Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Exp Cell Res ; 369(2): 335-347, 2018 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-29864400

RESUMEN

The continuously decreasing willingness for liver donation aggravates treatment of end-stage liver diseases requiring organ transplantation as the only curative strategy. Cell therapy approaches using human hepatocytes or stem cell-derived hepatocyte-like cells may be a therapeutic option out of this dilemma. ABCB5-positive mesenchymal stromal cells from human skin featured promising potential to treat immune-mediated diseases. Since most of chronic liver diseases involve exaggerating immune mechanisms, it was the aim to demonstrate in this study, whether ABCB5+ stem cells may serve as a resource to generate hepatocytic cells for application in liver cell transplantation. Using an established single-step protocol, which had been successfully applied to differentiate mesenchymal stromal cells into the hepatocytic lineage, ABCB5+ skin-derived stem cells did not gain significant characteristics of hepatocytes. Yet, upon culture in hepatocytic differentiation medium, ABCB5+ stem cells secreted immunomodulatory and anti-fibrotic factors as well as proteins, which may prompt hepatic morphogenesis besides others. Hepatic transplantation of ABCB5+ stem cells, which had been prior cultured in hepatocytic differentiation medium, did not cause any obvious deterioration of liver architecture suggesting their safe application. Thus, human ABCB5+ skin-derived stem cells secreted putative hepatotropic factors after culture in hepatocytic differentiation medium.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Hepatocitos/citología , Hepatocitos/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Piel/citología , Piel/metabolismo , Subfamilia B de Transportador de Casetes de Unión a ATP , Animales , Biomarcadores/metabolismo , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular/fisiología , Linaje de la Célula , Medios de Cultivo , Hepatectomía , Hepatocitos/trasplante , Humanos , Regeneración Hepática , Trasplante de Hígado , Masculino , Ratones , Ratones Noqueados , Modelos Animales , Trasplantes/citología , Trasplantes/metabolismo
2.
Arch Toxicol ; 93(9): 2645-2660, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31435712

RESUMEN

Although liver transplantation is a potential effective cure for patients with end-stage liver diseases, this strategy has several drawbacks including high cost, long waiting list, and limited availability of liver organs. Therefore, stem cell-based therapy is presented as an alternative option, which showed promising results in animal models of acute and chronic liver injuries. ABCB5+ cells isolated from skin dermis represent an easy accessible and expandable source of homogenous stem cell populations. In addition, ABCB5+ cells showed already promising results in the treatment of corneal and skin injury. To date, the effect of these cells on liver injury is still unknown. In the current study, sixteen weeks old Mdr2KO mice were i.v. injected with 500,000 ABCB5+ cells using different experimental setups. The effects of cellular therapy on inflammation, fibrosis, apoptosis, and proliferation were analyzed in the collected liver tissues. Toxicity of ABCB5+ cells was additionally investigated in mice with partial liver resection. In vitro, the fibrosis- and inflammatory-modulating effects of supernatant from ABCB5+ cells were examined in the human hepatic stellate cell line (LX-2). Cell injections into fibrotic Mdr2KO mice as well as into mice upon partial liver resection have no signs of toxicity with regard to cell transformation, cellular damage, fibrosis or inflammation as compared to controls. We next investigated the effects of ABCB5+ cells on established biliary liver fibrosis in the Mdr2KO mice. ABCB5+ cells to some extent influenced the shape of the liver inflammatory response and significantly reduced the amount of collagen deposition, as estimated from quantification of sirius red staining. Furthermore, reduced apoptosis and enhanced death compensatory proliferation resulted from ABCB5+ cell transformation. The stem cells secreted several trophic factors that activated TGF-ß family signaling in cultured LX-2 hepatic stellate cells (HSCs), therewith shaping cell fate to an αSMAhigh, Vimentinlow phenotype. Taken together, ABCB5+ cells can represent a safe and feasible strategy to support liver regeneration and to reduce liver fibrosis in chronic liver diseases.


Asunto(s)
Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Cirrosis Hepática/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/metabolismo , Animales , Modelos Animales de Enfermedad , Células Estrelladas Hepáticas/citología , Células Estrelladas Hepáticas/metabolismo , Humanos , Inyecciones Intravenosas , Cirrosis Hepática/metabolismo , Pruebas de Función Hepática , Células Madre Mesenquimatosas/citología , Ratones Endogámicos BALB C , Ratones Noqueados , Miembro 4 de la Subfamilia B de Casete de Unión a ATP
3.
Arch Toxicol ; 93(12): 3669-3670, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31664497

RESUMEN

We wish to submit a corrigendum to the above-mentioned article. Thank you very much for consideration and publication.

4.
Am J Pathol ; 187(3): 553-569, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28088007

RESUMEN

Hepatocyte transplantation is an alternative to whole liver transplantation. Yet, efficient liver repopulation by transplanted hepatocytes is low in livers of old animals. This restraint might be because of the poor proliferative capacity of aged donor hepatocytes or the regenerative impairment of the recipient livers. The age-dependent liver repopulation by transplanted wild-type hepatocytes was investigated in juvenile and senescent rats deficient in dipeptidyl-peptidase IV. Repopulation was quantified by flow cytometry and histochemical estimation of dipeptidyl-peptidase IV enzyme activity of donor cells in the negative host liver. As a potential pathway involved, expression of cell cycle proteins was assessed. Irrespective of the age of the donor hepatocytes, large cell clusters appeared in juvenile, but only small clusters in senescent host livers. Because juvenile and senescent donor hepatocytes were likewise functional, host-derived factor(s) impaired senescent host liver repopulation. Growth hormone levels were significantly higher in juvenile than in senescent rats, suggesting that growth hormone might promote host liver repopulation. Indeed, short-term treatment with growth hormone augmented senescent host liver repopulation involving the growth hormone-mediated release of the transcriptional blockade of genes associated with cell cycle progression. Short-term growth hormone substitution might improve liver repopulation by transplanted hepatocytes, thus augmenting the therapeutic benefit of clinical hepatocyte transplantation in older patients.


Asunto(s)
Envejecimiento/fisiología , Hormona del Crecimiento/farmacología , Hepatocitos/trasplante , Hígado/citología , Animales , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Análisis por Conglomerados , Citocromo P-450 CYP2E1/metabolismo , Citocromo P-450 CYP3A/metabolismo , Dipeptidil Peptidasa 4/metabolismo , Perfilación de la Expresión Génica , Ontología de Genes , Hormona del Crecimiento/sangre , Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Humanos , Masculino , Ratas Endogámicas F344
5.
Cytometry A ; 93(1): 32-49, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28906582

RESUMEN

Mesenchymal stromal/stem cells (MSC) are promising candidates for cellular therapy of different diseases in humans and in animals. Following the guidelines of the International Society for Cell Therapy, human MSC may be identified by expression of a specific panel of cell surface markers (CD105+, CD73+, CD90+, CD34-, CD14-, or CD11b-, CD79- or CD19-, HLA-DR-). In addition, multiple differentiation potential into at least the osteogenic, adipogenic, and chondrogenic lineage is a main criterion for MSC definition. Human MSC and MSC of a variety of mammals isolated from different tissues meet these criteria. In addition to the abovementioned, they express many more cell surface markers. Yet, these are not uniquely expressed by MSC. The gross phenotypic appearance like marker expression and differentiation potential is similar albeit not identical for MSC from different tissues and species. Similarly, MSC may feature different biological characteristics depending on the tissue source and the isolation and culture procedures. Their versatile biological qualities comprising immunomodulatory, anti-inflammatory, and proregenerative capacities rely largely on the migratory and secretory capabilities of MSC. They are attracted to sites of tissue lesion and secrete factors to promote self-repair of the injured tissue. This is a big perspective for clinical MSC applications in both veterinary and human medicine. Phase I/II clinical trials have been initiated to assess safety and feasibility of MSC therapies in acute and chronic disease settings. Yet, since the mode of MSC action in a specific disease environment is still unknown at large, it is mandatory to unravel the response of MSC from a given source onto a specific disease environment in suitable animal models prior to clinical applications. © 2017 International Society for Advancement of Cytometry.


Asunto(s)
Células Madre Mesenquimatosas/citología , Animales , Antígenos CD/metabolismo , Diferenciación Celular , Movimiento Celular , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Ensayos Clínicos como Asunto , Humanos , Inmunomodulación , Mamíferos , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/inmunología , Células Madre Mesenquimatosas/fisiología , Regeneración , Seguridad , Especificidad de la Especie
6.
Cytotherapy ; 19(12): 1462-1473, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29074333

RESUMEN

BACKGROUND AIMS: Portal hypertension is the main cause of complications in cirrhosis caused primarily by extensive fibrosis. Both anti-fibrotic and pro-fibrotic properties of mesenchymal stromal cells (MSCs) have been described in various animal models of liver fibrosis. Therefore, the impact of MSCs on portal hypertension and fibrosis should be investigated in an animal model of liver cirrhosis. METHODS: The effect of systemic treatment with adipose tissue-derived MSCs, pre-differentiated into hepatocytic cells, was investigated in a rat model of liver cirrhosis induced by chronic inhalation of carbon tetrachloride. RESULTS: Chronic intoxication with carbon tetrachloride increased the portal venous pressure, which was significantly attenuated by the treatment with MSCs. Consistent with the increase in portal and sinusoidal resistance in the cirrhotic liver, the splenic weight increased, which was again attenuated by the MSCs. The cells had no impact on the spontaneous improvement of liver dysfunction after cessation of treatment with carbon tetrachloride. However, fibrosis was significantly improved as assessed by image quantification of collagen stained with Sirius red. However, hydroxyproline was unchanged indicating that fibrillary collagen content was not affected. That was in line with the finding that the activation of hepatic stellate cells, mainly contributing to excess collagen production in liver cirrhosis, was not affected by the MSCs. The expression of metalloproteinases and their inhibitors did also not change. DISCUSSION: It is suggested that hepatocytic differentiated MSCs improved portal blood flow in the cirrhotic liver rather by the physical reestablishment of liver architecture than by biochemical repair.


Asunto(s)
Tejido Adiposo/citología , Hipertensión Portal/terapia , Cirrosis Hepática/fisiopatología , Trasplante de Células Madre Mesenquimatosas/métodos , Animales , Tetracloruro de Carbono/toxicidad , Diferenciación Celular , Colágeno/metabolismo , Modelos Animales de Enfermedad , Fibrosis/terapia , Células Estrelladas Hepáticas/metabolismo , Hepatocitos/patología , Hipertensión Portal/etiología , Hígado/patología , Cirrosis Hepática/inducido químicamente , Masculino , Células Madre Mesenquimatosas/citología , Ratas Endogámicas F344
7.
Ann Surg ; 263(3): 546-56, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25775061

RESUMEN

OBJECTIVE: To prevent posthepatectomy acute liver failure after extended resection by treatment with mesenchymal stem cells (MSCs). BACKGROUND: Liver tumors often require extended liver resection, overburdening metabolic and regenerative capacities of the remnant organ. Resulting dysfunction and failure may be improved by the proregenerative characteristics of MSCs. METHODS: Extended liver resection was performed in (DPPIV)-deficient F344-Fischer rats. Wild-type animals served as donors of peritoneal adipose-derived MSCs. These were predifferentiated in vitro into hepatocytic cells and delivered to the liver by splenic application. Liver-related blood parameters (international normalized ratio, bilirubin, aspartate aminotransferase, alanine aminotransferase) and liver histology (hematoxylin-eosin, Sudan III) were determined to monitor liver function. Metabolic changes were assessed by metabolomic analyses in the remnant liver and the serum. Liver damage and regeneration were quantified by determination of the apoptotic and proliferation rates. RESULTS: MSCs supported survival after partial hepatectomy. They decreased liver-related blood parameters indicative for the improvement of liver function. The extensive lipid accumulation in hepatocytes illustrating the metabolic overload after resection was attenuated. Treatment with MSCs normalized imbalance of amino acids, acylcarnitines, sphingolipids, and glycerophospholipids in the liver and blood. Furthermore, MSCs decreased the apoptotic rate and increased the proliferation rate. The experimental time period (48 hours) was too short to allow for integration of MSCs into the host liver. Thus, the mode of action was probably indirect. CONCLUSIONS: MSCs ameliorated hepatic dysfunction and improved liver regeneration after extended resection by paracrine mechanisms. They may represent a new therapeutic option to treat posthepatectomy acute liver failure.


Asunto(s)
Fallo Hepático Agudo/metabolismo , Fallo Hepático Agudo/terapia , Trasplante de Células Madre Mesenquimatosas , Tejido Adiposo/citología , Animales , Apoptosis , Diferenciación Celular , Hepatectomía , Técnicas para Inmunoenzimas , Etiquetado Corte-Fin in Situ , Pruebas de Función Hepática , Regeneración Hepática/fisiología , Metabolómica , Ratas , Ratas Endogámicas F344 , Coloración y Etiquetado
8.
Cell Mol Life Sci ; 72(13): 2599-612, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25687506

RESUMEN

Both acute and chronic liver diseases are associated with ample re-modeling of the liver parenchyma leading to functional impairment, which is thus obviously the cause or the consequence of the disruption of the epithelial integrity. It was, therefore, the aim of this study to investigate the distribution of the adherens junction components E- and N-cadherin, which are important determinants of tissue cohesion. E-cadherin was expressed in periportal but not in perivenous hepatocytes. In contrast, N-cadherin was more enriched towards the perivenous hepatocytes. In agreement, ß-catenin, which links both cadherins via α-catenin to the actin cytoskeleton, was expressed ubiquitously. This zonal expression of cadherins was preserved in acute liver injury after treatment with acetaminophen or partial hepatectomy, but disrupted in chronic liver damage like in non-alcoholic steatohepatitis (NASH) or α1-antitrypsin deficiency. Hepatocyte proliferation during acetaminophen-induced liver damage was predominant at the boundary between the damaged perivenous and the intact periportal parenchyma indicating a minor contribution of periportal hepatocytes to liver regeneration. In NASH livers, an oval cell reaction was observed pointing to massive tissue damage coinciding with the gross impairment of hepatocyte proliferation. In the liver parenchyma, metabolic functions are distributed heterogeneously. For example, the expression of phosphoenolpyruvate carboxykinase and E-cadherin overlapped in periportal hepatocytes. Thus, during liver regeneration after acute damage, the intact periportal parenchyma might sustain essential metabolic support like glucose supply or ammonia detoxification. However, disruption of epithelial integrity during chronic challenges may increase susceptibility to metabolic liver diseases such as NASH or vice versa. This might suggest the regulatory integration of tissue cohesion and metabolic functions in the liver.


Asunto(s)
Uniones Adherentes/metabolismo , Cadherinas/metabolismo , Hepatopatías/metabolismo , Hígado/metabolismo , Modelos Biológicos , Citoesqueleto de Actina/metabolismo , Animales , Western Blotting , Proteínas de Unión al ADN/genética , Técnica del Anticuerpo Fluorescente , Inmunohistoquímica , Hepatopatías/patología , Ratones , Ratones Noqueados , Fosfoenolpiruvato Carboxiquinasa (ATP)/metabolismo , beta Catenina/metabolismo
9.
Int J Mol Sci ; 17(7)2016 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-27409608

RESUMEN

BACKGROUND: The beneficial impact of mesenchymal stem cells (MSC) on both acute and chronic liver diseases has been confirmed, although the molecular mechanisms behind it remain elusive. We aim to identify factors secreted by undifferentiated and hepatocytic differentiated MSC in vitro in order to delineate liver repair pathways potentially targeted by MSC. METHODS: Secreted factors were determined by protein arrays and related pathways identified by biomathematical analyses. RESULTS: MSC from adipose tissue and bone marrow expressed a similar pattern of surface markers. After hepatocytic differentiation, CD54 (intercellular adhesion molecule 1, ICAM-1) increased and CD166 (activated leukocyte cell adhesion molecule, ALCAM) decreased. MSC secreted different factors before and after differentiation. These comprised cytokines involved in innate immunity and growth factors regulating liver regeneration. Pathway analysis revealed cytokine-cytokine receptor interactions, chemokine signalling pathways, the complement and coagulation cascades as well as the Januskinase-signal transducers and activators of transcription (JAK-STAT) and nucleotide-binding oligomerization domain-like receptor (NOD-like receptor) signalling pathways as relevant networks. Relationships to transforming growth factor ß (TGF-ß) and hypoxia-inducible factor 1-α (HIF1-α) signalling seemed also relevant. CONCLUSION: MSC secreted proteins, which differed depending on cell source and degree of differentiation. The factors might address inflammatory and growth factor pathways as well as chemo-attraction and innate immunity. Since these are prone to dysregulation in most liver diseases, MSC release hepatotropic factors, potentially supporting liver regeneration.


Asunto(s)
Molécula de Adhesión Celular del Leucocito Activado/metabolismo , Citocinas/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Hígado/metabolismo , Células Madre Mesenquimatosas/metabolismo , Tejido Adiposo/citología , Células de la Médula Ósea/citología , Diferenciación Celular , Células Cultivadas , Quimiocinas/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Hígado/citología , Regeneración Hepática/fisiología , Células Madre Mesenquimatosas/citología , Proteínas NLR/metabolismo , Fenotipo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo
10.
Exp Cell Res ; 321(2): 267-75, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24200501

RESUMEN

STUDY BACKGROUND: Extended liver resection is the only curative treatment option of liver cancer. Yet, the residual liver may not accomplish the high metabolic and regenerative capacity needed, which frequently leads to acute liver failure. Because of their anti-inflammatory and -apoptotic as well as pro-proliferative features, mesenchymal stem cells differentiated into hepatocyte-like cells might provide functional and regenerative compensation. Clinical translation of basic research requires pre-clinical approval in large animals. Therefore, we characterized porcine mesenchymal stem cells (MSC) from adipose tissue and bone marrow and their hepatocyte differentiation potential for future assessment of functional liver support after surgical intervention in the pig model. METHODS: Mesenchymal surface antigens and multi-lineage differentiation potential of porcine MSC isolated by collagenase digestion either from bone marrow or adipose tissue (subcutaneous/visceral) were assessed by flow cytometry. Morphology and functional properties (urea-, glycogen synthesis and cytochrome P450 activity) were determined during culture under differentiation conditions and compared with primary porcine hepatocytes. RESULTS: MSC from porcine adipose tissue and from bone marrow express the typical mesenchymal markers CD44, CD29, CD90 and CD105 but not haematopoietic markers. MSC from both sources displayed differentiation into the osteogenic as well as adipogenic lineage. After hepatocyte differentiation, expression of CD105 decreased significantly and cells adopted the typical polygonal morphology of hepatocytes. Glycogen storage was comparable in adipose tissue- and bone marrow-derived cells. Urea synthesis was about 35% lower in visceral than in subcutaneous adipose tissue-derived MSC. Cytochrome P450 activity increased significantly during differentiation and was twice as high in hepatocyte-like cells generated from bone marrow as from adipose tissue. CONCLUSION: The hepatocyte differentiation of porcine adipose tissue-derived MSC was shown for the first time yielding hepatocyte-like cells with specific functions similar in bone marrow and subcutaneous adipose tissue-derived MSC. That makes them good pre-clinical candidates for supportive approaches after liver resection in the pig.


Asunto(s)
Tejido Adiposo/citología , Diferenciación Celular , Hepatocitos/fisiología , Células Madre Mesenquimatosas/fisiología , Investigación Biomédica Traslacional , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/fisiología , Células Cultivadas , Femenino , Grasa Intraabdominal/citología , Células Madre Mesenquimatosas/citología , Fenotipo , Grasa Subcutánea/citología , Porcinos , Investigación Biomédica Traslacional/métodos
11.
Exp Cell Res ; 326(2): 230-9, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-24786317

RESUMEN

Non-alcoholic steatohepatitis (NASH) is a frequent clinical picture characterised by hepatic inflammation, lipid accumulation and fibrosis. When untreated, NASH bears a high risk of developing liver cirrhosis and consecutive hepatocellular carcinoma requiring liver transplantation in its end-stage. However, donor organ scarcity has prompted the search for alternatives, of which hepatocyte or stem cell-derived hepatocyte transplantation are regarded auspicious options of treatment. Mesenchymal stem cells (MSC) are able to differentiate into hepatocyte-like cells and thus may represent an alternative cell source to primary hepatocytes. In addition these cells feature anti-inflammatory and pro-regenerative characteristics, which might favour liver recovery from NASH. The aim of this study was to investigate the potential benefit of hepatocyte-like cells derived from human bone marrow MSC in a mouse model of diet-induced NASH. Seven days post-transplant, human hepatocyte-like cells were found in the mouse liver parenchyma. Triglyceride depositions were lowered in the liver but restored to normal in the blood. Hepatic inflammation was attenuated as verified by decreased expression of the acute phase protein serum amyloid A, inflammation-associated markers (e.g. lipocalin 2), as well as the pro-inflammatory cytokine TNFα. Moreover, the proliferation of host hepatocytes that indicate the regenerative capacity in livers receiving cell transplants was enhanced. Transplantation of MSC-derived human hepatocyte-like cells corrects NASH in mice by restoring triglyceride depositions, reducing inflammation and augmenting the regenerative capacity of the liver.


Asunto(s)
Hígado Graso/terapia , Trasplante de Células Madre Mesenquimatosas , Animales , Diferenciación Celular , Proliferación Celular , Modelos Animales de Enfermedad , Hígado Graso/metabolismo , Hígado Graso/patología , Hepatocitos/patología , Xenoinjertos , Humanos , Hígado/metabolismo , Hígado/patología , Regeneración Hepática , Masculino , Células Madre Mesenquimatosas/patología , Ratones , Ratones Noqueados , Enfermedad del Hígado Graso no Alcohólico , Triglicéridos/metabolismo
12.
Arch Toxicol ; 88(5): 1161-83, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24748404

RESUMEN

Histological alterations often constitute a fingerprint of toxicity and diseases. The extent to which these alterations are cause or consequence of compromised organ function, and the underlying mechanisms involved is a matter of intensive research. In particular, liver disease is often associated with altered tissue microarchitecture, which in turn may compromise perfusion and functionality. Research in this field requires the development and orchestration of new techniques into standardized processing pipelines that can be used to reproducibly quantify tissue architecture. Major bottlenecks include the lack of robust staining, and adequate reconstruction and quantification techniques. To bridge this gap, we established protocols employing specific antibody combinations for immunostaining, confocal imaging, three-dimensional reconstruction of approximately 100-µm-thick tissue blocks and quantification of key architectural features. We describe a standard procedure termed 'liver architectural staining' for the simultaneous visualization of bile canaliculi, sinusoidal endothelial cells, glutamine synthetase (GS) for the identification of central veins, and DAPI as a nuclear marker. Additionally, we present a second standard procedure entitled 'S-phase staining', where S-phase-positive and S-phase-negative nuclei (stained with BrdU and DAPI, respectively), sinusoidal endothelial cells and GS are stained. The techniques include three-dimensional reconstruction of the sinusoidal and bile canalicular networks from the same tissue block, and robust capture of position, size and shape of individual hepatocytes, as well as entire lobules from the same tissue specimen. In addition to the protocols, we have also established image analysis software that allows relational and hierarchical quantifications of different liver substructures (e.g. cells and vascular branches) and events (e.g. cell proliferation and death). Typical results acquired for routinely quantified parameters in adult mice (C57Bl6/N) include the hepatocyte volume (5,128.3 ± 837.8 µm(3)) and the fraction of the hepatocyte surface in contact with the neighbouring hepatocytes (67.4 ± 6.7 %), sinusoids (22.1 ± 4.8 %) and bile canaliculi (9.9 ± 3.8 %). Parameters of the sinusoidal network that we also routinely quantify include the radius of the sinusoids (4.8 ± 2.25 µm), the branching angle (32.5 ± 11.2°), the length of intersection branches (23.93 ± 5.9 µm), the number of intersection nodes per mm(3) (120.3 × 103 ± 42.1 × 10(3)), the average length of sinusoidal vessel per mm(3) (5.4 × 10(3) ± 1.4 × 10(3)mm) and the percentage of vessel volume in relation to the whole liver volume (15.3 ± 3.9) (mean ± standard deviation). Moreover, the provided parameters of the bile canalicular network are: length of the first-order branches (7.5 ± 0.6 µm), length of the second-order branches (10.9 ± 1.8 µm), length of the dead-end branches (5.9 ± 0.7 µm), the number of intersection nodes per mm(3) (819.1 × 10(3) ± 180.7 × 10(3)), the number of dead-end branches per mm(3) (409.9 × 10(3) ± 95.6 × 10(3)), the length of the bile canalicular network per mm(3) (9.4 × 10(3) ± 0.7 × 10(3) mm) and the percentage of the bile canalicular volume with respect to the total liver volume (3.4 ± 0.005). A particular strength of our technique is that quantitative parameters of hepatocytes and bile canalicular as well as sinusoidal networks can be extracted from the same tissue block. Reconstructions and quantifications performed as described in the current protocols can be used for quantitative mathematical modelling of the underlying mechanisms. Furthermore, protocols are presented for both human and pig livers. The technique is also applicable for both vibratome blocks and conventional paraffin slices.


Asunto(s)
Canalículos Biliares/citología , Procesamiento de Imagen Asistido por Computador/métodos , Hígado/irrigación sanguínea , Coloración y Etiquetado/métodos , Animales , Especificidad de Anticuerpos , Dipeptidil Peptidasa 4/inmunología , Hepatocitos/citología , Humanos , Procesamiento de Imagen Asistido por Computador/instrumentación , Hígado/ultraestructura , Masculino , Ratones Endogámicos C57BL , Microcirculación , Adhesión en Parafina , Control de Calidad , Reproducibilidad de los Resultados , Porcinos
13.
Int J Mol Sci ; 15(4): 7004-28, 2014 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-24758938

RESUMEN

Mesenchymal stem cells from human bone marrow (hMSC) have the potential to differentiate into hepatocyte-like cells in vitro and continue to maintain important hepatocyte functions in vivo after transplantation into host mouse livers. Here, hMSC were differentiated into hepatocyte-like cells in vitro (hMSC-HC) and transplanted into livers of immunodeficient Pfp/Rag2⁻/⁻ mice treated with a sublethal dose of acetaminophen (APAP) to induce acute liver injury. APAP induced a time- and dose-dependent damage of perivenous areas of the liver lobule. Serum levels of aspartate aminotransferase (AST) increased to similar levels irrespective of hMSC-HC transplantation. Yet, hMSC-HC resided in the damaged perivenous areas of the liver lobules short-term preventing apoptosis and thus progress of organ destruction. Disturbance of metabolic protein expression was lower in the livers receiving hMSC-HC. Seven weeks after APAP treatment, hepatic injury had completely recovered in groups both with and without hMSC-HC. Clusters of transplanted cells appeared predominantly in the periportal portion of the liver lobule and secreted human albumin featuring a prominent quality of differentiated hepatocytes. Thus, hMSC-HC attenuated the inflammatory response and supported liver regeneration after acute injury induced by acetaminophen. They hence may serve as a novel source of hepatocyte-like cells suitable for cell therapy of acute liver diseases.


Asunto(s)
Células de la Médula Ósea/citología , Enfermedad Hepática Inducida por Sustancias y Drogas/terapia , Hepatocitos/trasplante , Células Madre Mesenquimatosas/citología , Acetaminofén/toxicidad , Animales , Aspartato Aminotransferasas/sangre , Diferenciación Celular , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Hepatocitos/citología , Humanos , Regeneración Hepática , Masculino , Ratones , Factores de Tiempo
14.
Cells ; 13(6)2024 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-38534373

RESUMEN

Extended liver resection carries the risk of post-surgery liver failure involving thrombospondin-1-mediated aggravation of hepatic epithelial plasticity and function. Mesenchymal stromal cells (MSCs), by interfering with thrombospondin-1 (THBS1), counteract hepatic dysfunction, though the mechanisms involved remain unknown. Herein, two-thirds partial hepatectomy in mice increased hepatic THBS1, downstream transforming growth factor-ß3, and perturbation of liver tissue homeostasis. All these events were ameliorated by hepatic transfusion of human bone marrow-derived MSCs. Treatment attenuated platelet and macrophage recruitment to the liver, both major sources of THBS1. By mitigating THBS1, MSCs muted surgery-induced tissue deterioration and dysfunction, and thus supported post-hepatectomy regeneration. After liver surgery, patients displayed increased tissue THBS1, which is associated with functional impairment and may indicate a higher risk of post-surgery complications. Since liver dysfunction involving THBS1 improves with MSC treatment in various animal models, it seems feasible to also modulate THBS1 in humans to impede post-surgery acute liver failure.


Asunto(s)
Hepatopatías , Células Madre Mesenquimatosas , Humanos , Ratones , Animales , Hepatectomía , Regeneración Hepática/fisiología , Trombospondinas
15.
Cytometry A ; 83(1): 90-102, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23184585

RESUMEN

The liver has an enormous potential to restore the parenchymal tissue loss due to injury. This is accomplished by the proliferation of either the hepatocytes or liver progenitor cells in cases where massive damage prohibits hepatocytes from entering the proliferative response. Under debate is still whether hepatic stem cells are involved in liver tissue maintenance and regeneration or even whether they exist at all. The definition of an adult tissue-resident stem cell comprises basic functional stem cell criteria like the potential of self-renewal, multipotent, i.e. at least bipotent differentiation capacity and serial transplantability featuring the ability of functional tissue repopulation. The relationship between a progenitor and its progeny should exemplify the lineage commitment from the putative stem cell to the differentiated cell. This is mainly assessed by lineage tracing and immunohistochemical identification of markers specific to progenitors and their descendants. Flow cytometry approaches revealed that the liver stem cell population in animals is likely to be heterogeneous giving rise to progeny with different molecular signatures, depending on the stimulus to activate the putative stem cell compartment. The stem cell criteria are met by a variety of cells identified in the fetal and adult liver both under normal and injury conditions. It is the purpose of this review to verify hepatic stem cell candidates in the light of the stem cell definition criteria mentioned. Also from this point of view adult stem cells from non-hepatic tissues such as bone marrow, umbilical cord blood or adipose tissue, have the potential to differentiate into cells featuring functional hepatocyte characteristics. This has great impact because it opens the possibility of generating hepatocyte-like cells from adult stem cells in a sufficient amount and quality for their therapeutical application to treat end-stage liver diseases by stem cell-based hepatocytes in place of whole organ transplantation.


Asunto(s)
Células Madre Adultas/fisiología , Regeneración Hepática/fisiología , Hígado/citología , Hígado/fisiología , Células Madre Adultas/trasplante , Diferenciación Celular/fisiología , Proliferación Celular , Trasplante de Células , Hepatocitos/citología , Hepatocitos/fisiología , Humanos , Hepatopatías/terapia
16.
Exp Cell Res ; 318(3): 276-87, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22108588

RESUMEN

Non-alcoholic staetohepatitis (NASH) is associated with fat deposition in the liver favoring inflammatory processes and development of fibrosis, cirrhosis and finally hepatocellular cancer. In Western lifestyle countries, NASH has reached a 20% prevalence in the obese population with escalating tendency in the future. Very often, liver transplantation is the only therapeutic option. Recently, transplantation of hepatocyte-like cells differentiated from mesenchymal stem cells was suggested a feasible alternative to whole organ transplantation to ameliorate donor organ shortage. Hence, in the present work an animal model of NASH was established in immunodeficient mice to investigate the feasibility of human stem cell-derived hepatocyte-like cell transplantation. NASH was induced by feeding a methionine/choline-deficient diet (MCD-diet) for up to 5 weeks. Animals developed a fatty liver featuring fibrosis and elevation of the proinflammatory markers serum amyloid A (SAA) and tumor necrosis factor alpha (TNFα). Hepatic triglycerides were significantly increased as well as alanine aminotransferase demonstrating inflammation-linked hepatocyte damage. Elevation of αSMA mRNA and collagen I as well as liver architecture deterioation indicated massive fibrosis. Both short- and long-term post-transplantation human hepatocyte-like cells resided in the mouse host liver indicating parenchymal penetration and most likely functional engraftment. Hence, the NASH model in the immunodeficient mouse is the first to allow for the assessment of the therapeutic impact of human stem cell-derived hepatocyte transplantation.


Asunto(s)
Deficiencia de Colina/complicaciones , Dieta/efectos adversos , Modelos Animales de Enfermedad , Hígado Graso/etiología , Hígado Graso/terapia , Trasplante de Hígado , Metionina/deficiencia , Animales , Proteínas de Unión al ADN/genética , Hígado Graso/genética , Hígado Graso/patología , Estudios de Factibilidad , Regulación de la Expresión Génica , Masculino , Ratones , Ratones SCID , Ratones Transgénicos , Enfermedad del Hígado Graso no Alcohólico , Fenotipo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
17.
Hepatology ; 53(4): 1388-90, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21480341

RESUMEN

Cancer stem cells (CSCs) are generally dormant or slowly cycling tumor cells that have the ability to reconstitute tumors. They are thought to be involved in tumor resistance to chemo/radiation therapy and tumor relapse and progression. However, neither their existence nor their identity within many cancers has been well defined. Here, we have demonstrated that CD13 is a marker for semiquiescent CSCs in human liver cancer cell lines and clinical samples and that targeting these cells might provide a way to treat this disease. CD13+ cells predominated in the G0 phase of the cell cycle and typically formed cellular clusters in cancer foci. Following treatment, these cells survived and were enriched along the fibrous capsule where liver cancers usually relapse. Mechanistically, CD13 reduced ROS-induced DNA damage after genotoxic chemo/radiation stress and protected cells from apoptosis. In mouse xenograft models, combination of a CD13 inhibitor and the genotoxic chemotherapeutic fluorouracil (5-FU) drastically reduced tumor volume compared with either agent alone. 5-FU inhibited CD90+ proliferating CSCs, some of which produce CD13+ semiquiescent CSCs, while CD13 inhibition suppressed the self-renewing and tumor-initiating ability of dormant CSCs. Therefore, combining a CD13 inhibitor with a ROS-inducing chemo/radiation therapy may improve the treatment of liver cancer.

18.
Cells ; 11(11)2022 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-35681524

RESUMEN

Mesenchymal stromal cells (MSC) increasingly emerge as an option to ameliorate non-alcoholic steatohepatitis (NASH), a serious disease, which untreated may progress to liver cirrhosis and cancer. Before clinical translation, the mode of action of MSC needs to be established. Here, we established NASH in an immune-deficient mouse model by feeding a high fat diet. Human bone-marrow-derived MSC were delivered to the liver via intrasplenic transplantation. As verified by biochemical and image analyses, human mesenchymal stromal cells improved high-fat-diet-induced NASH in the mouse liver by decreasing hepatic lipid content and inflammation, as well as by restoring tissue homeostasis. MSC-mediated changes in gene expression indicated the switch from lipid storage to lipid utilization. It was obvious that host mouse hepatocytes harbored human mitochondria. Thus, it is feasible that resolution of NASH in mouse livers involved the donation of human mitochondria to the mouse hepatocytes. Therefore, human MSC might provide oxidative capacity for lipid breakdown followed by restoration of metabolic and tissue homeostasis.


Asunto(s)
Células Madre Mesenquimatosas , Enfermedad del Hígado Graso no Alcohólico , Animales , Dieta Alta en Grasa/efectos adversos , Humanos , Lípidos , Células Madre Mesenquimatosas/metabolismo , Ratones , Mitocondrias/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo
19.
Sci Rep ; 12(1): 21825, 2022 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-36528753

RESUMEN

Little is known about the impact of morphological disorders in distinct zones on metabolic zonation. It was described recently that periportal fibrosis did affect the expression of CYP proteins, a set of pericentrally located drug-metabolizing enzymes. Here, we investigated whether periportal steatosis might have a similar effect. Periportal steatosis was induced in C57BL6/J mice by feeding a high-fat diet with low methionine/choline content for either two or four weeks. Steatosis severity was quantified using image analysis. Triglycerides and CYP activity were quantified in photometric or fluorometric assay. The distribution of CYP3A4, CYP1A2, CYP2D6, and CYP2E1 was visualized by immunohistochemistry. Pharmacokinetic parameters of test drugs were determined after injecting a drug cocktail (caffeine, codeine, and midazolam). The dietary model resulted in moderate to severe mixed steatosis confined to periportal and midzonal areas. Periportal steatosis did not affect the zonal distribution of CYP expression but the activity of selected CYPs was associated with steatosis severity. Caffeine elimination was accelerated by microvesicular steatosis, whereas midazolam elimination was delayed in macrovesicular steatosis. In summary, periportal steatosis affected parameters of pericentrally located drug metabolism. This observation calls for further investigations of the highly complex interrelationship between steatosis and drug metabolism and underlying signaling mechanisms.


Asunto(s)
Hígado Graso , Midazolam , Ratones , Animales , Midazolam/farmacología , Cafeína/farmacocinética , Tasa de Depuración Metabólica , Sistema Enzimático del Citocromo P-450/metabolismo
20.
Cell Mol Gastroenterol Hepatol ; 13(4): 1041-1055, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34954190

RESUMEN

BACKGROUND & AIMS: The endocannabinoid system is involved in the modulation of inflammatory, fibrotic, metabolic, and carcinogenesis-associated signaling pathways via cannabinoid receptor (CB)1 and CB2. We hypothesized that the pharmacologic antagonization of CB1 receptor improves cholestasis in Abcb4-/- mice. METHODS: After weaning, male Abcb4-/- mice were treated orally with rimonabant (a specific antagonist of CB1) or ACEA (an agonist of CB1) until up to 16 weeks of age. Liver tissue and serum were isolated and examined by means of serum analysis, quantitative real time polymerase chain reaction, Western blot, immunohistochemistry, and enzyme function. Untreated Abcb4-/- and Bagg Albino Mouse/c wild-type mice served as controls. RESULTS: Cholestasis-induced symptoms such as liver damage, bile duct proliferation, and enhanced circulating bile acids were improved by CB1 antagonization. Rimonabant treatment also improved Phosphoenolpyruvat-Carboxykinase expression and reduced inflammation and the acute-phase response. The carcinogenesis-associated cellular-Jun N-terminal kinase/cellular-JUN and signal transducer and activator of transcription 3 signaling pathways activated in Abcb4-/- mice were reduced to wild-type level by CB1 antagonization. CONCLUSIONS: We showed a protective effect of oral CB1 antagonization in chronic cholestasis using the established Abcb4-/- model. Our results suggest that pharmacologic antagonization of the CB1 receptor could have a therapeutic benefit in cholestasis-associated metabolic changes, liver damage, inflammation, and carcinogenesis.


Asunto(s)
Colestasis , Receptor Cannabinoide CB1 , Animales , Carcinogénesis , Colestasis/tratamiento farmacológico , Inflamación , Masculino , Ratones , Rimonabant/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA