Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Annu Rev Physiol ; 85: 469-493, 2023 02 10.
Article in English | MEDLINE | ID: mdl-36270290

ABSTRACT

Liver regeneration occurs in response to diverse injuries and is capable of functionally reestablishing the lost parenchyma. This phenomenon has been known since antiquity, encapsulated in the Greek myth where Prometheus was to be punished by Zeus for sharing the gift of fire with humanity by having an eagle eat his liver daily, only to have the liver regrow back, thus ensuring eternal suffering and punishment. Today, this process is actively leveraged clinically during living donor liver transplantation whereby up to a two-thirds hepatectomy (resection or removal of part of the liver) on a donor is used for transplant to a recipient. The donor liver rapidly regenerates to recover the lost parenchymal mass to form a functional tissue. This astonishing regenerative process and unique capacity of the liver are examined in further detail in this review.


Subject(s)
Liver Transplantation , Animals , Humans , Living Donors , Liver , Hepatectomy , Liver Regeneration/physiology , Homeostasis , Mammals
2.
Am J Physiol Gastrointest Liver Physiol ; 327(1): G1-G15, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38651949

ABSTRACT

The progress of research focused on cholangiocytes and the biliary tree during development and following injury is hindered by limited available quantitative methodologies. Current techniques include two-dimensional standard histological cell-counting approaches, which are rapidly performed, error prone, and lack architectural context or three-dimensional analysis of the biliary tree in opacified livers, which introduce technical issues along with minimal quantitation. The present study aims to fill these quantitative gaps with a supervised machine-learning model (BiliQML) able to quantify biliary forms in the liver of anti-keratin 19 antibody-stained whole slide images. Training utilized 5,019 researcher-labeled biliary forms, which following feature selection, and algorithm optimization, generated an F score of 0.87. Application of BiliQML on seven separate cholangiopathy models [genetic (Afp-CRE;Pkd1l1null/Fl, Alb-CRE;Rbp-jkfl/fl, and Albumin-CRE;ROSANICD), surgical (bile duct ligation), toxicological (3,5-diethoxycarbonyl-1,4-dihydrocollidine), and therapeutic (Cyp2c70-/- with ileal bile acid transporter inhibition)] allowed for a means to validate the capabilities and utility of this platform. The results from BiliQML quantification revealed biological and pathological differences across these seven diverse models, indicating a highly sensitive, robust, and scalable methodology for the quantification of distinct biliary forms. BiliQML is the first comprehensive machine-learning platform for biliary form analysis, adding much-needed morphologic context to standard immunofluorescence-based histology, and provides clinical and basic science researchers with a novel tool for the characterization of cholangiopathies.NEW & NOTEWORTHY BiliQML is the first comprehensive machine-learning platform for biliary form analysis in whole slide histopathological images. This platform provides clinical and basic science researchers with a novel tool for the improved quantification and characterization of biliary tract disorders.


Subject(s)
Liver , Supervised Machine Learning , Liver/pathology , Liver/metabolism , Animals , Mice , Biliary Tract/pathology , Biliary Tract/metabolism , Image Processing, Computer-Assisted/methods , Bile Ducts/pathology , Bile Ducts/metabolism , Bile Duct Diseases/pathology , Bile Duct Diseases/metabolism , Disease Models, Animal
3.
Hepatology ; 78(5): 1337-1351, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37021797

ABSTRACT

BACKGROUND AND AIMS: Paucity of intrahepatic bile ducts (BDs) is caused by various etiologies and often leads to cholestatic liver disease. For example, in patients with Alagille syndrome (ALGS), which is a genetic disease primarily caused by mutations in jagged 1 ( JAG1) , BD paucity often results in severe cholestasis and liver damage. However, no mechanism-based therapy exists to restore the biliary system in ALGS or other diseases associated with BD paucity. Based on previous genetic observations, we investigated whether postnatal knockdown of the glycosyltransferase gene protein O -glucosyltransferase 1 ( Poglut1) can improve the ALGS liver phenotypes in several mouse models generated by removing one copy of Jag1 in the germline with or without reducing the gene dosage of sex-determining region Y-box 9 in the liver. APPROACH AND RESULTS: Using an ASO established in this study, we show that reducing Poglut1 levels in postnatal livers of ALGS mouse models with moderate to profound biliary abnormalities can significantly improve BD development and biliary tree formation. Importantly, ASO injections prevent liver damage in these models without adverse effects. Furthermore, ASO-mediated Poglut1 knockdown improves biliary tree formation in a different mouse model with no Jag1 mutations. Cell-based signaling assays indicate that reducing POGLUT1 levels or mutating POGLUT1 modification sites on JAG1 increases JAG1 protein level and JAG1-mediated signaling, suggesting a likely mechanism for the observed in vivo rescue. CONCLUSIONS: Our preclinical studies establish ASO-mediated POGLUT1 knockdown as a potential therapeutic strategy for ALGS liver disease and possibly other diseases associated with BD paucity.


Subject(s)
Alagille Syndrome , Glycosyltransferases , Liver , Oligonucleotides, Antisense , Animals , Mice , Alagille Syndrome/genetics , Alagille Syndrome/metabolism , Alagille Syndrome/pathology , Bile Ducts, Intrahepatic/metabolism , Bile Ducts, Intrahepatic/pathology , Calcium-Binding Proteins/genetics , Cholestasis/genetics , Cholestasis/metabolism , Gene Silencing , Glucosyltransferases/genetics , Glucosyltransferases/metabolism , Glycosyltransferases/genetics , Glycosyltransferases/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Jagged-1 Protein/genetics , Jagged-1 Protein/metabolism , Liver/metabolism , Liver/pathology , Membrane Proteins/genetics , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/metabolism , Phenotype , Serrate-Jagged Proteins/genetics , Serrate-Jagged Proteins/metabolism
4.
Nature ; 557(7704): 247-251, 2018 05.
Article in English | MEDLINE | ID: mdl-29720662

ABSTRACT

Transdifferentiation is a complete and stable change in cell identity that serves as an alternative to stem-cell-mediated organ regeneration. In adult mammals, findings of transdifferentiation have been limited to the replenishment of cells lost from preexisting structures, in the presence of a fully developed scaffold and niche1. Here we show that transdifferentiation of hepatocytes in the mouse liver can build a structure that failed to form in development-the biliary system in a mouse model that mimics the hepatic phenotype of human Alagille syndrome (ALGS)2. In these mice, hepatocytes convert into mature cholangiocytes and form bile ducts that are effective in draining bile and persist after the cholestatic liver injury is reversed, consistent with transdifferentiation. These findings redefine hepatocyte plasticity, which appeared to be limited to metaplasia, that is, incomplete and transient biliary differentiation as an adaptation to cell injury, based on previous studies in mice with a fully developed biliary system3-6. In contrast to bile duct development7-9, we show that de novo bile duct formation by hepatocyte transdifferentiation is independent of NOTCH signalling. We identify TGFß signalling as the driver of this compensatory mechanism and show that it is active in some patients with ALGS. Furthermore, we show that TGFß signalling can be targeted to enhance the formation of the biliary system from hepatocytes, and that the transdifferentiation-inducing signals and remodelling capacity of the bile-duct-deficient liver can be harnessed with transplanted hepatocytes. Our results define the regenerative potential of mammalian transdifferentiation and reveal opportunities for the treatment of ALGS and other cholestatic liver diseases.


Subject(s)
Biliary Tract/cytology , Biliary Tract/metabolism , Cell Transdifferentiation , Hepatocytes/cytology , Transforming Growth Factor beta/metabolism , Alagille Syndrome/pathology , Animals , Bile Ducts/cytology , Bile Ducts/metabolism , Cell Proliferation , Epithelial Cells/cytology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Receptors, Notch/metabolism , Signal Transduction
5.
Hepatology ; 71(4): 1331-1349, 2020 04.
Article in English | MEDLINE | ID: mdl-31469182

ABSTRACT

BACKGROUND AND AIMS: Alagille syndrome (ALGS) is a multisystem developmental disorder characterized by bile duct (BD) paucity, caused primarily by haploinsufficiency of the Notch ligand jagged1. The course of the liver disease is highly variable in ALGS. However, the genetic basis for ALGS phenotypic variability is unknown. Previous studies have reported decreased expression of the transcription factor SOX9 (sex determining region Y-box 9) in late embryonic and neonatal livers of Jag1-deficient mice. Here, we investigated the effects of altering the Sox9 gene dosage on the severity of liver disease in an ALGS mouse model. APPROACH AND RESULTS: Conditional removal of one copy of Sox9 in Jag1+/- livers impairs the biliary commitment of cholangiocytes and enhances the inflammatory reaction and liver fibrosis. Loss of both copies of Sox9 in Jag1+/- livers further worsens the phenotypes and results in partial lethality. Ink injection experiments reveal impaired biliary tree formation in the periphery of P30 Jag1+/- livers, which is improved by 5 months of age. Sox9 heterozygosity worsens the P30 biliary tree phenotype and impairs the partial recovery in 5-month-old animals. Notably, Sox9 overexpression improves BD paucity and liver phenotypes in Jag1+/- mice without ectopic hepatocyte-to-cholangiocyte transdifferentiation or long-term liver abnormalities. Notch2 expression in the liver is increased following Sox9 overexpression, and SOX9 binds the Notch2 regulatory region in the liver. Histological analysis shows a correlation between the level and pattern of SOX9 expression in the liver and outcome of the liver disease in patients with ALGS. CONCLUSIONS: Our results establish Sox9 as a dosage-sensitive modifier of Jag1+/- liver phenotypes with a permissive role in biliary development. Our data further suggest that liver-specific increase in SOX9 levels is a potential therapeutic approach for BD paucity in ALGS.


Subject(s)
Alagille Syndrome/genetics , Alagille Syndrome/pathology , Liver/pathology , SOX9 Transcription Factor/genetics , Animals , Bile Ducts/abnormalities , Cell Transdifferentiation/genetics , Child , Child, Preschool , Disease Models, Animal , Hepatocytes/cytology , Heterozygote , Humans , Infant , Jagged-1 Protein/genetics , Liver/abnormalities , Liver/metabolism , Mice , Mice, Inbred C57BL , Receptors, Notch/genetics , Receptors, Notch/metabolism , Severity of Illness Index , Signal Transduction
6.
Am J Physiol Gastrointest Liver Physiol ; 314(5): G547-G558, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29388792

ABSTRACT

Biliary atresia (BA), a neonatal liver disease, is characterized by obstruction of extrahepatic bile ducts with subsequent cholestasis, inflammation, and progressive liver fibrosis. To gain insights into the pathophysiology of BA, we focused attention on GATA6, a transcription factor implicated in biliary development. Early in fetal development GATA6 expression is evident in cholangiocytes and hepatocytes, but by late gestation it is extinguished in hepatocytes. Utilizing a unique set of BA liver samples collected before and after successful portoenterostomy (PE), we found that GATA6 expression is markedly upregulated in hepatocytes of patients with BA compared with healthy and cholestatic disease controls. This upregulation is recapitulated in two murine models simulating bile duct obstruction and intrahepatic bile ductule expansion. GATA6 expression in BA livers correlates with two established negative prognostic indicators (age at PE, degree of intrahepatic bile ductule expansion) and decreases after normalization of serum bilirubin by PE. GATA6 expression in BA livers correlates with expression of known regulators of cholangiocyte differentiation ( JAGGED1, HNF1ß, and HNF6). These same genes are upregulated after enforced expression of GATA6 in human hepatocyte cell models. In conclusion, GATA6 is a novel marker and a putative driver of hepatocyte-cholangiocyte metaplasia in BA, and its expression in hepatocytes is downregulated after successful PE. NEW & NOTEWORTHY A pathological hallmark in the liver of patients with biliary atresia is ductular reaction, an expansion of new bile ductules that are thought to arise from conversion of mature hepatocytes. Here, we show that transcription factor GATA6 is a marker and potential driver of hepatocyte ductal metaplasia in biliary atresia. Hepatocyte GATA6 expression is elevated in biliary atresia, correlates with bile duct expansion, and decreases after successful portoenterostomy.


Subject(s)
Bile Ducts, Extrahepatic/pathology , Biliary Atresia , GATA6 Transcription Factor/metabolism , Hepatocytes/metabolism , Liver/pathology , Animals , Biliary Atresia/metabolism , Biliary Atresia/pathology , Biliary Atresia/surgery , Biomarkers/metabolism , Cell Transdifferentiation/physiology , Cholestasis/metabolism , Disease Models, Animal , Hep G2 Cells , Humans , Metaplasia/metabolism , Metaplasia/pathology , Mice , Portoenterostomy, Hepatic/methods
7.
Arterioscler Thromb Vasc Biol ; 35(7): 1597-605, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26023079

ABSTRACT

OBJECTIVE: Calcific aortic valve disease (CAVD) is a significant cardiovascular disorder, and controversy exists as to whether it is primarily a dystrophic or osteogenic process in vivo. In this study, we sought to clarify the mechanism of CAVD by assessing a genetic mutation, Notch1 heterozygosity, which leads to CAVD with 100% penetrance in humans. APPROACH AND RESULTS: Murine immortalized Notch1(+/-) aortic valve interstitial cells (AVICs) were isolated and expanded in vitro. Molecular signaling of wild-type and Notch1(+/-) AVICs were compared to identify changes in pathways that have been linked to CAVD-transforming growth factor-ß1/bone morphogenetic protein, mitogen-activated protein kinase, and phosphoinositide 3-kinase/protein kinase B-and assessed for calcification potential. Additionally, AVIC mechanobiology was studied in a physiologically relevant, dynamic mechanical environment (10% cyclic strain) to investigate differences in responses between the cell types. We found that Notch1(+/-) AVICs resembled a myofibroblast-like phenotype expressing higher amounts of cadherin-11, a known mediator of dystrophic calcification, and decreased Runx2, a known osteogenic marker. We determined that cadherin-11 expression is regulated by Akt activity, and inhibition of Akt phosphorylation significantly reduced cadherin-11 expression. Moreover, in the presence of cyclic strain, Notch1(+/-) AVICs exhibited significantly upregulated phosphorylation of Akt at Ser473 and smooth muscle α-actin expression, indicative of a fully activated myofibroblast. Finally, these Notch1-mediated alterations led to enhanced dystrophic calcific nodule formation. CONCLUSIONS: This study presents novel insights in our understanding of Notch1-mediated CAVD by demonstrating that the mutation leads to AVICs that are fully activated myofibroblasts, resulting in dystrophic, but not osteogenic, calcification.


Subject(s)
Aortic Valve Stenosis/genetics , Aortic Valve Stenosis/metabolism , Aortic Valve/pathology , Calcinosis/genetics , Calcinosis/metabolism , Mechanotransduction, Cellular/genetics , Mutation , Myofibroblasts/metabolism , Receptor, Notch1/genetics , Animals , Aortic Valve/metabolism , Cadherins/metabolism , Core Binding Factor Alpha 1 Subunit/metabolism , Humans , MAP Kinase Kinase 2/metabolism , Mice , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism
8.
Dev Dyn ; 244(3): 497-506, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25370311

ABSTRACT

Background The liver is a complex organ with a variety of tissue components that require a precise architecture for optimal function of metabolic and detoxification processes. As a result of the delicate orchestration required between the various hepatic tissues, it is not surprising that impairment of hepatic function can be caused by a variety of factors leading to chronic liver disease. Results Despite the growing rate of chronic liver disease, there are currently few effective treatment options besides orthotopic liver transplantation. Better therapeutic options reside in the potential for genetic and cellular therapies that promote progenitor cell activation aiding de novo epithelial and vascular regeneration, cell replacement, or population of bioartificial hepatic devices. In order to explore this area of new therapeutic potential, it is crucial to understand the factors that promote hepatic function through regulating cell identities and tissue architecture. Conclusions In this commentary, we review the signals regulating liver cell fates during development and regeneration and highlight the importance of patterning the hepatic vascular systems to set the groundwork for the macro and micro hepatic architecture of the epithelium.


Subject(s)
Liver Regeneration/physiology , Liver/blood supply , Liver/embryology , Neovascularization, Physiologic/physiology , Animals , Humans
9.
Curr Opin Organ Transplant ; 21(6): 581-587, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27755169

ABSTRACT

PURPOSE OF REVIEW: Although the liver possesses a unique, innate ability to regenerate through mass compensation, transplantation remains the only therapy when damage outpaces regeneration, or liver metabolic capacity is irreversibly impacted. Recent insight from developmental biology has greatly influenced the advancement of alternative options to transplantation in these settings. RECENT FINDINGS: Factors known to direct liver cell specification, expansion, and differentiation have been used to generate hepatocyte-like cells from stem and somatic cells for developing cell therapies. Additionally, interactions between hepatic epithelial and nonepithelial cells key to establishing hepatic architecture have been used in tissue engineering approaches to advance self-organizing hepatic organoids and bioartificial liver devices. Simultaneously, recent clinically applicable advances in human hepatocyte transplantation and promotion of innate hepatic regeneration have been limited. SUMMARY: Although mature hepatocytes have the potential to bridge to, or replace whole organ transplantation, limits in the ability to obtain healthy cells, stabilize in-vitro expansion, cryopreserve, and alleviate rejection, still exist. Alternative sources for generating hepatocytes hold promise for cell therapy and tissue engineering. These may allow generation of autologous or universal donor cells that eliminate the need for immunosuppression; however, limits exist regarding hepatocyte maturity and efficacy at liver repopulation, as well as applicability to human chronic liver disease.


Subject(s)
Liver Regeneration , Organogenesis , Tissue Engineering , Animals , Hepatocytes , Humans , Liver/cytology , Liver/physiology , Liver Diseases/therapy , Liver, Artificial
10.
Am J Pathol ; 184(5): 1479-88, 2014 May.
Article in English | MEDLINE | ID: mdl-24631193

ABSTRACT

The potential for intrahepatic bile duct (IHBD) regeneration in patients with bile duct insufficiency diseases is poorly understood. Notch signaling and Hnf6 have each been shown to be important for the morphogenesis of IHBDs in mice. One congenital pediatric liver disease characterized by reduced numbers of IHBDs, Alagille syndrome, is associated with mutations in Notch signaling components. Therefore, we investigated whether liver cell plasticity could contribute to IHBD regeneration in mice with disruptions in Notch signaling and Hnf6. We studied a mouse model of bile duct insufficiency with liver epithelial cell-specific deficiencies in Hnf6 and Rbpj, a mediator of canonical Notch signaling. Albumin-Cre Hnf6(flox/flox)Rbpj(flox/flox) mice initially developed no peripheral bile ducts. The evolving postnatal liver phenotype was analyzed using IHBD resin casting, immunostaining, and serum chemistry. With age, Albumin-Cre Hnf6(flox/flox)Rbpj(flox/flox) mice mounted a ductular reaction extending through the hepatic tissue and then regenerated communicating peripheral IHBD branches. Rbpj and Hnf6 were determined to remain absent from biliary epithelial cells constituting the ductular reaction and the regenerated peripheral IHBDs. We report the expression of Sox9, a marker of biliary epithelial cells, in cells expressing hepatocyte markers. Tissue analysis indicates that reactive ductules did not arise directly from preexisting hilar IHBDs. We conclude that liver cell plasticity is competent for regeneration of IHBDs independent of Notch signaling via Rbpj and Hnf6.


Subject(s)
Bile Ducts, Intrahepatic/physiology , Hepatocyte Nuclear Factor 6/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Receptors, Notch/metabolism , Regeneration/physiology , Animals , Epithelial Cells/metabolism , Hepatocyte Nuclear Factor 6/deficiency , Hepatocytes/metabolism , Imaging, Three-Dimensional , Immunoglobulin J Recombination Signal Sequence-Binding Protein/deficiency , Immunohistochemistry , Keratin-19/metabolism , Mice, Knockout , Plant Lectins/metabolism , Portal Vein/metabolism , SOX9 Transcription Factor/metabolism
11.
Hum Mol Genet ; 21(6): 1374-83, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22156581

ABSTRACT

JAGGED1 mutations cause Alagille syndrome, comprising a constellation of clinical findings, including biliary, cardiac and craniofacial anomalies. Jagged1, a ligand in the Notch signaling pathway, has been extensively studied during biliary and cardiac development. However, the role of JAGGED1 during craniofacial development is poorly understood. Patients with Alagille syndrome have midface hypoplasia giving them a characteristic 'inverted V' facial appearance. This study design determines the requirement of Jagged1 in the cranial neural crest (CNC) cells, which encompass the majority of mesenchyme present during craniofacial development. Furthermore, with this approach, we identify the autonomous and non-autonomous requirement of Jagged1 in a cell lineage-specific approach during midface development. Deleting Jagged1 in the CNC using Wnt1-cre; Jag1 Flox/Flox recapitulated the midfacial hypoplasia phenotype of Alagille syndrome. The Wnt1-cre; Jag1 Flox/Flox mice die at postnatal day 30 due to inability to masticate owing to jaw misalignment and poor occlusion. The etiology of midfacial hypoplasia in the Wnt1-cre; Jag1 Flox/Flox mice was a consequence of reduced cellular proliferation in the midface, aberrant vasculogenesis with decreased productive vessel branching and reduced extracellular matrix by hyaluronic acid staining, all of which are associated with midface anomalies and aberrant craniofacial growth. Deletion of Notch1 from the CNC using Wnt1-cre; Notch1 F/F mice did not recapitulate the midface hypoplasia of Alagille syndrome. These data demonstrate the requirement of Jagged1, but not Notch1, within the midfacial CNC population during development. Future studies will investigate the mechanism in which Jagged1 acts in a cell autonomous and cell non-autonomous manner.


Subject(s)
Alagille Syndrome/physiopathology , Calcium-Binding Proteins/physiology , Craniofacial Abnormalities/etiology , Intercellular Signaling Peptides and Proteins/physiology , Membrane Proteins/physiology , Neural Crest/metabolism , Receptor, Notch1/physiology , Animals , Blotting, Western , Cells, Cultured , Craniofacial Abnormalities/metabolism , Craniofacial Abnormalities/pathology , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Female , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Integrases/metabolism , Jagged-1 Protein , Mesoderm/metabolism , Mesoderm/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Morphogenesis , Neural Crest/cytology , Phenotype , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Serrate-Jagged Proteins , Wnt1 Protein/genetics , Wnt1 Protein/metabolism
12.
Am J Physiol Gastrointest Liver Physiol ; 306(10): G849-62, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24650547

ABSTRACT

Vascular endothelial growth factor (VEGF) is crucial for vascular development in several organs. However, the specific contribution of epithelial-VEGF signaling in the liver has not been tested. We used a mouse model to specifically delete Vegf from the liver epithelial lineages during midgestational development and assessed the cell identities and architectures of epithelial and endothelial tissues. We find that without epithelial-derived VEGF, the zonal endothelial and hepatocyte cell identities are altered. We also find decreased portal vein and hepatic artery branching coincident with an increase in hepatic hypoxia postnatally. Together, these data indicate that VEGF secreted from the hepatic epithelium is required for normal differentiation of cells and establishment of three-dimensional vascular branching and zonal architectures in both epithelial and endothelial hepatic tissues.


Subject(s)
Hepatocytes/metabolism , Liver/embryology , Vascular Endothelial Growth Factor A/metabolism , Animals , Carbamoyl-Phosphate Synthase (Ammonia)/biosynthesis , Cell Differentiation/drug effects , Endothelial Cells/metabolism , Endothelium/metabolism , Glutamate-Ammonia Ligase/biosynthesis , Hepatocytes/pathology , Hypoxia/pathology , Liver/blood supply , Liver/physiopathology , Mice , Mice, Knockout
13.
bioRxiv ; 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38659780

ABSTRACT

Background and Aims: Since the role of hepatic progenitor cells (HPCs) constituting ductular reactions in pathogenesis remains ambiguous, we aimed to establish the in vivo cause-and-effect relationship between HPCs and angiogenesis, a process associated with chronic liver disease progression. We previously demonstrated that peritumoral ductules are associated with angiogenesis in liver tumors and forkhead box L1 (Foxl1)- expressing murine HPCs secrete angiogenic factors in vitro. Therefore, we hypothesized that HPCs are capable of remodeling the vascular microenvironment and this function of HPCs is dependent on recombination signal binding protein for immunoglobulin kappa J region (RBPJ), a key effector of the Notch signaling pathway. Approach and Results: We generated HPC-specific Rbpj conditional knockout mice using Foxl1-Cre and treated them with the 3,5-diethoxycarbonyl-1,4-dihydrocollidine-supplemented diet to induce cholestatic liver disease. Knockout mice displayed significant reduction of HPC proliferation and ductular reactions as well as attenuated vascular and fibrotic areas compared to control mice. Assessment of vascular endothelial growth factor A-positive areas in vivo and the effects of Rbpj shRNAs in vitro indicated that Rbpj knockout in HPCs reduces the total number of angiogenic factor-expressing cells rather than affecting angiogenic factor expression within HPCs. Single-nucleus RNA sequencing analysis indicated that conditional Rbpj knockout in HPCs induces transcriptional changes in endothelial cells and alters expression of genes involved in various functions of the endothelium. Conclusion: Our findings indicate that HPCs regulate endothelial responses to cholestatic liver disease and Rbpj deletion in HPCs attenuates these responses, identifying novel targets for modulating angiogenesis during disease progression.

14.
bioRxiv ; 2024 May 21.
Article in English | MEDLINE | ID: mdl-38826326

ABSTRACT

Fibrosing cholangiopathies, including biliary atresia and primary sclerosing cholangitis, involve immune-mediated bile duct epithelial injury and hepatic bile acid (BA) retention (cholestasis). Regulatory T-cells (Tregs) can prevent auto-reactive lymphocyte activation, yet the effects of BA on this CD4 lymphocyte subset are unknown. Gene regulatory networks for hepatic CD4 lymphocytes in a murine cholestasis model revealed Tregs are polarized to Th17 during cholestasis. Following bile duct ligation, Stat3 deletion in CD4 lymphocytes preserved hepatic Treg responses. While pharmacological reduction of hepatic BA in MDR2-/- mice prompted Treg expansion and diminished liver injury, this improvement subsided with Treg depletion. A cluster of patients diagnosed with biliary atresia showed both increased hepatic Treg responses and improved 2-year native liver survival, supporting that Tregs might protect against neonatal bile duct obstruction. Together, these findings suggest liver BA determine Treg function and should be considered as a therapeutic target to restore protective hepatic immune responses.

15.
Hepatology ; 55(1): 233-43, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21898486

ABSTRACT

UNLABELLED: Notch signaling and hepatocyte nuclear factor-6 (HNF-6) are two genetic factors known to affect lineage commitment in the bipotential hepatoblast progenitor cell (BHPC) population. A genetic interaction involving Notch signaling and HNF-6 in mice has been inferred through separate experiments showing that both affect BHPC specification and bile duct morphogenesis. To define the genetic interaction between HNF-6 and Notch signaling in an in vivo mouse model, we examined the effects of BHPC-specific loss of HNF-6 alone and within the background of BHPC-specific loss of recombination signal binding protein immunoglobulin kappa J (RBP-J), the common DNA-binding partner of all Notch receptors. Isolated loss of HNF-6 in this mouse model fails to demonstrate a phenotypic variance in bile duct development compared to control. However, when HNF-6 loss is combined with RBP-J loss, a phenotype consisting of cholestasis, hepatic necrosis, and fibrosis is observed that is more severe than the phenotype seen with Notch signaling loss alone. This phenotype is associated with significant intrahepatic biliary system abnormalities, including an early decrease in biliary epithelial cells, evolving to ductular proliferation and a decrease in the density of communicating peripheral bile duct branches. In this in vivo model, simultaneous loss of both HNF-6 and RBP-J results in down-regulation of both HNF-1ß and Sox9 (sex determining region Y-related HMG box transcription factor 9). CONCLUSION: HNF-6 and Notch signaling interact in vivo to control expression of downstream mediators essential to the normal development of the intrahepatic biliary system. This study provides a model to investigate genetic interactions of factors important to intrahepatic bile duct development and their effect on cholestatic liver disease phenotypes.


Subject(s)
Bile Ducts, Intrahepatic/growth & development , Bile Ducts, Intrahepatic/physiology , Hepatocyte Nuclear Factor 6/genetics , Hepatocytes/physiology , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Receptors, Notch/metabolism , Signal Transduction/genetics , Animals , Bile Ducts, Intrahepatic/cytology , Cell Lineage/physiology , Cholestasis/genetics , Cholestasis/metabolism , Cholestasis/physiopathology , Gene Expression Regulation, Developmental/physiology , Hepatocyte Nuclear Factor 1-beta/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Immunoglobulin kappa-Chains/genetics , Integrases/genetics , Mice , Mice, Inbred Strains , Mice, Knockout , SOX9 Transcription Factor/genetics
16.
EMBO J ; 27(7): 1017-28, 2008 Apr 09.
Article in English | MEDLINE | ID: mdl-18354499

ABSTRACT

Histone deacetylase 3 (Hdac3) is an enzymatic component of transcriptional repression complexes recruited by the nuclear hormone receptors. Inactivation of Hdac3 in cancer cell lines triggered apoptosis, and removal of Hdac3 in the germ line of mice caused embryonic lethality. Therefore, we deleted Hdac3 in the postnatal mouse liver. These mice developed hepatomegaly, which was the result of hepatocyte hypertrophy, and these morphological changes coincided with significant imbalances between carbohydrate and lipid metabolism. Loss of Hdac3 triggered changes in gene expression consistent with inactivation of repression mediated by nuclear hormone receptors. Loss of Hdac3 also increased the levels of Ppar gamma2, and treatment of these mice with a Ppar gamma antagonist partially reversed the lipid accumulation in the liver. In addition, gene expression analysis identified mammalian target of rapamycin signalling as being activated after deletion of Hdac3, and inhibition by rapamycin affected the accumulation of neutral lipids in Hdac3-null livers. Thus, Hdac3 regulates metabolism through multiple signalling pathways in the liver, and deletion of Hdac3 disrupts normal metabolic homeostasis.


Subject(s)
Gene Deletion , Gene Regulatory Networks , Histone Deacetylases/deficiency , Liver/enzymology , Acetylation/drug effects , Animals , Animals, Newborn , Cholesterol/biosynthesis , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Gene Regulatory Networks/drug effects , Hepatocytes/drug effects , Hepatocytes/enzymology , Histone Deacetylases/metabolism , Homeostasis/drug effects , Hypertrophy , Hypoglycemia/enzymology , Insulin/blood , Integrases/metabolism , Lipid Metabolism/drug effects , Liver/drug effects , Liver/pathology , Mice , Organ Specificity/drug effects , PPAR gamma/metabolism , Protein Kinases/metabolism , TOR Serine-Threonine Kinases
18.
Bioorg Med Chem ; 20(6): 1869-77, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22261025

ABSTRACT

Extract prepared from Xenopus eggs represents a cell-free system that has been shown to recapitulate a multitude of cellular processes, including cell cycle regulation, DNA replication/repair, and cytoskeletal dynamics. In addition, this system has been used to successfully reconstitute the Wnt pathway. Xenopus egg extract, which can be biochemically manipulated, offers an ideal medium in which small molecule screening can be performed in near native milieu. Thus, the use of Xenopus egg extract for small molecule screening represents an ideal bridge between targeted and phenotypic screening approaches. This review focuses on the use of this system for small molecules modulators of major signal transduction pathways (Notch, Hedgehog, and Wnt) that are critical for the development of the early Xenopus embryo. We describe the properties of Xenopus egg extract and our own high throughput screen for small molecules that modulate the Wnt pathway using this cell-free system. We propose that Xenopus egg extract could similarly be adapted for screening for modulators of the Notch and Hedgehog pathways.


Subject(s)
Drug Evaluation, Preclinical/methods , Ovum/chemistry , Ovum/drug effects , Signal Transduction/drug effects , Small Molecule Libraries/pharmacology , Xenopus laevis/metabolism , Animals , Hedgehog Proteins/metabolism , High-Throughput Screening Assays/methods , Receptors, Notch/metabolism , Small Molecule Libraries/chemistry , Wnt Signaling Pathway/drug effects
19.
Hepatology ; 51(4): 1391-400, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20069650

ABSTRACT

UNLABELLED: Alagille syndrome, a chronic hepatobiliary disease, is characterized by paucity of intrahepatic bile ducts (IHBDs). To determine the impact of Notch signaling specifically on IHBD arborization, we studied the influence of both chronic gain and loss of Notch function on the intact three-dimensional IHBD structure using a series of mutant mouse models and a resin casting method. Impaired Notch signaling in bipotential hepatoblast progenitor cells (BHPCs) dose-dependently decreased the density of peripheral IHBDs, whereas activation of Notch1 results in an increased density of peripheral IHBDs. Although Notch2 has a dominant role in IHBD formation, there is also a redundant role for other Notch receptors in determining the density of peripheral IHBDs. Because changes in IHBD density do not appear to be due to changes in cellular proliferation of bile duct progenitors, we suggest that Notch plays a permissive role in cooperation with other factors to influence lineage decisions of BHPCs and sustain peripheral IHBDs. CONCLUSION: There is a threshold requirement for Notch signaling at multiple steps, including IHBD tubulogenesis and maintenance, during hepatic development that determines the density of three-dimensional peripheral IHBD architecture.


Subject(s)
Bile Ducts, Intrahepatic/embryology , Receptor, Notch1/physiology , Receptor, Notch2/physiology , Signal Transduction/physiology , Animals , Keratin-19/analysis , Mice , Mice, Knockout
20.
Stem Cell Reports ; 16(2): 309-323, 2021 02 09.
Article in English | MEDLINE | ID: mdl-33450190

ABSTRACT

The bile salt export pump (BSEP) is responsible for the export of bile acid from hepatocytes. Impaired transcellular transport of bile acids in hepatocytes with mutations in BSEP causes cholestasis. Compensatory mechanisms to regulate the intracellular bile acid concentration in human hepatocytes with BSEP deficiency remain unclear. To define pathways that prevent cytotoxic accumulation of bile acid in hepatocytes, we developed a human induced pluripotent stem cell-based model of isogenic BSEP-deficient hepatocytes in a Transwell culture system. Induced hepatocytes (i-Heps) exhibited defects in the apical export of bile acids but maintained a low intracellular bile acid concentration by inducing basolateral export. Modeling the autoregulation of bile acids on hepatocytes, we found that BSEP-deficient i-Heps suppressed de novo bile acid synthesis using the FXR pathway via basolateral uptake and export without apical export. These observations inform the development of therapeutic targets to reduce the overall bile acid pool in patients with BSEP deficiency.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 11/physiology , Bile Acids and Salts/metabolism , Hepatocytes/cytology , Hepatocytes/metabolism , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Biological Transport , CRISPR-Cas Systems , Cell Culture Techniques/methods , Cell Differentiation , Cells, Cultured , Gene Editing , Humans , Models, Biological , Mutation
SELECTION OF CITATIONS
SEARCH DETAIL