM in the phenotype and maturation features of hiPSC-CM. Human ECM was isolated from myocardium tissue through a physical decellularization approach. The cardiac tissue decellularization process reduced DNA content significantly while maintaining ECM composition in terms of sulfated glycosaminoglycans (s-GAG) and collagen content. These ECM particles were successfully incorporated in three-dimensional (3D) hiPSC-CM aggregates (CM+ECM) with no impact on viability and metabolic activity throughout 20 days in 3D culture conditions. Also, CM+ECM aggregates displayed organized and longer sarcomeres, with improved calcium handling when compared to hiPSC-CM aggregates. This study shows that human cardiac ECM functionalization of hiPSC-based cardiac tissues improves cardiomyocyte maturation. The knowledge generated herein provides essential insights to streamline the application of ECM in the development of hiPSC-based therapies targeting cardiac diseases.publishersversionpublishe

    Assessment of potato peel and agro-forestry biochars supplementation on in vitro ruminal fermentation

    Get PDF
    UIDB/50006/2020 UIDB/04033/2020 grant ref. PDE/BDE/114434/2016 DL 57/2016 -Norma transitória.Background. The awareness of environmental and socio-economic impacts caused by greenhouse gas emissions from the livestock sector leverages the adoption of strategies to counteract it. Feed supplements can play an important role in the reduction of the main greenhouse gas produced by ruminants-methane (CH4). In this context, this study aims to assess the effect of two biochar sources and inclusion levels on rumen fermentation parameters in vitro. Methods. Two sources of biochar (agro-forestry residues, AFB, and potato peel, PPB) were added at two levels (5 and 10%, dry matter (DM) basis) to two basal substrates (haylage and corn silage) and incubated 24-h with rumen inocula to assess the effects on CH4 production and main rumen fermentation parameters in vitro. Results. AFB and PPB were obtained at different carbonization conditions resulting in different apparent surface areas, ash content, pH at the point of zero charge (pHpzc), and elemental analysis. Relative to control (0% biochar), biochar supplementation kept unaffected total gas production and yield (mL and mL/g DM, pD0.140 and pD0.240, respectively) and fermentation pH (p D 0.666), increased CH4 production and yield (mL and mL/g DM, respectively, pD0.001) and ammonia-N (NH3-N, pD0.040), and decreased total volatile fatty acids (VFA) production (p < 0.001) and H2 generated and consumed (p ≤ 0.001). Biochar sources and inclusion levels had no negative effect on most of the fermentation parameters and efficiency. Acetic.propionic acid ratio (pD0.048) and H2 consumed (pD0.019) were lower with AFB inclusion when compared to PPB. Biochar inclusion at 10% reduced H2 consumed (p < 0.001) and tended to reduce total gas production (pD0.055). Total VFA production (pD0.019), acetic acid proportion (pD0.011) and H2 generated (pD0.048) were the lowest with AFB supplemented at 10%, no differences being observed among the other treatments. The basal substrate affected most fermentation parameters independently of biochar source and level used. Discussion. Biochar supplementation increased NH3-N content, iso-butyric, iso-valeric and valeric acid proportions, and decreased VFA production suggesting a reduced energy supply for microbial growth, higher proteolysis and deamination of substrate N, and a decrease of NH3-N incorporation into microbial protein. No interaction was found between substrate and biochar source or level on any of the parameters measured. Although AFB and PPB had different textural and compositional characteristics, their effects on the rumen fermentation parameters were similar, the only observed effects being due to AFB included at 10%. Biochar supplementation promoted CH4 production regardless of the source and inclusion level, suggesting that there may be other effects beyond biomass and temperature of production of biochar, highlighting the need to consider other characteristics to better identify the mechanism by which biochar may influence CH4 production.publishersversionpublishe

    Intensifying the manufacture of hiPSC therapy products through metabolic and process understanding

    Get PDF
    In vitro differentiation of human induced pluripotent stem cells into specific lineages such as cardiomyocytes (hPSC-CM) and hepatocytes (hPCS-Hep) is a crucial process to enable their application in cell therapy and drug discovery. Nevertheless, despite the remarkable efforts over the last decade towards the implementation of protocols for hPSC expansion and differentiation, there are some technological challenges remaining include the low scalability and differentiation yields. Additionally, generated cells are still immature, closely reminiscent of fetal/embryonic cells in what regards phenotype and function. In this study, we aim to overcome this hurdle by devising bioinspired and integrated strategies to improve the generation and functionality of these hiPSC-derivatives. We also applied robust multi-parametric techniques including proteomics, transcriptomics, metabolomics and fluxomics as complementary analytical tools to support bioprocess optimization and product characterization. We cultured hiPSC as 3D aggregates in stirred-tank bioreactors (STB) operated in perfusion and used a capacitance probe for in situ monitoring of cell growth/differentiation. After cell expansion, the hepatic differentiation step was integrated by addition of key soluble factors and controlling the dissolved oxygen concentration at various stages of the process to generate populations enriched for definitive endoderm, hepatocyte progenitors and mature hepatocytes. The analyses of hepatic markers expression throughout the stages of the differentiation confirmed that hepatocyte differentiation was improved in 3D spheroids when compared to 2D culture. Noteworthy, these hiPSC-HLC exhibited functional characteristics typical of hepatocytes (albumin production, glycogen storage and CYP450 activity). We also demonstrate the potential of dielectric spectroscopy to monitor cell expansion and hepatic differentiation in STB. For CM differentiation, we relied on the aggregation of hPSC-derived cardiac progenitors to establish a scalable differentiation protocol capable of generating highly pure CM aggregate cultures. We assessed if alteration of culture medium composition to mimic in vivo substrate usage during cardiac development improved further hPSC-CM maturation in vitro. Our results showed that shifting hPSC-CMs from glucose-containing to galactose- and fatty acid-containing medium promotes their fast maturation into adult-like CMs with higher oxidative metabolism, transcriptional signatures closer to those of adult ventricular tissue, higher myofibril density and alignment, improved calcium handling, enhanced contractility, and more physiological action potential kinetics. “-Omics” analyses showed that addition of galactose to culture medium and culturing the cells under perfusion improves total oxidative capacity of the cells and ameliorates fatty acid oxidation. This study demonstrated that metabolic shifts during differentiation/maturation of hPSC-CM are a cause, rather than a consequence, of the phenotypic and functional alterations observed. The metabolic-based strategy established herein holds technical and economic advantages over the existing protocols due to its scalability, simplicity and ease of application. Funding: This work was supported by FCT-funded projects NETDIAMOND (SAICTPAC/0047/2015), MetaCardio (Ref.032566) and FCT/ERA-Net (ERAdicatPH; Ref. E-Rare3/0002/2015). iNOVA4Health Research Unit (LISBOA-01-0145-FEDER-007344) is also acknowledged

    Maspin differential expression patterns as a potential marker for targeted screening of esophageal adenocarcinoma/gastroesophageal junction adenocarcinoma.

    Get PDF
    Barrett's esophagus (BE) is a predisposing factor of esophageal adenocarcinoma/gastroesophageal junction adenocarcinoma (ECA/GEJ Aca). BE patients are stratified and subsequently monitored according to the risk of malignant progression by the combination of endoscopy and biopsy. This study is to evaluate the maspin expression patterns as early diagnostic markers of malignancy in BE patients. Immunohistochemistry (IHC) staining was performed on 62 archival core biopsies from 35 patients, including BE without dysplasia (intestinal metaplasia, IM), BE with low grade dysplasia, BE with high grade dysplasia, carcinoma in situ, and well to poorly differentiated ECA/GEJ Aca (PD-ECA/GEJ Aca). The intensity and the subcellular distribution of immunoreactivity were evaluated microscopically. Statistical analysis was performed using the χ2 and Fisher exact tests. The level of epithelial-specific tumor suppressor maspin protein inversely correlated with the progression from IM to PD-ECA/GEJ Aca. Lesions of each pathological grade could be divided into subtypes that exhibited distinct maspin subcellular distribution patterns, including nuclear only (Nuc), combined nuclear and cytoplasmic (Nuc+Cyt), cytoplasmic only (Cyt) and overall negligible (Neg). The Cyt subtype, which was minor in both IM and dysplasia (approximately 10%), was predominant in ECA/GEJ Aca as early as well-differentiated lesions (more than 50%: p = 0.0092). In comparison, nuclear staining of the tumor suppressor TP53 was heterogeneous in dysplasia, and did not correlate with the differentiation grades of ECA/GEJ Aca. The Cyt subtype of maspin expression pattern in core biopsies of BE patients may serve as a molecular marker for early diagnosis of ECA/GEJ Aca.This work was supported by the NIH grant P30CA022453 (to the Karmanos Cancer Institute with Sheng, S. as a program leader), the Ruth Sager Memorial Fund (to Sheng, S.), the Karmanos Cancer Institute Pilot Project Grant 25S5Z (to Sheng, S.), and the Karmanos Cancer Institute Prostate Cancer Research Pilot Project Grant (to Sheng, S.)

    CRUSE®-An innovative mobile application for patient monitoring and management in chronic spontaneous urticaria

    Get PDF
    Background: Chronic spontaneous urticaria (CSU) is unpredictable and can severely impair patients' quality of life. Patients with CSU need a convenient, user-friendly platform to complete patient-reported outcome measures (PROMs) on their mobile devices. CRUSE ®, the Chronic Urticaria Self Evaluation app, aims to address this unmet need. Methods: CRUSE ® was developed by an international steering committee of urticaria specialists. Priorities for the app based on recent findings in CSU were defined to allow patients to track and record their symptoms and medication use over time and send photographs. The CRUSE ® app collects patient data such as age, sex, disease onset, triggers, medication, and CSU characteristics that can be sent securely to physicians, providing real-time insights. Additionally, CRUSE ® contains PROMs to assess disease activity and control, which are individualised to patient profiles and clinical manifestations. Results: CRUSE ® was launched in Germany in March 2022 and is now available for free in 17 countries. It is adapted to the local language and displays a country-specific list of available urticaria medications. English and Ukrainian versions are available worldwide. From July 2022 to June 2023, 25,710 observations were documented by 2540 users; 72.7% were females, with a mean age of 39.6 years. At baseline, 93.7% and 51.3% of users had wheals and angioedema, respectively. Second-generation antihistamines were used in 74.0% of days. Conclusions: The initial data from CRUSE ® show the wide use and utility of effectively tracking patients' disease activity and control, paving the way for personalised CSU management.</p
    corecore