Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Hepatol ; 70(6): 1170-1179, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30654068

RESUMO

BACKGROUND & AIMS: Inherited abnormalities in apolipoprotein E (ApoE) or low-density lipoprotein receptor (LDLR) function result in early onset cardiovascular disease and death. Currently, the only curative therapy available is liver transplantation. Hepatocyte transplantation is a potential alternative; however, physiological levels of hepatocyte engraftment and repopulation require transplanted cells to have a competitive proliferative advantage of over host hepatocytes. Herein, we aimed to test the efficacy and safety of a novel preparative regimen for hepatocyte transplantation. METHODS: Herein, we used an ApoE-deficient mouse model to test the efficacy of a new regimen for hepatocyte transplantation. We used image-guided external-beam hepatic irradiation targeting the median and right lobes of the liver to enhance cell transplant engraftment. This was combined with administration of the hepatic mitogen GC-1, a thyroid hormone receptor-ß agonist mimetic, which was used to promote repopulation. RESULTS: The non-invasive preparative regimen of hepatic irradiation and GC-1 was well-tolerated in ApoE-/- mice. This regimen led to robust liver repopulation by transplanted hepatocytes, which was associated with significant reductions in serum cholesterol levels after transplantation. Additionally, in mice receiving this regimen, ApoE was detected in the circulation 4 weeks after treatment and did not induce an immunological response. Importantly, the normalization of serum cholesterol prevented the formation of atherosclerotic plaques in this model. CONCLUSIONS: Significant hepatic repopulation and the cure of dyslipidemia in this model, using a novel and well-tolerated preparative regimen, demonstrate the clinical potential of applying this method to the treatment of inherited metabolic diseases of the liver. LAY SUMMARY: Hepatocyte transplantation is a promising alternative to liver transplantation for the treatment of liver diseases. However, it is inefficient, as restricted growth of transplanted cells in the liver limits its therapeutic benefits. Preparative treatments improve the efficiency of this procedure, but no clinically-feasible options are currently available. In this study we develop a novel well-tolerated preparative treatment to improve growth of cells in the liver and then demonstrate that this treatment completely cures an inherited lipid disorder in a mouse model.


Assuntos
Apolipoproteínas E/deficiência , Aterosclerose/prevenção & controle , Dislipidemias/terapia , Hepatócitos/transplante , Hiperlipoproteinemia Tipo II/terapia , Acetatos/farmacologia , Animais , Apolipoproteínas E/sangue , Colesterol/sangue , Modelos Animais de Doenças , Feminino , Hepatócitos/efeitos da radiação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fenóis/farmacologia
2.
J Am Soc Nephrol ; 28(3): 876-887, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27738124

RESUMO

Hyperoxaluria is a major risk factor for kidney stones and has no specific therapy, although Oxalobacter formigenes colonization is associated with reduced stone risk. O. formigenes interacts with colonic epithelium and induces colonic oxalate secretion, thereby reducing urinary oxalate excretion, via an unknown secretagogue. The difficulties in sustaining O. formigenes colonization underscore the need to identify the derived factors inducing colonic oxalate secretion. We therefore evaluated the effects of O. formigenes culture conditioned medium (CM) on apical 14C-oxalate uptake by human intestinal Caco-2-BBE cells. Compared with control medium, O. formigenes CM significantly stimulated oxalate uptake (>2.4-fold), whereas CM from Lactobacillus acidophilus did not. Treating the O. formigenes CM with heat or pepsin completely abolished this bioactivity, and selective ultrafiltration of the CM revealed that the O. formigenes-derived factors have molecular masses of 10-30 kDa. Treatment with the protein kinase A inhibitor H89 or the anion exchange inhibitor 4,4'-diisothiocyano-2,2'-stilbenedisulfonic acid completely blocked the CM-induced oxalate transport. Knockdown of the oxalate transporter SLC26A6 also significantly restricted the induction of oxalate transport by CM. In a mouse model of primary hyperoxaluria type 1, rectal administration of O. formigenes CM significantly reduced (>32.5%) urinary oxalate excretion and stimulated (>42%) distal colonic oxalate secretion. We conclude that O. formigenes-derived bioactive factors stimulate oxalate transport in intestinal cells through mechanisms including PKA activation. The reduction in urinary oxalate excretion in hyperoxaluric mice treated with O. formigenes CM reflects the in vivo retention of biologic activity and the therapeutic potential of these factors.


Assuntos
Fatores Biológicos/farmacologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Oxalatos/metabolismo , Oxalobacter formigenes , Animais , Humanos , Hiperoxalúria/metabolismo , Transporte de Íons , Masculino , Camundongos
3.
J Hepatol ; 66(5): 987-1000, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28027971

RESUMO

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.


Assuntos
Rejeição de Enxerto , Hepatócitos/transplante , Fígado/efeitos da radiação , Condicionamento Pré-Transplante , Adulto , Animais , Feminino , Humanos , Hepatopatias/terapia , Macaca fascicularis , Masculino , Suínos , Transplante Heterólogo
4.
Hum Mol Genet ; 23(2): 368-82, 2014 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-24009312

RESUMO

Post-transcriptional processing of some long non-coding RNAs (lncRNAs) reveals that they are a source of miRNAs. We show that the 268-nt non-coding RNA component of mitochondrial RNA processing endoribonuclease, (RNase MRP), is the source of at least two short (∼20 nt) RNAs designated RMRP-S1 and RMRP-S2, which function as miRNAs. Point mutations in RNase MRP cause human cartilage-hair hypoplasia (CHH), and several disease-causing mutations map to RMRP-S1 and -S2. SHAPE chemical probing identified two alternative secondary structures altered by disease mutations. RMRP-S1 and -S2 are significantly reduced in two fibroblast cell lines and a B-cell line derived from CHH patients. Tests of gene regulatory activity of RMRP-S1 and -S2 identified over 900 genes that were significantly regulated, of which over 75% were down-regulated, and 90% contained target sites with seed complements of RMRP-S1 and -S2 predominantly in their 3' UTRs. Pathway analysis identified regulated genes that function in skeletal development, hair development and hematopoietic cell differentiation including PTCH2 and SOX4 among others, linked to major CHH phenotypes. Also, genes associated with alternative RNA splicing, cell proliferation and differentiation were highly targeted. Therefore, alterations RMRP-S1 and -S2, caused by point mutations in RMRP, are strongly implicated in the molecular mechanism of CHH.


Assuntos
Endorribonucleases/genética , Cabelo/anormalidades , Doença de Hirschsprung/genética , Síndromes de Imunodeficiência/genética , Fígado/metabolismo , MicroRNAs/genética , Osteocondrodisplasias/congênito , Interferência de RNA , RNA Longo não Codificante/genética , Processamento Alternativo , Linhagem Celular , Células HEK293 , Hematopoese/genética , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Fígado/patologia , Conformação de Ácido Nucleico , Osteocondrodisplasias/genética , Receptores Patched , Receptor Patched-2 , Fenótipo , Doenças da Imunodeficiência Primária , Receptores de Superfície Celular/metabolismo , Fatores de Transcrição SOXC/metabolismo
5.
Hepatology ; 62(1): 147-57, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25690322

RESUMO

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.


Assuntos
Células-Tronco Pluripotentes Induzidas/metabolismo , Hepatopatias/etiologia , Deficiência de alfa 1-Antitripsina/complicações , Células Cultivadas , Retículo Endoplasmático Rugoso/metabolismo , Humanos , Hepatopatias/metabolismo , alfa 1-Antitripsina/metabolismo
6.
Hepatol Commun ; 7(3): e0070, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36848094

RESUMO

BACKGROUND: α1-antitrypsin deficiency is most commonly caused by a mutation in exon-7 of SERPINA1 (SA1-ATZ), resulting in hepatocellular accumulation of a misfolded variant (ATZ). Human SA1-ATZ-transgenic (PiZ) mice exhibit hepatocellular ATZ accumulation and liver fibrosis. We hypothesized that disrupting the SA1-ATZ transgene in PiZ mice by in vivo genome editing would confer a proliferative advantage to the genome-edited hepatocytes, enabling them to repopulate the liver. METHODS: To create a targeted DNA break in exon-7 of the SA1-ATZ transgene, we generated 2 recombinant adeno-associated viruses (rAAV) expressing a zinc-finger nuclease pair (rAAV-ZFN), and another rAAV for gene correction by targeted insertion (rAAV-TI). PiZ mice were injected i.v. with rAAV-TI alone or the rAAV-ZFNs at a low (7.5×1010vg/mouse, LD) or a high dose (1.5×1011vg/mouse, HD), with or without rAAV-TI. Two weeks and 6 months after treatment, livers were harvested for molecular, histological, and biochemical analyses. RESULTS: Two weeks after treatment, deep sequencing of the hepatic SA1-ATZ transgene pool showed 6%±3% or 15%±4% nonhomologous end joining in mice receiving LD or HD rAAV-ZFN, respectively, which increased to 36%±12% and 36%±12%, respectively, 6 months after treatment. Two weeks postinjection of rAAV-TI with LD or HD of rAAV-ZFN, repair by targeted insertion occurred in 0.10%±0.09% and 0.25%±0.14% of SA1-ATZ transgenes, respectively, which increased to 5.2%±5.0% and 33%±13%, respectively, 6 months after treatment. Six months after rAAV-ZFN administration, there was a marked clearance of ATZ globules from hepatocytes, and resolution of liver fibrosis, along with reduction of hepatic TAZ/WWTR1, hedgehog ligands, Gli2, a TIMP, and collagen content. CONCLUSIONS: ZFN-mediated SA1-ATZ transgene disruption provides a proliferative advantage to ATZ-depleted hepatocytes, enabling them to repopulate the liver and reverse hepatic fibrosis.


Assuntos
Edição de Genes , Nucleases de Dedos de Zinco , Humanos , Animais , Camundongos , Cirrose Hepática/genética , Cirrose Hepática/terapia , Hepatócitos , Modelos Animais de Doenças , Peptídeos e Proteínas de Sinalização Intracelular
8.
Gastroenterology ; 136(3): 990-9, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19026649

RESUMO

BACKGROUND & AIMS: The ability to obtain unlimited numbers of human hepatocytes would improve the development of cell-based therapies for liver diseases, facilitate the study of liver biology, and improve the early stages of drug discovery. Embryonic stem cells are pluripotent, potentially can differentiate into any cell type, and therefore could be developed as a source of human hepatocytes. METHODS: To generate human hepatocytes, human embryonic stem cells were differentiated by sequential culture in fibroblast growth factor 2 and human activin-A, hepatocyte growth factor, and dexamethasone. Functional hepatocytes were isolated by sorting for surface asialoglycoprotein-receptor expression. Characterization was performed by real-time polymerase chain reaction, immunohistochemistry, immunoblot, functional assays, and transplantation. RESULTS: Embryonic stem cell-derived hepatocytes expressed liver-specific genes, but not genes representing other lineages, secreted functional human liver-specific proteins similar to those of primary human hepatocytes, and showed human hepatocyte cytochrome P450 metabolic activity. Serum from rodents given injections of embryonic stem cell-derived hepatocytes contained significant amounts of human albumin and alpha1-antitrypsin. Colonies of cytokeratin-18 and human albumin-expressing cells were present in the livers of recipient animals. CONCLUSIONS: Human embryonic stem cells can be differentiated into cells with many characteristics of primary human hepatocytes. Hepatocyte-like cells can be enriched and recovered based on asialoglycoprotein-receptor expression and potentially could be used in drug discovery research and developed as therapeutics.


Assuntos
Técnicas de Cultura de Células/métodos , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/transplante , Hepatócitos/citologia , Transplante de Células-Tronco , Ativinas/farmacologia , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Linhagem Celular , Dexametasona/farmacologia , Células-Tronco Embrionárias/ultraestrutura , Fator 2 de Crescimento de Fibroblastos/farmacologia , Expressão Gênica , Proteína Relacionada a TNFR Induzida por Glucocorticoide/farmacologia , Fator de Crescimento de Hepatócito/farmacologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Microscopia Eletrônica , Fenótipo
9.
Liver Transpl ; 16(9): 1086-97, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20818747

RESUMO

We recently reported the isolation and characterization of a novel population of progenitor cells called liver-derived progenitor cells (LDPCs), which could differentiate into functional hepatocytes in vitro. However, our original studies resulted in relatively low and variable hepatic differentiation efficiency without validation of in vivo potential of LDPCs. Here, we report an efficient and robust hepatic differentiation of LDPCs under well-defined culture conditions and in vivo differentiation of LDPCs to mature hepatocytes. In addition to morphological studies, we performed reverse-transcription polymerase chain reaction (RT-PCR) and microRNA analyses of the in vitro hepatic differentiation of LDPCs to substantiate the efficiency of the differentiation process. The histological studies on the differentiated LDPCs showed that more than 50% of the cells were positive for albumin, cytokeratin 18, and hepatocyte nuclear factor 1 alpha and contained glycogen particles, all consistent with differentiation to functional hepatocytes. We also demonstrated by RT-PCR that upon differentiation, they expressed several markers found in mature hepatocytes and the microRNA profile of LDPCs became similar to the profile of fresh hepatocytes, confirming our morphological findings. Finally, the transplantation of LDPCs in a dipeptidyl peptidase IV-deficient (DPPIV(-/-)) rat model showed that LDPCs were able to engraft and form mature hepatocytes in the livers of the DPPIV(-/-) rats. In summary, LDPCs are a unique population of liver progenitor cells capable of hepatic differentiation both in vitro and in vivo, which makes them a potentially valuable resource for important applications such as pharmacological studies and cell therapies for a variety of liver disorders.


Assuntos
Diferenciação Celular , Hepatócitos/metabolismo , Fígado/metabolismo , MicroRNAs/metabolismo , Células-Tronco/metabolismo , Albuminas/metabolismo , Animais , Biomarcadores/metabolismo , Diferenciação Celular/genética , Proliferação de Células , Forma Celular , Células Cultivadas , Dipeptidil Peptidase 4/deficiência , Dipeptidil Peptidase 4/genética , Feminino , Imunofluorescência , Regulação da Expressão Gênica , Glicogênio/metabolismo , Fator 1-alfa Nuclear de Hepatócito/metabolismo , Hepatócitos/transplante , Queratina-18/metabolismo , Fígado/citologia , Fenótipo , Ratos , Ratos Endogâmicos F344 , Ratos Sprague-Dawley , Ratos Transgênicos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transplante de Células-Tronco
10.
Hepatology ; 50(3): 815-24, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19585550

RESUMO

UNLABELLED: Asialoglycoprotein receptor (ASGPR)-mediated endocytosis has been used to target genes to hepatocytes in vivo. However, the level and duration of transgene expression have been low because of lysosomal translocation and degradation of the DNA and lack of its integration into the host genome. In this study we packaged the DNA of interest in proteoliposomes containing the fusogenic galactose-terminated F-glycoprotein of the Sendai virus (FPL) for targeted delivery to hepatocytes. After the FPL binds to ASGPR on the hepatocyte surface, fusogenic activity of the F-protein delivers the DNA into the cytosol, bypassing the endosomal pathway. For transgene integration we designed plasmids containing one transcription unit expressing the Sleeping Beauty transposase (SB) and another expressing human uridinediphosphoglucuronate glucuronosyltransferase-1A1 (pSB-hUGT1A1). The latter was flanked by inverted/direct repeats that are substrates of SB. In cell culture, FPL-mediated delivery of the E. coli beta-galactosidase gene (LacZ) resulted in transduction of ASGPR-positive cells (rat hepatocytes or Hepa1 cell line), but not of ASGPR-negative 293 cells. Intravenous injection of the FPL-entrapped pSB-hUGT1A1 (4-8 microg/day, 1-4 doses) into UGT1A1-deficient hyperbilirubinemic Gunn rats (model of Crigler-Najjar syndrome type 1) resulted in hUGT1A1 expression in 5%-10% of hepatocytes, but not in other cell types. Serum bilirubin levels declined by 30% +/- 4% in 2 weeks and remained at that level throughout the 7-month study duration. With histidine containing FPL, serum bilirubin was reduced by 40% +/- 5%, and bilirubin glucuronides were excreted into bile. No antibodies were detectable in the recipient rats against the F-protein or human UGT1A1. CONCLUSION: FPL is an efficient hepatocyte-targeted gene delivery platform in vivo that warrants further exploration toward clinical application.


Assuntos
Receptor de Asialoglicoproteína/administração & dosagem , Icterícia/terapia , Proteolipídeos/administração & dosagem , Transposases/administração & dosagem , Animais , Síndrome de Crigler-Najjar/terapia , Modelos Animais de Doenças , Técnicas de Transferência de Genes , Terapia Genética/métodos , Glucuronosiltransferase/administração & dosagem , Hepatócitos/efeitos dos fármacos , Humanos , Hiperbilirrubinemia/terapia , Ratos , Ratos Gunn , Proteínas Virais de Fusão/administração & dosagem
11.
Hepatology ; 49(1): 258-67, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19003915

RESUMO

Engraftment of donor hepatocytes is a critical step that determines the success of hepatocyte transplantation. Rapid and efficient integration of donor cells would enable prompt liver repopulation of these cells in response to selective proliferative stimuli offered by a preparative regimen. We have earlier demonstrated that hepatic irradiation (HIR) in combination with a variety of hepatotrophic growth signals, such as partial hepatectomy and hepatocyte growth factor, can be used as a preparative regimen for liver repopulation of transplanted hepatocytes. In this study, we investigated the effects of HIR on engraftment of transplanted dipeptidyl peptidase IV (DPPIV)-positive hepatocytes in congeneic DPPIV-deficient rats. HIR-induced apoptosis of hepatic sinusoidal endothelial cells (SEC) within 6 hours of HIR resulted in dehiscence of the SEC lining in 24 hours. Although there was no change of the number of Kupffer cells after HIR, colloidal carbon clearance decreased 24 hours post HIR, indicating a suppression of phagocytic function. DPPIV+ donor cells were transplanted 24 hours after HIR (0-50 Gy). There was an HIR dose-dependent increase in the donor hepatocyte mass engrafted in the liver parenchyma. The number of viable transplanted hepatocytes present in hepatic sinusoids or integrated in the parenchyma was greater in the HIR-treated group at 3 and 7 days after transplantation compared with the sham controls. Finally, we validated these rodent studies in cynomolgus monkeys, demonstrating that a single 10-Gy dose of HIR was sufficient to enhance engraftment of donor porcine hepatocytes. These data indicate that transient disruption of the SEC barrier and inhibition of the phagocytic function of Kupffer cells by HIR enhances hepatocyte engraftment and the integrated donor cell mass. Thus, preparative HIR could be potentially useful to augment hepatocyte transplantation.


Assuntos
Hepatócitos/efeitos da radiação , Hepatócitos/transplante , Fígado/cirurgia , Animais , Apoptose , Proliferação de Células/efeitos da radiação , Dipeptidil Peptidase 4/genética , Endotélio Vascular/patologia , Endotélio Vascular/fisiologia , Feminino , Sobrevivência de Enxerto , Hepatectomia , Células de Kupffer/fisiologia , Células de Kupffer/efeitos da radiação , Fígado/citologia , Regeneração Hepática/fisiologia , Macaca fascicularis , Masculino , Fagocitose/efeitos da radiação , Ratos , Ratos Endogâmicos F344 , Suínos , Condicionamento Pré-Transplante/métodos , Transplante Heterólogo
12.
Dev Cell ; 4(5): 607-8, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12737794

RESUMO

Two nuclear receptors of xenobiotic drugs, PXR and CAR, are central regulators of detoxification enzymes. New studies extend the role of these receptors to a natural detoxification process. They coordinate induction of proteins for storage, glucuronidation, and canalicular transport of bilirubin.


Assuntos
Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores de Esteroides/metabolismo , Fatores de Transcrição/metabolismo , Animais , Bilirrubina/metabolismo , Receptor Constitutivo de Androstano , Glucuronosiltransferase/metabolismo , Humanos , Inativação Metabólica , Receptor de Pregnano X
13.
Methods Mol Biol ; 481: 117-40, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19096805

RESUMO

Ex vivo gene transfer into hepatocytes could serve several purposes in the context of gene therapy or cell transplantation: (1) isolated hepatocytes can be transduced in culture with therapeutic genes and then transplanted into the recipient; (2) marker genes can be introduced for subsequent identification of transplanted cells and their progeny; (3) gene transfer can be used for conditional immortalization of hepatocytes for expansion in culture; (4) immunomodulatory genes can be transferred into hepatocytes to prevent allograft rejection. Gene transfer into cultured hepatocytes can be achieved using DNA that is not incorporated into recombinant viruses. In such systems, transgene integration into the host cell genome can be enhanced using transposon systems, such as "sleeping beauty." In addition to using the conventional reagents, such as cationic liposomes, DNA transfer into hepatocytes can be achieved by Nucleofection or special hepatocyte-targeted carriers such as proteoliposomes containing galactose-terminated glycoproteins (e.g. the F protein of the Sendai virus). Alternatively, genes can be transferred using recombinant viruses, such as adenoviral vectors that are episomal or retroviral vectors (including lentiviruses) that permit integration of the transgene into the host genome. Gene transfer using lentiviral vectors has been achieved in both attached and suspended hepatocytes. Transduction efficiency of lentiviral vectors can be enhanced using magnetic nanoparticles (Magnetofection).


Assuntos
Técnicas de Transferência de Genes , Hepatócitos/metabolismo , Animais , Linhagem Celular Transformada/citologia , Terapia Genética/métodos , Hepatócitos/citologia , Hepatócitos/transplante , Humanos , Transplante de Fígado/métodos , Magnetismo/métodos , Modelos Biológicos , Nanopartículas/uso terapêutico , Coloração e Rotulagem/métodos , Condicionamento Pré-Transplante/métodos
14.
Int J Radiat Oncol Biol Phys ; 103(4): 922-934, 2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30503786

RESUMO

Orthotopic liver transplantation (OLT) has been effective in managing end-stage liver disease since the advent of cyclosporine immunosuppression therapy in 1980. The major limitations of OLT are organ supply, monetary cost, and the burden of lifelong immunosuppression. Hepatocyte transplantation, as a substitute for OLT, has been an exciting topic of investigation for several decades. HT is potentially minimally invasive and can serve as a vehicle for delivery of personalized medicine through autologous cell transplant after modification ex vivo. However, 3 major hurdles have prevented large-scale clinical application: (1) availability of transplantable cells; (2) safe and efficient ex vivo gene therapy methods; and (3) engraftment and repopulation efficiency. This review will discuss new sources for transplantable liver cells obtained by lineage reprogramming, clinically acceptable methods of genetic manipulation, and the development of hepatic irradiation-based preparative regimens for enhancing engraftment and repopulation of transplanted hepatocytes. We will also review the results of the first 3 patients with genetic liver disorders who underwent preparative hepatic irradiation before hepatocyte transplantation.


Assuntos
Hepatócitos/citologia , Transplante de Fígado/métodos , Animais , Proliferação de Células , Terapia Genética , Humanos , Transplante de Fígado/efeitos adversos , Segurança
15.
Hepatol Commun ; 3(1): 129-146, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30620000

RESUMO

Hepatocyte transplantation is an attractive alternative to liver transplantation. Thus far, however, extensive liver repopulation by adult hepatocytes has required ongoing genetic, physical, or chemical injury to host liver. We hypothesized that providing a regulated proliferative and/or survival advantage to transplanted hepatocytes should enable repopulation in a normal liver microenvironment. Here, we repopulated livers of DPPIV- (dipeptidyl peptidase-4) rats and Ugt1a1 (uridinediphosphoglucuronate glucuronosyltransferase 1a1)-deficient Gunn rats (model of Crigler-Najjar syndrome type 1), both models without underlying liver injury, for up to 1 year by transplanting lenti-hYAP-ERT2 (mutated estrogen receptor ligand-binding domain 2)-transduced hepatocytes (YAP-Hc). Yap (yes-associated protein) nuclear translocation/function in YAP-Hc was regulated by tamoxifen. Repopulating YAP-Hc and host hepatocytes were fluorescence-activated cell sorting-purified and their transcriptomic profiles compared by RNAseq. After 1 year of liver repopulation, YAP-Hc clusters exhibited normal morphology, integration into hepatic plates and hepatocyte-specific gene expression, without dysplasia, dedifferentiation, or tumorigenesis. RNAseq analysis showed up-regulation of 145 genes promoting cell proliferation and 305 genes suppressing apoptosis, including hepatocyte growth factor and connective tissue growth factor among the top 30 in each category and provided insight into the mechanism of cell competition that enabled replacement of host hepatocytes by YAP-Hc. In Gunn rats transplanted with YAP-Hc+tamoxifen, there was a 65%-81% decline in serum bilirubin over 6 months versus 8%-20% with control-Hc, representing a 3-4-fold increase in therapeutic response. This correlated with liver repopulation as demonstrated by the presence of Ugt1a1-positive hepatocyte clusters in livers and western blot analysis of tissue homogenates. Conclusion: Tamoxifen-regulated nuclear translocation/function of hYAP-ERT2 enabled long-term repopulation of DPPIV-/- and Gunn rat livers by hYAP-ERT2-transduced hepatocytes without tumorigenesis. This cell transplantation strategy may offer a potential therapy for most of the inherited monogenic liver diseases that do not exhibit liver injury.

16.
Transplantation ; 85(9): 1253-60, 2008 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-18475180

RESUMO

BACKGROUND: Primary hyperoxaluria type-1 (PH1) is an autosomal recessive disease characterized by excessive oxalate production by hepatocytes caused by the deficiency of peroxisomal alanine-glyoxylate aminotransferase (AGT) activity. Persistent hyperoxaluria causes nephrocalcinosis and urolithiasis, leading to renal failure, followed by tissue oxalosis with life-threatening complications. Combined liver-kidney transplantation is the only definitive treatment of PH1. Hepatocyte transplantation, which is much less invasive, could have offered an attractive alternative. However, because the AGT-deficient hepatocytes overproduce oxalate, a large fraction of the mutant host hepatocytes must be replaced by AGT-competent cells, which is beyond the capacity of current hepatocyte transplantation procedures. Here, we have evaluated a preparative irradiation-based method of liver repopulation in an Agxt-deleted mouse model of PH1 (Agxt-/-). MATERIALS AND METHODS: Hepatocytes (10(6) viable cells) isolated from congeneic mice ([ROSA]26 C57BL/6J) expressing Escherichia coli beta-galactosidase were transplanted into Agxt-/- mice by intrasplenic injection. The preparative regimen consisted of X-irradiation of the host liver and mitotic stimulation of the hepatocytes by adenovector-based expression of hepatocyte growth factor. RESULTS: The procedure resulted in progressive replacement of the mutant host hepatocytes with the AGT-competent hepatocytes, leading to correction of urinary oxalate excretion. Oral ethylene glycol challenge (0.7% for 1 week) resulted in nephrocalcinosis and microlithiasis in untreated Agxt-/- mice, but not in the mice after hepatic repopulation. CONCLUSION: The results indicate that hepatocyte transplantation after appropriate preparative regimens may permit sufficient repopulation of the liver to ameliorate hyperoxaluria, and therefore should be evaluated further as a potential treatment of PH1.


Assuntos
Hepatócitos/transplante , Hiperoxalúria Primária/cirurgia , Animais , Modelos Animais de Doenças , Etilenoglicol/administração & dosagem , Hepatócitos/enzimologia , Regeneração Hepática , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nefrocalcinose/etiologia , Transaminases/deficiência , Transplante Homólogo , beta-Galactosidase/metabolismo
18.
Clin Pharmacol Drug Dev ; 6(2): 140-146, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28263463

RESUMO

Hyperbilirubinemia is a common finding in individuals with a history of substance abuse. Although this may indicate a serious disorder of liver function, this is not always the case. An understanding of bilirubin formation, metabolism, and transport can provide a helpful approach to dealing with these patients. This is typified by studies of patients treated with the antiretroviral drug atazanavir. Atazanavir has been associated with hyperbilirubinemia in as many as one-third of individuals for whom it has been prescribed, evoking concerns of hepatotoxicity. The studies in this report were designed to determine mechanisms by which this occurs. The data show that this drug inhibits the enzyme UDP-glucuronosyl transferase-1A1, responsible for conjugating bilirubin with glucuronic acid. This conjugation step is required for bilirubin excretion into bile, and when it is inhibited, bilirubin refluxes from the liver into the circulation, causing unconjugated hyperbilirubinemia. Other parameters of bilirubin formation, binding to albumin in the circulation, uptake into hepatocytes, and intracellular protein binding in hepatocytes were unaffected by atazanavir. The effect of atazanavir on serum bilirubin levels is reversible, consistent with lack of structural damage to the liver.


Assuntos
Sulfato de Atazanavir/efeitos adversos , Bilirrubina/sangue , Inibidores da Protease de HIV/efeitos adversos , Hiperbilirrubinemia/induzido quimicamente , Animais , Sulfato de Atazanavir/administração & dosagem , Bilirrubina/metabolismo , Células Cultivadas , Feminino , Glucuronosiltransferase/antagonistas & inibidores , Glutationa Transferase/metabolismo , Inibidores da Protease de HIV/administração & dosagem , Hepatócitos/metabolismo , Humanos , Hiperbilirrubinemia/sangue , Masculino , Ratos Wistar , Ritonavir/administração & dosagem , Transtornos Relacionados ao Uso de Substâncias/complicações
19.
Hepatol Int ; 11(1): 54-69, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27530815

RESUMO

The discovery that coordinated expression of a limited number of genes can reprogram differentiated somatic cells to induced pluripotent stem cells (iPSC) has opened novel possibilities for developing cell-based models of diseases and regenerative medicine utilizing cell reprogramming or cell transplantation. Directed differentiation of iPSCs can potentially generate differentiated cells belonging to any germ layer, including cells with hepatocyte-like morphology and function. Such cells, termed iHeps, can be derived by sequential cell signaling using available information on embryological development or by forced expression of hepatocyte-enriched transcription factors. In addition to the translational aspects of iHeps, the experimental findings have provided insights into the mechanisms of cell plasticity that permit one cell type to transition to another. However, iHeps generated by current methods do not fully exhibit all characteristics of mature hepatocytes, highlighting the need for additional research in this area. Here we summarize the current approaches and achievements in this field and discuss some existing hurdles and emerging approaches for improving iPSC differentiation, as well as maintaining such cells in culture for increasing their utility in disease modeling and drug development.


Assuntos
Técnicas de Cultura de Células/métodos , Hepatócitos/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Animais , Diferenciação Celular/fisiologia , Reprogramação Celular/fisiologia , Humanos , Medicina Regenerativa , Transdução de Sinais
20.
Methods Mol Biol ; 1506: 131-147, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27830550

RESUMO

Liver transplantation has been established as a curative therapy for acute and chronic liver failure, as well as liver-based inherited metabolic diseases. Because of the complexity of organ transplantation and the worldwide shortage of donor organs, hepatocyte transplantation is being developed as a bridging therapy until donor organs become available, or for amelioration of inherited liver-based diseases. The Gunn rat is a molecular and metabolic model of Crigler-Najjar syndrome type 1, which is characterized by lifelong unconjugated hyperbilirubinemia due to the lack of uridinediphosphoglucuronate glucuronosyltransferase-1 (UGT1A1)-mediated bilirubin glucuronidation. Gunn rats are convenient for evaluating the effect of hepatocyte transplantation or gene therapy, because the extent of UGT1A1 replacement can be assessed by serial determination of serum bilirubin levels, and excretion of bilirubin glucuronides in bile provide definitive evidence of the function of the transplanted hepatocytes or the effect of gene therapy. The core techniques involved in hepatocyte transplantation in Gunn rats are discussed in this chapter.


Assuntos
Transplante de Células/métodos , Síndrome de Crigler-Najjar/cirurgia , Técnicas de Transferência de Genes , Hepatócitos/transplante , Hepatopatias/cirurgia , Animais , Bile/química , Pigmentos Biliares/análise , Bilirrubina/análogos & derivados , Bilirrubina/sangue , Bilirrubina/metabolismo , Separação Celular/instrumentação , Separação Celular/métodos , Cromatografia Líquida de Alta Pressão , Síndrome de Crigler-Najjar/sangue , Modelos Animais de Doenças , Feminino , Terapia Genética/métodos , Glucuronosiltransferase/genética , Glucuronosiltransferase/metabolismo , Hepatócitos/metabolismo , Heterozigoto , Homozigoto , Humanos , Hiperbilirrubinemia/sangue , Fígado/metabolismo , Fígado/cirurgia , Hepatopatias/metabolismo , Testes de Função Hepática , Masculino , Ratos , Ratos Gunn
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA