Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Int J Mol Sci ; 22(2)2021 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-33419219

RESUMO

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.


Assuntos
Células Epiteliais Alveolares/efeitos dos fármacos , Isquemia Fria/métodos , Meios de Cultivo Condicionados/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Traumatismo por Reperfusão/tratamento farmacológico , Células A549 , Células Epiteliais Alveolares/metabolismo , Âmnio/citologia , Apoptose/efeitos dos fármacos , Apoptose/genética , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Células Cultivadas , Citocinas/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , NF-kappa B/genética , Traumatismo por Reperfusão/fisiopatologia
2.
Biochem Biophys Res Commun ; 522(1): 171-176, 2020 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-31757423

RESUMO

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.


Assuntos
Âmnio/citologia , Técnicas de Cultura de Células/instrumentação , Células-Tronco Mesenquimais/citologia , Placenta/citologia , Indutores da Angiogênese/metabolismo , Materiais Biocompatíveis/química , Adesão Celular , Agregação Celular , Técnicas de Cultura de Células/métodos , Movimento Celular , Sobrevivência Celular , Células Imobilizadas/citologia , Células Imobilizadas/metabolismo , Feminino , Células Endoteliais da Veia Umbilical Humana , Humanos , Fatores Imunológicos/metabolismo , Células-Tronco Mesenquimais/metabolismo , Gravidez
3.
Differentiation ; 106: 9-14, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30826473

RESUMO

The aim of this review is to summarize and give an overview on the findings of signaling between hepatic and hematopoietic progenitors of the liver. To date, there are not many findings published in the field, and the aim of this review is to cover all current publications in this area. The liver is the main site of hematopoiesis during fetal development. However, little is known about how hepatic and other non-hematopoietic progenitors potentially influence hematopoiesis and vice versa. The concurrent peaks of hepatic and hematopoietic progenitor proliferation during development indicate interactions that could possibly be mediated through cell-cell contact, extracellular matrices, cytokines and growth factors, or other signaling molecules. For example, hepatic progenitors, such as hepatic stem cells and hepatoblasts, possess characteristic surface markers that can be cleaved, giving rise to fragments of various lengths. A surface molecule of hepatoblasts has been demonstrated to play an essential role in hematopoiesis. Particularly, these effects on hematopoiesis were distinct, depending on whether it was membrane-bound or cleaved. In this review, the various hepatic and hematopoietic progenitor cell types are concisely described, and the current findings of their potential interactions are summarized.


Assuntos
Diferenciação Celular , Feto/citologia , Células-Tronco Hematopoéticas/citologia , Fígado/citologia , Animais , Feto/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Humanos , Fígado/fisiologia
4.
J Cell Physiol ; 233(6): 4841-4851, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29150960

RESUMO

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.


Assuntos
Células-Tronco Adultas/metabolismo , Proliferação de Células , Neoplasias Colorretais/metabolismo , Molécula de Adesão da Célula Epitelial/metabolismo , Células-Tronco Fetais/metabolismo , Fígado/metabolismo , Células-Tronco Neoplásicas/metabolismo , Fragmentos de Peptídeos/metabolismo , Transdução de Sinais , Proteína ADAM17/genética , Proteína ADAM17/metabolismo , Células-Tronco Adultas/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Molécula de Adesão da Célula Epitelial/genética , Células-Tronco Fetais/efeitos dos fármacos , Regulação da Expressão Gênica , Glicosilação , Células HT29 , Humanos , Ácidos Hidroxâmicos/farmacologia , Fígado/citologia , Fígado/efeitos dos fármacos , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/patologia , Cultura Primária de Células , Domínios Proteicos , Transdução de Sinais/efeitos dos fármacos
5.
J Vasc Res ; 54(6): 376-385, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29166642

RESUMO

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.


Assuntos
Diferenciação Celular , Linhagem da Célula , Durapatita/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Engenharia Tecidual/métodos , Alicerces Teciduais , Biomarcadores/metabolismo , Proliferação de Células , Separação Celular/métodos , Células Cultivadas , Citometria de Fluxo , Regulação da Expressão Gênica , Humanos , Fenótipo , Porosidade , Propriedades de Superfície , Fatores de Tempo
6.
Xenotransplantation ; 24(2)2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28130881

RESUMO

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.


Assuntos
Sobrevivência de Enxerto/imunologia , Hepatócitos/imunologia , Transplante Heterólogo , Animais , Animais Geneticamente Modificados , Anticorpos/imunologia , Formação de Anticorpos/genética , Formação de Anticorpos/imunologia , Antígenos/imunologia , Sobrevivência de Enxerto/efeitos dos fármacos , Hepatócitos/transplante , Terapia de Imunossupressão/métodos , Imunossupressores/farmacologia , Papio hamadryas/imunologia , Suínos , Transplante Heterólogo/métodos
7.
Differentiation ; 89(5): 137-45, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26142155

RESUMO

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.


Assuntos
Diferenciação Celular , Epiderme/transplante , Queratinócitos/citologia , Adulto , Proliferação de Células , Tamanho Celular , Transplante de Células , Terapia Baseada em Transplante de Células e Tecidos , Células Cultivadas , Humanos , Queratinócitos/metabolismo , Queratinócitos/transplante , Transplante de Células-Tronco , Células-Tronco/citologia
8.
J Med Biol Eng ; 36: 153-167, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27231463

RESUMO

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.

9.
Am J Physiol Gastrointest Liver Physiol ; 308(4): G233-50, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-25477371

RESUMO

Epithelial cell adhesion molecule (EpCAM) is a transmembrane glycoprotein, which is frequently and highly expressed on carcinomas, tumor-initiating cells, selected tissue progenitors, and embryonic and adult stem cells. During liver development, EpCAM demonstrates a dynamic expression, since it can be detected in fetal liver, including cells of the parenchyma, whereas mature hepatocytes are devoid of EpCAM. Liver regeneration is associated with a population of EpCAM-positive cells within ductular reactions, which gradually lose the expression of EpCAM along with maturation into hepatocytes. EpCAM can be switched on and off through a wide panel of strategies to fine-tune EpCAM-dependent functional and differentiative traits. EpCAM-associated functions relate to cell-cell adhesion, proliferation, maintenance of a pluripotent state, regulation of differentiation, migration, and invasion. These functions can be conferred by the full-length protein and/or EpCAM-derived fragments, which are generated upon regulated intramembrane proteolysis. Control by EpCAM therefore not only depends on the presence of full-length EpCAM at cellular membranes but also on varying rates of the formation of EpCAM-derived fragments that have their own regulatory properties and on changes in the association of EpCAM with interaction partners. Thus spatiotemporal localization of EpCAM in immature liver progenitors, transit-amplifying cells, and mature liver cells will decisively impact the regulation of EpCAM functions and might be one of the triggers that contributes to the adaptive processes in stem/progenitor cell lineages. This review will summarize EpCAM-related molecular events and how they relate to hepatobiliary differentiation and regeneration.


Assuntos
Antígenos de Neoplasias/metabolismo , Moléculas de Adesão Celular/metabolismo , Hepatócitos/metabolismo , Fígado/metabolismo , Transdução de Sinais , Células-Tronco/metabolismo , Animais , Antígenos de Neoplasias/genética , Moléculas de Adesão Celular/genética , Diferenciação Celular , Linhagem da Célula , Proliferação de Células , Molécula de Adesão da Célula Epitelial , Regulação da Expressão Gênica no Desenvolvimento , Hepatócitos/patologia , Humanos , Fígado/patologia , Fígado/fisiopatologia , Hepatopatias/metabolismo , Hepatopatias/patologia , Hepatopatias/fisiopatologia , Regeneração Hepática , Células-Tronco/patologia
10.
Biotechnol Bioeng ; 112(4): 801-10, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25335987

RESUMO

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.


Assuntos
Reatores Biológicos , Medula Óssea , Técnicas de Cultura de Órgãos/métodos , Adulto , Fenômenos Fisiológicos Celulares , Durapatita , Humanos , Alicerces Teciduais
11.
Xenotransplantation ; 22(4): 239-48, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25950141

RESUMO

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.


Assuntos
Hepatócitos/transplante , Transplante Heterólogo/métodos , Animais , Animais Geneticamente Modificados , Sobrevivência de Enxerto , Xenoenxertos , Humanos , Modelos Animais , Suínos , Transplante Heterólogo/tendências
12.
Hepatol Res ; 45(8): 919-32, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25195540

RESUMO

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.

14.
J Exp Med ; 204(8): 1973-87, 2007 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-17664288

RESUMO

Human hepatic stem cells (hHpSCs), which are pluripotent precursors of hepatoblasts and thence of hepatocytic and biliary epithelia, are located in ductal plates in fetal livers and in Canals of Hering in adult livers. They can be isolated by immunoselection for epithelial cell adhesion molecule-positive (EpCAM+) cells, and they constitute approximately 0.5-2.5% of liver parenchyma of all donor ages. The self-renewal capacity of hHpSCs is indicated by phenotypic stability after expansion for >150 population doublings in a serum-free, defined medium and with a doubling time of approximately 36 h. Survival and proliferation of hHpSCs require paracrine signaling by hepatic stellate cells and/or angioblasts that coisolate with them. The hHpSCs are approximately 9 microm in diameter, express cytokeratins 8, 18, and 19, CD133/1, telomerase, CD44H, claudin 3, and albumin (weakly). They are negative for alpha-fetoprotein (AFP), intercellular adhesion molecule (ICAM) 1, and for markers of adult liver cells (cytochrome P450s), hemopoietic cells (CD45), and mesenchymal cells (vascular endothelial growth factor receptor and desmin). If transferred to STO feeders, hHpSCs give rise to hepatoblasts, which are recognizable by cordlike colony morphology and up-regulation of AFP, P4503A7, and ICAM1. Transplantation of freshly isolated EpCAM+ cells or of hHpSCs expanded in culture into NOD/SCID mice results in mature liver tissue expressing human-specific proteins. The hHpSCs are candidates for liver cell therapies.


Assuntos
Técnicas de Cultura de Células/métodos , Fígado/citologia , Fígado/embriologia , Células-Tronco/citologia , Adesão Celular , Membrana Celular/metabolismo , Meios de Cultura Livres de Soro/metabolismo , Células Epiteliais/citologia , Células-Tronco Hematopoéticas/metabolismo , Hepatócitos/metabolismo , Humanos , Molécula 1 de Adesão Intercelular/metabolismo , Antígenos Comuns de Leucócito/biossíntese , Fígado/metabolismo , Mesoderma/metabolismo , Transdução de Sinais , alfa-Fetoproteínas/metabolismo
15.
Liver Transpl ; 18(2): 226-37, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22034152

RESUMO

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.


Assuntos
Separação Celular/métodos , Doença Hepática Terminal/cirurgia , Células-Tronco Fetais/transplante , Transplante de Células-Tronco Hematopoéticas , Hepatite C/complicações , Cirrose Hepática/cirurgia , Fígado/embriologia , Transplante de Células-Tronco Mesenquimais , Perfusão , Biomarcadores/metabolismo , Técnicas de Cultura de Células , Sobrevivência Celular , Colagenases/metabolismo , Doença Hepática Terminal/virologia , Células-Tronco Fetais/metabolismo , Idade Gestacional , Células Hep G2 , Humanos , Imunossupressores/uso terapêutico , Fígado/irrigação sanguínea , Fígado/metabolismo , Cirrose Hepática/virologia , Masculino , Pessoa de Meia-Idade , Fenótipo , Veia Porta/embriologia , Resultado do Tratamento
16.
Clin Exp Hepatol ; 7(1): 101-110, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-34027122

RESUMO

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.

17.
J Biomed Mater Res A ; 109(6): 859-868, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-32997885

RESUMO

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.


Assuntos
Materiais Biocompatíveis/química , Cálcio/química , Durapatita/química , Substitutos Ósseos , Carbonato de Cálcio/química , Compostos de Cálcio/química , Células Cultivadas , Emulsões , Células Endoteliais , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Nitratos/química , Solubilidade , Microtomografia por Raio-X
18.
Cells Tissues Organs ; 192(1): 39-49, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20197653

RESUMO

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.


Assuntos
Técnicas de Cultura de Células/métodos , Células-Tronco Embrionárias/citologia , Animais , Reatores Biológicos , Técnicas de Cultura de Células/instrumentação , Processos de Crescimento Celular/fisiologia , Células Cultivadas , Humanos , Camundongos , Perfusão
19.
Artif Organs ; 34(6): 462-72, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20456323

RESUMO

Various metabolic tests were compared for the performance characterization of a liver cell bioreactor as a routine function assessment of cultures in a standby for patient application in clinical studies. Everyday quality assessment (QA) is essential to ensure a continuous level of cellular functional capacity in the development of hepatic progenitor cell expansion systems providing cells for regenerative medicine research; it is also of interest to meet safety requirements in bioartificial extracorporeal liver support systems under clinical evaluation. Quality criteria for the description of bioreactor cultures were developed using primary porcine liver cells as a model. Porcine liver cells isolated by collagenase perfusion with an average of 3 x 10(9) primary cells were used in 39 bioreactors for culture periods up to 33 days. Measurements of monoethylglycinexylidide synthesis and elimination of lidocaine, galactose elimination, and sorbitol elimination proved to be useful for routine QA of primary liver cell cultures. We demonstrate two methods for dispensing test substances, bolus administration and continuous, steady-state administration. Bolus test data were grouped in Standard, Therapy, Infection/Contamination, and Cell-free control groups. Statistical analyses show significant differences among all groups for every test substance. Post hoc comparisons indicated significant differences between Standard and Cell-free groups for all elimination parameters. For continuous tests, results were categorized according to number of culture days and time-dependent changes were analyzed. Continuous administration enables a better view of culture health and the time dependency of cellular function, whereas bolus administration is more flexible. Both procedures can be used to define cell function. Assessment of cellular function and bioreactor quality can contribute significantly to the quality of experimental or clinical studies in the field of hepatic bioreactor development.


Assuntos
Galactose/metabolismo , Hepatócitos/metabolismo , Lidocaína/análogos & derivados , Lidocaína/metabolismo , Fígado Artificial , Sorbitol/metabolismo , Animais , Reatores Biológicos , Técnicas de Cultura de Células/métodos , Células Cultivadas , Hepatócitos/citologia , Humanos , Fígado/citologia , Testes de Função Hepática , Suínos
20.
Biomed Res Int ; 2020: 9847579, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32309444

RESUMO

Mesenchymal stem cells (MSCs) and their secreted extracellular vesicles have been used effectively in different lung disease animal models and clinical trials. Their specific beneficial effects, the potential differences between MSCs derived from different organs, and interactions between MSC products and target cells still need to be studied further. Therefore, we investigated the effects of secreted products of human MSCs derived from the bone marrow and adipose tissue on human lung small airway epithelial (AE) cells in vitro. AE cells were cocultured with MSCs in inserts that allowed the free exchange of medium but did not allow direct cell-to-cell contact. We examined the effects on AE cell viability, proliferation, cell numbers, expression of AE cell-specific genes, and CD54 (intercellular adhesion molecule 1 (ICAM1)) surface positivity, as well as the secretion/uptake of growth factors relevant for AE cell. We found that coculture increased the viability of AE cells. The majority of AE cells expressed CD54 on their surface, but the percentage of cells being positive for CD54 did not increase in coculture. However, ICAM1 gene expression was increased in coculture. Also, we observed increased gene expression of mucin (MUC1), a lung-enriched cell surface glycoprotein. These observed effects were the same between bone marrow and adipose tissue MSCs. However, MSCs derived from adipose tissue reduced angiopoietin concentrations in coculture, whereas those from the bone marrow did not. Conclusively, MSCs influenced AE cells positively by increasing their viability and affecting gene expression, with some effects being specific for the tissue origin of MSCs.


Assuntos
Técnicas de Cocultura/métodos , Células Epiteliais/citologia , Pulmão , Células-Tronco Mesenquimais/citologia , Tecido Adiposo/citologia , Animais , Aquaporina 5 , Células da Medula Óssea/citologia , Células Cultivadas , Modelos Animais de Doenças , Expressão Gênica , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA