Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 194
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 175(6): 1607-1619.e15, 2018 11 29.
Article in English | MEDLINE | ID: mdl-30500539

ABSTRACT

In the healthy adult liver, most hepatocytes proliferate minimally. However, upon physical or chemical injury to the liver, hepatocytes proliferate extensively in vivo under the direction of multiple extracellular cues, including Wnt and pro-inflammatory signals. Currently, liver organoids can be generated readily in vitro from bile-duct epithelial cells, but not hepatocytes. Here, we show that TNFα, an injury-induced inflammatory cytokine, promotes the expansion of hepatocytes in 3D culture and enables serial passaging and long-term culture for more than 6 months. Single-cell RNA sequencing reveals broad expression of hepatocyte markers. Strikingly, in vitro-expanded hepatocytes engrafted, and significantly repopulated, the injured livers of Fah-/- mice. We anticipate that tissue repair signals can be harnessed to promote the expansion of otherwise hard-to-culture cell-types, with broad implications.


Subject(s)
Antigens, Differentiation/biosynthesis , Cell Culture Techniques , Cell Proliferation/drug effects , Hepatocytes/metabolism , Spheroids, Cellular/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Animals , Cell Line, Transformed , Hep G2 Cells , Hepatocytes/transplantation , Human Umbilical Vein Endothelial Cells , Humans , Liver/injuries , Liver/metabolism , Mice, Knockout , Spheroids, Cellular/transplantation , Time Factors
2.
Nature ; 613(7943): 355-364, 2023 01.
Article in English | MEDLINE | ID: mdl-36599988

ABSTRACT

DNA methylation is a fundamental epigenetic mark that governs gene expression and chromatin organization, thus providing a window into cellular identity and developmental processes1. Current datasets typically include only a fraction of methylation sites and are often based either on cell lines that underwent massive changes in culture or on tissues containing unspecified mixtures of cells2-5. Here we describe a human methylome atlas, based on deep whole-genome bisulfite sequencing, allowing fragment-level analysis across thousands of unique markers for 39 cell types sorted from 205 healthy tissue samples. Replicates of the same cell type are more than 99.5% identical, demonstrating the robustness of cell identity programmes to environmental perturbation. Unsupervised clustering of the atlas recapitulates key elements of tissue ontogeny and identifies methylation patterns retained since embryonic development. Loci uniquely unmethylated in an individual cell type often reside in transcriptional enhancers and contain DNA binding sites for tissue-specific transcriptional regulators. Uniquely hypermethylated loci are rare and are enriched for CpG islands, Polycomb targets and CTCF binding sites, suggesting a new role in shaping cell-type-specific chromatin looping. The atlas provides an essential resource for study of gene regulation and disease-associated genetic variants, and a wealth of potential tissue-specific biomarkers for use in liquid biopsies.


Subject(s)
Cells , DNA Methylation , Epigenesis, Genetic , Epigenome , Humans , Cell Line , Cells/classification , Cells/metabolism , Chromatin/genetics , Chromatin/metabolism , CpG Islands/genetics , DNA/genetics , DNA/metabolism , Embryonic Development , Enhancer Elements, Genetic , Organ Specificity , Polycomb-Group Proteins/metabolism , Whole Genome Sequencing
3.
Cell ; 153(2): 283-4, 2013 Apr 11.
Article in English | MEDLINE | ID: mdl-23582318

ABSTRACT

In the liver, the hepatocyte mass is kept stable through a tight balance between hepatocyte death and proliferation that is frequently lost upon acute or chronic liver injury. Wuestefeld et al. (2013) now identify a potentially druggable target that enhances hepatocyte proliferation and promotes liver regeneration, thereby preventing liver failure.

4.
Hepatology ; 79(5): 1088-1097, 2024 May 01.
Article in English | MEDLINE | ID: mdl-37824086

ABSTRACT

BACKGROUND AND AIMS: Hepatocyte transplantation for genetic liver diseases has several potential advantages over gene therapy. However, the low efficiency of cell engraftment has limited its clinical implementation. This problem could be overcome by selectively expanding transplanted donor cells until they replace enough of the liver mass to achieve therapeutic benefit. We previously described a gene therapy method to selectively expand hepatocytes deficient in cytochrome p450 reductase (Cypor) using acetaminophen (APAP). Because Cypor is required for the transformation of APAP to a hepatotoxic metabolite, Cypor-deficient cells are protected from toxicity and are able to expand following APAP-induced liver injury. Here, we apply this selection system to correct a mouse model of phenylketonuria by cell transplantation. APPROACH AND RESULTS: Hepatocytes from a wild-type donor animal were edited in vitro to create Cypor deficiency and then transplanted into phenylketonuric animals. Following selection with APAP, blood phenylalanine concentrations were fully normalized and remained stable following APAP withdrawal. Cypor-deficient hepatocytes expanded from < 1% to ~14% in corrected animals, and they showed no abnormalities in blood chemistries, liver histology, or drug metabolism. CONCLUSIONS: We conclude that APAP-mediated selection of transplanted hepatocytes is a potential therapeutic for phenylketonuria with long-term efficacy and a favorable safety profile.


Subject(s)
Chemical and Drug Induced Liver Injury , Phenylketonurias , Mice , Animals , Acetaminophen , Hepatocytes/metabolism , Liver/pathology , Phenylketonurias/metabolism , Phenylketonurias/pathology , Disease Models, Animal , Chemical and Drug Induced Liver Injury/pathology , Mice, Inbred C57BL
5.
Nature ; 567(7746): 43-48, 2019 03.
Article in English | MEDLINE | ID: mdl-30760930

ABSTRACT

Cell-identity switches, in which terminally differentiated cells are converted into different cell types when stressed, represent a widespread regenerative strategy in animals, yet they are poorly documented in mammals. In mice, some glucagon-producing pancreatic α-cells and somatostatin-producing δ-cells become insulin-expressing cells after the ablation of insulin-secreting ß-cells, thus promoting diabetes recovery. Whether human islets also display this plasticity, especially in diabetic conditions, remains unknown. Here we show that islet non-ß-cells, namely α-cells and pancreatic polypeptide (PPY)-producing γ-cells, obtained from deceased non-diabetic or diabetic human donors, can be lineage-traced and reprogrammed by the transcription factors PDX1 and MAFA to produce and secrete insulin in response to glucose. When transplanted into diabetic mice, converted human α-cells reverse diabetes and continue to produce insulin even after six months. Notably, insulin-producing α-cells maintain expression of α-cell markers, as seen by deep transcriptomic and proteomic characterization. These observations provide conceptual evidence and a molecular framework for a mechanistic understanding of in situ cell plasticity as a treatment for diabetes and other degenerative diseases.


Subject(s)
Diabetes Mellitus/pathology , Diabetes Mellitus/therapy , Glucagon-Secreting Cells/cytology , Glucagon-Secreting Cells/metabolism , Glucose/metabolism , Insulin/metabolism , Islets of Langerhans/pathology , Animals , Biomarkers/analysis , Cell Lineage/drug effects , Cellular Reprogramming/drug effects , Diabetes Mellitus/immunology , Diabetes Mellitus/metabolism , Disease Models, Animal , Female , Glucagon/metabolism , Glucagon-Secreting Cells/drug effects , Glucagon-Secreting Cells/transplantation , Glucose/pharmacology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Islets of Langerhans/drug effects , Islets of Langerhans/immunology , Islets of Langerhans/metabolism , Maf Transcription Factors, Large/genetics , Maf Transcription Factors, Large/metabolism , Male , Mice , Organ Specificity/drug effects , Pancreatic Polypeptide/metabolism , Pancreatic Polypeptide-Secreting Cells/cytology , Pancreatic Polypeptide-Secreting Cells/drug effects , Pancreatic Polypeptide-Secreting Cells/metabolism , Proteomics , Sequence Analysis, RNA , Trans-Activators/genetics , Trans-Activators/metabolism , Transcriptome , Transduction, Genetic
6.
Dev Biol ; 504: 120-127, 2023 12.
Article in English | MEDLINE | ID: mdl-37813160

ABSTRACT

The current gold-standard for genetic lineage tracing in transgenic mice is based on cell-type specific expression of Cre recombinase. As an alternative, we developed a cell-type specific CRISPR/spCas9 system for lineage tracing. This method relies on RNA polymerase II promoter driven self-cleaving guide RNAs (scgRNA) to achieve tissue-specificity. To demonstrate proof-of-principle for this approach a transgenic mouse was generated harbouring a knock-in of a scgRNA into the Cytokeratin 14 (Krt14) locus. Krt14 expression marks the stem cells of squamous epithelium in the skin and oral mucosa. The scgRNA targets a Stop cassette preceding a fluorescent reporter in the Ai9-tdtomato mouse. Ai9-tdtomato reporter mice harbouring this allele along with a spCas9 transgene demonstrated precise marking of the Krt14 lineage. We conclude that RNA polymerase II promoter driven scgRNAs enable the use of CRISPR/spCas9 for genetic lineage tracing.


Subject(s)
CRISPR-Cas Systems , RNA Polymerase II , Animals , Mice , CRISPR-Cas Systems/genetics , Integrases/genetics , Keratin-14/genetics , Keratin-14/metabolism , Mice, Transgenic , Promoter Regions, Genetic/genetics , RNA Polymerase II/genetics , RNA Polymerase II/metabolism
7.
Hepatology ; 78(2): 486-502, 2023 08 01.
Article in English | MEDLINE | ID: mdl-36037289

ABSTRACT

BACKGROUND AND AIMS: Assessing mammalian gene function in vivo has traditionally relied on manipulation of the mouse genome in embryonic stem cells or perizygotic embryos. These approaches are time-consuming and require extensive breeding when simultaneous mutations in multiple genes is desired. The aim of this study is to introduce a rapid in vivo multiplexed editing (RIME) method and provide proof of concept of this system. APPROACH AND RESULTS: RIME, a system wherein CRISPR/caspase 9 technology, paired with adeno-associated viruses (AAVs), permits the inactivation of one or more genes in the adult mouse liver. The method is quick, requiring as little as 1 month from conceptualization to knockout, and highly efficient, enabling editing in >95% of target cells. To highlight its use, we used this system to inactivate, alone or in combination, genes with functions spanning metabolism, mitosis, mitochondrial maintenance, and cell proliferation. CONCLUSIONS: RIME enables the rapid, efficient, and inexpensive analysis of multiple genes in the mouse liver in vivo .


Subject(s)
CRISPR-Cas Systems , Gene Editing , Mice , Animals , Gene Editing/methods , Clustered Regularly Interspaced Short Palindromic Repeats , Liver , Mammals
8.
Pediatr Blood Cancer ; 71(8): e31030, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38733122

ABSTRACT

Fanconi anemia (FA) is a disease caused by defective deoxyribonucleic acid (DNA) repair that manifests as bone marrow failure, cancer predisposition, and developmental defects. We previously reported that monotherapy with either metformin (MET) or oxymetholone (OXM) improved peripheral blood (PB) counts and the number and functionality of bone marrow hematopoietic stem progenitor cells (HSPCs) number in Fancd2-/- mice. To evaluate whether the combination treatment of these drugs has a synergistic effect to prevent bone marrow failure in FA, we treated cohorts of Fancd2-/- mice and wildtype controls with either MET alone, OXM alone, MET+OXM, or placebo diet from age 3 weeks to 18 months. The OXM treated animals showed modest improvements in blood parameters including platelet count (p = .01) and hemoglobin levels (p < .05). In addition, the percentage of quiescent hematopoietic stem cell (HSC) (LSK [Lin-Sca+c-Kit+]) was significantly increased (p = .001) by long-term treatment with MET alone. The combination of metformin and oxymetholone did not result in a significant synergistic effect in any hematopoietic parameter. Gene expression analysis of liver tissue from these animals showed that some of the expression changes caused by Fancd2 deletion were partially normalized by metformin treatment. Importantly, no adverse effects of the individual or combination therapies were observed, despite the long-term administration. We conclude that androgen therapy is not a contraindication to concurrent metformin administration in clinical trials. HIGHLIGHTS: Long-term coadministration of metformin in combination with oxymetholone is well tolerated by Fancd2-/- mice. Hematopoietic stem cell quiescence in mutant mice was enhanced by treatment with metformin alone. Metformin treatment caused a partial normalization of gene expression in the livers of mutant mice.


Subject(s)
Disease Models, Animal , Drug Therapy, Combination , Fanconi Anemia , Metformin , Oxymetholone , Animals , Metformin/pharmacology , Metformin/administration & dosage , Mice , Fanconi Anemia/drug therapy , Fanconi Anemia Complementation Group D2 Protein/genetics , Mice, Knockout , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism
9.
Int J Cancer ; 153(1): 183-196, 2023 07 01.
Article in English | MEDLINE | ID: mdl-36912284

ABSTRACT

Fanconi anemia (FA) is a heritable malformation, bone marrow failure and cancer predisposition syndrome that confers an exceptionally high risk of squamous carcinomas. These carcinomas originate in epithelia lining the mouth, proximal esophagus, vulva and anus: their origins are not understood, and no effective ways have been identified to prevent or delay their appearance. Many FA-associated carcinomas are also therapeutically challenging: they may be multi-focal and stage-advanced at diagnosis, and most individuals with FA cannot tolerate standard-of-care systemic therapies such as DNA cross-linking drugs or ionizing radiation due to constitutional DNA damage hypersensitivity. We developed the Fanconi Anemia Cancer Cell Line Resource (FA-CCLR) to foster new work on the origins, treatment and prevention of FA-associated carcinomas. The FA-CCLR consists of Fanconi-isogenic head and neck squamous cell carcinoma (HNSCC) cell line pairs generated from five individuals with FA-associated HNSCC, and five individuals with sporadic HNSCC. Sporadic, isogenic HNSCC cell line pairs were generated in parallel with FA patient-derived isogenic cell line pairs to provide comparable experimental material to use to identify cell and molecular phenotypes driven by germline or somatic loss of Fanconi pathway function, and the subset of these FA-dependent phenotypes that can be modified, complemented or suppressed. All 10 FANC-isogenic cell line pairs are available to academic, non-profit and industry investigators via the "Fanconi Anemia Research Materials" Resource and Repository at Oregon Health & Sciences University, Portland OR.


Subject(s)
Carcinoma, Squamous Cell , Fanconi Anemia , Head and Neck Neoplasms , Female , Humans , Squamous Cell Carcinoma of Head and Neck , Fanconi Anemia/genetics , Fanconi Anemia/complications , Fanconi Anemia/pathology , Translational Science, Biomedical , Head and Neck Neoplasms/genetics , Carcinoma, Squamous Cell/genetics , Cell Line, Tumor
10.
Am J Hum Genet ; 105(6): 1237-1253, 2019 12 05.
Article in English | MEDLINE | ID: mdl-31785787

ABSTRACT

We report an early-onset autosomal-recessive neurological disease with cerebellar atrophy and lysosomal dysfunction. We identified bi-allelic loss-of-function (LoF) variants in Oxidative Resistance 1 (OXR1) in five individuals from three families; these individuals presented with a history of severe global developmental delay, current intellectual disability, language delay, cerebellar atrophy, and seizures. While OXR1 is known to play a role in oxidative stress resistance, its molecular functions are not well established. OXR1 contains three conserved domains: LysM, GRAM, and TLDc. The gene encodes at least six transcripts, including some that only consist of the C-terminal TLDc domain. We utilized Drosophila to assess the phenotypes associated with loss of mustard (mtd), the fly homolog of OXR1. Strong LoF mutants exhibit late pupal lethality or pupal eclosion defects. Interestingly, although mtd encodes 26 transcripts, severe LoF and null mutations can be rescued by a single short human OXR1 cDNA that only contains the TLDc domain. Similar rescue is observed with the TLDc domain of NCOA7, another human homolog of mtd. Loss of mtd in neurons leads to massive cell loss, early death, and an accumulation of aberrant lysosomal structures, similar to what we observe in fibroblasts of affected individuals. Our data indicate that mtd and OXR1 are required for proper lysosomal function; this is consistent with observations that NCOA7 is required for lysosomal acidification.


Subject(s)
Atrophy/pathology , Cerebellar Diseases/pathology , Lysosomes/pathology , Mitochondrial Proteins/metabolism , Nervous System Diseases/pathology , Oxidative Stress , Adolescent , Adult , Animals , Atrophy/genetics , Atrophy/metabolism , Cerebellar Diseases/genetics , Cerebellar Diseases/metabolism , Child , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Lysosomes/metabolism , Male , Mitochondrial Proteins/genetics , Nervous System Diseases/genetics , Nervous System Diseases/metabolism , Pedigree , Phenotype , Young Adult
11.
Hepatology ; 74(1): 444-457, 2021 07.
Article in English | MEDLINE | ID: mdl-33423324

ABSTRACT

BACKGROUND AND AIMS: Following liver injury, a fraction of hepatocytes adopt features of biliary epithelial cells (BECs) in a process known as biliary reprogramming. The aim of this study was to elucidate the molecular events accompanying this dramatic shift in cellular identity. APPROACH AND RESULTS: We applied the techniques of bulk RNA-sequencing (RNA-seq), single-cell RNA-seq, and assay for transposase-accessible chromatin with high-throughput sequencing to define the epigenetic and transcriptional changes associated with biliary reprogramming. In addition, we examined the role of TGF-ß signaling by profiling cells undergoing reprogramming in mice with hepatocyte-specific deletion in the downstream TGF-ß signaling component mothers against decapentaplegic homolog 4 (Smad4). Biliary reprogramming followed a stereotyped pattern of altered gene expression consisting of robust induction of biliary genes and weaker repression of hepatocyte genes. These changes in gene expression were accompanied by corresponding modifications at the chromatin level. Although some reprogrammed cells had molecular features of "fully differentiated" BECs, most lacked some biliary characteristics and retained some hepatocyte characteristics. Surprisingly, single-cell analysis of Smad4 mutant mice revealed a dramatic increase in reprogramming. CONCLUSION: Hepatocytes undergo widespread chromatin and transcriptional changes during biliary reprogramming, resulting in epigenetic and gene expression profiles that are similar to, but distinct from, native BECs. Reprogramming involves a progressive accumulation of biliary molecular features without discrete intermediates. Paradoxically, canonical TGF-ß signaling through Smad4 appears to constrain biliary reprogramming, indicating that TGF-ß can either promote or inhibit biliary differentiation depending on which downstream components of the pathway are engaged. This work has implications for the formation of BECs and bile ducts in the adult liver.


Subject(s)
Cell Plasticity/genetics , Liver Regeneration/genetics , Liver/physiology , Animals , Bile Ducts/cytology , Cell Differentiation/genetics , Epigenesis, Genetic , Epithelial Cells/physiology , Hepatocytes/physiology , Hepatocytes/transplantation , Humans , Liver/cytology , Male , Mice , Mice, Transgenic , RNA-Seq , Single-Cell Analysis , Smad4 Protein/genetics
12.
Immunity ; 39(5): 874-84, 2013 Nov 14.
Article in English | MEDLINE | ID: mdl-24184056

ABSTRACT

Severe sepsis remains a poorly understood systemic inflammatory condition with high mortality rates and limited therapeutic options in addition to organ support measures. Here we show that the clinically approved group of anthracyclines acts therapeutically at a low dose regimen to confer robust protection against severe sepsis in mice. This salutary effect is strictly dependent on the activation of DNA damage response and autophagy pathways in the lung, as demonstrated by deletion of the ataxia telangiectasia mutated (Atm) or the autophagy-related protein 7 (Atg7) specifically in this organ. The protective effect of anthracyclines occurs irrespectively of pathogen burden, conferring disease tolerance to severe sepsis. These findings demonstrate that DNA damage responses, including the ATM and Fanconi Anemia pathways, are important modulators of immune responses and might be exploited to confer protection to inflammation-driven conditions, including severe sepsis.


Subject(s)
Anthracyclines/pharmacology , Anti-Bacterial Agents/pharmacology , DNA Repair/drug effects , Lung/drug effects , Peritonitis/drug therapy , Sepsis/prevention & control , Adenoviridae Infections/immunology , Animals , Anthracyclines/therapeutic use , Anti-Bacterial Agents/therapeutic use , Ataxia Telangiectasia Mutated Proteins/deficiency , Ataxia Telangiectasia Mutated Proteins/physiology , Autophagy-Related Protein 7 , Cecum/injuries , DNA Damage , Epirubicin/administration & dosage , Epirubicin/pharmacology , Epirubicin/therapeutic use , Fanconi Anemia Complementation Group D2 Protein/physiology , Inflammation , Inflammation Mediators/analysis , Injections, Intraperitoneal , Lung/metabolism , Meropenem , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/deficiency , Microtubule-Associated Proteins/physiology , Organ Specificity , Peritonitis/etiology , Peritonitis/genetics , Peritonitis/immunology , Peritonitis/physiopathology , Respiratory Tract Infections/immunology , Shock, Septic/prevention & control , Thienamycins/therapeutic use , Whole-Body Irradiation
13.
Mol Ther ; 29(10): 2898-2909, 2021 10 06.
Article in English | MEDLINE | ID: mdl-34461297

ABSTRACT

Recombinant adeno-associated viral (rAAV) vectors are considered promising tools for gene therapy directed at the liver. Whereas rAAV is thought to be an episomal vector, its single-stranded DNA genome is prone to intra- and inter-molecular recombination leading to rearrangements and integration into the host cell genome. Here, we ascertained the integration frequency of rAAV in human hepatocytes transduced either ex vivo or in vivo and subsequently expanded in a mouse model of xenogeneic liver regeneration. Chromosomal rAAV integration events and vector integrity were determined using the capture-PacBio sequencing approach, a long-read next-generation sequencing method that has not previously been used for this purpose. Chromosomal integrations were found at a surprisingly high frequency of 1%-3% both in vitro and in vivo. Importantly, most of the inserted rAAV sequences were heavily rearranged and were accompanied by deletions of the host genomic sequence at the integration site.


Subject(s)
Dependovirus/physiology , Hepatocytes/transplantation , Liver Regeneration , Animals , Cells, Cultured , Chromosomes/genetics , Dependovirus/genetics , Disease Models, Animal , Genetic Therapy , Genetic Vectors/administration & dosage , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Mice , Transduction, Genetic , Virus Integration
14.
Mol Ther ; 29(2): 680-690, 2021 02 03.
Article in English | MEDLINE | ID: mdl-33554867

ABSTRACT

Adeno-associated virus (AAV) integrates into host genomes at low frequency, but when integration occurs in oncogenic hotspots it can cause hepatocellular carcinoma (HCC). Given the possibility of recombinant AAV (rAAV) integration leading to HCC, common causes of liver inflammation like non-alcoholic fatty liver disease (NAFLD) may increase the risk of rAAV-induced HCC. A rAAV targeting the oncogenic mouse Rian locus was used, and as expected led to HCC in all mice infected as neonates, likely due to growth-related hepatocyte proliferation in young mice. Mice infected with rAAV as adults did not develop HCC unless they were fed a diet leading to NAFLD, with increased inflammation and hepatocyte proliferation. Female mice were less susceptible to rAAV-induced HCC, and male mice with NAFLD treated with estrogen exhibited less inflammation and immune exhaustion associated with oncogenesis compared to those without estrogen. Adult NAFLD mice infected with a non-targeted control rAAV also developed HCC, though only half as frequently as those exposed to the Rian targeted rAAV. This study shows that adult mice exposed to rAAV gene therapy in the context of chronic liver disease developed HCC at high frequency, and thus warrants further study in humans given the high prevalence of NAFLD in the population.


Subject(s)
Carcinoma, Hepatocellular/etiology , Dependovirus/genetics , Genetic Therapy/adverse effects , Genetic Vectors/genetics , Liver Diseases/complications , Liver Diseases/etiology , Liver Neoplasms/etiology , Animals , Carcinoma, Hepatocellular/diagnosis , Disease Models, Animal , Genetic Therapy/methods , Incidence , Liver Diseases/pathology , Liver Neoplasms/diagnosis , Mice
15.
Hepatology ; 72(2): 656-670, 2020 08.
Article in English | MEDLINE | ID: mdl-31785104

ABSTRACT

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


Subject(s)
Benzoates/pharmacokinetics , Benzylamines/pharmacokinetics , Cholesterol/metabolism , Hepatocytes/metabolism , Lipoproteins/metabolism , Liver X Receptors/agonists , Liver/metabolism , Animals , Hepatocytes/transplantation , Humans , Liver/surgery , Male , Mice , Mice, Knockout
16.
Gastroenterology ; 157(2): 552-568, 2019 08.
Article in English | MEDLINE | ID: mdl-31029706

ABSTRACT

BACKGROUND & AIMS: Acetaminophen (APAP) overdose is a major cause of acute liver failure (ALF). Mitochondrial SH3BP5 (also called SAB) and phosphorylation of c-Jun N-terminal kinase (JNK) mediate the hepatotoxic effects of APAP. We investigated the involvement of steroidogenic acute regulatory protein (STARD1), a mitochondrial cholesterol transporter, in this process and sensitization by valproic acid (VPA), which depletes glutathione and stimulates steroidogenesis. METHODS: Nonfasted C57BL/6J mice (control) and mice with liver-specific deletion of STARD1 (Stard1ΔHep), SAB (SabΔHep), or JNK1 and JNK2 (Jnk1+2ΔHep) were given VPA with or without APAP. Liver tissues were collected and analyzed by histology and immunohistochemistry and for APAP metabolism, endoplasmic reticulum (ER) stress, and mitochondrial function. Adult human hepatocytes were transplanted into Fah-/-/Rag2-/-/Il2rg-/-/NOD (FRGN) mice to create mice with humanized livers. RESULTS: Administration of VPA before administration of APAP increased the severity of liver damage in control mice. The combination of VPA and APAP increased expression of CYP2E1, formation of NAPQI-protein adducts, and depletion of glutathione from liver tissues of control mice, resulting in ER stress and the upregulation of STARD1. Livers from control mice given VPA and APAP accumulated cholesterol in the mitochondria and had sustained mitochondrial depletion of glutathione and mitochondrial dysfunction. Inhibition of ER stress, by administration of tauroursodeoxycholic acid to control mice, prevented upregulation of STARD1 in liver and protected the mice from hepatoxicity following administration of VPA and APAP. Administration of N-acetylcysteine to control mice prevented VPA- and APAP-induced ER stress and liver injury. Stard1ΔHep mice were resistant to induction of ALF by VPA and APAP, despite increased mitochondrial levels of glutathione and phosphorylated JNK; we made similar observations in fasted Stard1ΔHep mice given APAP alone. SabΔHep mice or Jnk1+2ΔHep mice did not develop ALF following administration of VPA and APAP. The ability of VPA to increase the severity of APAP-induced liver damage was observed in FRGN mice with humanized liver. CONCLUSIONS: In studies of mice, we found that upregulation of STARD1 following ER stress mediates APAP hepatoxicity via SH3BP5 and phosphorylation of JNK1 and JNK2.


Subject(s)
Acetaminophen/toxicity , Chemical and Drug Induced Liver Injury/pathology , Endoplasmic Reticulum Stress/drug effects , Hepatocytes/pathology , Phosphoproteins/metabolism , Adult , Animals , Chemical and Drug Induced Liver Injury/etiology , Disease Models, Animal , Drug Overdose/complications , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/transplantation , Humans , Lipogenesis/drug effects , Liver/cytology , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphoproteins/genetics , Steroids/metabolism , Transplantation Chimera , Up-Regulation , Valproic Acid/administration & dosage
17.
Nature ; 506(7488): 382-6, 2014 Feb 20.
Article in English | MEDLINE | ID: mdl-24390344

ABSTRACT

Recombinant adeno-associated viral (rAAV) vectors have shown early promise in clinical trials. The therapeutic transgene cassette can be packaged in different AAV capsid pseudotypes, each having a unique transduction profile. At present, rAAV capsid serotype selection for a specific clinical trial is based on effectiveness in animal models. However, preclinical animal studies are not always predictive of human outcome. Here, in an attempt to further our understanding of these discrepancies, we used a chimaeric human-murine liver model to compare directly the relative efficiency of rAAV transduction in human versus mouse hepatocytes in vivo. As predicted from preclinical and clinical studies, rAAV2 vectors functionally transduced mouse and human hepatocytes at equivalent but relatively low levels. However, rAAV8 vectors, which are very effective in many animal models, transduced human hepatocytes rather poorly-approximately 20 times less efficiently than mouse hepatocytes. In light of the limitations of the rAAV vectors currently used in clinical studies, we used the same murine chimaeric liver model to perform serial selection using a human-specific replication-competent viral library composed of DNA-shuffled AAV capsids. One chimaeric capsid composed of five different parental AAV capsids was found to transduce human primary hepatocytes at high efficiency in vitro and in vivo, and provided species-selected transduction in primary liver, cultured cells and a hepatocellular carcinoma xenograft model. This vector is an ideal clinical candidate and a reagent for gene modification of human xenotransplants in mouse models of human diseases. More importantly, our results suggest that humanized murine models may represent a more precise approach for both selecting and evaluating clinically relevant rAAV serotypes for gene therapeutic applications.


Subject(s)
Dependovirus/genetics , Genetic Therapy/methods , Genetic Vectors/genetics , Heterografts/metabolism , Liver/metabolism , Transduction, Genetic/methods , Transgenes/genetics , Animals , Capsid/metabolism , Capsid Proteins/genetics , Capsid Proteins/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cells, Cultured , Chimera/genetics , Chimera/metabolism , Clinical Trials as Topic , Dependovirus/isolation & purification , Disease Models, Animal , Female , Hepatocytes/cytology , Hepatocytes/metabolism , Hepatocytes/pathology , Hepatocytes/transplantation , Humans , Liver/cytology , Liver/pathology , Male , Mice , Species Specificity
18.
Pediatr Blood Cancer ; 66(1): e27460, 2019 01.
Article in English | MEDLINE | ID: mdl-30255556

ABSTRACT

BACKGROUND: Fanconi anemia is an inherited bone marrow failure disorder associated with a high incidence of leukemia and solid tumors. Currently, no interventions to prevent or delay the formation of solid tumors are available. PROCEDURE: Two of the most important hallmarks of Fanconi anemia are inflammation and oxidative stress. In this study, we administrated the antioxidant atorvastatin and the anti-inflammatory drug celecoxib to cohorts of Fancd2-/- /Trp53+/- mice, a model of Fanconi anemia. Treatment started at weaning and continued until the mice developed a palpable mass or suffered from >20% weight loss. Tumor samples and selected tissues were subjected to histopathological examination. χ2 test was performed to analyze tumor incidence, and Kaplan-Meier survival curves were evaluated with log-rank test. In addition, a small cohort of mice was monitored for the safety of the drugs. RESULTS: The combined oral administration of both drugs significantly delayed tumor onset in Fancd2-/- /Trp53+/- mice. Specifically, the treatment delayed the onset of ovarian tumors in Fancd2-/- /Trp53+/- mice and increased the mean ovarian tumor-free survival time by 17%, whereas this combinatorial drug regimen did not have a significant effect on other tumor types. In addition, no detrimental effects on hematopoiesis from the drug treatment were observed during a 12-month safety monitoring. CONCLUSIONS: The data presented here suggest that a combination of atorvastatin and celecoxib may be a good candidate for chemoprevention in Fanconi anemia.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Disease Models, Animal , Fanconi Anemia Complementation Group D2 Protein/physiology , Fanconi Anemia/drug therapy , Tumor Suppressor Protein p53/physiology , Animals , Atorvastatin/administration & dosage , Celecoxib/administration & dosage , Fanconi Anemia/pathology , Female , Male , Mice , Mice, Knockout , Survival Rate
19.
Nature ; 494(7436): 247-50, 2013 Feb 14.
Article in English | MEDLINE | ID: mdl-23354049

ABSTRACT

The Wnt target gene Lgr5 (leucine-rich-repeat-containing G-protein-coupled receptor 5) marks actively dividing stem cells in Wnt-driven, self-renewing tissues such as small intestine and colon, stomach and hair follicles. A three-dimensional culture system allows long-term clonal expansion of single Lgr5(+) stem cells into transplantable organoids (budding cysts) that retain many characteristics of the original epithelial architecture. A crucial component of the culture medium is the Wnt agonist RSPO1, the recently discovered ligand of LGR5. Here we show that Lgr5-lacZ is not expressed in healthy adult liver, however, small Lgr5-LacZ(+) cells appear near bile ducts upon damage, coinciding with robust activation of Wnt signalling. As shown by mouse lineage tracing using a new Lgr5-IRES-creERT2 knock-in allele, damage-induced Lgr5(+) cells generate hepatocytes and bile ducts in vivo. Single Lgr5(+) cells from damaged mouse liver can be clonally expanded as organoids in Rspo1-based culture medium over several months. Such clonal organoids can be induced to differentiate in vitro and to generate functional hepatocytes upon transplantation into Fah(-/-) mice. These findings indicate that previous observations concerning Lgr5(+) stem cells in actively self-renewing tissues can also be extended to damage-induced stem cells in a tissue with a low rate of spontaneous proliferation.


Subject(s)
Hepatocytes/cytology , Hepatocytes/metabolism , Receptors, G-Protein-Coupled/metabolism , Regeneration , Stem Cells/cytology , Stem Cells/metabolism , Wnt Signaling Pathway , Alleles , Animals , Bile Ducts/cytology , Bile Ducts/metabolism , Cell Lineage , Clone Cells/cytology , Clone Cells/metabolism , Culture Media/chemistry , Culture Media/metabolism , Disease Models, Animal , Female , Gene Knock-In Techniques , Hepatocytes/pathology , Hydrolases/deficiency , Hydrolases/genetics , Liver/cytology , Liver/metabolism , Liver/pathology , Liver Diseases/metabolism , Liver Diseases/pathology , Male , Mice , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Organoids/cytology , Organoids/transplantation , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/genetics , Thrombospondins/deficiency , Thrombospondins/genetics , Thrombospondins/metabolism , Tyrosinemias/metabolism , Tyrosinemias/pathology
20.
Mol Ther ; 26(5): 1327-1342, 2018 05 02.
Article in English | MEDLINE | ID: mdl-29550076

ABSTRACT

Direct lineage reprogramming can convert readily available cells in the body into desired cell types for cell replacement therapy. This is usually achieved through forced activation or repression of lineage-defining factors or pathways. In particular, reprogramming toward the pancreatic ß cell fate has been of great interest in the search for new diabetes therapies. It has been suggested that cells from various endodermal lineages can be converted to ß-like cells. However, it is unclear how closely induced cells resemble endogenous pancreatic ß cells and whether different cell types have the same reprogramming potential. Here, we report in vivo reprogramming of pancreatic ductal cells through intra-ductal delivery of an adenoviral vector expressing the transcription factors Pdx1, Neurog3, and Mafa. Induced ß-like cells are mono-hormonal, express genes essential for ß cell function, and correct hyperglycemia in both chemically and genetically induced diabetes models. Compared with intrahepatic ducts and hepatocytes treated with the same vector, pancreatic ducts demonstrated more rapid activation of ß cell transcripts and repression of donor cell markers. This approach could be readily adapted to humans through a commonly performed procedure, endoscopic retrograde cholangiopancreatography (ERCP), and provides potential for cell replacement therapy in type 1 diabetes patients.


Subject(s)
Cellular Reprogramming , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Pancreatic Ducts/cytology , ATPases Associated with Diverse Cellular Activities/genetics , ATPases Associated with Diverse Cellular Activities/metabolism , Adenoviridae/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Biomarkers , Cellular Reprogramming/genetics , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation , Genetic Vectors/genetics , Hepatocytes/metabolism , Humans , Insulin/genetics , Insulin/metabolism , Maf Transcription Factors, Large/genetics , Maf Transcription Factors, Large/metabolism , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Single-Cell Analysis , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL