Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cells ; 8(8)2019 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-31349725

RESUMEN

Aging is a risk factor for adipose tissue dysfunction, which is associated with inflammatory innate immune mechanisms. Since the adipose tissue/liver axis contributes to hepatosteatosis, we sought to determine age-related adipose tissue dysfunction in the context of the activation of the innate immune system fostering fatty liver phenotypes. Using wildtype and immune-deficient mice, we compared visceral adipose tissue and liver mass as well as hepatic lipid storage in young (ca. 14 weeks) and adult (ca. 30 weeks) mice. Adipocyte size was determined as an indicator of adipocyte function and liver steatosis was quantified by hepatic lipid content. Further, lipid storage was investigated under normal and steatosis-inducing culture conditions in isolated hepatocytes. The physiological age-related increase in body weight was associated with a disproportionate increase in adipose tissue mass in immune-deficient mice, which coincided with higher triglyceride storage in the liver. Lipid storage was similar in isolated hepatocytes from wildtype and immune-deficient mice under normal culture conditions but was significantly higher in immune-deficient than in wildtype hepatocytes under steatosis-inducing culture conditions. Immune-deficient mice also displayed increased inflammatory, adipogenic, and lipogenic markers in serum and adipose tissue. Thus, the age-related increase in body weight coincided with an increase in adipose tissue mass and hepatic steatosis. In association with a (pro-)inflammatory milieu, aging thus promotes hepatosteatosis, especially in immune-deficient mice.


Asunto(s)
Tejido Adiposo/metabolismo , Proteínas de Unión al ADN/deficiencia , Metabolismo de los Lípidos/genética , Hígado/metabolismo , Adipocitos , Tejido Adiposo/patología , Animales , Hígado Graso/genética , Hígado Graso/metabolismo , Hígado Graso/patología , Femenino , Perfilación de la Expresión Génica , Hepatocitos , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/inmunología , Síndromes de Inmunodeficiencia/metabolismo , Síndromes de Inmunodeficiencia/patología , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo
2.
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
3.
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
4.
Sci Rep ; 7(1): 2617, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28572613

RESUMEN

In patients, acute kidney injury (AKI) is often due to haemodynamic impairment associated with hepatic decompensation following extended liver surgery. Mesenchymal stem cells (MSCs) supported tissue protection in a variety of acute and chronic diseases, and might hence ameliorate AKI induced by extended liver resection. Here, 70% liver resection was performed in male pigs. MSCs were infused through a central venous catheter and haemodynamic parameters as well as markers of acute kidney damage were monitored under intensive care conditions for 24 h post-surgery. Cytokine profiles were established to anticipate the MSCs' potential mode of action. After extended liver resection, hyperdynamic circulation, associated with hyponatraemia, hyperkalaemia, an increase in serum aldosterone and low urine production developed. These signs of hepatorenal dysfunction and haemodynamic impairment were corrected by MSC treatment. MSCs elevated PDGF levels in the serum, possibly contributing to circulatory homeostasis. Another 14 cytokines were increased in the kidney, most of which are known to support tissue regeneration. In conclusion, MSCs supported kidney and liver function after extended liver resection. They probably acted through paracrine mechanisms improving haemodynamics and tissue homeostasis. They might thus provide a promising strategy to prevent acute kidney injury in the context of post-surgery acute liver failure.


Asunto(s)
Lesión Renal Aguda/prevención & control , Hemodinámica , Hígado/lesiones , Trasplante de Células Madre Mesenquimatosas , Lesión Renal Aguda/sangre , Lesión Renal Aguda/etiología , Animales , Presión Sanguínea , Modelos Animales de Enfermedad , Hígado/cirugía , Masculino , Comunicación Paracrina , Sus scrofa
5.
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
6.
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
7.
Trends Mol Med ; 21(11): 673-686, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26476857

RESUMEN

Hepatocyte transplantation aims to provide a functional substitution of liver tissue lost due to trauma or toxins. Chronic liver diseases are associated with inflammation, deterioration of tissue homeostasis, and deprivation of metabolic capacity. Recent advances in liver biology have focused on the pro-regenerative features of mesenchymal stem cells (MSCs). We argue that MSCs represent an attractive therapeutic option to treat liver disease. Indeed, their pleiotropic actions include the modulation of immune reactions, the stimulation of cell proliferation, and the attenuation of cell death responses. These characteristics are highly warranted add-ons to their capacity for hepatocyte differentiation. Undoubtedly, the elucidation of the regenerative mechanisms of MSCs in different liver diseases will promote their versatile and disease-specific therapeutic use.


Asunto(s)
Hepatopatías/terapia , Hígado/citología , Hígado/fisiología , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Animales , Hepatocitos/citología , Hepatocitos/trasplante , Humanos , Células Madre Mesenquimatosas/metabolismo
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.
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
10.
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
11.
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
12.
Front Physiol ; 3: 78, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22485094

RESUMEN

Without therapeutic intervention acute liver failure (ALF) is the consequence of a progredient destruction of the liver parenchyma due to metabolic exhaustion of the hepatocytes. Perivenous hepatocytes are responsible for the detoxification of noxious compounds via the cytochrome P450 enzyme system. Liver transplantation is the only remaining therapeutic option in the end-stage of the disease. Assuming that metabolic capacity could be provided by healthy hepatocytes and thus substitute for the genuine parenchymal cells hepatocyte transplantation since quite some time is considered to be an alternative to whole liver transplantation. While this hypothesis achieved proof-of-concept in animal trials clinical breakthrough is still awaiting success, the reasons of which are ongoing matter of debate. In recent times mesenchymal stem cells (MSC) came into focus as a transplantable cell source to treat ALF. Interestingly, as demonstrated in various rodent animal models their mode of action is rather based on trophic support of hepatocytes remaining in the damaged host parenchyma rather than substitution of tissue loss. Mechanistically, either direct or indirect paracrine effects from the transplanted cells acting pro-proliferative, anti-apoptotic, and anti-inflammatory seem to trigger the regenerative response of the residual healthy hepatocytes in the otherwise lethally injured liver parenchyma. Thus, allogeneic MSC may be the best choice for the treatment of ALF taking advantage of their short-term benefit to sustain the critical phase of the acute insult avoiding long-term immunosuppression.

13.
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
14.
Methods Mol Biol ; 698: 315-30, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21431529

RESUMEN

The hepatocyte is the smallest functional entity of the liver and executes the majority of this organ's -metabolic functions. Hence, hepatocyte transplantation has become a versatile alternative to whole organ liver transplantation. This novel treatment option is based on the assumption that transplanted -hepatocytes integrate into the host liver, proliferate at the site of tissue damage, take over the long-term hepatic -synthetic capacity, and thus substitute for the diseased host tissue. However, clinical success is still waiting for a breakthrough, likely because of two major reasons including (1) the scarcity of cadaveric donor livers and (2) the largely poor quality of cells isolated from marginal quality donor organs. Therefore, alternative cell sources have to be established to further prompt the clinical success of hepatocyte transplantation. Due to their multiple differentiation potential and nearly unlimited availability, stem cells are an attractive -alternate resource. Because of both clinical and ethical objections, adult stem cells are often preferred over embryonic stem cells as a starting material. Recent studies have demonstrated the ability of mesenchymal stem cells derived from various tissues to differentiate into hepatocyte-like cells in vitro as well as showing specific hepatocyte functions in vivo after transplantation into the livers of mice or rats.


Asunto(s)
Hígado/citología , Trasplante de Células Madre Mesenquimatosas/métodos , Tejido Adiposo/citología , Animales , Células de la Médula Ósea/citología , Separación Celular , Dipeptidil Peptidasa 4/metabolismo , Citometría de Flujo , Regulación de la Expresión Génica , Humanos , Inmunohistoquímica , Células Madre Mesenquimatosas/metabolismo , Ratones , Modelos Animales , Perfusión , Vena Porta , Ratas , Bazo/metabolismo
15.
Nat Protoc ; 5(4): 617-27, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20224562

RESUMEN

Donor organ shortage is still the major obstacle for the clinical application of hepatocyte transplantation in the treatment of liver diseases. However, generation of hepatocyte-like cells from mesenchymal stem cells (MSCs) has become a real alternative to the isolation of primary hepatocytes. MSCs are extracted from the tissue by collagenase digestion and enriched by their capacity to grow on plastic surfaces. Enriched cells display distinct mesenchymal surface markers and are capable of multiple lineage differentiation. In the presence of specific growth conditions, the cells adopt functional features of differentiated hepatocytes. After orthotopic transplantation, differentiated human stem cells engraft in the host liver parenchyma of immunocompromised mice. This protocol describes the in vitro differentiation of stem cells from human bone marrow and their transplantation into livers of immunodeficient mice. The cell culture procedures take about 4-5 weeks, and cells engrafted in the mouse liver may be detected 2-3 months after transplantation.


Asunto(s)
Hepatocitos/citología , Hepatocitos/trasplante , Trasplante de Hígado/métodos , Células Madre Mesenquimatosas/citología , Albúminas/metabolismo , Animales , Células de la Médula Ósea/citología , Diferenciación Celular , Separación Celular , Hepatocitos/metabolismo , Humanos , Huésped Inmunocomprometido , Ratones , Ratones Noqueados , Modelos Animales , Trasplante Heterólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...