Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 168
Filtrar
2.
Int J Mol Sci ; 23(23)2022 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-36499207

RESUMEN

Three decades of hepatocyte transplantation have confirmed such a cell-based approach as an adjunct or alternative treatment to solid organ transplantation. Donor cell survival and engraftment were indirectly measured by hepatospecific secretive or released metabolites, such as ammonia metabolism in urea cycle defects. In cases of sepsis or viral infection, ammonia levels can significantly and abruptly increase in these recipients, erroneously implying rejection. Pro-inflammatory cytokines associated with viral or bacterial infections are known to affect many liver functions, including drug-metabolizing enzymes and hepatic transport activities. We examined the influence of pro-inflammatory cytokines in primary human hepatocytes, isolated from both normal donors or patients with metabolic liver diseases. Different measures of hepatocyte functions, including ammonia metabolism and phase 1-3 metabolism, were performed. All the hepatic functions were profoundly and significantly suppressed after exposure to concentrations of from 0.1 to 10 ng/mL of different inflammatory cytokines, alone and in combination. Our data indicate that, like phase I metabolism, suppression of phase II/III and ammonia metabolism occurs in hepatocytes exposed to pro-inflammatory cytokines in the absence of cell death. Such inflammatory events do not necessarily indicate a rejection response or loss of the cell graft, and these systemic inflammatory signals should be carefully considered when the immunosuppressant regiment is reduced or relieved in a hepatocyte transplantation recipient in response to such alleged rejection.


Asunto(s)
Hepatopatías , Enfermedades Metabólicas , Humanos , Citocinas/metabolismo , Amoníaco/metabolismo , Hepatocitos/metabolismo , Hígado/metabolismo , Hepatopatías/metabolismo , Enfermedades Metabólicas/metabolismo
3.
Mol Psychiatry ; 27(8): 3533-3543, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35418601

RESUMEN

Liver-generated plasma apolipoprotein E (apoE) does not enter the brain but nonetheless correlates with Alzheimer's disease (AD) risk and AD biomarker levels. Carriers of APOEε4, the strongest genetic AD risk factor, exhibit lower plasma apoE and altered brain integrity already at mid-life versus non-APOEε4 carriers. Whether altered plasma liver-derived apoE or specifically an APOEε4 liver phenotype promotes neurodegeneration is unknown. Here we investigated the brains of Fah-/-, Rag2-/-, Il2rg-/- mice on the Non-Obese Diabetic (NOD) background (FRGN) with humanized-livers of an AD risk-associated APOE ε4/ε4 versus an APOE ε2/ε3 genotype. Reduced endogenous mouse apoE levels in the brains of APOE ε4/ε4 liver mice were accompanied by various changes in markers of synaptic integrity, neuroinflammation and insulin signaling. Plasma apoE4 levels were associated with unfavorable changes in several of the assessed markers. These results propose a previously unexplored role of the liver in the APOEε4-associated risk of neurodegenerative disease.


Asunto(s)
Enfermedad de Alzheimer , Enfermedades Neurodegenerativas , Animales , Ratones , Apolipoproteína E4/genética , Ratones Endogámicos NOD , Apolipoproteínas E/genética , Encéfalo/metabolismo , Enfermedad de Alzheimer/genética , Genotipo , Biomarcadores , Hígado/metabolismo
4.
Cell Transplant ; 31: 9636897211069900, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35094608

RESUMEN

Hepatocyte transplantation is a promising treatment for liver failure and inborn metabolic liver diseases, but progress has been hampered by a scarcity of available organs. Here, hepatocytes isolated from livers procured for a neonatal hepatocyte donation program within a research setting were assessed for metabolic function and suitability for transplantation. Organ donation was considered for infants who died in neonatal intensive care in the Stockholm region during 2015-2021. Inclusion was assessed when a decision to discontinue life-sustaining treatment had been made and hepatectomy performed after declaration of death. Hepatocyte isolation was performed by three-step collagenase perfusion. Hepatocyte viability, yield, and function were assessed using fresh and cryopreserved cells. Engraftment and maturation of cryopreserved neonatal hepatocytes were assessed by transplantation into an immunodeficient mouse model and analysis of the gene expression of phase I, phase II, and liver-specific enzymes and proteins. Twelve livers were procured. Median warm ischemia time (WIT) was 190 [interquartile range (IQR): 80-210] minutes. Median viability was 86% (IQR: 71%-91%). Median yield was 6.9 (IQR: 3.4-12.8) x106 viable hepatocytes/g. Transplantation into immunodeficient mice resulted in good engraftment and maturation of hepatocyte-specific proteins and enzymes. A neonatal organ donation program including preterm born infants was found to be feasible. Hepatocytes isolated from neonatal donors had good viability, function, and engraftment despite prolonged WIT. Therefore, neonatal livers should be considered as a donor source for clinical hepatocyte transplantation, even in cases with extended WIT.


Asunto(s)
Trasplante de Hígado , Obtención de Tejidos y Órganos , Animales , Hepatocitos/metabolismo , Humanos , Recién Nacido , Hígado/metabolismo , Trasplante de Hígado/métodos , Ratones , Donantes de Tejidos
5.
Int J Mol Sci ; 22(3)2021 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-33530582

RESUMEN

Urea cycle disorders are enzymopathies resulting from inherited deficiencies in any genes of the cycle. In severe cases, currently available therapies are marginally effective, with liver transplantation being the only definitive treatment. Donor liver availability can limit even this therapy. Identification of novel therapeutics for genetic-based liver diseases requires models that provide measurable hepatic functions and phenotypes. Advances in stem cell and genome editing technologies could provide models for the investigation of cell-based genetic diseases, as well as the platforms for drug discovery. This report demonstrates a practical, and widely applicable, approach that includes the successful reprogramming of somatic cells from a patient with a urea cycle defect, their genetic correction and differentiation into hepatic organoids, and the subsequent demonstration of genetic and phenotypic change in the edited cells consistent with the correction of the defect. While individually rare, there is a large number of other genetic-based liver diseases. The approach described here could be applied to a broad range and a large number of patients with these hepatic diseases where it could serve as an in vitro model, as well as identify successful strategies for corrective cell-based therapy.


Asunto(s)
Edición Génica , Hepatocitos/metabolismo , Redes y Vías Metabólicas/genética , Organoides/citología , Células Madre/metabolismo , Urea/metabolismo , Biomarcadores , Sistemas CRISPR-Cas , Diferenciación Celular , Células Cultivadas , Susceptibilidad a Enfermedades , Perfilación de la Expresión Génica , Estudios de Asociación Genética , Variación Genética , Hepatocitos/citología , Humanos , Inmunohistoquímica , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Modelos Biológicos , Células Madre/citología
6.
Mol Ther ; 29(5): 1903-1917, 2021 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-33484963

RESUMEN

Ornithine transcarbamylase deficiency (OTCD) is a monogenic disease of ammonia metabolism in hepatocytes. Severe disease is frequently treated by orthotopic liver transplantation. An attractive approach is the correction of a patient's own cells to regenerate the liver with gene-repaired hepatocytes. This study investigates the efficacy and safety of ex vivo correction of primary human hepatocytes. Hepatocytes isolated from an OTCD patient were genetically corrected ex vivo, through the deletion of a mutant intronic splicing site achieving editing efficiencies >60% and the restoration of the urea cycle in vitro. The corrected hepatocytes were transplanted into the liver of FRGN mice and repopulated to high levels (>80%). Animals transplanted and liver repopulated with genetically edited patient hepatocytes displayed normal ammonia, enhanced clearance of an ammonia challenge and OTC enzyme activity, as well as lower urinary orotic acid when compared to mice repopulated with unedited patient hepatocytes. Gene expression was shown to be similar between mice transplanted with unedited or edited patient hepatocytes. Finally, a genome-wide screening by performing CIRCLE-seq and deep sequencing of >70 potential off-targets revealed no unspecific editing. Overall analysis of disease phenotype, gene expression, and possible off-target editing indicated that the gene editing of a severe genetic liver disease was safe and effective.


Asunto(s)
Edición Génica/métodos , Hepatocitos/trasplante , Mutación , Enfermedad por Deficiencia de Ornitina Carbamoiltransferasa/terapia , Ornitina Carbamoiltransferasa/genética , Adulto , Anciano , Amoníaco/metabolismo , Animales , Células Cultivadas , Niño , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Hepatocitos/química , Hepatocitos/citología , Humanos , Intrones , Masculino , Ratones , Enfermedad por Deficiencia de Ornitina Carbamoiltransferasa/genética , Ácido Orótico/orina , Empalme del ARN
7.
Mech Ageing Dev ; 194: 111426, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33385396

RESUMEN

Advanced age is the major risk factor for idiopathic Parkinson's disease (PD), but to date the biological relationship between PD and ageing remains elusive. Here we describe the rationale and the design of the H2020 funded project "PROPAG-AGEING", whose aim is to characterize the contribution of the ageing process to PD development. We summarize current evidences that support the existence of a continuum between ageing and PD and justify the use of a Geroscience approach to study PD. We focus in particular on the role of inflammaging, the chronic, low-grade inflammation characteristic of elderly physiology, which can propagate and transmit both locally and systemically. We then describe PROPAG-AGEING design, which is based on the multi-omic characterization of peripheral samples from clinically characterized drug-naïve and advanced PD, PD discordant twins, healthy controls and "super-controls", i.e. centenarians, who never showed clinical signs of motor disability, and their offspring. Omic results are then validated in a large number of samples, including in vitro models of dopaminergic neurons and healthy siblings of PD patients, who are at higher risk of developing PD, with the final aim of identifying the molecular perturbations that can deviate the trajectories of healthy ageing towards PD development.


Asunto(s)
Envejecimiento/metabolismo , Investigación Biomédica , Encéfalo/metabolismo , Geriatría , Mediadores de Inflamación/metabolismo , Neuronas/metabolismo , Enfermedad de Parkinson/metabolismo , Factores de Edad , Anciano , Anciano de 80 o más Años , Envejecimiento/genética , Envejecimiento/patología , Encéfalo/patología , Encéfalo/fisiopatología , Estudios de Casos y Controles , Europa (Continente) , Femenino , Genómica , Humanos , Masculino , Metabolómica , Actividad Motora , Degeneración Nerviosa , Neuronas/patología , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Enfermedad de Parkinson/fisiopatología , Proyectos de Investigación , Transducción de Señal , Estudios en Gemelos como Asunto
8.
Cells ; 9(7)2020 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-32679793

RESUMEN

Perinatal stem cells and epithelial cells isolated from full term amnion membrane, in particular, have attracted interest over the last decade, as a promising source of multipotent cells for cellular therapies. Human amnion epithelial cells (hAEC) have been used to treat monogenetic liver disease such as maple syrup urine disease or fibrosis of the liver in preclinical studies. In most studies xeno-transplants of hAEC were conducted without providing immunosuppression to recipients, reflecting the tolerogenic properties of hAEC. For many cell types, successful cryopreservation is critical for providing a readily available, off-the-shelf product. In this study, hAEC were isolated from full-term human placenta from 14 different donors, cryopreserved using a protocol and reagents commonly adopted for epithelial cell preservation. The cells were analyzed in terms of survival, recovery, and homogeneity, profiled for surface markers characteristic of epithelial, mesenchymal, endothelial, or hematopoietic cells. There were no significant differences observed in the percentage of cells with epithelial cell markers before and after cryopreservation. The relative proportion of stromal and hematopoietic cells was significantly reduced in hAEC preparations after cryopreservation. The expression of stem cell and immunomodulatory molecules were confirmed in the final product. Since multipotent cells are readily available from full-term placenta, this novel cell source might significantly increase the number of patients eligible to receive cellular therapies for liver and other diseases.


Asunto(s)
Amnios/citología , Criopreservación , Células Epiteliales/citología , Supervivencia Celular , Células Epiteliales/metabolismo , Matriz Extracelular/metabolismo , Humanos , Inmunomodulación , Control de Calidad , Células Madre/citología , Células Madre/metabolismo
9.
Sci Rep ; 10(1): 7052, 2020 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-32341402

RESUMEN

Alpha 1-antitrypsin (AAT) deficiency arises from an inherited mutation in the SERPINA1 gene. The disease causes damage in the liver where the majority of the AAT protein is produced. Lack of functioning circulating AAT protein also causes uninhibited elastolytic activity in the lungs leading to AAT deficiency-related emphysema. The only therapy apart from liver transplantation is augmentation with human AAT protein pooled from sera, which is only reserved for patients with advanced lung disease caused by severe AAT deficiency. We tested modified mRNA encoding human AAT in primary human hepatocytes in culture, including hepatocytes from AAT deficient patients. Both expression and functional activity were investigated. Secreted AAT protein increased from 1,14 to 3,43 µg/ml in media from primary human hepatocytes following mRNA treatment as investigated by ELISA and western blot. The translated protein showed activity and protease inhibitory function as measured by elastase activity assay. Also, mRNA formulation in lipid nanoparticles was assessed for systemic delivery in both wild type mice and the NSG-PiZ transgenic mouse model of AAT deficiency. Systemic intravenous delivery of modified mRNA led to hepatic uptake and translation into a functioning protein in mice. These data support the use of systemic mRNA therapy as a potential treatment for AAT deficiency.


Asunto(s)
ARN Mensajero/metabolismo , Deficiencia de alfa 1-Antitripsina/genética , Deficiencia de alfa 1-Antitripsina/terapia , Animales , Western Blotting , Células Cultivadas , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Humanos , Nanopartículas/química , alfa 1-Antitripsina/genética , alfa 1-Antitripsina/fisiología
10.
Nutrients ; 12(2)2020 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-31973116

RESUMEN

A growing amount of evidence suggests that the downregulation of protein synthesis is an adaptive response during physiological aging, which positively contributes to longevity and can be modulated by nutritional interventions like caloric restriction (CR). The expression of ribosomal RNA (rRNA) is one of the main determinants of translational rate, and epigenetic modifications finely contribute to its regulation. Previous reports suggest that hypermethylation of ribosomal DNA (rDNA) locus occurs with aging, although with some species- and tissue- specificity. In the present study, we experimentally measured DNA methylation of three regions (the promoter, the 5' of the 18S and the 5' of 28S sequences) in the rDNA locus in liver tissues from rats at two, four, 10, and 18 months. We confirm previous findings, showing age-related hypermethylation, and describe, for the first time, that this gain in methylation also occurs in human hepatocytes. Furthermore, we show that age-related hypermethylation is enhanced in livers of rat upon CR at two and 10 months, and that at two months a trend towards the reduction of rRNA expression occurs. Collectively, our results suggest that CR modulates age-related regulation of methylation at the rDNA locus, thus providing an epigenetic readout of the pro-longevity effects of CR.


Asunto(s)
Envejecimiento/metabolismo , Restricción Calórica , Metilación de ADN/fisiología , Sitios Genéticos/fisiología , ARN Ribosómico/metabolismo , Animales , ADN Ribosómico/metabolismo , Epigénesis Genética , Humanos , Hígado/metabolismo , Longevidad/fisiología , Masculino , Regiones Promotoras Genéticas/fisiología , Ratas
11.
Hepatology ; 72(2): 656-670, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-31785104

RESUMEN

BACKGROUND AND AIMS: Genetically modified mice have been used extensively to study human disease. However, the data gained are not always translatable to humans because of major species differences. Liver-humanized mice (LHM) are considered a promising model to study human hepatic and systemic metabolism. Therefore, we aimed to further explore their lipoprotein metabolism and to characterize key hepatic species-related, physiological differences. APPROACH AND RESULTS: Fah-/- , Rag2-/- , and Il2rg-/- knockout mice on the nonobese diabetic (FRGN) background were repopulated with primary human hepatocytes from different donors. Cholesterol lipoprotein profiles of LHM showed a human-like pattern, characterized by a high ratio of low-density lipoprotein to high-density lipoprotein, and dependency on the human donor. This pattern was determined by a higher level of apolipoprotein B100 in circulation, as a result of lower hepatic mRNA editing and low-density lipoprotein receptor expression, and higher levels of circulating proprotein convertase subtilisin/kexin type 9. As a consequence, LHM lipoproteins bind to human aortic proteoglycans in a pattern similar to human lipoproteins. Unexpectedly, cholesteryl ester transfer protein was not required to determine the human-like cholesterol lipoprotein profile. Moreover, LHM treated with GW3965 mimicked the negative lipid outcomes of the first human trial of liver X receptor stimulation (i.e., a dramatic increase of cholesterol and triglycerides in circulation). Innovatively, LHM allowed the characterization of these effects at a molecular level. CONCLUSIONS: LHM represent an interesting translatable model of human hepatic and lipoprotein metabolism. Because several metabolic parameters displayed donor dependency, LHM may also be used in studies for personalized medicine.


Asunto(s)
Benzoatos/farmacocinética , Bencilaminas/farmacocinética , Colesterol/metabolismo , Hepatocitos/metabolismo , Lipoproteínas/metabolismo , Receptores X del Hígado/agonistas , Hígado/metabolismo , Animales , Hepatocitos/trasplante , Humanos , Hígado/cirugía , Masculino , Ratones , Ratones Noqueados
12.
Stem Cells Dev ; 28(14): 907-919, 2019 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-31122128

RESUMEN

Differentiation of stem cells to hepatocyte-like cells (HLCs) holds great promise for basic research, drug and toxicological investigations, and clinical applications. There are currently no protocols for the production of HLCs from stem cells, such as embryonic stem cells or induced pluripotent stem cells, that produce fully mature hepatocytes with a wide range of mature hepatic functions. This report describes a standard method to assess the maturation of stem cell-derived HLCs with a moderately high-throughput format, by analysing liver gene expression by quantitative RT-qPCR. This method also provides a robust data set of the expression of 62 genes expressed in normal liver, generated from 17 fetal and 25 mature human livers, so that investigators can quickly and easily compare the expression of these genes in their stem cell-derived HLCs with the values obtained in authentic fetal and mature human liver. The simple methods described in this study will provide a quick and accurate assessment of the efficacy of a differentiation protocol and will help guide the optimization of differentiation conditions.


Asunto(s)
Diferenciación Celular , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Hepatocitos/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Hígado/metabolismo , Hepatocitos/citología , Humanos , Células Madre Pluripotentes Inducidas/citología , Hígado/citología
13.
PLoS One ; 14(4): e0215490, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31022207

RESUMEN

Induced pluripotent stem cell (iPSC)-technology is an important platform in medicine and disease modeling. Physiological degeneration and disease onset are common occurrences in the aging population. iPSCs could offer regenerative medical options for age-related degeneration and disease in the elderly. However, reprogramming somatic cells from the elderly is inefficient when successful at all. Perhaps due to their low rates of replication in culture, traditional transduction and reprogramming approaches with centenarian fibroblasts met with little success. A simple and reproducible reprogramming process is reported here which enhances interactions of the cells with the viral vectors that leads to improved iPSC generation. The improved methods efficiently generates fully reprogrammed iPSC lines from 105-107 years old subjects in feeder-free conditions using an episomal, Sendai-Virus (SeV) reprogramming vector expressing four reprogramming factors. In conclusion, dermal fibroblasts from human subjects older than 100 years can be efficiently and reproducibly reprogrammed to fully pluripotent cells with minor modifications to the standard reprogramming procedures. Efficient generation of iPSCs from the elderly may provide a source of cells for the regeneration of tissues and organs with autologous cells as well as cellular models for the study of aging, longevity and age-related diseases.


Asunto(s)
Técnicas de Reprogramación Celular/métodos , Reprogramación Celular , Fibroblastos/fisiología , Células Madre Pluripotentes Inducidas/fisiología , Adulto , Factores de Edad , Anciano de 80 o más Años , Células Cultivadas , Vectores Genéticos/genética , Humanos , Hidrodinámica , Recién Nacido , Cultivo Primario de Células , Reproducibilidad de los Resultados , Virus Sendai/genética , Piel/citología , Envejecimiento de la Piel/fisiología , Transfección/métodos , Trasplante Autólogo/métodos
14.
J Inherit Metab Dis ; 42(6): 1054-1063, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-30843237

RESUMEN

A liver-humanized mouse model for CPS1-deficiency was generated by the high-level repopulation of the mouse liver with CPS1-deficient human hepatocytes. When compared with mice that are highly repopulated with CPS1-proficient human hepatocytes, mice that are repopulated with CPS1-deficient human hepatocytes exhibited characteristic symptoms of human CPS1 deficiency including an 80% reduction in CPS1 metabolic activity, delayed clearance of an ammonium chloride infusion, elevated glutamine and glutamate levels, and impaired metabolism of [15 N]ammonium chloride into urea, with no other obvious phenotypic differences. Because most metabolic liver diseases result from mutations that alter critical pathways in hepatocytes, a model that incorporates actual disease-affected, mutant human hepatocytes is useful for the investigation of the molecular, biochemical, and phenotypic differences induced by that mutation. The model is also expected to be useful for investigations of modified RNA, gene, and cellular and small molecule therapies for CPS1-deficiency. Liver-humanized models for this and other monogenic liver diseases afford the ability to assess the therapy on actual disease-affected human hepatocytes, in vivo, for long periods of time and will provide data that are highly relevant for investigations of the safety and efficacy of gene-editing technologies directed to human hepatocytes and the translation of gene-editing technology to the clinic.


Asunto(s)
Carbamoil-Fosfato Sintasa (Amoniaco)/genética , Enfermedad por Deficiencia de Carbamoil-Fosfato Sintasa I/genética , Enfermedad por Deficiencia de Carbamoil-Fosfato Sintasa I/patología , Hepatocitos/trasplante , Hidrolasas/genética , Hígado/metabolismo , Animales , Carbamoil-Fosfato Sintasa (Amoniaco)/metabolismo , Células Cultivadas , Niño , Modelos Animales de Enfermedad , Femenino , Hepatocitos/metabolismo , Humanos , Hidrolasas/metabolismo , Lactante , Recién Nacido , Hígado/patología , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Especificidad de Órganos/genética
15.
J Gerontol A Biol Sci Med Sci ; 74(1): 1-8, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29554203

RESUMEN

The feasibility of liver transplantation from old healthy donors suggests that this organ is able to preserve its functionality during aging. To explore the biological basis of this phenomenon, we characterized the epigenetic profile of liver biopsies collected from 45 healthy liver donors ranging from 13 to 90 years old using the Infinium HumanMethylation450 BeadChip. The analysis indicates that a large remodeling in DNA methylation patterns occurs, with 8,823 age-associated differentially methylated CpG probes. Notably, these age-associated changes tended to level off after the age of 60, as confirmed by Horvath's clock. Using stringent selection criteria, we further identified a DNA methylation signature of aging liver including 75 genomic regions. We demonstrated that this signature is specific for liver compared to other tissues and that it is able to detect biological age-acceleration effects associated with obesity. Finally, we combined DNA methylation measurements with available expression data. Although the intersection between the two omic characterizations was low, both approaches suggested a previously unappreciated role of epithelial-mesenchymal transition and Wnt-signaling pathways in the aging of human liver.


Asunto(s)
Envejecimiento/metabolismo , Epigénesis Genética , Trasplante de Hígado , Hígado/metabolismo , ARN/genética , Transcriptoma/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Biopsia , Metilación de ADN , Femenino , Humanos , Hígado/citología , Masculino , Persona de Mediana Edad , Donantes de Tejidos , Adulto Joven
16.
Int J Cancer ; 144(10): 2613-2624, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30488605

RESUMEN

Hepatocellular carcinoma (HCC) is a highly malignant tumor that responds very poorly to existing therapies, most probably due to its extraordinary inter- and intra-tumor molecular heterogeneity. The modest therapeutic response to molecular targeted agents underlines the need for new therapeutic approaches for HCC. In our study, we took advantage of well-characterized human HCC cell lines, differing in transcriptomic subtypes, DNA mutation and amplification alterations, reflecting the heterogeneity of primary HCCs, to provide a preclinical evaluation of the specific heat shock protein 90 (HSP90) inhibitor AUY922 (luminespib). Indeed, HSP90 is highly expressed in different tumor types, but its role in hepatocarcinogenesis remains unclear. Here, we analyzed HSP90 expression in primary human HCC tissues and evaluated the antitumor effects of AUY922 in vitro as well as in vivo. HSP90 expression was significantly higher in HCC tissues than in cirrhotic peritumoral liver tissues. AUY922 treatment reduced the cell proliferation and viability of HCC cells in a dose-dependent manner, but did not do so for normal human primary hepatocytes. AUY922 treatment led to the upregulation of HSP70 and the simultaneous depletion of HSP90 client proteins. In addition, in a cell type-dependent manner, treatment induced either both caspase-dependent ß-catenin cleavage and the upregulation of p53, or Mcl-1 expression, or NUPR1 expression, which contributed to the increased efficacy of, or resistance to, treatment. Finally, in vivo AUY922 inhibited tumor growth in a xenograft model. In conclusion, HSP90 is a promising therapeutic target in HCC, and AUY922 could be a drug candidate for its treatment.


Asunto(s)
Antineoplásicos/uso terapéutico , Carcinoma Hepatocelular/tratamiento farmacológico , Proteínas HSP90 de Choque Térmico/metabolismo , Isoxazoles/uso terapéutico , Neoplasias Hepáticas/tratamiento farmacológico , Resorcinoles/uso terapéutico , Bibliotecas de Moléculas Pequeñas/uso terapéutico , Adulto , Anciano , Anciano de 80 o más Años , Animales , Carcinoma Hepatocelular/metabolismo , Caspasas/metabolismo , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células Hep G2 , Humanos , Neoplasias Hepáticas/metabolismo , Masculino , Ratones Desnudos , Persona de Mediana Edad , Mutación/genética , Transcriptoma/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , beta Catenina/metabolismo
17.
Cytotherapy ; 21(1): 113-124, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30409699

RESUMEN

Placenta is a non-controversial and promising source of cells for the treatment of several liver diseases. We previously reported that transplanted human amnion epithelial cells (hAECs) differentiate into hepatocyte-like cells, resulting in correction of mouse models of metabolic liver disease or acute hepatic failure. As part of preclinical safety studies, we investigated the distribution of hAECs using two routes of administration to efficiently deliver hAECs to the liver. Optical imaging is commonly used because it can provide fast, high-throughput, whole-body imaging, thus DiR-labeled hAECs were injected into immunodeficient mice, via the spleen or the tail vein. The cell distribution was monitored using an in vivo imaging system over the next 24 h. After splenic injection, the DiR signal was detected in liver and spleen at 1, 3 and 24 h post-transplant. The distribution was confirmed by analysis of human DNA content at 24 h post-transplant and human-specific cytokeratin 8/18 staining. Tail vein infusion resulted in cell engraftment mainly in the lungs, with minimal detection in the liver. Delivery of cells to the portal vein, via the spleen, resulted in efficient delivery of hAECs to the liver, with minimal, off-target distribution to lungs or other organs.


Asunto(s)
Amnios/citología , Trasplante de Células/métodos , Células Epiteliales/trasplante , Hepatopatías/terapia , Placenta/citología , Animales , Células Cultivadas , Femenino , Humanos , Hígado/metabolismo , Pulmón , Ratones , Modelos Animales , Vena Porta , Embarazo , Bazo , Vena Esplénica
18.
J Immunol ; 202(3): 724-735, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30587530

RESUMEN

This study investigates the mechanism(s) underlying the immunoregulatory activities of placenta-derived human amnion epithelial cells (hAEC). The working hypothesis is that NAD+ and ATP, along with ectoenzymes involved in their metabolism, play a significant role in hAEC-mediated immune regulation. Proof of principle of the hypothesis was obtained by analyzing the interactions between hAEC and the main human leukocyte populations. The results obtained indicate that hAEC constitutively express a unique combination of functional ectoenzymes, driving the production of adenosine (ADO) via canonical (CD39, CD73) and alternative (CD38, CD203a/PC-1, CD73) pathways. Further, the picture is completed by the observation that hAEC express A1, A2a, and A2b ADO receptors as well as ADO deaminase, the enzyme involved in ADO catabolism. The contribution of the purinergic mediator to immunomodulation was confirmed by exposing in vitro different immune effector cells to the action of primary hAECs. B cells showed an enhanced proliferation and diminished spontaneous apoptosis when in contact with hAEC. T cell proliferation was partially inhibited by hAEC through ADO production, as confirmed by using specific ectoenzyme inhibitors. Further, hAEC induced an expansion of both T and B regulatory cells. Last, hAEC inhibited NK cell proliferation. However, the involvement of ADO-producing ectoenzymes is less apparent in this context. In conclusion, hAEC exert different in vitro immunoregulatory effects, per se, as a result of interactions with different populations of immune effector cells. These results support the view that hAEC are instrumental for regenerative medicine as well as in therapeutic applications for immune-related diseases.


Asunto(s)
Adenosina Desaminasa/metabolismo , Adenosina/biosíntesis , Amnios/citología , Proliferación Celular , Células Epiteliales/enzimología , Adenosina Desaminasa/genética , Linfocitos B/citología , Células Cultivadas , Células Epiteliales/inmunología , Humanos , Células Asesinas Naturales/citología , Activación de Linfocitos , Redes y Vías Metabólicas , Receptor de Adenosina A1/metabolismo , Receptor de Adenosina A2A/metabolismo , Linfocitos T/citología
19.
Int J Oncol ; 51(2): 533-544, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28656311

RESUMEN

The beneficial health properties of the Mediter-ranean diet are well recognized. The principle source of fat in Mediterranean diet is extra-virgin olive oil (EVOO). Oleocanthal (OC) is a naturally occurring minor phenolic compound isolated from EVOO, which has shown a potent anti-inflammatory activity, by means of its ability to inhibit the cyclooxygenase (COX) enzymes COX-1 and COX-2. A large body of evidence indicates that phenols exhibit anticancer activities. The aim of the present study was to evaluate the potential anticancer effects of OC in hepatocellular carcinoma (HCC) and colorectal carcinoma (CRC) models. A panel of human HCC (HepG2, Huh7, Hep3B and PLC/PRF/5) and CRC (HT29, SW480) cell lines was used. Cells were treated with OC, and cell viability and apoptosis were evaluated. Compared with classical commercially available COX inhibitors (ibuprofen, indomethacin, nimesulide), OC was more effective in inducing cell growth inhibition in HCC and CRC cells. Moreover, OC inhibited colony formation and induced apoptosis, as confirmed by PARP cleavage, activation of caspases 3/7 and chromatin condensation. OC treatment in a dose dependent-manner induced expression of γH2AX, a marker of DNA damage, increased intracellular ROS production and caused mitochondrial depolarization. Moreover, the effects of OC were suppressed by the ROS scavenger N-acetyl-L-cysteine. Finally, OC was not toxic in primary normal human hepatocytes. In conclusion, OC treatment was found to exert a potent anticancer activity against HCC and CRC cells. Taken together, our findings provide preclinical support of the chemotherapeutic potential of EVOO against cancer.


Asunto(s)
Aldehídos/administración & dosificación , Carcinoma Hepatocelular/dietoterapia , Neoplasias Colorrectales/dietoterapia , Neoplasias Hepáticas/dietoterapia , Fenoles/administración & dosificación , Aldehídos/química , Apoptosis/efectos de los fármacos , Carcinoma Hepatocelular/enzimología , Carcinoma Hepatocelular/patología , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/enzimología , Neoplasias Colorrectales/patología , Inhibidores de la Ciclooxigenasa/administración & dosificación , Monoterpenos Ciclopentánicos , Daño del ADN/efectos de los fármacos , Células Hep G2 , Humanos , Neoplasias Hepáticas/enzimología , Neoplasias Hepáticas/patología , Aceite de Oliva/administración & dosificación , Aceite de Oliva/química , Fenoles/química , Especies Reactivas de Oxígeno/metabolismo
20.
PLoS One ; 12(5): e0177279, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28486562

RESUMEN

The main goal of the therapy with purified human plasma alpha1-antitrypsin (A1AT) is to increase A1AT levels and to prevent lungs from elastolytic activity in patients with PiZZ (Glu342Lys) A1AT deficiency-related emphysema. Potential hepatic gains of this therapy are unknown. Herein, we investigated the effect of A1AT therapy on SERPINA1 (gene encoding A1AT) expression. The expression of SERPINA1 was determined in A1AT or A1AT plus Oncostatin M (OSM) treated primary human hepatocytes isolated from liver tissues from A1AT deficient patients and control liver tissues. In addition, SERPINA1 mRNA was assessed in lung tissues from PiZZ emphysema patients with and without A1AT therapy, and in adherent human peripheral blood mononuclear cells (PBMC) isolated from healthy PiMM donors. In a dose-dependent manner purified A1AT lowered SERPINA1 expression in hepatocytes. This latter effect was more prominent in hepatocytes stimulated with OSM. Although it did not reach statistical significance (P = 0.0539)-analysis of lung tissues showed lower SERPINA1 expression in PiZZ emphysema patients receiving augmentation therapy relative to those without therapy. Finally, exogenously added purified A1AT (1mg/ml) reduced SERPINA1 expression in naïve as well as in lipopolysaccharide (LPS)-stimulated human adherent PBMCs. Exogenous A1AT protein reduces its own endogenous expression. Hence, augmentation with native M-A1AT protein and a parallel reduction in expression of dysfunctional mutant Z-A1AT may be beneficial for PiZZ liver, and this motivates further studies.


Asunto(s)
Regulación hacia Abajo/efectos de los fármacos , alfa 1-Antitripsina/farmacología , Adolescente , Adulto , Células Cultivadas , Preescolar , Femenino , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , ARN Mensajero/genética , Adulto Joven , alfa 1-Antitripsina/genética , alfa 1-Antitripsina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...