Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Mol Genet Metab ; 139(4): 107653, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37463544

RESUMO

Classical homocystinuria (HCU) is a rare inborn error of amino acid metabolism characterized by accumulation of homocysteine, an intermediate product of methionine metabolism, leading to significant systemic toxicities, particularly within the vascular, skeletal, and ocular systems. Most patients require lifelong dietary therapy with severe restriction of natural protein to minimize methionine intake, and many patients still struggle to maintain healthy homocysteine levels. Since eliminating methionine from the diet reduces homocysteine levels, we hypothesized that an enzyme that can degrade methionine within the gastrointestinal (GI) tract could help HCU patients maintain healthy levels while easing natural protein restrictions. We describe the preclinical development of CDX-6512, a methionine gamma lyase (MGL) enzyme that was engineered for stability and activity within the GI tract for oral administration to locally degrade methionine. CDX-6512 is stable to low pH and intestinal proteases, enabling it to survive the harsh GI environment without enteric coating and to degrade methionine freed from dietary protein within the small intestine. Administering CDX-6512 to healthy non-human primates following a high protein meal led to a dose-dependent suppression of plasma methionine. In Tg-I278T Cbs-/- mice, an animal model that recapitulates aspects of HCU disease including highly elevated serum homocysteine levels, oral dosing of CDX-6512 after a high protein meal led to suppression in serum levels of both methionine and homocysteine. When animals received a daily dose of CDX-6512 with a high protein meal for two weeks, the Tg-I278T Cbs-/- mice maintained baseline homocysteine levels, whereas homocysteine levels in untreated animals increased by 39%. These preclinical data demonstrate the potential of CDX-6512 as an oral enzyme therapy for HCU.


Assuntos
Homocistinúria , Humanos , Camundongos , Animais , Homocistinúria/tratamento farmacológico , Homocistinúria/genética , Metionina/metabolismo , Homocisteína , Cistationina beta-Sintase/genética , Cistationina beta-Sintase/metabolismo , Racemetionina , Trato Gastrointestinal/metabolismo
2.
J Inherit Metab Dis ; 46(6): 1089-1103, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37494004

RESUMO

Maple syrup urine disease (MSUD) is an inborn error of branched-chain amino acid metabolism affecting several thousand individuals worldwide. MSUD patients have elevated levels of plasma leucine and its metabolic product α-ketoisocaproate (KIC), which can lead to severe neurotoxicity, coma, and death. Patients must maintain a strict diet of protein restriction and medical formula, and periods of noncompliance or illness can lead to acute metabolic decompensation or cumulative neurological impairment. Given the lack of therapeutic options for MSUD patients, we sought to develop an oral enzyme therapy that can degrade leucine within the gastrointestinal tract prior to its systemic absorption and thus enable patients to maintain acceptable plasma leucine levels while broadening their access to natural protein. We identified a highly active leucine decarboxylase enzyme from Planctomycetaceae bacterium and used directed evolution to engineer the enzyme for stability to gastric and intestinal conditions. Following high-throughput screening of over 12 000 enzyme variants over 9 iterative rounds of evolution, we identified a lead variant, LDCv10, which retains activity following simulated gastric or intestinal conditions in vitro. In intermediate MSUD mice or healthy nonhuman primates given a whey protein meal, oral treatment with LDCv10 suppressed the spike in plasma leucine and KIC and reduced the leucine area under the curve in a dose-dependent manner. Reduction in plasma leucine correlated with decreased brain leucine levels following oral LDCv10 treatment. Collectively, these data support further development of LDCv10 as a potential new therapy for MSUD patients.


Assuntos
Doença da Urina de Xarope de Bordo , Humanos , Camundongos , Animais , Leucina , Aminoácidos de Cadeia Ramificada , Proteínas , Terapia Enzimática , Primatas/metabolismo
3.
Sci Rep ; 13(1): 4748, 2023 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-36959353

RESUMO

Fabry disease is caused by a deficiency of α-galactosidase A (GLA) leading to the lysosomal accumulation of globotriaosylceramide (Gb3) and other glycosphingolipids. Fabry patients experience significant damage to the heart, kidney, and blood vessels that can be fatal. Here we apply directed evolution to generate more stable GLA variants as potential next generation treatments for Fabry disease. GLAv05 and GLAv09 were identified after screening more than 12,000 GLA variants through 8 rounds of directed evolution. Both GLAv05 and GLAv09 exhibit increased stability at both lysosomal and blood pH, stability to serum, and elevated enzyme activity in treated Fabry fibroblasts (19-fold) and GLA-/- podocytes (10-fold). GLAv05 and GLAv09 show improved pharmacokinetics in mouse and non-human primates. In a Fabry mouse model, the optimized variants showed prolonged half-lives in serum and relevant tissues, and a decrease of accumulated Gb3 in heart and kidney. To explore the possibility of diminishing the immunogenic potential of rhGLA, amino acid residues in sequences predicted to bind MHC II were targeted in late rounds of GLAv09 directed evolution. An MHC II-associated peptide proteomics assay confirmed a reduction in displayed peptides for GLAv09. Collectively, our findings highlight the promise of using directed evolution to generate enzyme variants for more effective treatment of lysosomal storage diseases.


Assuntos
Doença de Fabry , Humanos , Camundongos , Animais , Doença de Fabry/tratamento farmacológico , Doença de Fabry/genética , alfa-Galactosidase/genética , alfa-Galactosidase/metabolismo , Rim/metabolismo , Modelos Animais de Doenças , Fibroblastos/metabolismo
4.
JCI Insight ; 5(20)2020 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-33055427

RESUMO

Phenylalanine hydroxylase-deficient (PAH-deficient) phenylketonuria (PKU) results in systemic hyperphenylalaninemia, leading to neurotoxicity with severe developmental disabilities. Dietary phenylalanine (Phe) restriction prevents the most deleterious effects of hyperphenylalaninemia, but adherence to diet is poor in adult and adolescent patients, resulting in characteristic neurobehavioral phenotypes. Thus, an urgent need exists for new treatments. Additionally, rodent models of PKU do not adequately reflect neurocognitive phenotypes, and thus there is a need for improved animal models. To this end, we have developed PAH-null pigs. After selection of optimal CRISPR/Cas9 genome-editing reagents by using an in vitro cell model, zygote injection of 2 sgRNAs and Cas9 mRNA demonstrated deletions in preimplantation embryos, with embryo transfer to a surrogate leading to 2 founder animals. One pig was heterozygous for a PAH exon 6 deletion allele, while the other was compound heterozygous for deletions of exon 6 and of exons 6-7. The affected pig exhibited hyperphenylalaninemia (2000-5000 µM) that was treatable by dietary Phe restriction, consistent with classical PKU, along with juvenile growth retardation, hypopigmentation, ventriculomegaly, and decreased brain gray matter volume. In conclusion, we have established a large-animal preclinical model of PKU to investigate pathophysiology and to assess new therapeutic interventions.


Assuntos
Fígado/metabolismo , Fenilalanina Hidroxilase/genética , Fenilalanina/genética , Fenilcetonúrias/genética , Adolescente , Adulto , Animais , Sistemas CRISPR-Cas/genética , Dieta , Modelos Animais de Doenças , Edição de Genes , Humanos , Fígado/efeitos dos fármacos , Fenótipo , Fenilalanina/metabolismo , Fenilalanina/farmacologia , Fenilcetonúrias/dietoterapia , Fenilcetonúrias/metabolismo , Fenilcetonúrias/patologia , Suínos
5.
Curr Protoc Toxicol ; 67: 14.13.1-14.13.27, 2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-26828329

RESUMO

Liver disease is a major global health concern. Liver cirrhosis is one of the leading causes of death in the world and currently the only therapeutic option for end-stage liver disease (e.g., acute liver failure, cirrhosis, chronic hepatitis, cholestatic diseases, metabolic diseases, and malignant neoplasms) is orthotropic liver transplantation. Transplantation of hepatocytes has been proposed and used as an alternative to whole organ transplant to stabilize and prolong the lives of patients in some clinical cases. Although these experimental therapies have demonstrated promising and beneficial results, their routine use remains a challenge due to the shortage of donor livers available for cell isolation, variable quality of those tissues, the potential need for lifelong immunosuppression in the transplant recipient, and high costs. Therefore, new therapeutic strategies and more reliable clinical treatments are urgently needed. Recent and continuous technological advances in the development of stem cells suggest they may be beneficial in this respect. In this review, we summarize the history of stem cell and induced pluripotent stem cell (iPSC) technology in the context of hepatic differentiation and discuss the potential applications the technology may offer for human liver disease modeling and treatment. This includes developing safer drugs and cell-based therapies to improve the outcomes of patients with currently incurable health illnesses. We also review promising advances in other disease areas to highlight how the stem cell technology could be applied to liver diseases in the future. © 2016 by John Wiley & Sons, Inc.


Assuntos
Hepatócitos/transplante , Células-Tronco Pluripotentes Induzidas , Hepatopatias , Regeneração Hepática , Medicina Regenerativa/métodos , Transplante de Células-Tronco , Células-Tronco Adultas/citologia , Células-Tronco Adultas/transplante , Animais , Diferenciação Celular , Modelos Animais de Doenças , Hepatócitos/citologia , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/transplante , Hepatopatias/etiologia , Hepatopatias/terapia
6.
Curr Protoc Toxicol ; 62: 14.12.1-23, 2014 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-25378242

RESUMO

Orthotopic liver transplantation remains the only curative treatment for many end-stage liver diseases, yet the number of patients receiving liver transplants remains limited by the number of organs available for transplant. There is a need for alternative therapies for liver diseases. The transplantation of isolated hepatocytes (liver cells) has been used as an experimental therapy for liver disease in a limited number of cases. Recently, the 100th case of hepatocyte transplantation was reported. This review discusses the history of the hepatocyte transplant field, the major discoveries that supported and enabled the first hepatocyte transplants, and reviews the cases and outcomes of the first 100 clinical transplants. Some of the problems that limit the application or efficacy of hepatocyte transplantation are discussed, as are possible solutions to these problems. In conclusion, hepatocyte transplants have proven effective particularly in cases of metabolic liver disease where reversal or amelioration of the characteristic symptoms of the disease is easily quantified. However, no patients have been completely corrected of a metabolic liver disease for a significant amount of time by hepatocyte transplantation alone. It is likely that future developments in new sources of cells for transplantation will be required before this cellular therapy can be fully implemented and available for large numbers of patients.


Assuntos
Hepatócitos/citologia , Hepatopatias/terapia , Humanos , Hepatopatias/patologia , Transplante de Fígado
7.
Cell Transplant ; 23(9): 1143-51, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-23768881

RESUMO

Transplantation of human hepatocytes is gaining recognition as a bridge or an alternative to orthotopic liver transplantation for patients with acute liver failure and genetic defects. Since most patients require multiple cell infusions over an extended period of time, we investigated hepatic functions in cells maintained in University of Wisconsin solution at 4°C up to 72 h. Eleven different assessments of hepatic viability and function were investigated both pre- and posthypothermic storage, including plating efficiency, caspase-3/7 activity, ammonia metabolism, and drug-metabolizing capacity of isolated hepatocytes. Long-term function, basal, and induced cytochrome P450 activities were measured after exposure to prototypical inducing agents. Cells from 47 different human liver specimens were analyzed. Viability significantly decreased in cells cold stored in UW solution, while apoptosis level and plating efficiency were not significantly different from fresh cells. Luminescent and fluorescent methods assessed phases I and II activities both pre- and post-24-72 h of cold preservation. A robust induction (up to 200-fold) of phase I enzymes was observed in cultured cells. Phase II and ammonia metabolism remained stable during hypothermic storage, although the inductive effect of culture on each metabolic activity was eventually lost. Using techniques that characterize 11 measurements of hepatic viability and function from plating efficiency, to ammonia metabolism, to phases I and II drug metabolism, it was determined that while viability decreased, the remaining viable cells in cold-stored suspensions retained critical hepatic functions for up to 48 h at levels not significantly different from those observed in freshly isolated cells.


Assuntos
Criopreservação/métodos , Hepatócitos/citologia , Adenosina/química , Adenosina/farmacologia , Adolescente , Adulto , Idoso , Alopurinol/química , Alopurinol/farmacologia , Amônia/metabolismo , Caspase 3/metabolismo , Caspase 7/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Criança , Pré-Escolar , Temperatura Baixa , Sistema Enzimático do Citocromo P-450/metabolismo , Feminino , Glutationa/química , Glutationa/farmacologia , Hepatócitos/metabolismo , Hepatócitos/transplante , Humanos , Lactente , Insulina/química , Insulina/farmacologia , Masculino , Pessoa de Meia-Idade , Soluções para Preservação de Órgãos/química , Soluções para Preservação de Órgãos/farmacologia , Rafinose/química , Rafinose/farmacologia , Adulto Jovem
8.
Cell Transplant ; 23(1): 27-38, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23394081

RESUMO

Hepatocyte transplantation has been used to treat liver disease. The availability of cells for these procedures is quite limited. Human embryonic stem cells (hESCs) and induced pluripotent stem cells (hiPSCs) may be a useful source of hepatocytes for basic research and transplantation if efficient and effective differentiation protocols were developed and problems with tumorigenicity could be overcome. Recent evidence suggests that the cell of origin may affect hiPSC differentiation. Thus, hiPSCs generated from hepatocytes may differentiate back to hepatocytes more efficiently than hiPSCs from other cell types. We examined the efficiency of reprogramming adult and fetal human hepatocytes. The present studies report the generation of 40 hiPSC lines from primary human hepatocytes under feeder-free conditions. Of these, 37 hiPSC lines were generated from fetal hepatocytes, 2 hiPSC lines from normal hepatocytes, and 1 hiPSC line from hepatocytes of a patient with Crigler-Najjar syndrome, type 1. All lines were confirmed reprogrammed and expressed markers of pluripotency by gene expression, flow cytometry, immunocytochemistry, and teratoma formation. Fetal hepatocytes were reprogrammed at a frequency over 50-fold higher than adult hepatocytes. Adult hepatocytes were only reprogrammed with six factors, while fetal hepatocytes could be reprogrammed with three (OCT4, SOX2, NANOG) or four factors (OCT4, SOX2, NANOG, LIN28 or OCT4, SOX2, KLF4, C-MYC). The increased reprogramming efficiency of fetal cells was not due to increased transduction efficiency or vector toxicity. These studies confirm that hiPSCs can be generated from adult and fetal hepatocytes including those with genetic diseases. Fetal hepatocytes reprogram much more efficiently than adult hepatocytes, although both could serve as useful sources of hiPSC-derived hepatocytes for basic research or transplantation.


Assuntos
Reprogramação Celular/fisiologia , Células-Tronco Embrionárias/fisiologia , Hepatócitos/fisiologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Adulto , Animais , Técnicas de Cultura de Células , Diferenciação Celular/fisiologia , Processos de Crescimento Celular/fisiologia , Células Cultivadas , Células-Tronco Embrionárias/citologia , Feminino , Hepatócitos/citologia , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Lactente , Recém-Nascido , Fator 4 Semelhante a Kruppel , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Adulto Jovem
9.
Stem Cells Dev ; 22 Suppl 1: 96-102, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24304085

RESUMO

Cellular therapy for liver disease has been available in the clinic for more than 20 years, yet remarkably few patients receive this experimental therapy. Reasons for the small number of transplants performed are partially related to access to useful liver tissue and the difficulty with the isolation of viable cells. Stem cell sources of hepatocytes could theoretically relieve these obstacles to therapy if large numbers of functional hepatocytes could be generated and transplanted without risk of tumorigenicity. To date, there are no reports of stem cell sources with all of these characteristics, despite claims otherwise. Here we report the results of preclinical studies with appropriate animals models of metabolic liver disease and acute liver failure, and their correction by the transplantation of human amnion epithelial stem cells. The encouraging results of the preclinical studies have motivated the movement of isolation and banking of these cells to good manufacturing practice conditions so that the cells can be used in the clinic for transplantation of patients with liver disease.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos , Hepatopatias/economia , Falência Hepática Aguda/economia , Transplante de Células-Tronco/economia , Âmnio/citologia , Hepatócitos/citologia , Humanos , Hepatopatias/terapia , Falência Hepática Aguda/terapia , Pesquisa Translacional Biomédica
10.
Stem Cell Res ; 11(1): 563-73, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23644508

RESUMO

UNLABELLED: Domino liver transplantation is a method used to increase the number of liver grafts available for orthotopic liver transplantation (OLT). Reports indicate that livers from patients with metabolic liver disease can be safely transplanted into select recipients if the donor's defect and the recipient's metabolic needs are carefully considered. The liver of patients with many types of metabolic liver disease is morphologically and biochemically normal, except for the mutation that characterizes that disease. Other biochemical functions normally performed by the liver are present and presumably "normal" in these hepatocytes. Hepatocytes were isolated from the liver of 35 organ donors and 35 liver tissues taken at OLT from patients with liver disease were analyzed for 9 different measures of viability and function. The data indicate that cells isolated from some diseased livers performed as well or better than those isolated from organ donors with respect to viability, cell yield, plating efficiency and in assays of liver function, including drug metabolism, conjugation reactions and ammonia metabolism. Cells from metabolic diseased livers rapidly and efficiently repopulated a mouse liver upon transplantation. CONCLUSIONS: As with domino liver transplantation, domino cell transplantation deserves consideration as method to extend the pool of available organs and cells for transplantation.


Assuntos
Hepatócitos/transplante , Hepatopatias/patologia , Transplante de Fígado/métodos , Fígado/patologia , Adolescente , Adulto , Animais , Criança , Pré-Escolar , Feminino , Hepatócitos/citologia , Hepatócitos/metabolismo , Humanos , Lactente , Fígado/metabolismo , Fígado/cirurgia , Hepatopatias/metabolismo , Hepatopatias/cirurgia , Masculino , Camundongos , Adulto Jovem
11.
Mol Genet Metab ; 109(2): 132-8, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23566440

RESUMO

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.


Assuntos
Aminoácidos/sangue , Âmnio/citologia , Células Epiteliais/transplante , Doença da Urina de Xarope de Bordo/terapia , Neurotransmissores/metabolismo , Animais , Encéfalo/metabolismo , Ciclo do Ácido Cítrico , Humanos , Doença da Urina de Xarope de Bordo/sangue , Camundongos , Camundongos Transgênicos
12.
Hepatology ; 57(3): 1017-23, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23175463

RESUMO

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.


Assuntos
Âmnio/citologia , Células Epiteliais/transplante , Doença da Urina de Xarope de Bordo/terapia , Placenta/citologia , Transplante de Células-Tronco/métodos , 3-Metil-2-Oxobutanoato Desidrogenase (Lipoamida)/genética , 3-Metil-2-Oxobutanoato Desidrogenase (Lipoamida)/metabolismo , Aminoácidos de Cadeia Ramificada/genética , Animais , Animais Recém-Nascidos , Peso Corporal/fisiologia , Encéfalo/crescimento & desenvolvimento , Encéfalo/patologia , Diferenciação Celular/fisiologia , Modelos Animais de Doenças , Células Epiteliais/citologia , Feminino , Hepatócitos/citologia , Humanos , Doença da Urina de Xarope de Bordo/genética , Doença da Urina de Xarope de Bordo/patologia , Camundongos , Camundongos Mutantes , Gravidez , Transplante Heterólogo
13.
Cell Transplant ; 21(6): 1245-60, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22080793

RESUMO

Human hepatocyte transplantation is gaining acceptance for the treatment of liver diseases. However, the reagents used to isolate hepatocytes from liver tissue are not standardized and show lot-to-lot variability in enzyme activity and endotoxin contamination. For clinical application, highly purified reagents are preferable to crude digest preparations. A purified tissue dissociating enzyme (TDE) preparation (CIzyme(TM) purified enzymes) was developed based on the enzyme compositions found in a superior lot of collagenase previously used by our group for human hepatocyte isolation. The performance of this enzyme preparation was compared to collagenase type XI on 110 liver cases by assessing hepatocyte yield, viability, and seven other functional assays that included plating efficiency, basal and induced CYP450 activities, phase II conjugation activity, and ammonia metabolism. No statistically significant difference was observed between these TDEs when they were used to isolate hepatocytes from liver resections or organ donor tissue on 54 hepatocyte isolations with type XI enzyme and 56 isolations using CIzyme(TM). These results show that a highly purified and defined TDE preparation can be formulated that provides excellent performance with respect to viability, yield, and functional activity of the isolated cells. In addition to reproducible formulation, these purified enzyme products have only 2-3% of the endotoxin of crude enzyme preparations. These results show that purified enzymes such as CIzyme(TM) will be a safe and effective for the isolation of human hepatocytes for clinical transplants.


Assuntos
Colagenases/metabolismo , Hepatócitos/citologia , Separação Celular , Cromatografia Líquida de Alta Pressão , Colagenases/análise , Citocromo P-450 CYP1A1/metabolismo , Citocromo P-450 CYP1A2/metabolismo , Citocromo P-450 CYP3A/metabolismo , Hepatócitos/metabolismo , Humanos , Testosterona/metabolismo
14.
Hepatology ; 53(5): 1719-29, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21374689

RESUMO

UNLABELLED: Hepatocyte transplantation to treat liver disease is largely limited by the availability of useful cells. Human amniotic epithelial cells (hAECs) from term placenta express surface markers and gene characteristics of embryonic stem cells and have the ability to differentiate into all three germ layers, including tissues of endodermal origin (i.e., liver). Thus, hAECs could provide a source of stem cell-derived hepatocytes for transplantation. We investigated the differentiation of hAECs in vitro and after transplantation into the livers of severe combined immunodeficient (SCID)/beige mice. Moreover, we tested the ability of rat amniotic epithelial cells (rAECs) to replicate and differentiate upon transplantation into a syngenic model of liver repopulation. In vitro results indicate that the presence of extracellular matrix proteins together with a mixture of growth factors, cytokines, and hormones are required for differentiation of hAECs into hepatocyte-like cells. Differentiated hAECs expressed hepatocyte markers at levels comparable to those of fetal hepatocytes. They were able to metabolize ammonia, testosterone, and 17α-hydroxyprogesterone caproate, and expressed inducible fetal cytochromes. After transplantation into the liver of retrorsine (RS)-treated SCID/beige mice, naïve hAECs differentiated into hepatocyte-like cells that expressed mature liver genes such as cytochromes, plasma proteins, transporters, and other hepatic enzymes at levels equal to adult liver tissue. When transplanted in a syngenic animal pretreated with RS, rAECs were able to engraft and generate a progeny of cells with morphology and protein expression typical of mature hepatocytes. CONCLUSION: Amniotic epithelial cells possess the ability to differentiate into cells with characteristics of functional hepatocytes both in vitro and in vivo, thus representing a useful and noncontroversial source of cells for transplantation.


Assuntos
Âmnio/citologia , Diferenciação Celular , Células Epiteliais/citologia , Hepatócitos/citologia , Animais , Células Cultivadas , Humanos , Camundongos , Camundongos Endogâmicos C57BL
15.
Biochim Biophys Acta ; 1792(10): 1004-10, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19699299

RESUMO

Skvorak et al. [1] demonstrated the therapeutic efficacy of HTx in a murine model of iMSUD, confirming significant metabolic improvement and survival. To determine the effect of HTx on extrahepatic organs, we examined the metabolic effects of HTx in brain from iMSUD animals. Amino acid analysis revealed that HTx corrected increased ornithine, partially corrected depleted glutamine, and revealed a trend toward alloisoleucine correction. For amino acid and monoamine neurotransmitters, decreased GABA was partially corrected with HTx, while the l-histidine dipeptide of GABA, homocarnosine, was decreased in iMSUD mice and hypercorrected following HTx. Elevated branched-chain amino acids (BCAA; leucine, isoleucine, and valine) in MSUD can deplete brain tyrosine and tryptophan (the precursors of monoamine neurotransmitters, dopamine (DA) and serotonin (5-hydroxytryptamine; 5-HT)) through competition via the large neutral amino acid transporter. HTx corrected decreased DA levels and the DA metabolite, 3-methoxytyramine, and partially corrected the DA intermediate 3,4-dihydroxyphenylacetate (DOPAC) and 5-HT levels, despite normal tyrosine and tryptophan levels in iMSUD mouse brain. We further observed enhanced intracellular turnover of both DA and 5-HT in iMSUD mouse brain, both of which partially corrected with HTx. Our results suggest new pathomechanisms of neurotransmitter metabolism in this disorder and support the therapeutic relevance of HTx in iMSUD mice, while providing proof-of-principle that HTx has corrective potential in extrahepatic organs.


Assuntos
Encéfalo/anormalidades , Encéfalo/metabolismo , Hepatócitos/transplante , Doença da Urina de Xarope de Bordo/patologia , Doença da Urina de Xarope de Bordo/terapia , Animais , Encéfalo/patologia , Carnosina/análogos & derivados , Carnosina/metabolismo , Humanos , Fígado/patologia , Camundongos , Modelos Biológicos , Neurotransmissores/metabolismo , Ácido gama-Aminobutírico/metabolismo
16.
Mol Ther ; 17(7): 1266-73, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19436271

RESUMO

Maple syrup urine disease (MSUD; OMIM 248600) is an inborn error of metabolism of the branched chain alpha-ketoacid dehydrogenase (BCKDH) complex that is treated primarily by dietary manipulation of branched-chain amino acids (BCAA). Dietary restriction is lifelong and compliance is difficult. Liver transplantation significantly improves outcomes; however, alternative therapies are needed. To test novel therapies such as hepatocyte transplantation (HTx), we previously created a murine model of intermediate MSUD (iMSUD), which closely mimics human iMSUD. LacZ-positive murine donor hepatocytes were harvested and directly injected (10(5) cells/50 microl) into liver of iMSUD mice (two injections at 1-10 days of age). Donor hepatocytes engrafted into iMSUD recipient liver, increased liver BCKDH activity, improved blood total BCAA/alanine ratio, increased body weight at weaning, and extended the lifespan of HTx-treated iMSUD mice compared to phosphate-buffered saline (PBS)-treated and untreated iMSUD mice. Based on these data demonstrating partial metabolic correction of iMSUD in a murine model, coupled to the fact that multiple transplants are possible to enhance these results, we suggest that HTx represents a promising therapeutic intervention for MSUD that warrants further investigation.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/métodos , Hepatócitos/transplante , Doença da Urina de Xarope de Bordo/mortalidade , Doença da Urina de Xarope de Bordo/terapia , 3-Metil-2-Oxobutanoato Desidrogenase (Lipoamida)/metabolismo , Animais , Peso Corporal , Modelos Animais de Doenças , Fígado/metabolismo , Doença da Urina de Xarope de Bordo/patologia , Camundongos , Fenótipo , Reação em Cadeia da Polimerase , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sobrevida
17.
Brain ; 132(Pt 4): 903-18, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19293241

RESUMO

Maple syrup urine disease (MSUD) is an inherited disorder of branched-chain amino acid metabolism presenting with life-threatening cerebral oedema and dysmyelination in affected individuals. Treatment requires life-long dietary restriction and monitoring of branched-chain amino acids to avoid brain injury. Despite careful management, children commonly suffer metabolic decompensation in the context of catabolic stress associated with non-specific illness. The mechanisms underlying this decompensation and brain injury are poorly understood. Using recently developed mouse models of classic and intermediate maple syrup urine disease, we assessed biochemical, behavioural and neuropathological changes that occurred during encephalopathy in these mice. Here, we show that rapid brain leucine accumulation displaces other essential amino acids resulting in neurotransmitter depletion and disruption of normal brain growth and development. A novel approach of administering norleucine to heterozygous mothers of classic maple syrup urine disease pups reduced branched-chain amino acid accumulation in milk as well as blood and brain of these pups to enhance survival. Similarly, norleucine substantially delayed encephalopathy in intermediate maple syrup urine disease mice placed on a high protein diet that mimics the catabolic stress shown to cause encephalopathy in human maple syrup urine disease. Current findings suggest two converging mechanisms of brain injury in maple syrup urine disease including: (i) neurotransmitter deficiencies and growth restriction associated with branched-chain amino acid accumulation and (ii) energy deprivation through Krebs cycle disruption associated with branched-chain ketoacid accumulation. Both classic and intermediate models appear to be useful to study the mechanism of brain injury and potential treatment strategies for maple syrup urine disease. Norleucine should be further tested as a potential treatment to prevent encephalopathy in children with maple syrup urine disease during catabolic stress.


Assuntos
Edema Encefálico/etiologia , Doença da Urina de Xarope de Bordo/complicações , Aminoácidos/metabolismo , Animais , Comportamento Animal , Encéfalo/patologia , Edema Encefálico/patologia , Edema Encefálico/prevenção & controle , Mapeamento Encefálico/métodos , Proteínas Alimentares/administração & dosagem , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos/métodos , Transtornos do Crescimento/etiologia , Transtornos do Crescimento/metabolismo , Cetoácidos/metabolismo , Imageamento por Ressonância Magnética/métodos , Doença da Urina de Xarope de Bordo/tratamento farmacológico , Doença da Urina de Xarope de Bordo/patologia , Camundongos , Camundongos Knockout , Norleucina/uso terapêutico , Análise de Sobrevida
18.
Genesis ; 44(7): 345-53, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16832820

RESUMO

Genetically engineered mice with point mutations in endogenous genes (i.e., knockin mice) are extremely useful tools for dissecting gene function. Currently available methodologies for creating knockin mice are limited in that the introduced mutation is globally present in all cells of the animal from conception through adulthood. In this report, we describe a strategy for creating mice in which a point mutant allele replaces the wild type allele in a conditional manner, e.g., in a tissue-specific and/or temporally restricted pattern. As proof of concept, we created mice that conditionally harbor a point mutated gamma-aminobutyric acid receptor subunit. In the absence of Cre recombinase, the engineered allele produces only wild type product with no evidence of expression of the mutant. In contrast, following Cre-mediated recombination, only the point mutant product is produced. By restricting Cre expression to subpopulations of neurons of postnatal animals, we demonstrate tissue-specific regulation of the point mutant knockin. This strategy will be useful for a wide variety of studies that require precise conditional replacement of an endogenous wild type gene with a point mutant.


Assuntos
Análise Mutacional de DNA/métodos , Mutação Puntual , Receptores de GABA-A/genética , Animais , Marcação de Genes , Genótipo , Integrases/metabolismo , Camundongos , Camundongos Knockout , Neurônios/metabolismo , Especificidade de Órgãos , Fatores de Tempo
19.
BMC Med Genet ; 7: 33, 2006 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-16579849

RESUMO

BACKGROUND: Maple Syrup Urine Disease (MSUD) is an inborn error of metabolism caused by a deficiency of branched-chain keto acid dehydrogenase. MSUD has several clinical phenotypes depending on the degree of enzyme deficiency. Current treatments are not satisfactory and require new approaches to combat this disease. A major hurdle in developing new treatments has been the lack of a suitable animal model. METHODS: To create a murine model of classic MSUD, we used gene targeting and embryonic stem cell technologies to create a mouse line that lacked a functional E2 subunit gene of branched-chain keto acid dehydrogenase. To create a murine model of intermediate MSUD, we used transgenic technology to express a human E2 cDNA on the knockout background. Mice of both models were characterized at the molecular, biochemical, and whole animal levels. RESULTS: By disrupting the E2 subunit gene of branched-chain keto acid dehydrogenase, we created a gene knockout mouse model of classic MSUD. The homozygous knockout mice lacked branched-chain keto acid dehydrogenase activity, E2 immunoreactivity, and had a 3-fold increase in circulating branched-chain amino acids. These metabolic derangements resulted in neonatal lethality. Transgenic expression of a human E2 cDNA in the liver of the E2 knockout animals produced a model of intermediate MSUD. Branched-chain keto acid dehydrogenase activity was 5-6% of normal and was sufficient to allow survival, but was insufficient to normalize circulating branched-chain amino acids levels, which were intermediate between wildtype and the classic MSUD mouse model. CONCLUSION: These mice represent important animal models that closely approximate the phenotype of humans with the classic and intermediate forms of MSUD. These animals provide useful models to further characterize the pathogenesis of MSUD, as well as models to test novel therapeutic strategies, such as gene and cellular therapies, to treat this devastating metabolic disease.


Assuntos
Modelos Animais de Doenças , Doença da Urina de Xarope de Bordo/genética , Camundongos/genética , 3-Metil-2-Oxobutanoato Desidrogenase (Lipoamida)/metabolismo , Aciltransferases/análise , Aciltransferases/genética , Aminoácidos de Cadeia Ramificada/sangue , Aminoácidos de Cadeia Ramificada/urina , Animais , Marcação de Genes , Humanos , Doença da Urina de Xarope de Bordo/enzimologia , Doença da Urina de Xarope de Bordo/metabolismo , Camundongos Knockout , Camundongos Transgênicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA