Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 118
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
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
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.
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
4.
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
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
7.
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
8.
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
9.
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
10.
J Hepatol ; 66(5): 987-1000, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28027971

RESUMEN

BACKGROUND & AIMS: Hepatocyte transplantation partially corrects genetic disorders and has been associated anecdotally with reversal of acute liver failure. Monitoring for graft function and rejection has been difficult, and has contributed to limited graft survival. Here we aimed to use preparative liver-directed radiation therapy, and continuous monitoring for possible rejection in an attempt to overcome these limitations. METHODS: Preparative hepatic irradiation was examined in non-human primates as a strategy to improve engraftment of donor hepatocytes, and was then applied in human subjects. T cell immune monitoring was also examined in human subjects to assess adequacy of immunosuppression. RESULTS: Porcine hepatocyte transplants engrafted and expanded to comprise up to 15% of irradiated segments in immunosuppressed monkeys preconditioned with 10Gy liver-directed irradiation. Two patients with urea cycle deficiencies had early graft loss following hepatocyte transplantation; retrospective immune monitoring suggested the need for additional immunosuppression. Preparative radiation, anti-lymphocyte induction, and frequent immune monitoring were instituted for hepatocyte transplantation in a 27year old female with classical phenylketonuria. Post-transplant liver biopsies demonstrated multiple small clusters of transplanted cells, multiple mitoses, and Ki67+ hepatocytes. Mean peripheral blood phenylalanine (PHE) level fell from pre-transplant levels of 1343±48µM (normal 30-119µM) to 854±25µM (treatment goal ≤360µM) after transplant (36% decrease; p<0.0001), despite transplantation of only half the target number of donor hepatocytes. PHE levels remained below 900µM during supervised follow-up, but graft loss occurred after follow-up became inconsistent. CONCLUSIONS: Radiation preconditioning and serial rejection risk assessment may produce better engraftment and long-term survival of transplanted hepatocytes. Hepatocyte xenografts engraft for a period of months in non-human primates and may provide effective therapy for patients with acute liver failure. LAY SUMMARY: Hepatocyte transplantation can potentially be used to treat genetic liver disorders but its application in clinical practice has been impeded by inefficient hepatocyte engraftment and the inability to monitor rejection of transplanted liver cells. In this study, we first show in non-human primates that pretreatment of the host liver with radiation improves the engraftment of transplanted liver cells. We then used this knowledge in a series of clinical hepatocyte transplants in patients with genetic liver disorders to show that radiation pretreatment and rejection risk monitoring are safe and, if optimized, could improve engraftment and long-term survival of transplanted hepatocytes in patients.


Asunto(s)
Rechazo de Injerto , Hepatocitos/trasplante , Hígado/efectos de la radiación , Acondicionamiento Pretrasplante , Adulto , Animales , Femenino , Humanos , Hepatopatías/terapia , Macaca fascicularis , Masculino , Porcinos , Trasplante Heterólogo
11.
Hepatology ; 62(1): 147-57, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25690322

RESUMEN

UNLABELLED: In the classical form of α1-antitrypsin deficiency (ATD), aberrant intracellular accumulation of misfolded mutant α1-antitrypsin Z (ATZ) in hepatocytes causes hepatic damage by a gain-of-function, "proteotoxic" mechanism. Whereas some ATD patients develop severe liver disease (SLD) that necessitates liver transplantation, others with the same genetic defect completely escape this clinical phenotype. We investigated whether induced pluripotent stem cells (iPSCs) from ATD individuals with or without SLD could model these personalized variations in hepatic disease phenotypes. Patient-specific iPSCs were generated from ATD patients and a control and differentiated into hepatocyte-like cells (iHeps) having many characteristics of hepatocytes. Pulse-chase and endoglycosidase H analysis demonstrate that the iHeps recapitulate the abnormal accumulation and processing of the ATZ molecule, compared to the wild-type AT molecule. Measurements of the fate of intracellular ATZ show a marked delay in the rate of ATZ degradation in iHeps from SLD patients, compared to those from no liver disease patients. Transmission electron microscopy showed dilated rough endoplasmic reticulum in iHeps from all individuals with ATD, not in controls, but globular inclusions that are partially covered with ribosomes were observed only in iHeps from individuals with SLD. CONCLUSION: iHeps model the individual disease phenotypes of ATD patients with more rapid degradation of misfolded ATZ and lack of globular inclusions in cells from patients who have escaped liver disease. The results support the concept that "proteostasis" mechanisms, such as intracellular degradation pathways, play a role in observed variations in clinical phenotype and show that iPSCs can potentially be used to facilitate predictions of disease susceptibility for more precise and timely application of therapeutic strategies.


Asunto(s)
Células Madre Pluripotentes Inducidas/metabolismo , Hepatopatías/etiología , Deficiencia de alfa 1-Antitripsina/complicaciones , Células Cultivadas , Retículo Endoplásmico Rugoso/metabolismo , Humanos , Hepatopatías/metabolismo , alfa 1-Antitripsina/metabolismo
12.
Drug Metab Dispos ; 44(7): 1027-37, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26921389

RESUMEN

Variability in drug-metabolizing enzyme developmental trajectories contributes to interindividual differences in susceptibility to chemical toxicity and adverse drug reactions, particularly in the first years of life. Factors linked to these interindividual differences are largely unknown, but molecular mechanisms regulating ontogeny are likely involved. To evaluate chromatin structure dynamics as a likely contributing mechanism, age-dependent changes in modified and variant histone occupancy were evaluated within known CYP3A4 and 3A7 regulatory domains. Chromatin immunoprecipitation using fetal or postnatal human hepatocyte chromatin pools followed by quantitative polymerase chain reaction DNA amplification was used to determine relative chromatin occupancy by modified and variant histones. Chromatin structure representing a poised transcriptional state (bivalent chromatin), indicated by the occupancy by modified histones associated with both active and repressed transcription, was observed for CYP3A4 and most 3A7 regulatory regions in both postnatal and fetal livers. However, the CYP3A4 regulatory regions had significantly greater occupancy by modified histones associated with repressed transcription in the fetal liver. Conversely, some modified histones associated with active transcription exhibited greater occupancy in the postnatal liver. CYP3A7 regulatory regions also had significantly greater occupancy by modified histones associated with repressed transcription in the fetus. The observed occupancy by modified histones is consistent with chromatin structural dynamics contributing to CYP3A4 ontogeny, although the data are less conclusive regarding CYP3A7. Interpretation of the latter data may be confounded by cell-type heterogeneity in the fetal liver.


Asunto(s)
Ensamble y Desensamble de Cromatina , Cromatina/metabolismo , Citocromo P-450 CYP3A/metabolismo , Histonas/metabolismo , Hígado/enzimología , Adulto , Factores de Edad , Anciano , Anciano de 80 o más Años , Sitios de Unión , Niño , Preescolar , Cromatina/química , Cromatina/genética , Citocromo P-450 CYP3A/genética , Regulación del Desarrollo de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Edad Gestacional , Hepatocitos/enzimología , Histonas/química , Histonas/genética , Humanos , Lactante , Hígado/embriología , Persona de Mediana Edad , Conformación de Ácido Nucleico , Regiones Promotoras Genéticas , Conformación Proteica , Relación Estructura-Actividad , Transcripción Genética
13.
J Biol Chem ; 289(30): 20583-93, 2014 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-24895126

RESUMEN

The mitochondrial branched-chain α-ketoacid dehydrogenase complex (BCKDC) is negatively regulated by reversible phosphorylation.BCKDC kinase (BDK) inhibitors that augment BCKDC flux have been shown to reduce branched-chain amino acid (BCAA) concentrations in vivo. In the present study, we employed high-throughput screens to identify compound 3,6- dichlorobenzo[b]thiophene-2-carboxylic acid (BT2) as a novel BDK inhibitor (IC(50) = 3.19 µM). BT2 binds to the same site in BDK as other known allosteric BDK inhibitors, including (S)-α-cholorophenylproprionate ((S)-CPP). BT2 binding to BDK triggers helix movements in the N-terminal domain, resulting in the dissociation of BDK from the BCKDC accompanied by accelerated degradation of the released kinase in vivo. BT2 shows excellent pharmacokinetics (terminal T(1/2) = 730 min) and metabolic stability (no degradation in 240 min), which are significantly better than those of (S)-CPP. BT2, its analog 3-chloro-6-fluorobenzo[ b]thiophene-2-carboxylic acid (BT2F), and a prodrug of BT2 (i.e. N-(4-acetamido-1,2,5-oxadiazol-3-yl)-3,6-dichlorobenzo[ b]thiophene-2-carboxamide (BT3)) significantly increase residual BCKDC activity in cultured cells and primary hepatocytes from patients and a mouse model of maple syrup urine disease. Administration of BT2 at 20 mg/kg/day to wild-type mice for 1 week leads to nearly complete dephosphorylation and maximal activation of BCKDC in heart, muscle, kidneys, and liver with reduction in plasma BCAA concentrations. The availability of benzothiophene carboxylate derivatives as stable BDK inhibitors may prove useful for the treatment of metabolic disease caused by elevated BCAA concentrations.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Hepatocitos/enzimología , Complejo Cetoglutarato Deshidrogenasa/antagonistas & inhibidores , Complejo Cetoglutarato Deshidrogenasa/metabolismo , Proteolisis/efectos de los fármacos , Tiofenos/farmacología , Regulación Alostérica/efectos de los fármacos , Animales , Línea Celular , Relación Dosis-Respuesta a Droga , Estabilidad de Enzimas/efectos de los fármacos , Estabilidad de Enzimas/genética , Hepatocitos/patología , Humanos , Complejo Cetoglutarato Deshidrogenasa/genética , Ratones , Ratones Noqueados , Tiofenos/farmacocinética
14.
Eur Surg Res ; 54(3-4): 162-77, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25633583

RESUMEN

Since the first human hepatocyte transplants (HTx) in 1992, clinical studies have clearly established proof of principle for this therapy as a treatment for patients with acquired or inherited liver disease. Although major accomplishments have been made, there are still some specific limitations to this technology, which, if overcome, could greatly enhance the efficacy and implementation of this therapy. Here, we describe what in our view are the most significant obstacles to the clinical application of HTx and review the solutions currently proposed. The obstacles of significance include the limited number and quality of liver tissues as a cell source, the lack of clinical grade reagents, quality control evaluation of hepatocytes prior to transplantation, hypothermic storage of cells prior to transplantation, preconditioning treatments to enhance engraftment and proliferation of donor cells, tracking or monitoring cells after transplantation, and the optimal immunosuppression protocols for transplant recipients.


Asunto(s)
Hepatocitos/trasplante , Hepatopatías/terapia , Separación Celular , Humanos , Terapia de Inmunosupresión , Hígado/citología , Hígado/fisiología , Hígado/cirugía , Preservación Biológica
15.
Hepatology ; 57(3): 1017-23, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23175463

RESUMEN

UNLABELLED: There is improved survival and partial metabolic correction of a mouse intermediate maple syrup urine disease (iMSUD) model after allogenic hepatocyte transplantation, confirming that a small number of enzyme-proficient liver-engrafted cells can improve phenotype. However, clinical shortages of suitable livers for hepatocyte isolation indicate a need for alternative cell sources. Human amnion epithelial cells (hAECs) share stem cell characteristics without the latter's safety and ethical concerns and differentiate to hepatocyte-like cells. Eight direct hepatic hAEC transplantations were performed in iMSUD mice over the first 35 days beginning at birth; animals were provided a normal protein diet and sacrificed at 35 and 100 days. Treatment at the neonatal stage is clinically relevant for MSUD and may offer a donor cell engraftment advantage. Survival was significantly extended and body weight was normalized in iMSUD mice receiving hAEC transplantations compared with untreated iMSUD mice, which were severely cachectic and died ≤28 days after birth. Branched chain α-keto acid dehydrogenase enzyme activity was significantly increased in transplanted livers. The branched chain amino acids leucine, isoleucine, valine, and alloisoleucine were significantly improved in serum and brain, as were other large neutral amino acids. CONCLUSION: Placental-derived stem cell transplantation lengthened survival and corrected many amino acid imbalances in a mouse model of iMSUD. This highlights the potential for their use as a viable alternative clinical therapy for MSUD and other liver-based metabolic diseases.


Asunto(s)
Amnios/citología , Células Epiteliales/trasplante , Enfermedad de la Orina de Jarabe de Arce/terapia , Placenta/citología , Trasplante de Células Madre/métodos , 3-Metil-2-Oxobutanoato Deshidrogenasa (Lipoamida)/genética , 3-Metil-2-Oxobutanoato Deshidrogenasa (Lipoamida)/metabolismo , Aminoácidos de Cadena Ramificada/genética , Animales , Animales Recién Nacidos , Peso Corporal/fisiología , Encéfalo/crecimiento & desarrollo , Encéfalo/patología , Diferenciación Celular/fisiología , Modelos Animales de Enfermedad , Células Epiteliales/citología , Femenino , Hepatocitos/citología , Humanos , Enfermedad de la Orina de Jarabe de Arce/genética , Enfermedad de la Orina de Jarabe de Arce/patología , Ratones , Ratones Mutantes , Embarazo , Trasplante Heterólogo
16.
Drug Metab Dispos ; 42(4): 695-9, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24430612

RESUMEN

When investigating the potential for xanthine oxidase (XO)-mediated metabolism of a new chemical entity in vitro, selective chemical inhibition experiments are typically used. Most commonly, these inhibition experiments are performed using the inhibitor allopurinol (AP) and commercially prepared human liver cytosol (HLC) as the enzyme source. For reasons detailed herein, it is also a common practice to perfuse livers with solutions containing AP prior to liver harvest. The exposure to AP in HLC preparations could obviously pose a problem for measuring in vitro XO activity. To investigate this potential problem, an HPLC-MS/MS assay was developed to determine whether AP and its primary metabolite, oxypurinol, are retained within the cytosol for livers that were treated with AP during liver harvest. Differences in enzymatic activity for XO and aldehyde oxidase (AO) in human cytosol that can be ascribed to AP exposure were also evaluated. The results confirmed the presence of residual AP (some) and oxypurinol (all) human liver cytosol preparations that had been perfused with an AP-containing solution. In every case where oxypurinol was detected, XO activity was not observed. In contrast, the presence of AP and oxypurinol did not appear to have an impact on AO activity. Pooled HLC that was purchased from a commercial source also contained residual oxypurinol and did not show any XO activity. In the future, it is recommended that each HLC batch is screened for oxypurinol and/or XO activity prior to testing for XO-mediated metabolism of a new chemical entity.


Asunto(s)
Alopurinol/farmacología , Citosol/enzimología , Inhibidores Enzimáticos/farmacología , Hígado/enzimología , Oxipurinol/farmacología , Xantina Oxidasa/metabolismo , Aldehído Oxidasa/metabolismo , Alopurinol/análisis , Alopurinol/metabolismo , Cromatografía Líquida de Alta Presión , Citosol/efectos de los fármacos , Inhibidores Enzimáticos/análisis , Inhibidores Enzimáticos/metabolismo , Femenino , Humanos , Límite de Detección , Hígado/efectos de los fármacos , Masculino , Oxipurinol/análisis , Oxipurinol/metabolismo , Perfusión , Espectrometría de Masas en Tándem , Técnicas de Cultivo de Tejidos/métodos , Xantina Oxidasa/antagonistas & inhibidores
17.
Gastroenterology ; 142(1): 25-8, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22057114

RESUMEN

Murine hepatocytes become polyploid and then undergo ploidy reversal and become aneuploid in a dynamic process called the ploidy conveyor. Although polyploidization occurs in some types of human cells, the degree of aneuploidy in human hepatocytes is not known. We isolated hepatocytes derived from healthy human liver samples and determined chromosome number and identity using traditional karyotyping and fluorescence in situ hybridization. Similar to murine hepatocytes, human hepatocytes are highly aneuploid. Moreover, imaging studies revealed multipolar spindles and chromosome segregation defects in dividing human hepatocytes. Aneuploidy therefore does not necessarily predispose liver cells to transformation but might promote genetic diversity among hepatocytes.


Asunto(s)
Aneuploidia , Cromosomas Humanos , Variación Genética , Hepatocitos/patología , Adolescente , Adulto , Anciano , Animales , Niño , Preescolar , Femenino , Hepatocitos/trasplante , Humanos , Hidrolasas/deficiencia , Hidrolasas/genética , Hibridación Fluorescente in Situ , Lactante , Cariotipificación , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Quimera por Trasplante , Adulto Joven
18.
Genome Res ; 20(8): 1020-36, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20538623

RESUMEN

Liver cytochrome P450s (P450s) play critical roles in drug metabolism, toxicology, and metabolic processes. Despite rapid progress in the understanding of these enzymes, a systematic investigation of the full spectrum of functionality of individual P450s, the interrelationship or networks connecting them, and the genetic control of each gene/enzyme is lacking. To this end, we genotyped, expression-profiled, and measured P450 activities of 466 human liver samples and applied a systems biology approach via the integration of genetics, gene expression, and enzyme activity measurements. We found that most P450s were positively correlated among themselves and were highly correlated with known regulators as well as thousands of other genes enriched for pathways relevant to the metabolism of drugs, fatty acids, amino acids, and steroids. Genome-wide association analyses between genetic polymorphisms and P450 expression or enzyme activities revealed sets of SNPs associated with P450 traits, and suggested the existence of both cis-regulation of P450 expression (especially for CYP2D6) and more complex trans-regulation of P450 activity. Several novel SNPs associated with CYP2D6 expression and enzyme activity were validated in an independent human cohort. By constructing a weighted coexpression network and a Bayesian regulatory network, we defined the human liver transcriptional network structure, uncovered subnetworks representative of the P450 regulatory system, and identified novel candidate regulatory genes, namely, EHHADH, SLC10A1, and AKR1D1. The P450 subnetworks were then validated using gene signatures responsive to ligands of known P450 regulators in mouse and rat. This systematic survey provides a comprehensive view of the functionality, genetic control, and interactions of P450s.


Asunto(s)
Sistema Enzimático del Citocromo P-450/genética , Sistema Enzimático del Citocromo P-450/metabolismo , Regulación Enzimológica de la Expresión Génica , Genómica , Hígado/enzimología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Niño , Preescolar , Femenino , Expresión Génica , Estudio de Asociación del Genoma Completo , Humanos , Lactante , Recién Nacido , Masculino , Ratones , Persona de Mediana Edad , Preparaciones Farmacéuticas/metabolismo , Polimorfismo de Nucleótido Simple , Ratas , Biología de Sistemas , Transcripción Genética , Adulto Joven
19.
Mol Genet Metab ; 109(2): 132-8, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23566440

RESUMEN

Orthotopic liver transplant (OLT) significantly improves patient outcomes in maple syrup urine disease (MSUD; OMIM: 248600), yet organ shortages point to the need for alternative therapies. Hepatocyte transplantation has shown both clinical and preclinical efficacy as an intervention for metabolic liver diseases, yet the availability of suitable livers for hepatocyte isolation is also limited. Conversely, human amnion epithelial cells (hAEC) may have utility as a hepatocyte substitute, and they share many of the characteristics of pluripotent embryonic stem cells while lacking their safety and ethical concerns. We reported that like hepatocytes, transplantation of hAEC significantly improved survival and lifespan, normalized body weight, and significantly improved branched-chain amino acid (BCAA) levels in sera and brain in a transgenic murine model of intermediate maple syrup urine disease (imsud). In the current report, we detail the neural and peripheral metabolic improvements associated with hAEC transplant in imsud mice, including amino acids associated with bioenergetics, the urea cycle, as well as the neurotransmitter systems for serotonin, dopamine, and gamma-aminobutyric acid (GABA). This stem cell therapy results in significant global correction of the metabolic profile that characterizes the disease, both in the periphery and the central nervous system, the target organ for toxicity in iMSUD. The significant correction of the disease phenotype, coupled with the theoretical benefits of hAEC, particularly their lack of immunogenicity and tumorigenicity, suggests that human amnion epithelial cells deserve serious consideration for clinical application to treat metabolic liver diseases.


Asunto(s)
Aminoácidos/sangre , Amnios/citología , Células Epiteliales/trasplante , Enfermedad de la Orina de Jarabe de Arce/terapia , Neurotransmisores/metabolismo , Animales , Encéfalo/metabolismo , Ciclo del Ácido Cítrico , Humanos , Enfermedad de la Orina de Jarabe de Arce/sangre , Ratones , Ratones Transgénicos
20.
Drug Metab Dispos ; 41(10): 1843-51, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23913028

RESUMEN

Erlotinib is approved for the treatment of non-small cell lung and pancreatic cancers, and is metabolized by CYP3A4. Inducers and inhibitors of CYP3A enzymes such as ritonavir and efavirenz, respectively, may be used as part of the highly active antiretroviral therapy drugs to treat patients with human immunodeficiency virus (HIV). When HIV patients with a malignancy need treatment with erlotinib, there is a potential of as-yet-undefined drug-drug interaction. We evaluated these interactions using human hepatocytes benchmarked against the interaction of erlotinib with ketoconazole and rifampin, the archetype cytochrome P450 inhibitor and inducer, respectively. Hepatocytes were treated with vehicle [0.1% dimethylsulfoxide, ritonavir (10 µM)], ketoconazole (10 µM), efavirenz (10 µM), or rifampin (10 µM) for 4 days. On day 5, erlotinib (5 µM) was incubated with the above agents for another 24-48 hours. Concentrations of erlotinib and O-desmethyl erlotinib were quantitated in collected samples (combined lysate and medium) using liquid chromatography and tandem mass spectrometry. The half-life (t(½)) of erlotinib increased from 10.6 ± 2.6 to 153 ± 80 and 23.9 ± 4.8 hours, respectively, upon treatment with ritonavir and ketoconazole. The apparent intrinsic clearance (C(Lint, app)) of erlotinib was lowered 16-fold by ritonavir and 1.9-fold by ketoconazole. Efavirenz and rifampin decreased t1/2 of erlotinib from 10.3 ± 1.1 to 5.0 ± 1.5 and 3.4 ± 0.2 hours, respectively. Efavirenz and rifampin increased the C(Lint, app) of erlotinib by 2.2- and 2-fold, respectively. Our results suggest that to achieve desired drug exposure, the clinically used dose (150 mg daily) of erlotinib may have to be significantly reduced (25 mg every other day) or increased (300 mg daily), respectively, when ritonavir or efavirenz is coadministered.


Asunto(s)
Benzoxazinas/uso terapéutico , Interacciones Farmacológicas/fisiología , Infecciones por VIH/tratamiento farmacológico , Hepatocitos/metabolismo , Neoplasias/tratamiento farmacológico , Quinazolinas/uso terapéutico , Ritonavir/uso terapéutico , Inhibidores de 14 alfa Desmetilasa/uso terapéutico , Adulto , Anciano , Alquinos , Fármacos Anti-VIH/metabolismo , Fármacos Anti-VIH/uso terapéutico , Ciclopropanos , Clorhidrato de Erlotinib , Femenino , Infecciones por VIH/metabolismo , Infecciones por VIH/virología , Inhibidores de la Proteasa del VIH/metabolismo , Inhibidores de la Proteasa del VIH/uso terapéutico , Semivida , Hepatocitos/efectos de los fármacos , Humanos , Cetoconazol/uso terapéutico , Masculino , Persona de Mediana Edad , Neoplasias/metabolismo , Neoplasias/virología , Inhibidores de la Síntesis del Ácido Nucleico/uso terapéutico , Quinazolinas/metabolismo , Rifampin/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA