Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Appl Toxicol ; 44(3): 333-343, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37699698

ABSTRACT

The HUMIMIC skin-liver Chip2 microphysiological systems model using the epidermal model, EpiDerm™, was reported previously to mimic application route-dependent metabolism of the hair dye, 4-amino-2-hydroxytoluene (AHT). Therefore, we evaluated the use of alternative skin models-SkinEthic™, EpiDermFT™ and PhenionFT™-for the same purpose. In static incubations, AHT permeation was similar using SkinEthic™ and EpiDerm™ models. Older Day 21 (D21) SkinEthic™ models with a thicker stratum corneum did not exhibit a greater barrier to AHT (overall permeation was the same in D17 and D21 models). All epidermal models metabolised AHT, with the EpiDerm™ exhibiting higher N-acetylation than SkinEthic™ models. AHT metabolism by D21 SkinEthic™ models was lower than that by D17 SkinEthic™ and EpiDerm™ models, thus a thicker stratum corneum was associated with fewer viable cells and a lower metabolic activity. AHT permeation was much slower using PhenionFT™ compared to epidermal models and better reflected permeation of AHT through native human skin. This model also extensively metabolised AHT to N-acetyl-AHT. After a single topical or systemic application of AHT to Chip2 model with PhenionFT™, medium was analysed for parent and metabolites over 5 days. The first-pass metabolism of AHT was demonstrated, and the introduction of a wash step after 30 min decreased the exposure to AHT and its metabolites by 33% and 40%-43%, respectively. In conclusion, epidermal and FT skin models used in the Chip2 can mimic the first-pass skin metabolism of AHT. This highlights the flexibility of the Chip2 to incorporate different skin models according to the purpose.


Subject(s)
Cresols , Hair Dyes , Humans , Hair Dyes/metabolism , Skin/metabolism , Aniline Compounds/metabolism , Liver
2.
J Appl Toxicol ; 44(2): 287-300, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37700462

ABSTRACT

The HUMMIC skin-liver Chip2 microphysiological system using EpiDerm™ and HepaRG and stellate liver spheroids was used to evaluate the route-specific metabolism and toxicodynamic effects of genistein. Human-relevant exposure levels were compared: 60 nM representing the plasma concentration expected after topical application of a cosmetic product and 1 µM representing measured plasma concentrations after ingesting soya products. Genistein was applied as single and repeated topical and/or systemic doses. The kinetics of genistein and its metabolites were measured over 5 days. Toxicodynamic effects were measured using transcriptional analyses of skin and liver organoids harvested on Days 2 and 5. Route-specific differences in genistein's bioavailability were observed, with first-pass metabolism (sulfation) occurring in the skin after topical application. Only repeated application of 1 µM, resembling daily oral intake of soya products, induced statistically significant changes in gene expression in liver organoids only. This was concomitant with a much higher systemic concentration of genistein which was not reached in any other dosing scenario. This suggests that single or low doses of genistein are rapidly metabolised which limits its toxicodynamic effects on the liver and skin. Therefore, by facilitating longer and/or repeated applications, the Chip2 can support safety assessments by linking relevant gene modulation with systemically available parent or metabolite(s). The rate of metabolism was in accordance with the short half-life observed in in vivo in humans, thus supporting the relevance of the findings. In conclusion, the skin-liver Chip2 provides route-specific information on metabolic fate and toxicodynamics that may be relevant to safety assessment.


Subject(s)
Genistein , Skin , Humans , Genistein/toxicity , Toxicokinetics , Liver
3.
PLoS Comput Biol ; 18(10): e1010587, 2022 10.
Article in English | MEDLINE | ID: mdl-36260620

ABSTRACT

Microphysiological systems (MPS) are powerful tools for emulating human physiology and replicating disease progression in vitro. MPS could be better predictors of human outcome than current animal models, but mechanistic interpretation and in vivo extrapolation of the experimental results remain significant challenges. Here, we address these challenges using an integrated experimental-computational approach. This approach allows for in silico representation and predictions of glucose metabolism in a previously reported MPS with two organ compartments (liver and pancreas) connected in a closed loop with circulating medium. We developed a computational model describing glucose metabolism over 15 days of culture in the MPS. The model was calibrated on an experiment-specific basis using data from seven experiments, where HepaRG single-liver or liver-islet cultures were exposed to both normal and hyperglycemic conditions resembling high blood glucose levels in diabetes. The calibrated models reproduced the fast (i.e. hourly) variations in glucose and insulin observed in the MPS experiments, as well as the long-term (i.e. over weeks) decline in both glucose tolerance and insulin secretion. We also investigated the behaviour of the system under hypoglycemia by simulating this condition in silico, and the model could correctly predict the glucose and insulin responses measured in new MPS experiments. Last, we used the computational model to translate the experimental results to humans, showing good agreement with published data of the glucose response to a meal in healthy subjects. The integrated experimental-computational framework opens new avenues for future investigations toward disease mechanisms and the development of new therapies for metabolic disorders.


Subject(s)
Diabetes Mellitus , Insulin , Animals , Humans , Insulin/metabolism , Glucose/metabolism , Diabetes Mellitus/metabolism , Liver/metabolism , Insulin Secretion , Blood Glucose/metabolism
4.
Annu Rev Pharmacol Toxicol ; 58: 65-82, 2018 01 06.
Article in English | MEDLINE | ID: mdl-29029591

ABSTRACT

Enhancing the early detection of new therapies that are likely to carry a safety liability in the context of the intended patient population would provide a major advance in drug discovery. Microphysiological systems (MPS) technology offers an opportunity to support enhanced preclinical to clinical translation through the generation of higher-quality preclinical physiological data. In this review, we highlight this technological opportunity by focusing on key target organs associated with drug safety and metabolism. By focusing on MPS models that have been developed for these organs, alongside other relevant in vitro models, we review the current state of the art and the challenges that still need to be overcome to ensure application of this technology in enhancing drug discovery.


Subject(s)
Drug Discovery/methods , Pharmaceutical Preparations/chemistry , Animals , Drug Evaluation, Preclinical/methods , Humans
5.
J Appl Toxicol ; 41(10): 1553-1567, 2021 10.
Article in English | MEDLINE | ID: mdl-33594739

ABSTRACT

We used TissUse's HUMIMIC Chip2 microfluidic model, incorporating reconstructed skin models and liver spheroids, to investigate the impact of consumer-relevant application scenarios on the metabolic fate of the hair dye, 4-amino-2-hydroxytoluene (AHT). After a single topical or systemic application of AHT to Chip2 models, medium was analysed for parent and metabolites over 5 days. The metabolic profile of a high dose (resulting in a circuit concentration of 100 µM based on 100% bioavailability) of AHT was the same after systemic and topical application to 96-well EpiDerm™ models. Additional experiments indicated that metabolic capacity of EpiDerm™ models were saturated at this dose. At 2.5 µM, concentrations of AHT and several of its metabolites differed between application routes. Topical application resulted in a higher Cmax and a 327% higher area under the curve (AUC) of N-acetyl-AHT, indicating a first-pass effect in the EpiDerm™ models. In accordance with in vivo observations, there was a concomitant decrease in the Cmax and AUC of AHT-O-sulphate after topical, compared with systemic application. A similar alteration in metabolite ratios was observed using a 24-well full-thickness skin model, EpiDermFT™, indicating that a first-pass effect was also possible to detect in a more complex model. In addition, washing the EpiDermFT™ after 30 min, thus reflecting consumer use, decreased the systemic exposure to AHT and its metabolites. In conclusion, the skin-liver Chip2 model can be used to (a) recapitulate the first-pass effect of the skin and alterations in the metabolite profile of AHT observed in vivo and (b) provide consumer-relevant data regarding leave-on/rinse-off products.


Subject(s)
Aniline Compounds/metabolism , Aniline Compounds/toxicity , Cresols/metabolism , Cresols/toxicity , Hair Dyes/metabolism , Hair Dyes/toxicity , Liver/metabolism , Skin/metabolism , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Humans , Liver/drug effects , Organ Culture Techniques , Skin/drug effects
6.
Adv Exp Med Biol ; 856: 299-316, 2016.
Article in English | MEDLINE | ID: mdl-27671728

ABSTRACT

Equipment and device qualification and test assay validation in the field of tissue engineered human organs for substance assessment remain formidable tasks with only a few successful examples so far. The hurdles seem to increase with the growing complexity of the biological systems, emulated by the respective models. Controlled single tissue or organ culture in bioreactors improves the organ-specific functions and maintains their phenotypic stability for longer periods of time. The reproducibility attained with bioreactor operations is, per se, an advantage for the validation of safety assessment. Regulatory agencies have gradually altered the validation concept from exhaustive "product" to rigorous and detailed process characterization, valuing reproducibility as a standard for validation. "Human-on-a-chip" technologies applying micro-physiological systems to the in vitro combination of miniaturized human organ equivalents into functional human micro-organisms are nowadays thought to be the most elaborate solution created to date. They target the replacement of the current most complex models-laboratory animals. Therefore, we provide here a road map towards the validation of such "human-on-a-chip" models and qualification of their respective bioreactor and microchip equipment along a path currently used for the respective animal models.


Subject(s)
Bioreactors , Chemical Safety , Validation Studies as Topic , Humans , Lab-On-A-Chip Devices
7.
Commun Biol ; 7(1): 877, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39025915

ABSTRACT

Current research on metabolic disorders and diabetes relies on animal models because multi-organ diseases cannot be well studied with standard in vitro assays. Here, we have connected cell models of key metabolic organs, the pancreas and liver, on a microfluidic chip to enable diabetes research in a human-based in vitro system. Aided by mechanistic mathematical modeling, we demonstrate that hyperglycemia and high cortisone concentration induce glucose dysregulation in the pancreas-liver microphysiological system (MPS), mimicking a diabetic phenotype seen in patients with glucocorticoid-induced diabetes. In this diseased condition, the pancreas-liver MPS displays beta-cell dysfunction, steatosis, elevated ketone-body secretion, increased glycogen storage, and upregulated gluconeogenic gene expression. Conversely, a physiological culture condition maintains glucose tolerance and beta-cell function. This method was reproducible in two laboratories and was effective in multiple pancreatic islet donors. The model also provides a platform to identify new therapeutic proteins, as demonstrated with a combined transcriptome and proteome analysis.


Subject(s)
Cortisone , Glucose , Homeostasis , Liver , Pancreas , Humans , Liver/metabolism , Liver/drug effects , Cortisone/metabolism , Glucose/metabolism , Pancreas/metabolism , Lab-On-A-Chip Devices , Insulin-Secreting Cells/metabolism , Microphysiological Systems
8.
Mol Pharm ; 10(7): 2616-29, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23668542

ABSTRACT

Therapeutic efficacy of glycoproteins is affected by many factors, including molecular size and net charge; both are influenced by the presence and composition of glycan structures. Human alpha 1-antitrypsin (A1AT) was cloned and expressed in human embryonic kidney cells (HEK293) that are capable of mammalian glycosylation. Utilizing PCR-based site-directed mutagenesis, new A1AT variants were created with single, double, or triple additional N-glycosylation sites to the three naturally occurring N-glycosylation sites. Because of the supplementary N-glycans, the A1AT variants exhibited an increased molecular weight. Retention of inhibitory activity was shown via trypsin inhibitory assay. The A1AT variants were treated with PNGase F, and the resulting N-glycans were analyzed by MALDI-TOF mass spectrometry. The N-glycan profile of the recombinant A1AT variants was mostly composed of monofucosylated bi-, tri-, and tetraantennary complex-type N-glycans, with a tendency toward higher antennary structures compared to the wild-type. The relevance of N-glycosylation in A1AT for the circulatory serum half-life was demonstrated in CD1 mice. The A1AT neoglycoprotein with an additional N-glycosylation site at position N123 exhibited a 62% increase in serum half-life. Additionally, using a two-compartment model, the A1AT variants exhibited increased α-phase values, especially N123 (223%) and N201 (255%). The results suggest the recombinant A1AT neoglycoprotein as a serious alternative to A1AT derived from human plasma.


Subject(s)
Glycoproteins/blood , Glycoproteins/chemistry , alpha 1-Antitrypsin/blood , alpha 1-Antitrypsin/chemistry , Animals , Blotting, Western , Cell Line , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Female , Glycoproteins/pharmacokinetics , Glycosylation , Humans , Mice , Polysaccharides/blood , Polysaccharides/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , alpha 1-Antitrypsin/pharmacokinetics
9.
Analyst ; 138(14): 4066-9, 2013 Jul 21.
Article in English | MEDLINE | ID: mdl-23662302

ABSTRACT

A new simple method for non-invasive cell culture viability monitoring based on vital fluorescent stains is introduced, and its efficiency for long-term experiments on cells is demonstrated. In contrast to common methods for cell viability control, which are usually either destructive (like flow-type counters or dead cells coloring and counting), or hardly quantitative like fluorescent microscopy, the method described is automated, does not require the removal of cells from their growth area and is sensitive enough to deal with as low as tens of cells.


Subject(s)
Biosensing Techniques , Fibroblasts/cytology , Keratinocytes/cytology , Microscopy, Fluorescence , Optical Fibers , Skin/cytology , Cell Proliferation , Cell Survival , Cells, Cultured , Flow Cytometry , Humans
10.
ALTEX ; 40(1): 61-82, 2023.
Article in English | MEDLINE | ID: mdl-35536601

ABSTRACT

Thyroid hormones (THs) are crucial regulators of human metabolism and early development. During the safety assessment of plant protection products, the human relevance of chemically induced TH perturbations observed in test animals remains uncertain. European regulatory authorities request follow-up in vitro studies to elucidate human-relevant interferences on thyroid gland function or TH catabolism through hepatic enzyme induction. However, human in vitro assays based on single molecular initiating events poorly reflect the complex TH biology and related liver-thyroid axis. To address this complexity, we present human three-dimensional thyroid and liver organoids with key functions of TH metabolism. The thyroid model resembles in vivo-like follicular architecture and a TSH-dependent triiodothyronine synthesis over 21 days, which is inhibited by methimazole. The HepaRG-based liver model, secreting the critical TH-binding proteins albumin and thyroxine-binding globulin, emulates an active TH catabolism via the formation of glucuronidated and sulfated thyroxine (gT4/sT4). Activation of the nuclear receptors PXR and AHR was demonstrated via the induction of specific CYP isoenzymes by rifampicin, pregnenolone-16α-carbonitrile, and ß-naphthoflavone. However, this nuclear receptor activation, assumed to regulate UDP-glucuronosyltransferases and sulfotransferases, appeared to have no effect on gT4 and sT4 formation in this human-derived hepatic cell line model. Finally, established single-tissue models were successfully co-cultured in a perfused two-organ chip for 21 days. In conclusion, this model presents a first step towards a complex multimodular human platform that will help to identify both direct and indirect thyroid disruptors that are relevant from a human safety perspective.


Subject(s)
Chemical Safety , Thyroid Gland , Animals , Humans , Thyroid Gland/metabolism , Microfluidics , Thyroid Hormones/metabolism , Thyroid Hormones/pharmacology , Liver , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Cytoplasmic and Nuclear/pharmacology
11.
ALTEX ; 40(1): 83-102, 2023.
Article in English | MEDLINE | ID: mdl-35791291

ABSTRACT

Endocrine disruption by environmental chemicals continues to be a concern for human safety. The rat, a widely used model organism in toxicology, is very sensitive to chemical-induced thyroid perturbation, e.g., histopathological alterations in thyroid tissue. Species differences in the susceptibility to thyroid perturbation lead to uncertainty in human safety risk assessments. Hazard identification and characterization of chemically induced thyroid perturbation would therefore benefit from in vitro models addressing different mechanisms of action in a single functional assay, ideally across species. We here introduce a rat thyroid-liver chip that enables simultaneous identification of direct and indirect (liver-mediated) thyroid perturbation on organ-level functions in vitro. A second manuscript describes our work toward a human thyroid-liver chip (Kühnlenz et al., 2022). The presented microfluidic model consisting of primary rat thyroid follicles and liver 3D spheroids maintains a tissue-specific phenotype for up to 21 days. More precisely, the thyroid model exhibits a follicular architecture expressing basolateral and apical markers and secretes T4. Likewise, liver spheroids retain hepatocellular characteristics, e.g., a stable release of albumin and urea, the presence of bile canalicular networks, and the formation of T4-glucuronide. Experiments with reference chemicals demonstrated proficiency to detect direct and indirect mechanisms of thyroid perturbation through decreased thyroid hormone secretion and increased gT4 formation, respectively. Prospectively this rat thyroid-liver chip model, together with its human counterpart, may support a species-specific quantitative in vitro to in vivo extrapolation to improve a data-driven and evidence-based human safety risk assessment with significant contributions to the 3R principles.


Subject(s)
Rodentia , Thyroid Gland , Humans , Rats , Animals , Animal Testing Alternatives , Liver
12.
Front Pharmacol ; 14: 1076254, 2023.
Article in English | MEDLINE | ID: mdl-36843954

ABSTRACT

All cosmetic ingredients registered in Europe must be evaluated for their safety using non-animal methods. Microphysiological systems (MPS) offer a more complex higher tier model to evaluate chemicals. Having established a skin and liver HUMIMIC Chip2 model demonstrating how dosing scenarios impact the kinetics of chemicals, we investigated whether thyroid follicles could be incorporated to evaluate the potential of topically applied chemicals to cause endocrine disruption. This combination of models in the HUMIMIC Chip3 is new; therefore, we describe here how it was optimized using two chemicals known to inhibit thyroid production, daidzein and genistein. The MPS was comprised of Phenion® Full Thickness skin, liver spheroids and thyroid follicles co-cultured in the TissUse HUMIMIC Chip3. Endocrine disruption effects were determined according to changes in thyroid hormones, thyroxine (T4) and 3,3',5-triiodothyronine (T3). A main part of the Chip3 model optimization was the replacement of freshly isolated thyroid follicles with thyrocyte-derived follicles. These were used in static incubations to demonstrate the inhibition of T4 and T3 production by genistein and daidzein over 4 days. Daidzein exhibited a lower inhibitory activity than genistein and both inhibitory activities were decreased after a 24 h preincubation with liver spheroids, indicating metabolism was via detoxification pathways. The skin-liver-thyroid Chip3 model was used to determine a consumer-relevant exposure to daidzein present in a body lotion based on thyroid effects. A "safe dose" of 0.235 µg/cm2 i.e., 0.047% applied in 0.5 mg/cm2 of body lotion was the highest concentration of daidzein which does not result in changes in T3 and T4 levels. This concentration correlated well with the value considered safe by regulators. In conclusion, the Chip3 model enabled the incorporation of the relevant exposure route (dermal), metabolism in the skin and liver, and the bioactivity endpoint (assessment of hormonal balance i.e., thyroid effects) into a single model. These conditions are closer to those in vivo than 2D cell/tissue assays lacking metabolic function. Importantly, it also allowed the assessment of repeated doses of chemical and a direct comparison of systemic and tissue concentrations with toxicodynamic effects over time, which is more realistic and relevant for safety assessment.

13.
Adv Exp Med Biol ; 745: 26-46, 2012.
Article in English | MEDLINE | ID: mdl-22437811

ABSTRACT

Dynamic macroscale bioreactor systems are the most recent breakthrough in cell culture technology. This major achievement, at the beginning of the 21st century, fortunately coincided with an embarrassing gap in the measures to predict the safety and modes of action of chemicals, cosmetics, air particles and pharmaceuticals. The major hurdles to the translation of these breakthrough achievements of cell culture technology into meaningful solutions for predictive high throughput substance testing remain miniaturization from the milliliter to the microliter scale and the supply of relevant amounts of standardized human tissue. This chapter provides insights into the latest developments in this area, illustrates an original multi-micro-organ bioreactor concept and identifies highways for closing the gap.


Subject(s)
Cell Culture Techniques/methods , Bioreactors , Humans , Stem Cell Niche/drug effects , Toxicity Tests
14.
Altern Lab Anim ; 40(5): 235-57, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23215661

ABSTRACT

Various factors, including the phylogenetic distance between laboratory animals and humans, the discrepancy between current in vitro systems and the human body, and the restrictions of in silico modelling, have generated the need for new solutions to the ever-increasing worldwide dilemma of substance testing. This review provides a historical sketch on the accentuation of this dilemma, and highlights fundamental limitations to the countermeasures taken so far. It describes the potential of recently-introduced microsystems to emulate human organs in 'organ-on-a-chip' devices. Finally, it focuses on an in-depth analysis of the first devices that aimed to mimic human systemic organ interactions in 'human-on-a-chip' systems. Their potential to replace acute systemic toxicity testing in animals, and their inability to provide alternatives to repeated dose long-term testing, are discussed. Inspired by the latest discoveries in human biology, tissue engineering and micro-systems technology, this review proposes a paradigm shift to overcome the apparent challenges. A roadmap is outlined to create a new homeostatic level of biology in 'human-on-a-chip' systems in order to, in the long run, replace systemic repeated dose safety evaluation and disease modelling in animals.


Subject(s)
Animal Testing Alternatives , Animals, Laboratory , Microfluidic Analytical Techniques/methods , Toxicity Tests/methods , Animals , Humans , Stem Cell Research
15.
Pharmaceutics ; 14(8)2022 Jul 22.
Article in English | MEDLINE | ID: mdl-35893784

ABSTRACT

Pharmaceutical and personal care industries require human representative models for testing to ensure the safety of their products. A major route of penetration into our body after substance exposure is via the skin. Our aim was to generate robust culture conditions for a next generation human skin-on-chip model containing neopapillae and to establish proof-of-concept testing with the sensitizer, cinnamaldehyde. Reconstructed human skin consisting of a stratified and differentiated epidermis on a fibroblast populated hydrogel containing neopapillae spheroids (RhS-NP), were cultured air-exposed and under dynamic flow for 10 days. The robustness of three independent experiments, each with up to 21 intra-experiment replicates, was investigated. The epidermis was seen to invaginate into the hydrogel towards the neopapille spheroids. Daily measurements of lactate dehydrogenase (LDH) and glucose levels within the culture medium demonstrated high viability and stable metabolic activity throughout the culture period in all three independent experiments and in the replicates within an experiment. Topical cinnamaldehyde exposure to RhS-NP resulted in dose-dependent cytotoxicity (increased LDH release) and elevated cytokine secretion of contact sensitizer specific IL-18, pro-inflammatory IL-1ß, inflammatory IL-23 and IFN-γ, as well as anti-inflammatory IL-10 and IL-12p70. This study demonstrates the robustness and feasibility of complex next generation skin models for investigating skin immunotoxicity.

16.
Cells ; 11(20)2022 10 19.
Article in English | MEDLINE | ID: mdl-36291161

ABSTRACT

Significant advancements in the field of preclinical in vitro blood-brain barrier (BBB) models have been achieved in recent years, by developing monolayer-based culture systems towards complex multi-cellular assays. The coupling of those models with other relevant organoid systems to integrate the investigation of blood-brain barrier permeation in the larger picture of drug distribution and metabolization is still missing. Here, we report for the first time the combination of a human induced pluripotent stem cell (hiPSC)-derived blood-brain barrier model with a cortical brain and a liver spheroid model from the same donor in a closed microfluidic system (MPS). The two model compounds atenolol and propranolol were used to measure permeation at the blood-brain barrier and to assess metabolization. Both substances showed an in vivo-like permeation behavior and were metabolized in vitro. Therefore, the novel multi-organ system enabled not only the measurement of parent compound concentrations but also of metabolite distribution at the blood-brain barrier.


Subject(s)
Blood-Brain Barrier , Induced Pluripotent Stem Cells , Pharmaceutical Preparations , Humans , Atenolol/metabolism , Blood-Brain Barrier/metabolism , Brain , Induced Pluripotent Stem Cells/metabolism , Liver , Pharmaceutical Preparations/metabolism , Propranolol/metabolism
17.
Exp Biol Med (Maywood) ; 247(1): 1-75, 2022 01.
Article in English | MEDLINE | ID: mdl-34783606

ABSTRACT

There is an evolution and increasing need for the utilization of emerging cellular, molecular and in silico technologies and novel approaches for safety assessment of food, drugs, and personal care products. Convergence of these emerging technologies is also enabling rapid advances and approaches that may impact regulatory decisions and approvals. Although the development of emerging technologies may allow rapid advances in regulatory decision making, there is concern that these new technologies have not been thoroughly evaluated to determine if they are ready for regulatory application, singularly or in combinations. The magnitude of these combined technical advances may outpace the ability to assess fit for purpose and to allow routine application of these new methods for regulatory purposes. There is a need to develop strategies to evaluate the new technologies to determine which ones are ready for regulatory use. The opportunity to apply these potentially faster, more accurate, and cost-effective approaches remains an important goal to facilitate their incorporation into regulatory use. However, without a clear strategy to evaluate emerging technologies rapidly and appropriately, the value of these efforts may go unrecognized or may take longer. It is important for the regulatory science field to keep up with the research in these technically advanced areas and to understand the science behind these new approaches. The regulatory field must understand the critical quality attributes of these novel approaches and learn from each other's experience so that workforces can be trained to prepare for emerging global regulatory challenges. Moreover, it is essential that the regulatory community must work with the technology developers to harness collective capabilities towards developing a strategy for evaluation of these new and novel assessment tools.


Subject(s)
Biomedical Research , Computer Simulation , Humans
18.
Biotechnol Bioeng ; 108(9): 2118-28, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21495009

ABSTRACT

Human alpha-1-antitrypsin (A1AT) is a protease inhibitor that is involved in the protection of lungs from neutrophil elastase enzyme that drastically modifies tissue functioning. The glycoprotein consists of 394 amino acids and is N-glycosylated at Asn-46, Asn-83, and Asn-247. A1AT deficiency is currently treated with A1AT that is purified from human serum. In view of therapeutic applications, rA1AT was produced using a novel human neuronal cell line (AGE1.HN®) and we investigated the N-glycosylation pattern as well as the in vitro anti-inflammatory activity of the recombinant glycoprotein. rA1AT (300 mg/L) was biologically active as analyzed using elastase assay. The N-glycan pool, released by PNGase F digestion, was characterized using 2D-HPLC, MALDI-TOF mass spectrometry, and by exoglycosidase digestions. A total of 28 N-glycan structures were identified, ranging from diantennary to tetraantennary complex-type N-glycans. Most of the N-glycans were found to be (α1-6) core-fucosylated and part of them contain the Lewis X epitope. The two major compounds are a monosialylated diantennary difucosylated glycan and a disialylated diantennary core-fucosylated glycan, representing 25% and 18% of the total N-glycan pool, respectively. Analysis of the site-specificity revealed that Asn-247 was mainly occupied by diantennary N-glycans whereas Asn-46 was occupied by di-, and triantennary N-glycans. Asn-83 was exclusively occupied by sialylated tri- and tetraantennary N-glycans. Next, we evaluated the anti-inflammatory activity of rA1AT using A1AT purified from human serum as a reference. rA1AT was found to inhibit the production of TNF-α in neutrophils and monocytes as commercial A1AT does.


Subject(s)
Biotechnology/methods , Neurons/metabolism , Recombinant Proteins/metabolism , alpha 1-Antitrypsin/metabolism , Anti-Inflammatory Agents/metabolism , Anti-Inflammatory Agents/pharmacology , Cell Line , Cells, Cultured , Chromatography, High Pressure Liquid , Electrophoresis, Polyacrylamide Gel , Glycosylation , Humans , Leukocyte Elastase/antagonists & inhibitors , Leukocyte Elastase/metabolism , Monocytes/metabolism , Neutrophils/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Sialic Acids/analysis , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tumor Necrosis Factor-alpha/metabolism , alpha 1-Antitrypsin/biosynthesis , alpha 1-Antitrypsin/genetics , alpha 1-Antitrypsin/pharmacology
19.
Data Brief ; 37: 107140, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34136596

ABSTRACT

Integration-free induced pluripotent stem cells from related human donors' exhibit great potential to the ongoing development of organ models. Blood cells from two different human donors were isolated, purified and reprogrammed into induced pluripotent stem cells. These induced pluripotent stem cell lines were characterized precisely for pluripotency markers (with the PluriTest and flow cytometry analysis) and their differentiation capacities into meso-, ecto- and endoderm. The induced pluripotent stem cell lines are available for commercial use and are therefore of high interest for many groups working in stem cell research. A normal karyotype of the induced pluripotent stem cells was proven with the KaryoStat assay. In total 6 human donors that belong to one family donated blood for induced pluripotent stem cell reprogramming. In this "Data in Brief" publication, we show the dataset for the two male iPSC lines HUMIMIC TISSUi006-A (StemUse106) and TISSUi007-A (StemUse107). The main characterisation was recently published by Ramme et al. in Stem Cell Research [1]. All iPSC lines were also examined negative for any mycoplasma or bacteria contamination.

20.
Stem Cell Res ; 53: 102327, 2021 05.
Article in English | MEDLINE | ID: mdl-33901815

ABSTRACT

The integration-free iPSC lines TISSUi006-A and TISSUi007-A were generated by reprogramming blood cells with episomal vectors. The male human donors belong to a Caucasian family in which four additional family members donated and iPSC lines were generated. All iPSC lines within this family are approved for commercial use by donor consent. Those iPSC lines offer the opportunity to study the influence of affiliation within one family. In future, more iPSCs lines of many more family members can be created to understand the effects of relatives with different ages on the reprogramming into iPSCs and differentiation into specific cell types.


Subject(s)
Induced Pluripotent Stem Cells , Cell Differentiation , Cellular Reprogramming , Humans , Male , Plasmids , Tissue Donors
SELECTION OF CITATIONS
SEARCH DETAIL