Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
Add more filters

Publication year range
1.
Int J Mol Sci ; 22(2)2021 Jan 06.
Article in English | MEDLINE | ID: mdl-33419219

ABSTRACT

The clinical results of lung transplantation (LTx) are still less favorable than other solid organ transplants in both the early and long term. The fragility of the lungs limits the procurement rate and can favor the occurrence of ischemia-reperfusion injury (IRI). Ex vivo lung perfusion (EVLP) with Steen SolutionTM (SS) aims to address problems, and the implementation of EVLP to alleviate the activation of IRI-mediated processes has been achieved using mesenchymal stromal/stem cell (MSC)-based treatments. In this study, we investigated the paracrine effects of human amnion-derived MSCs (hAMSCs) in an in vitro model of lung IRI that includes cold ischemia and normothermic EVLP. We found that SS enriched by a hAMSC-conditioned medium (hAMSC-CM) preserved the viability and delayed the apoptosis of alveolar epithelial cells (A549) through the downregulation of inflammatory factors and the upregulation of antiapoptotic factors. These effects were more evident using the CM of 3D hAMSC cultures, which contained an increased amount of immunosuppressive and growth factors compared to both 2D cultures and encapsulated-hAMSCs. To conclude, we demonstrated an in vitro model of lung IRI and provided evidence that a hAMSC-CM attenuated IRI effects by improving the efficacy of EVLP, leading to strategies for a potential implementation of this technique.


Subject(s)
Alveolar Epithelial Cells/drug effects , Cold Ischemia/methods , Culture Media, Conditioned/pharmacology , Mesenchymal Stem Cells/drug effects , Reperfusion Injury/drug therapy , A549 Cells , Alveolar Epithelial Cells/metabolism , Amnion/cytology , Apoptosis/drug effects , Apoptosis/genetics , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Survival/drug effects , Cell Survival/genetics , Cells, Cultured , Cytokines/genetics , Gene Expression Regulation/drug effects , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , NF-kappa B/genetics , Reperfusion Injury/physiopathology
2.
Biochem Biophys Res Commun ; 522(1): 171-176, 2020 01 29.
Article in English | MEDLINE | ID: mdl-31757423

ABSTRACT

Different approaches have been studied in both preclinical and clinical settings to develop cell-based therapies and/or engineered cell-based therapies to better integrate grafts with the host. In these techniques, much attention is addressed to the use of adult stem cells such as mesenchymal stem cells (MSCs), but identifying and obtaining sufficient numbers of therapeutic cells, and the right route of administration, is often a challenge. In this study, we tested the feasibility of encapsulating human amnion-derived MSCs (hAMSCs) in a semipermeable and biocompatible fiber as a new approach for regenerative medicine. Our data showed that hAMSCs aggregated in the device constitutes an effective system for enhancing, or at least for maintaining, the paracrine activity of these cells in order to better promote tissue regeneration in an immune isolated state. In our new experimental approach, the hAMSCs retained their therapeutic potential, as shown by both the production of specific immunomodulatory/angiogenic factors and immunomodulatory and angiogenic ability observed in vitro. Unlike cell infusion methods, the use of encapsulated-cells leads to minimally invasive approaches, avoiding a direct interaction with the host. Therefore, the potentiality of an allograft or xenograft without the need for immunosuppression, and the lack of tumorigenesis is very intriguing.


Subject(s)
Amnion/cytology , Cell Culture Techniques/instrumentation , Mesenchymal Stem Cells/cytology , Placenta/cytology , Angiogenesis Inducing Agents/metabolism , Biocompatible Materials/chemistry , Cell Adhesion , Cell Aggregation , Cell Culture Techniques/methods , Cell Movement , Cell Survival , Cells, Immobilized/cytology , Cells, Immobilized/metabolism , Female , Human Umbilical Vein Endothelial Cells , Humans , Immunologic Factors/metabolism , Mesenchymal Stem Cells/metabolism , Pregnancy
3.
J Cell Physiol ; 233(6): 4841-4851, 2018 06.
Article in English | MEDLINE | ID: mdl-29150960

ABSTRACT

Epithelial Cell Adhesion Molecule (EpCAM), or CD326, is a trans-membrane glycoprotein expressed by multiple normal epithelia as well as carcinoma. Human hepatic stem cells and bile duct epithelium of the liver are EpCAM positive. In tumor cell lines, its intracellular domain can be released after cleavage of the extracellular domain. Within the cell nucleus, it induces cell proliferation, but cleavage depends on cell contact. Fragments of various lengths have been described in tumor cells. Despite its described important role in proliferation in tumor cells, there is not much known about the expression and role of EpCAM fragments in primary human liver cells. Here, we demonstrate that EpCAM protein fragments and function are considerable different between tumor cells, normal fetal and adult liver cells. Contrary to previously reported findings in tumor cells, gene knockdown or treatment with an inhibitor of the cleavage enzyme ADAM17 (TACE) rather increased cell numbers in primary human fetal liver-derived EpCAM-positive cells. EpCAM fragment sizes were not affected by treatment with inhibitor. Knockdown of EPCAM gene expression by siRNA in sorted cells did not significantly affect proliferation-associated genes or cell numbers. The intracellular domain could not be detected within cell nuclei of fetal and adult liver cells. In conclusion, signaling through the intracellular domain of EpCAM appears to be a mechanism that induces proliferation specifically in tumorigenic cells but not in normal primary EpCAM-positive liver cells.


Subject(s)
Adult Stem Cells/metabolism , Cell Proliferation , Colorectal Neoplasms/metabolism , Epithelial Cell Adhesion Molecule/metabolism , Fetal Stem Cells/metabolism , Liver/metabolism , Neoplastic Stem Cells/metabolism , Peptide Fragments/metabolism , Signal Transduction , ADAM17 Protein/genetics , ADAM17 Protein/metabolism , Adult Stem Cells/drug effects , Cell Proliferation/drug effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Epithelial Cell Adhesion Molecule/genetics , Fetal Stem Cells/drug effects , Gene Expression Regulation , Glycosylation , HT29 Cells , Humans , Hydroxamic Acids/pharmacology , Liver/cytology , Liver/drug effects , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Primary Cell Culture , Protein Domains , Signal Transduction/drug effects
4.
J Vasc Res ; 54(6): 376-385, 2017.
Article in English | MEDLINE | ID: mdl-29166642

ABSTRACT

Foamed hydroxyapatite offers a three-dimensional scaffold for the development of bone constructs, mimicking perfectly the in vivo bone structure. In vivo, calcium release at the surface is assumed to provide a locally increased gradient supporting the maintenance of the hematopoietic stem cells niche. We fabricated hydroxyapatite scaffolds with high surface calcium concentration by infiltration, and used human umbilical vein endothelial cells (HUVECs) as a model to study the effects on hematopoietic lineage direction. HUVECs are umbilical vein-derived and thus possess progenitor characteristics, with a prospective potential to give rise to hematopoietic lineages. HUVECs were cultured for long term on three-dimensional porous hydroxyapatite scaffolds, which were either infiltrated biphasic foams or untreated. Controls were cultured in two-dimensional dishes. The release of calcium into culture medium was determined, and cells were analyzed for typical hematopoietic and endothelial gene expressions, surface markers by flow cytometry, and hematopoietic potential using colony-forming unit assays. Our results indicate that the biphasic foams promoted a hematopoietic lineage direction of HUVECs, suggesting an improved in vivo-like scaffold for hematopoietic bone tissue engineering.


Subject(s)
Cell Differentiation , Cell Lineage , Durapatite/metabolism , Hematopoietic Stem Cells/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Tissue Engineering/methods , Tissue Scaffolds , Biomarkers/metabolism , Cell Proliferation , Cell Separation/methods , Cells, Cultured , Flow Cytometry , Gene Expression Regulation , Humans , Phenotype , Porosity , Surface Properties , Time Factors
5.
Xenotransplantation ; 24(2)2017 03.
Article in English | MEDLINE | ID: mdl-28130881

ABSTRACT

BACKGROUND: Some patients with acute or acute-on-chronic hepatic failure die before a suitable human liver allograft becomes available. Encouraging results have been achieved in such patients by the transplantation of human hepatocyte progenitor cells from fetal liver tissue. The aim of the study was to explore survival of hepatocytes from genetically engineered pigs after direct injection into the spleen and other selected sites in immunosuppressed baboons to monitor the immune response and the metabolic function and survival of the transplanted hepatocytes. METHODS: Baboons (n=3) were recipients of GTKO/hCD46 pig hepatocytes. All three baboons received anti-thymocyte globulin (ATG) induction and tapering methylprednisolone. Baboon 1 received maintenance immunosuppressive therapy with tacrolimus and rapamycin. Baboons 2 and 3 received an anti-CD40mAb/rapamycin-based regimen that prevents sensitization to pig solid organ grafts. The baboons were euthanized 4 or 5 weeks after hepatocyte transplantation. The baboon immune response was monitored by the measurement of anti-non-Gal IgM and IgG antibodies (by flow cytometry) and CFSE-mixed lymphocyte reaction. Monitoring for hepatocyte survival and function was by (i) real-time PCR detection of porcine DNA, (ii) real-time PCR for porcine gene expression, and (iii) pig serum albumin levels (by ELISA). The sites of hepatocyte injection were examined microscopically. RESULTS: Detection of porcine DNA and porcine gene expression was minimal at all sites of hepatocyte injection. Serum levels of porcine albumen were very low-500-1000-fold lower than in baboons with orthotopic pig liver grafts, and approximately 5000-fold lower than in healthy pigs. No hepatocytes or infiltrating immune cells were seen at any of the injection sites. Two baboons (Baboons 1 and 3) demonstrated a significant increase in anti-pig IgM and an even greater increase in IgG, indicating sensitization to pig antigens. DISCUSSION AND CONCLUSIONS: As a result of this disappointing experience, the following points need to be considered. (i) Were the isolated pig hepatocytes functionally viable? (ii) Are pig hepatocytes more immunogenic than pig hearts, kidneys, artery patch grafts, or islets? (iii) Does injection of pig cells (antigens) into the spleen and/or lymph nodes stimulate a greater immune response than when pig tissues are grafted at other sites? (iv) Did the presence of the recipient's intact liver prevent survival and proliferation of pig hepatocytes? (v) Is pig CD47-primate SIRP-α compatibility essential? In conclusion, the transplantation of genetically engineered pig hepatocytes into multiple sites in immunosuppressed baboons was associated with very early graft failure. Considerable further study is required before clinical trials should be undertaken.


Subject(s)
Graft Survival/immunology , Hepatocytes/immunology , Transplantation, Heterologous , Animals , Animals, Genetically Modified , Antibodies/immunology , Antibody Formation/genetics , Antibody Formation/immunology , Antigens/immunology , Graft Survival/drug effects , Hepatocytes/transplantation , Immunosuppression Therapy/methods , Immunosuppressive Agents/pharmacology , Papio hamadryas/immunology , Swine , Transplantation, Heterologous/methods
6.
Differentiation ; 89(5): 137-45, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26142155

ABSTRACT

An alternative approach for traditional clinical mesh grafting in burn wound treatment is the use of expanded autologous keratinocytes in suspension or sheets that are cultured over 2-4 weeks in a remote service facility. While a wound reepithelialization has been described, the functional and aesthetic outcome is under debate. Cell isolation from split-skin donor tissue aims to preserve the valuable stem cell progenitors from the basal epidermal layer and to provide patients with a rapid wound reepithelialization and a satisfying outcome. While the presence of epidermal progenitors in the cell graft is thought to enable an improved epidermal surface post reepithelialization, we investigated a feasible clinical approach involving cultured versus noncultured epidermal cells comparing the α6int(high)/K15(high)/FSC(low)/SSC(low) and α6int(high)/K5(high)/FSC(low)/SSC(low) keratinocyte progenitor subpopulations before and after in vitro culture process. Our results show a significant increase of cell size during in vitro passaging and a decrease of progenitor markers linked to a gradual differentiation. A provision of the regenerative epidermal progenitors, isolated from the split-skin biopsy and applied directly onto the wound in an on-site setting of isolation and cell spray grafting in the operation room, could be of interest when choosing options for skin wound care with autologous cells.


Subject(s)
Cell Differentiation , Epidermis/transplantation , Keratinocytes/cytology , Adult , Cell Proliferation , Cell Size , Cell Transplantation , Cell- and Tissue-Based Therapy , Cells, Cultured , Humans , Keratinocytes/metabolism , Keratinocytes/transplantation , Stem Cell Transplantation , Stem Cells/cytology
7.
J Med Biol Eng ; 36: 153-167, 2016.
Article in English | MEDLINE | ID: mdl-27231463

ABSTRACT

Advancement in thermal three-dimensional printing techniques has greatly increased the possible applications of various materials in medical applications and tissue engineering. Yet, potential toxic effects on primary human cells have been rarely investigated. Therefore, we compared four materials commonly used in thermal printing for bioengineering, namely thermally printed acrylonitrile butadiene styrene, MED610, polycarbonate, and polylactic acid, and investigated their effects on primary human adult skin epidermal keratinocytes and bone marrow mesenchymal stromal cells (BM-MSCs) in vitro. We investigated indirect effects on both cell types caused by potential liberation of soluble substances from the materials, and also analyzed BM-MSCs in direct contact with the materials. We found that even in culture without direct contact with the materials, the culture with MED610 (and to a lesser extent acrylonitrile butadiene styrene) significantly affected keratinocytes, reducing cell numbers and proliferation marker Ki67 expression, and increasing glucose consumption, lactate secretion, and expression of differentiation-associated genes. BM-MSCs had decreased metabolic activity, and exhibited increased cell death in direct culture on the materials. MED610 and acrylonitrile butadiene styrene induced the strongest expression of genes associated to differentiation and estrogen receptor activation. In conclusion, we found strong cell-type-specific effects of the materials, suggesting that materials for applications in regenerative medicine should be carefully selected not only based on their mechanical properties but also based on their cell-type-specific biological effects.

8.
Biotechnol Bioeng ; 112(4): 801-10, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25335987

ABSTRACT

The construction and long-term maintenance of three-dimensional in vitro bone marrow models is of great interest but still quite challenging. Here we describe the use of a multi-compartment hollow-fiber membrane based three-dimensional perfusion bioreactor for long-term culture of whole human bone marrow mononuclear cells. We also investigated bioreactors with incorporated open-porous foamed hydroxyapatite scaffolds, mimicking the in vivo bone matrix. Cells in bioreactors with and without scaffolds were cultured to 6 weeks and compared to Petri dish controls. Cells were analyzed for gene expression, surface markers by flow cytometry, metabolic activity, hematopoietic potential, viability, and attachment by immunocytochemistry. Cells in bioreactors were metabolic active during long-term culture. The percentages of hematopoietic stem cell and mature endothelial cell fractions were maintained in bioreactors. The expression of most of the analyzed genes stabilized and increased after long-term culture of 6 weeks. Compared to Petri dish culture controls, bioreactor perfusion culture improved in both the short and long-term, the colony formation unit capacity of hematopoietic progenitors. Cells attached to the ample surface area provided by hydroxyapatite scaffolds. The implementation of a hydroxyapatite scaffold did not influence colony formation capacity, percentages of cell type specific fractions, gene expression, cell viability or metabolic turnover when compared to control cells cultured in bioreactors without scaffolds. In conclusion, three-dimensional perfusion bioreactor culture enables long-term maintenance of primary human bone marrow cells, with hydroxyapatite scaffolds providing an in vivo-like scaffold for three-dimensional culture.


Subject(s)
Bioreactors , Bone Marrow , Organ Culture Techniques/methods , Adult , Cell Physiological Phenomena , Durapatite , Humans , Tissue Scaffolds
9.
Xenotransplantation ; 22(4): 239-48, 2015.
Article in English | MEDLINE | ID: mdl-25950141

ABSTRACT

Hepatocyte transplantation (Tx) is a potential therapy for certain diseases of the liver, including hepatic failure. However, there is a limited supply of human livers as a source of cells and, after isolation, human hepatocytes can be difficult to expand in culture, limiting the number available for Tx. Hepatocytes from other species, for example, the pig, have therefore emerged as a potential alternative source. We searched the literature through the end of 2014 to assess the current status of experimental research into hepatocyte xenoTx. The literature search identified 51 reports of in vivo cross-species Tx of hepatocytes in a variety of experimental models. Most studies investigated the Tx of human (n = 23) or pig (n = 19) hepatocytes. No studies explored hepatocytes from genetically engineered pigs. The spleen was the most common site of Tx (n = 23), followed by the liver (through the portal vein [n = 6]) and peritoneal cavity (n = 19). In 47 studies (92%), there was evidence of hepatocyte engraftment and function across a species barrier. The data provided by this literature search strengthen the hypothesis that xenoTx of hepatocytes is feasible and potentially successful as a clinical therapy for certain liver diseases, including hepatic failure. By excluding vascular structures, hepatocytes isolated from genetically engineered pig livers may address some of the immunological problems of xenoTx.


Subject(s)
Hepatocytes/transplantation , Transplantation, Heterologous/methods , Animals , Animals, Genetically Modified , Graft Survival , Heterografts , Humans , Models, Animal , Swine , Transplantation, Heterologous/trends
10.
Hepatol Res ; 45(8): 919-32, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25195540

ABSTRACT

AIM: The transcription factor CCAAT/enhancer-binding protein alpha (C/EBPα) has been shown to play an important role in liver development, cell proliferation and differentiation. It is, however, largely unknown if C/EBPα regulates cell differentiation and proliferation differently in the diverse cell types of the human liver. We investigated the role of C/EBPα in primary human fetal liver cells and liver cell subpopulations in vitro using a 3-D perfusion bioreactor as an advanced in vivo-like human organ culture model. METHODS: Human fetal liver cells were investigated in vitro. C/EBPα gene expression was knocked down using siRNA or overexpressed by plasmid transfection. Cell type-specific gene expression was studied, cell populations and their proliferation were investigated, and metabolic parameters were analyzed. RESULTS: When C/EBPα gene expression was knocked down, we observed a significantly reduced expression of typical endothelial, hematopoietic and mesenchymal genes such as CD31, vWF, CD90, CD45 and α-smooth muscle actin in fetal cells. The intracellular expression of hepatic proteins and genes for liver-specific serum proteins α-fetoprotein and albumin were reduced, their protein secretion was increased. Fetal endothelial cell numbers were reduced and hepatoblast numbers were increased. C/EBPα overexpression in fetal cells resulted in increased endothelial numbers, but did not affect mesenchymal cell types or hepatoblasts. CONCLUSION: We demonstrated that the effects of C/EBPα are specific for the different human fetal liver cell types, using an advanced 3-D perfusion bioreactor as a human in vivo-like model.

11.
Liver Transpl ; 18(2): 226-37, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22034152

ABSTRACT

Although hepatic cell transplantation (CT) holds the promise of bridging patients with end-stage chronic liver failure to whole liver transplantation, suitable cell populations are under debate. In addition to hepatic cells, mesenchymal stem cells (MSCs) and hematopoietic stem cells (HSCs) are being considered as alternative cell sources for initial clinical cell work. Fetal liver (FL) tissue contains potential progenitors for all these cell lineages. Based on the collagenase incubation of tissue fragments, traditional isolation techniques yield only a fraction of the number of available cells. We report a 5-step method in which a portal vein in situ perfusion technique is used for tissue from the late second trimester. This method results in the high viabilities known for adult liver vascular perfusion, addresses the low cell yields of conventional digestion methods, and reduces the exposure of the tissue to collagenase 4-fold. We used donated tissue from gestational weeks 18 to 22, which yielded 1.8 ± 0.7 × 10(9) cells with an average viability of 78%. Because HSC transplantation and MSC transplantation are of interest for the treatment of hepatic failure, we phenotypically confirmed that in addition to hepatic progenitors, the resulting cell preparation contained cells expressing typical MSC and HSC markers. The percentage of FL cells expressing proliferation markers was 45 times greater than the percentage of adult hepatocytes expressing these markers and was comparable to the percentage of immortalized HepG2 liver hepatocellular carcinoma cells; this indicated the strong proliferative capacity of fetal cells. We report a case of human FL CT with the described liver cell population for clinical end-stage chronic liver failure. The patient's Model for End-Stage Liver Disease (MELD) score improved from 15 to 10 within the first 18 months of observation. In conclusion, this human FL cell isolation protocol may be of interest for further clinical translation work on the development of liver cell-based therapies.


Subject(s)
Cell Separation/methods , End Stage Liver Disease/surgery , Fetal Stem Cells/transplantation , Hematopoietic Stem Cell Transplantation , Hepatitis C/complications , Liver Cirrhosis/surgery , Liver/embryology , Mesenchymal Stem Cell Transplantation , Perfusion , Biomarkers/metabolism , Cell Culture Techniques , Cell Survival , Collagenases/metabolism , End Stage Liver Disease/virology , Fetal Stem Cells/metabolism , Gestational Age , Hep G2 Cells , Humans , Immunosuppressive Agents/therapeutic use , Liver/blood supply , Liver/metabolism , Liver Cirrhosis/virology , Male , Middle Aged , Phenotype , Portal Vein/embryology , Treatment Outcome
12.
Biotechnol Bioeng ; 109(12): 3172-81, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22688505

ABSTRACT

Based on a hollow fiber perfusion technology with internal oxygenation, a miniaturized bioreactor with a volume of 0.5 mL for in vitro studies was recently developed. Here, the suitability of this novel culture system for pharmacological studies was investigated, focusing on the model drug diclofenac. Primary human liver cells were cultivated in bioreactors and in conventional monolayer cultures in parallel over 10 days. From day 3 on, diclofenac was continuously applied at a therapeutic concentration (6.4 µM) for analysis of its metabolism. In addition, the activity and gene expression of the cytochrome P450 (CYP) isoforms CYP1A2, CYP2B6, CYP2C9, CYP2D6, and CYP3A4 were assessed. Diclofenac was metabolized in bioreactor cultures with an initial conversion rate of 230 ± 57 pmol/h/10(6) cells followed by a period of stable conversion of about 100 pmol/h/10(6) cells. All CYP activities tested were maintained until day 10 of bioreactor culture. The expression of corresponding mRNAs correlated well with the degree of preservation. Immunohistochemical characterization showed the formation of neo-tissue with expression of CYP2C9 and CYP3A4 and the drug transporters breast cancer resistance protein (BCRP) and multidrug resistance protein 2 (MRP2) in the bioreactor. In contrast, monolayer cultures showed a rapid decline of diclofenac conversion and cells had largely lost activity and mRNA expression of the assessed CYP isoforms at the end of the culture period. In conclusion, diclofenac metabolism, CYP activities and gene expression levels were considerably more stable in bioreactor cultures, making the novel bioreactor a useful tool for pharmacological or toxicological investigations requiring a highly physiological in vitro representation of the liver.


Subject(s)
Bioreactors , Cell Culture Techniques/instrumentation , Hepatocytes/metabolism , Miniaturization/instrumentation , Pharmacology/methods , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Cells, Cultured , Cytochrome P-450 Enzyme System/analysis , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Diclofenac/pharmacokinetics , Gene Expression Regulation , Hepatocytes/chemistry , Hepatocytes/cytology , Humans , Immunohistochemistry , Pharmacokinetics , Pharmacology/instrumentation , Real-Time Polymerase Chain Reaction
13.
Artif Organs ; 36(4): 446-9, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22074237

ABSTRACT

Medical treatment of burns and chronic wounds remains a challenge. We discussed a therapy concept that combines skin cell spray transplantation with a novel wound dressing based on artificial hollow fiber membrane capillaries. In skin cell-based therapy development, autologous skin progenitor cells are isolated from a healthy skin area and sprayed onto the wound. A medical device was introduced that uses perfused capillaries, known from clinical plasma separation, as a temporarily applied extracorporeal wound capillary bed. The functions of the dressing are comparable with those of dialysis; the capillaries, however, are applied externally onto the wound. Perfusion with a clinical peripheral nutrition and buffer solution can provide wound irrigation, wound debris removal, cell nutrition, pH regulation, and electrolyte balance while potentially serving to address delivery of regenerative factors and antibiosis. An innovative active skin wound dressing that provides cell support and stimulates regeneration by wound irrigation is discussed.


Subject(s)
Skin Transplantation/methods , Skin, Artificial , Skin/cytology , Bandages , Burns/surgery , Capillaries/physiology , Humans , Transplantation, Autologous
14.
Drug Metab Dispos ; 39(7): 1131-8, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21436404

ABSTRACT

Reliable and stable in vitro cellular systems maintaining specific liver functions important for drug metabolism and disposition are urgently needed in preclinical drug discovery and development research. The cell line HepaRG exhibits promising properties such as expression and function of drug-metabolizing enzymes and transporter proteins, which resemble those found in freshly isolated human hepatocytes. In this study, HepaRG cells were cultured up to 68 days in a three-dimensional multicompartment capillary membrane bioreactor, which enables high-density cell culture under dynamic conditions. The activity of drug-metabolizing cytochrome P450 (P450) enzymes was investigated by a cocktail of substrates for CYP1A1/2 (phenacetin), CYP2C9 (diclofenac), CYP2B6 (bupropion), and CYP3A4 (midazolam). The model P450 substrates, which were introduced to the bioreactor system mimicking in vivo bolus doses, showed stable metabolism over the entire experimental period of several weeks with the exception of bupropion hydroxylase, which increased over time. Ketoconazole treatment decreased the CYP3A4 activity by 69%, and rifampicin induced the CYP3A4- and CYP2B6-dependent activity 6-fold, which predicts well the magnitude of changes observed in vivo. Moreover, polarity of transporter expression and formation of tissue-like structures including bile canaliculi were demonstrated by immune histochemistry. The long-lasting bioreactor system using HepaRG cells thus provides a promising and stable liver-like in vitro model for continuous investigations of the hepatic kinetics of drugs and of drug-drug interactions, which well predict the situation in vivo in humans.


Subject(s)
Bioreactors , Cytochrome P-450 Enzyme System/metabolism , Cell Line , Humans , Substrate Specificity
15.
J Mater Sci Mater Med ; 22(5): 1087-99, 2011 May.
Article in English | MEDLINE | ID: mdl-21461918

ABSTRACT

Although significant progress has been made in the field of orthotopic liver transplantation, cell-based therapies seem to be a promising alternative to whole-organ transplantation. The reasons are manifold but organ shortage is the main cause for this approach. However, many problems such as the question which cell type should be used or which application site is best for transplantation have been raised. In addition, some clinicians have had success by cultivating liver cells in bioreactors for temporary life support. Besides answering the question which cell type, which injection site or even which culture form should be used for liver support recent international harmonization of legal requirements is needed to be addressed by clinicians, scientists and companies dealing with cellular therapies. We here briefly summarize the possible cell types used to partially or temporarily correct liver diseases, the most recent development of bioreactor technology and important regulatory issues.


Subject(s)
Hepatocytes/transplantation , Liver Diseases/therapy , Liver Failure/therapy , Liver, Artificial , Liver/cytology , Bioreactors , Cell Culture Techniques , Child , Child, Preschool , Equipment Design , Female , Humans , Infant , Informed Consent/legislation & jurisprudence , Male , Middle Aged
16.
Altern Lab Anim ; 39(2): 147-71, 2011 May.
Article in English | MEDLINE | ID: mdl-21639679

ABSTRACT

Drug-induced liver injury is a common reason for drug attrition in late clinical phases, and even for post-launch withdrawals. As a consequence, there is a broad consensus in the pharmaceutical industry, and within regulatory authorities, that a significant improvement of the current in vitro test methodologies for accurate assessment and prediction of such adverse effects is needed. For this purpose, appropriate in vivo-like hepatic in vitro models are necessary, in addition to novel sources of human hepatocytes. In this report, we describe recent and ongoing research toward the use of human embryonic stem cell (hESC)-derived hepatic cells, in conjunction with new and improved test methods, for evaluating drug metabolism and hepatotoxicity. Recent progress on the directed differentiation of human embryonic stem cells to the functional hepatic phenotype is reported, as well as the development and adaptation of bioreactors and toxicity assay technologies for the testing of hepatic cells. The aim of achieving a testing platform for metabolism and hepatotoxicity assessment, based on hESC-derived hepatic cells, has advanced markedly in the last 2-3 years. However, great challenges still remain, before such new test systems could be routinely used by the industry. In particular, we give an overview of results from the Vitrocellomics project (EU Framework 6) and discuss these in relation to the current state-of-the-art and the remaining difficulties, with suggestions on how to proceed before such in vitro systems can be implemented in industrial discovery and development settings and in regulatory acceptance.


Subject(s)
Animal Testing Alternatives , Drug Evaluation, Preclinical/methods , Embryonic Stem Cells , Hepatocytes/drug effects , Toxicity Tests/methods , Animals , Bioreactors , Biotransformation , Cell Differentiation , Cell Line , Cell Respiration , Enzyme Induction , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Liver/drug effects , Liver/metabolism , Metabolic Networks and Pathways , Rats
17.
Clin Exp Hepatol ; 7(1): 101-110, 2021 Mar.
Article in English | MEDLINE | ID: mdl-34027122

ABSTRACT

AIM OF THE STUDY: CD326 has been used as a single marker to enrich for hepatic stem cell populations in the liver. However, bile duct epithelium is also positive for CD326, which impedes the selection of pure hepatic stem cell populations. Some markers have been proposed to be co-expressed by hepatic stem cells but these have not been systematically compared. Therefore, we determined the percentages and compared the characteristics of human liver cells expressing potential stem cell surface markers. MATERIAL AND METHODS: We analyzed CD326 expression in human liver tissues from fetal, neonatal, pediatric, and adult stages using immunohistochemistry. In flow cytometry, we quantified fetal liver cells for their co-expression of CD326 with CD56, CD117, CD44, CD90, CD49f, LGR5 and SSEA4. We analyzed the various fractions for their quantitative expression of genes typically associated with progenitors and hepatic lineages. RESULTS: 12.5% of cells were positive for CD326; of these, 63.5% co-expressed CD44. The lowest co-expression percentages were for SSEA4 (2.1%) and LGR5 (0.7%). Fractions revealed distinct gene expression patterns. Of all combinations, cells that co-expressed surface CD326 and SSEA4 demonstrated the highest gene expression for the proliferation marker MKi67 and hepatic markers DLK1, AFP and ALB, and were the only fraction negative for the biliary epithelial marker KRT19. Histology of adult and fetal liver showed cells positive for CD326 and SSEA4 but negative for CK19. CONCLUSIONS: CD326-positive cells represent a heterogeneous population, which in combination with SSEA4 potentially distinguishes bile duct epithelium from hepatic stem cells. These findings can help to further classify human hepatic progenitor stages.

18.
Bioengineering (Basel) ; 8(8)2021 Jul 23.
Article in English | MEDLINE | ID: mdl-34436107

ABSTRACT

Liver cells cultured in 3D bioreactors is an interesting option for temporary extracorporeal liver support in the treatment of acute liver failure and for animal models for preclinical drug screening. Bioreactor capacity to eliminate drugs is generally used for assessing cell metabolic competence in different bioreactors or to scale-up bioreactor design and performance for clinical or preclinical applications. However, drug adsorption and physical transport often disguise the intrinsic drug biotransformation kinetics and cell metabolic state. In this study, we characterized the intrinsic kinetics of lidocaine elimination and adsorption by porcine liver cells cultured in 3D four-compartment hollow fiber membrane network perfusion bioreactors. Models of lidocaine transport and biotransformation were used to extract intrinsic kinetic information from response to lidocaine bolus of bioreactor versus adhesion cultures. Different from 2D adhesion cultures, cells in the bioreactors are organized in liver-like aggregates. Adsorption on bioreactor constituents significantly affected lidocaine elimination and was effectively accounted for in kinetic analysis. Lidocaine elimination and cellular monoethylglicinexylidide biotransformation featured first-order kinetics with near-to-in vivo cell-specific capacity that was retained for times suitable for clinical assist and drug screening. Different from 2D cultures, cells in the 3D bioreactors challenged with lidocaine were exposed to close-to-physiological lidocaine and monoethylglicinexylidide concentration profiles. Kinetic analysis suggests bioreactor technology feasibility for preclinical drug screening and patient assist and that drug adsorption should be accounted for to assess cell state in different cultures and when laboratory bioreactor design and performance is scaled-up to clinical use or toxicological drug screening.

19.
J Biomed Mater Res A ; 109(6): 859-868, 2021 06.
Article in English | MEDLINE | ID: mdl-32997885

ABSTRACT

Hydroxyapatite (HA) is a well-known regenerative biomaterial. However, the slow degradation rate of HA is still an obstacle in clinical applications. In this study, we concentrated on investigating the degradation behavior of the calcium-rich HA foams, which had a demonstrated effect on blood differentiation in previous studies. The HA foams were processed by an emulsion method and were infiltrated with calcium nitrate to create a calcium carbonate second phase, heterogeneously distributed on and under the surface of the foam. During the 28-day solubility test, the calcium carbonate phase contributed to enhanced Ca2+ ion release into the saline compared to phase pure HA foams. Both types of foams were biocompatible as demonstrated by human endothelial cell culture on their surface. The release of calcium ions, the degradation behavior, and the endothelial cell differentiation behavior suggest this biphasic ceramic is a candidate for bone marrow in vitro culture and a possible bone substitute material.


Subject(s)
Biocompatible Materials/chemistry , Calcium/chemistry , Durapatite/chemistry , Bone Substitutes , Calcium Carbonate/chemistry , Calcium Compounds/chemistry , Cells, Cultured , Emulsions , Endothelial Cells , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Nitrates/chemistry , Solubility , X-Ray Microtomography
20.
Cells Tissues Organs ; 192(1): 39-49, 2010.
Article in English | MEDLINE | ID: mdl-20197653

ABSTRACT

We describe hollow fiber-based three-dimensional (3D) dynamic perfusion bioreactor technology for embryonic stem cells (ESC) which is scalable for laboratory and potentially clinical translation applications. We added 2 more compartments to the typical 2-compartment devices, namely an additional media capillary compartment for countercurrent 'arteriovenous' flow and an oxygenation capillary compartment. Each capillary membrane compartment can be perfused independently. Interweaving the 3 capillary systems to form repetitive units allows bioreactor scalability by multiplying the capillary units and provides decentralized media perfusion while enhancing mass exchange and reducing gradient distances from decimeters to more physiologic lengths of <1 mm. The exterior of the resulting membrane network, the cell compartment, is used as a physically active scaffold for cell aggregation; adjusting intercapillary distances enables control of the size of cell aggregates. To demonstrate the technology, mouse ESC (mESC) were cultured in 8- or 800-ml cell compartment bioreactors. We were able to confirm the hypothesis that this bioreactor enables mESC expansion qualitatively comparable to that obtained with Petri dishes, but on a larger scale. To test this, we compared the growth of 129/SVEV mESC in static two-dimensional Petri dishes with that in 3D perfusion bioreactors. We then tested the feasibility of scaling up the culture. In an 800-ml prototype, we cultured approximately 5 x 10(9) cells, replacing up to 800 conventional 100-mm Petri dishes. Teratoma formation studies in mice confirmed protein expression and gene expression results with regard to maintaining 'stemness' markers during cell expansion.


Subject(s)
Cell Culture Techniques/methods , Embryonic Stem Cells/cytology , Animals , Bioreactors , Cell Culture Techniques/instrumentation , Cell Growth Processes/physiology , Cells, Cultured , Humans , Mice , Perfusion
SELECTION OF CITATIONS
SEARCH DETAIL