Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 231
Filter
1.
Methods Mol Biol ; 2837: 199-206, 2024.
Article in English | MEDLINE | ID: mdl-39044086

ABSTRACT

Chimeric mouse models with a humanized liver (Hu-HEP mice) provide a unique tool to study human hepatotropic virus diseases, including viral infection, viral pathogenesis, and anti-viral therapy. Here, we describe a detailed protocol for studying hepatitis B infection in NRG-derived fumarylacetoacetate hydrolase (FAH) knockout mice repopulated with human hepatocytes (FRG-Hu HEP mice). The procedures include (1) maintenance and genotyping of the FRG mice, (2) intrasplenic injection of primary human hepatocytes (PHH), (3) 2-(2-nitro-4-fluoromethylbenzoyl)-1,3-cyclohexanedione (NTBC) drug reduction cycling to improve human hepatocyte repopulation, (4) human albumin detection, and (5) HBV infection and detection. The method is simple and allows for highly reproducible generation of FRG-Hu HEP mice for HBV infection and therapy investigations.


Subject(s)
Disease Models, Animal , Hepatitis B virus , Hepatitis B , Hepatocytes , Hydrolases , Liver , Mice, Knockout , Animals , Humans , Mice , Hydrolases/genetics , Hydrolases/metabolism , Hydrolases/deficiency , Hepatitis B/virology , Hepatitis B virus/genetics , Liver/virology , Liver/pathology , Hepatocytes/virology , Hepatocytes/transplantation , Mice, Inbred NOD , Interleukin Receptor Common gamma Subunit/genetics , Interleukin Receptor Common gamma Subunit/deficiency , Chimera , Cyclohexanones , Nitrobenzoates
2.
Cells ; 10(8)2021 08 10.
Article in English | MEDLINE | ID: mdl-34440809

ABSTRACT

Mitochondria play a key role in metabolic transitions involved in the reprogramming of somatic cells into induced pluripotent stem cells (iPSCs), but the underlying molecular mechanisms remain largely unexplored. To obtain new insight into the mechanisms of cellular reprogramming, we studied the role of FAH domain-containing protein 1 (FAHD1) in the reprogramming of murine embryonic fibroblasts (MEFs) into iPSCs and their subsequent differentiation into neuronal cells. MEFs from wild type (WT) and Fahd1-knock-out (KO) mice were reprogrammed into iPSCs and characterized for alterations in metabolic parameters and the expression of marker genes indicating mitochondrial biogenesis. Fahd1-KO MEFs showed a higher reprogramming efficiency accompanied by a significant increase in glycolytic activity as compared to WT. We also observed a strong increase of mitochondrial DNA copy number and expression of biogenesis marker genes in Fahd1-KO iPSCs relative to WT. Neuronal differentiation of iPSCs was accompanied by increased expression of mitochondrial biogenesis genes in both WT and Fahd1-KO neurons with higher expression in Fahd1-KO neurons. Together these observations establish a role of FAHD1 as a potential negative regulator of reprogramming and add additional insight into mechanisms by which FAHD1 modulates mitochondrial functions.


Subject(s)
Cellular Reprogramming , Glycolysis/physiology , Hydrolases/genetics , Animals , Cell Differentiation , Cell Line , DNA, Mitochondrial/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Hydrolases/deficiency , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/genetics , Mitochondria/metabolism , Neurons/cytology , Neurons/metabolism , Oxidative Phosphorylation
3.
Arch Pediatr ; 28(4): 338-341, 2021 May.
Article in English | MEDLINE | ID: mdl-33858731

ABSTRACT

The main clinical features of tyrosinemia type 1 usually appear in the first months of life, including fever, diarrhea, vomiting, liver involvement, growth failure, and renal proximal tubulopathy with subsequent hypophosphatemic rickets. An early diagnosis is crucial in order to provide specific management and to prevent complications. Here, we report on two cases referred primarily to pediatric nephrologists for the diagnosis of "neonatal tubulopathy" and management of "X-linked hypophosphatemia (XLH)," respectively. Our aim is to emphasize that (1) even a mixed tubulopathy can reveal tyrosinemia, and (2) tyrosinemia is a classic differential diagnosis of XLH that should not be forgotten, especially in the era of the anti-FGF23 burosumab.


Subject(s)
Familial Hypophosphatemic Rickets/diagnosis , Genetic Diseases, Inborn , Hydrolases/deficiency , Kidney Tubules, Proximal/physiopathology , Tyrosinemias/diagnosis , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Child, Preschool , Disease Management , Fanconi Syndrome , Female , Humans , Infant, Newborn
4.
Cells ; 10(2)2021 02 10.
Article in English | MEDLINE | ID: mdl-33578760

ABSTRACT

Adenosine diphosphate ribosylation (ADP-ribosylation; ADPr), the addition of ADP-ribose moieties onto proteins and nucleic acids, is a highly conserved modification involved in a wide range of cellular functions, from viral defence, DNA damage response (DDR), metabolism, carcinogenesis and neurobiology. Here we study MACROD1 and MACROD2 (mono-ADP-ribosylhydrolases 1 and 2), two of the least well-understood ADPr-mono-hydrolases. MACROD1 has been reported to be largely localized to the mitochondria, while the MACROD2 genomic locus has been associated with various neurological conditions such as autism, attention deficit hyperactivity disorder (ADHD) and schizophrenia; yet the potential significance of disrupting these proteins in the context of mammalian behaviour is unknown. Therefore, here we analysed both Macrod1 and Macrod2 gene knockout (KO) mouse models in a battery of well-defined, spontaneous behavioural testing paradigms. Loss of Macrod1 resulted in a female-specific motor-coordination defect, whereas Macrod2 disruption was associated with hyperactivity that became more pronounced with age, in combination with a bradykinesia-like gait. These data reveal new insights into the importance of ADPr-mono-hydrolases in aspects of behaviour associated with both mitochondrial and neuropsychiatric disorders.


Subject(s)
Behavior, Animal , Carboxylic Ester Hydrolases/deficiency , DNA Repair Enzymes/deficiency , Hydrolases/deficiency , Animals , Body Weight , Carboxylic Ester Hydrolases/metabolism , DNA Repair Enzymes/metabolism , Female , Gait , Gene Knockout Techniques , Genotype , Hydrolases/metabolism , Male , Mice, Knockout , Motor Activity , Reproducibility of Results
5.
Gastroenterology ; 158(4): 1044-1057.e17, 2020 03.
Article in English | MEDLINE | ID: mdl-31759059

ABSTRACT

BACKGROUND & AIMS: Liver regeneration is impaired in mice with hepatocyte-specific deficiencies in microRNA (miRNA) processing, but it is not clear which miRNAs regulate this process. We developed a high-throughput screen to identify miRNAs that regulate hepatocyte repopulation after toxic liver injury using fumarylacetoacetate hydrolase-deficient mice. METHODS: We constructed plasmid pools encoding more than 30,000 tough decoy miRNA inhibitors (hairpin nucleic acids designed to specifically inhibit interactions between miRNAs and their targets) to target hepatocyte miRNAs in a pairwise manner. The plasmid libraries were delivered to hepatocytes in fumarylacetoacetate hydrolase-deficient mice at the time of liver injury via hydrodynamic tail-vein injection. Integrated transgene-containing transposons were quantified after liver repopulation via high-throughput sequencing. Changes in polysome-bound transcripts after miRNA inhibition were determined using translating ribosome affinity purification followed by high-throughput sequencing. RESULTS: Analyses of tough decoy abundance in hepatocyte genomic DNA and input plasmid pools identified several thousand miRNA inhibitors that were significantly depleted or increased after repopulation. We classified a subset of miRNA binding sites as those that have strong effects on liver repopulation, implicating the targeted hepatocyte miRNAs as regulators of this process. We then generated a high-content map of pairwise interactions between 171 miRNA-binding sites and identified synergistic and redundant effects. CONCLUSIONS: We developed a screen to identify miRNAs that regulate liver repopulation after injury in live mice.


Subject(s)
High-Throughput Nucleotide Sequencing/methods , Liver Regeneration/genetics , Liver/injuries , MicroRNAs/analysis , Animals , Chromosome Mapping , Hepatocytes/physiology , Hydrolases/deficiency , Liver/physiopathology , Mice , MicroRNAs/antagonists & inhibitors , Plasmids , RNA-Binding Proteins/analysis
6.
Nutrients ; 11(10)2019 Oct 16.
Article in English | MEDLINE | ID: mdl-31623189

ABSTRACT

Tyrosinemia type 1 (TT1) is a rare metabolic disease caused by a defect in the tyrosine degradation pathway. Neurocognitive deficiencies have been described in TT1 patients, that have, among others, been related to changes in plasma large neutral amino acids (LNAA) that could result in changes in brain LNAA and neurotransmitter concentrations. Therefore, this project aimed to investigate plasma and brain LNAA, brain neurotransmitter concentrations and behavior in C57 Bl/6 fumarylacetoacetate hydrolase deficient (FAH-/-) mice treated with 2-(2-nitro-4-trifluoromethylbenoyl)-1,3-cyclohexanedione (NTBC) and/or diet and wild-type mice. Plasma and brain tyrosine concentrations were clearly increased in all NTBC treated animals, even with diet (p < 0.001). Plasma and brain phenylalanine concentrations tended to be lower in all FAH-/- mice. Other brain LNAA, were often slightly lower in NTBC treated FAH-/- mice. Brain neurotransmitter concentrations were usually within a normal range, although serotonin was negatively correlated with brain tyrosine concentrations (p < 0.001). No clear behavioral differences between the different groups of mice could be found. To conclude, this is the first study measuring plasma and brain biochemistry in FAH-/- mice. Clear changes in plasma and brain LNAA have been shown. Further research should be done to relate the biochemical changes to neurocognitive impairments in TT1 patients.


Subject(s)
Amino Acids, Neutral/blood , Behavior, Animal/drug effects , Biogenic Monoamines/metabolism , Brain/drug effects , Cyclohexanones/pharmacology , Diet, Protein-Restricted , Enzyme Inhibitors/pharmacology , Hydroxyindoleacetic Acid/metabolism , Nitrobenzoates/pharmacology , Tyrosinemias/therapy , Animal Feed , Animals , Biomarkers/blood , Brain/metabolism , Brain/physiopathology , Disease Models, Animal , Female , Hydrolases/deficiency , Hydrolases/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Tyrosinemias/blood , Tyrosinemias/physiopathology , Tyrosinemias/psychology
7.
Proc Natl Acad Sci U S A ; 116(44): 22229-22236, 2019 10 29.
Article in English | MEDLINE | ID: mdl-31611405

ABSTRACT

Fumarylacetoacetate hydrolase (FAH) is the last enzyme in tyrosine catabolism, and mutations in the FAH gene are associated with hereditary tyrosinemia type I (HT1 or TYRSN1) in humans. In a behavioral screen of N-ethyl-N-nitrosourea mutagenized mice we identified a mutant line which we named "swingshift" (swst, MGI:3611216) with a nonsynonymous point mutation (N68S) in Fah that caused age-dependent disruption of sleep-wake patterns. Mice homozygous for the mutation had an earlier onset of activity (several hours before lights off) and a reduction in total activity and body weight when compared with wild-type or heterozygous mice. Despite abnormal behavioral entrainment to light-dark cycles, there were no differences in the period or phase of the central clock in mutant mice, indicating a defect downstream of the suprachiasmatic nucleus. Interestingly, these behavioral phenotypes became milder as the mice grew older and were completely rescued by the administration of NTBC [2-(2-nitro-4-trifluoromethylbenzoyl)-1,3-cyclohexanedione], an inhibitor of 4-hydroxyphenylpyruvate dioxygenase, which is upstream of FAH. Mechanistically, the swst mutation had no effect on the enzymatic activity of FAH, but rather promoted the degradation of the mutant protein. This led to reduced FAH protein levels and enzymatic activity in the liver and kidney (but not the brain or fibroblasts) of homozygous mice. In addition, plasma tyrosine-but not methionine, phenylalanine, or succinylacetone-increased in homozygous mice, suggesting that swst mutants provide a model of mild, chronic HT1.


Subject(s)
Circadian Rhythm , Hydrolases/genetics , Mutation , Sleep , Tyrosinemias/genetics , 4-Hydroxyphenylpyruvate Dioxygenase/antagonists & inhibitors , Animals , Cells, Cultured , Cyclohexanones/therapeutic use , Enzyme Inhibitors/therapeutic use , Enzyme Stability , HEK293 Cells , Homozygote , Humans , Hydrolases/deficiency , Hydrolases/metabolism , Kidney/metabolism , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Nitrobenzoates/therapeutic use , Organ Specificity , Suprachiasmatic Nucleus/metabolism , Tyrosinemias/drug therapy , Tyrosinemias/physiopathology
8.
Biochem Biophys Res Commun ; 516(2): 591-598, 2019 08 20.
Article in English | MEDLINE | ID: mdl-31239154

ABSTRACT

In the fumarylacetoacetate hydrolase deficient (Fah-/-) mouse, massive liver repopulation can be easily obtained after transplanted hepatocytes. Understanding the mechanisms of complete liver repopulation in Fah-/- mice will be useful for future clinical application. Here, we found that the endogenous hepatocytes in liver of Fah-/- mice undertook senescence during the time of tyrosinemia symptoms. Increase of senescent hepatocytes in Fah-/- mice provided proliferative advantage to the transplanted hepatocytes. Importantly, senescent hepatocytes upregulated the expression of extracellular matrix enzyme, contributing to degradation of extracellular matrix components and weakness of cell adhesion and connection. The liver exhibiting a loose architecture provided the space for the engraftment and expansion of transplanted hepatocytes. These findings underscore the underlying mechanisms of completed liver repopulation in Fah-/- mice. Senescence followed by loose hepatic parenchyma is a preconditioning for liver repopulation, which would be a promising strategy to achieve therapeutic liver repopulation in clinical settings.


Subject(s)
Cellular Senescence , Hepatocytes/cytology , Liver/cytology , Animals , Cell Cycle Checkpoints , Cell Proliferation , Cyclohexanones , Hepatocytes/transplantation , Hydrolases/deficiency , Hydrolases/metabolism , Mice , Nitrobenzoates
9.
Cytogenet Genome Res ; 158(1): 25-31, 2019.
Article in English | MEDLINE | ID: mdl-31055587

ABSTRACT

Diagnosing a complex genetic syndrome and correctly assigning the concomitant phenotypic traits to a well-defined clinical form is often a medical challenge. In this work, we report the analysis of a family with complex phenotypes, including microcephaly, intellectual disability, dysmorphic features, and polydactyly in the proband, with the aim of adding new aspects for obtaining a clear diagnosis. We performed array-comparative genomic hybridization and quantitative reverse transcriptase PCR (qRT-PCR) analyses. We identified a deletion of chromosome 20p12.1 involving the macrodomain containing 2/mono-ADP ribosylhydrolase 2 gene (MACROD2) in several members of the family. This gene is actually not associated with a specific syndrome but with congenital anomalies of multiple organs. qRT-PCR showed higher levels of a MACROD2 mRNA isoform in the individuals carrying the deletion. Our results, together with other data reported in the literature, support the hypothesis that the deletion in MACROD2 can affect correct embryonic development and that the presence of another associated event, such as epigenetic modifications at the MACROD2 locus, can influence the level of severity of the pathology.


Subject(s)
Abnormalities, Multiple/genetics , DNA Repair Enzymes/genetics , Hydrolases/genetics , Intellectual Disability/genetics , Kidney/abnormalities , Microcephaly/genetics , Pancreas/abnormalities , Polydactyly/genetics , Sequence Deletion , Adult , Chromosomes, Human, Pair 20/genetics , Chromosomes, Human, Pair 20/ultrastructure , Comparative Genomic Hybridization , DNA Repair Enzymes/deficiency , DNA Repair Enzymes/physiology , Embryonic Development/genetics , Female , Humans , Hydrolases/deficiency , Hydrolases/physiology , Male , Pedigree , Phenotype , Psychomotor Disorders/genetics
10.
Cell ; 177(3): 608-621.e12, 2019 04 18.
Article in English | MEDLINE | ID: mdl-30955891

ABSTRACT

Normal tissues accumulate genetic changes with age, but it is unknown if somatic mutations promote clonal expansion of non-malignant cells in the setting of chronic degenerative diseases. Exome sequencing of diseased liver samples from 82 patients revealed a complex mutational landscape in cirrhosis. Additional ultra-deep sequencing identified recurrent mutations in PKD1, PPARGC1B, KMT2D, and ARID1A. The number and size of mutant clones increased as a function of fibrosis stage and tissue damage. To interrogate the functional impact of mutated genes, a pooled in vivo CRISPR screening approach was established. In agreement with sequencing results, examination of 147 genes again revealed that loss of Pkd1, Kmt2d, and Arid1a promoted clonal expansion. Conditional heterozygous deletion of these genes in mice was also hepatoprotective in injury assays. Pre-malignant somatic alterations are often viewed through the lens of cancer, but we show that mutations can promote regeneration, likely independent of carcinogenesis.


Subject(s)
Liver Diseases/pathology , Liver/metabolism , Regeneration , Animals , Chronic Disease , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Humans , Hydrolases/deficiency , Hydrolases/genetics , Liver/pathology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Liver Diseases/genetics , Male , Mice , Mice, Knockout , Middle Aged , Mutation , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Regeneration/physiology , TRPP Cation Channels/genetics , TRPP Cation Channels/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Exome Sequencing
11.
DNA Cell Biol ; 38(4): 314-321, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30762444

ABSTRACT

The shortage of human hepatocytes continues to be a significant limitation for the widespread application of hepatocyte transplantation and bioartificial liver (BAL) support therapy. Recombinant activation gene 2 (Rag2) and fumarylacetoacetate hydrolase (Fah)-deficient mice could be highly repopulated with human hepatocytes. However, Fah/Rag2-deficient mice can only produce up to 1 × 108 human hepatocytes per mouse. We hypothesized that 2-10 × 1010 human hepatocytes can be produced per Fah/Rag2-deficient pig, which is an adequate supply for hepatocyte transplantation and BAL therapy. In a novel approach, we used stably transfected Cas9 cells and single-guide RNA adenoviruses containing fluorescent reporters to enrich porcine cells with Fah/Rag2 dual gene mutations. This resulted in the construction of Fah/Rag2 double knockout porcine iliac artery endothelial cells, which were subsequently used for generating Fah/Rag2-deficient pigs.


Subject(s)
Adenoviridae/genetics , CRISPR-Cas Systems/genetics , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Gene Knockout Techniques/methods , Hydrolases/deficiency , Hydrolases/genetics , Animals , Base Sequence , Cell Line , Mutation , Swine , Time Factors
12.
Adv Mater ; 30(52): e1805308, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30368954

ABSTRACT

mRNA-mediated protein replacement represents a promising concept for the treatment of liver disorders. Children born with fumarylacetoacetate hydrolase (FAH) mutations suffer from Hepatorenal Tyrosinemia Type 1 (HT-1) resulting in renal dysfunction, liver failure, neurological impairments, and cancer. Protein replacement therapy using FAH mRNA offers tremendous potential to cure HT-1, but is currently hindered by the development of effective mRNA carriers that can function in diseased livers. Structure-guided, rational optimization of 5A2-SC8 mRNA-loaded dendrimer lipid nanoparticles (mDLNPs) increases delivery potency of FAH mRNA, resulting in functional FAH protein and sustained normalization of body weight and liver function in FAH-/- knockout mice. Optimization using luciferase mRNA produces DLNP carriers that are efficacious at mRNA doses as low as 0.05 mg kg-1 in vivo. mDLNPs transfect > 44% of all hepatocytes in the liver, yield high FAH protein levels (0.5 mg kg-1 mRNA), and are well tolerated in a knockout mouse model with compromised liver function. Genetically engineered FAH-/- mice treated with FAH mRNA mDLNPs have statistically equivalent levels of TBIL, ALT, and AST compared to wild type C57BL/6 mice and maintain normal weight throughout the month-long course of treatment. This study provides a framework for the rational optimization of LNPs to improve delivery of mRNA broadly and introduces a specific and viable DLNP carrier with translational potential to treat genetic diseases of the liver.


Subject(s)
Dendrimers , Hydrolases/genetics , Liver/metabolism , Nanoparticles , RNA, Messenger/administration & dosage , Tyrosinemias/therapy , Animals , Dendrimers/chemistry , Disease Models, Animal , Genetic Therapy , Hydrolases/deficiency , Hydrolases/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Nanoparticles/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , Random Allocation , Tyrosinemias/metabolism
13.
Circ Res ; 123(1): 33-42, 2018 06 22.
Article in English | MEDLINE | ID: mdl-29572206

ABSTRACT

RATIONALE: Neutrophils likely contribute to the thrombotic complications of human atheromata. In particular, neutrophil extracellular traps (NETs) could exacerbate local inflammation and amplify and propagate arterial intimal injury and thrombosis. PAD4 (peptidyl arginine deiminase 4) participates in NET formation, but an understanding of this enzyme's role in atherothrombosis remains scant. OBJECTIVE: This study tested the hypothesis that PAD4 and NETs influence experimental atherogenesis and in processes implicated in superficial erosion, a form of plaque complication we previously associated with NETs. METHODS AND RESULTS: Bone marrow chimeric Ldlr deficient mice reconstituted with either wild-type or PAD4-deficient cells underwent studies that assessed atheroma formation or procedures designed to probe mechanisms related to superficial erosion. PAD4 deficiency neither retarded fatty streak formation nor reduced plaque size or inflammation in bone marrow chimeric mice that consumed an atherogenic diet. In contrast, either a PAD4 deficiency in bone marrow-derived cells or administration of DNaseI to disrupt NETs decreased the extent of arterial intimal injury in mice with arterial lesions tailored to recapitulate characteristics of human atheroma complicated by erosion. CONCLUSIONS: These results indicate that PAD4 from bone marrow-derived cells and NETs do not influence chronic experimental atherogenesis, but participate causally in acute thrombotic complications of intimal lesions that recapitulate features of superficial erosion.


Subject(s)
Extracellular Traps/physiology , Hydrolases/physiology , Plaque, Atherosclerotic/etiology , Thrombosis/etiology , Animals , Bone Marrow Transplantation , Carotid Artery Diseases/etiology , Carotid Artery Diseases/pathology , Cell Death , Deoxyribonuclease I/pharmacology , Extracellular Traps/drug effects , Humans , Hydrolases/deficiency , Male , Mice , Mice, Inbred C57BL , Neutrophils/physiology , Osteomyelitis/etiology , Plaque, Atherosclerotic/pathology , Protein-Arginine Deiminase Type 4 , Thrombosis/prevention & control , Tunica Intima/injuries
14.
Am J Physiol Renal Physiol ; 314(5): F809-F819, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29357426

ABSTRACT

We previously demonstrated that kidney peptidylarginine deiminase-4 (PAD4) plays a critical role in ischemic acute kidney injury (AKI) in mice by promoting renal tubular inflammation and neutrophil infiltration (Ham A, Rabadi M, Kim M, Brown KM, Ma Z, D'Agati V, Lee HT. Am J Physiol Renal Physiol 307: F1052-F1062, 2014). Although the role of PAD4 in granulocytes including neutrophils is well known, we surprisingly observed profound renal proximal tubular PAD4 induction after renal ischemia-reperfusion (I/R) injury. Here we tested the hypothesis that renal proximal tubular PAD4 rather than myeloid-cell lineage PAD4 plays a critical role in exacerbating ischemic AKI by utilizing mice lacking PAD4 in renal proximal tubules (PAD4ff PEPCK Cre mice) or in granulocytes (PAD4ff LysM Cre mice). Mice lacking renal proximal tubular PAD4 were significantly protected against ischemic AKI compared with wild-type (PAD4ff) mice. Surprisingly, mice lacking PAD4 in myeloid cells were also protected against renal I/R injury although this protection was less compared with renal proximal tubular PAD4-deficient mice. Renal proximal tubular PAD4-deficient mice had profoundly reduced renal tubular apoptosis, whereas myeloid-cell PAD4-deficient mice showed markedly reduced renal neutrophil infiltration. Taken together, our studies suggest that both renal proximal tubular PAD4 as well as myeloid-cell lineage PAD4 play a critical role in exacerbating ischemic AKI. Renal proximal tubular PAD4 appears to contribute to ischemic AKI by promoting renal tubular apoptosis, whereas myeloid-cell PAD4 is preferentially involved in promoting neutrophil infiltration to the kidney and inflammation after renal I/R.


Subject(s)
Acute Kidney Injury/enzymology , Apoptosis , Hydrolases/metabolism , Kidney Tubules, Proximal/enzymology , Neutrophil Infiltration , Neutrophils/enzymology , Reperfusion Injury/enzymology , Acute Kidney Injury/genetics , Acute Kidney Injury/pathology , Acute Kidney Injury/prevention & control , Animals , Cytokines/metabolism , Hydrolases/deficiency , Hydrolases/genetics , Inflammation Mediators/metabolism , Kidney Tubules, Proximal/pathology , Mice, Inbred C57BL , Mice, Knockout , Protein-Arginine Deiminase Type 4 , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Reperfusion Injury/prevention & control , Signal Transduction
15.
Kidney Int ; 93(2): 365-374, 2018 02.
Article in English | MEDLINE | ID: mdl-29061334

ABSTRACT

Ischemia/reperfusion is a common cause of acute kidney injury (AKI). However, mechanisms underlying the sudden loss in kidney function and tissue injury remain to be fully elucidated. Here, we investigated the role of peptidyl arginine deiminase-4 (PAD4), which converts arginine to citrulline and plays a role in epigenetic regulation and inflammation, in renal ischemia/reperfusion injury. PAD4 expression was highly induced in infiltrating leukocytes 24 hours following renal ischemia and reperfusion. This induction was accompanied by citrullination of histone H3 and formation of neutrophil extracellular traps in kidneys of wild-type mice. By contrast, PAD4-deficient mice did not form neutrophil extracellular traps, expressed lower levels of pro-inflammatory cytokines and were partially protected from renal ischemia/reperfusion-induced AKI. Furthermore, PAD4-deficient mice recovered kidney function 48 hours after ischemia/reperfusion, whereas kidney function in the wild-type mice progressively worsened. Administration of DNase I, which degrades neutrophil extracellular traps or the PAD-specific inhibitor YW3-56 before ischemia, partially prevented renal ischemia/reperfusion-induced AKI. Notably, transfer of neutrophils from wild-type, but not from PAD4-deficient mice, was sufficient to restore renal neutrophil extracellular trap formation and impair kidney function following renal ischemia/reperfusion. Thus, neutrophil PAD4 plays a pivotal role in renal ischemia/reperfusion-induced AKI.


Subject(s)
Acute Kidney Injury/enzymology , Extracellular Traps/enzymology , Hydrolases/metabolism , Kidney/enzymology , Neutrophils/enzymology , Reperfusion Injury/enzymology , Acute Kidney Injury/pathology , Acute Kidney Injury/physiopathology , Acute Kidney Injury/prevention & control , Animals , Citrullination , Cytokines/metabolism , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Histiocytes/metabolism , Hydrolases/antagonists & inhibitors , Hydrolases/deficiency , Hydrolases/genetics , Inflammation Mediators/metabolism , Kidney/drug effects , Kidney/pathology , Kidney/physiopathology , Male , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration , Neutrophils/drug effects , Neutrophils/pathology , Neutrophils/transplantation , Protein-Arginine Deiminase Type 4 , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Reperfusion Injury/prevention & control
16.
Am J Physiol Renal Physiol ; 314(2): F293-F305, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29021225

ABSTRACT

We previously demonstrated that renal tubular peptidylarginine deiminase-4 (PAD4) is induced after ischemia-reperfusion (IR) injury and this induction of PAD4 exacerbates ischemic acute kidney injury (AKI) by promoting renal tubular inflammation and neutrophil infiltration. However, the mechanisms of renal tubular PAD4 induction after IR remain unknown. Here, we tested the hypothesis that ATP, a proinflammatory danger-associated molecular pattern (DAMP) ligand released from necrotic cells after IR injury, induces renal tubular PAD4 and exacerbates ischemic AKI via P2 purinergic receptor activation. ATP as well as ATPγS (a nonmetabolizable ATP analog) induced PAD4 mRNA, protein, and activity in human and mouse renal proximal tubule cells. Supporting the hypothesis that ATP induces renal tubular PAD4 via P2X7 receptor activation, A804598 (a selective P2X7 receptor antagonist) blocked the ATP-mediated induction of renal tubular PAD4 whereas BzATP (a selective P2X7 receptor agonist) mimicked the effects of ATP by inducing renal tubular PAD4 expression and activity. Moreover, ATP-mediated calcium influx in renal proximal tubule cells was blocked by A804598 and was mimicked by BzATP. P2X7 activation by BzATP also induced PAD4 expression and activity in mouse kidney in vivo. Finally, supporting a critical role for PAD4 in P2X7-mediated exacerbation of renal injury, BzATP exacerbated ischemic AKI in PAD4 wild-type mice but not in PAD4-deficient mice. Taken together, our studies show that ATP induces renal tubular PAD4 via P2X7 receptor activation to exacerbate renal tubular inflammation and injury after IR.


Subject(s)
Acute Kidney Injury/chemically induced , Adenosine Triphosphate/toxicity , Hydrolases/metabolism , Kidney Tubules, Proximal/drug effects , Protein-Arginine Deiminases/metabolism , Purinergic P2X Receptor Agonists/toxicity , Receptors, Purinergic P2X7/drug effects , Reperfusion Injury/chemically induced , Acute Kidney Injury/genetics , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Animals , Calcium Signaling/drug effects , Cell Line , Disease Models, Animal , Disease Progression , Humans , Hydrolases/deficiency , Hydrolases/genetics , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Mice, Inbred C57BL , Mice, Knockout , Necrosis , Neutrophil Infiltration/drug effects , Protein Kinase C/metabolism , Protein-Arginine Deiminase Type 4 , Protein-Arginine Deiminases/genetics , Receptors, Purinergic P2X7/metabolism , Reperfusion Injury/genetics , Reperfusion Injury/metabolism , Reperfusion Injury/pathology
17.
Stem Cell Reports ; 9(2): 499-512, 2017 08 08.
Article in English | MEDLINE | ID: mdl-28757167

ABSTRACT

Liver or hepatocytes transplantation is limited by the availability of donor organs. Functional hepatocytes independent of the donor sources may have wide applications in regenerative medicine and the drug industry. Recent studies have demonstrated that chemical cocktails may induce reprogramming of fibroblasts into a range of functional somatic cells. Here, we show that mouse fibroblasts can be transdifferentiated into the hepatocyte-like cells (iHeps) using only one transcription factor (TF) (Foxa1, Foxa2, or Foxa3) plus a chemical cocktail. These iHeps show typical epithelial morphology, express multiple hepatocyte-specific genes, and acquire hepatocyte functions. Genetic lineage tracing confirms the fibroblast origin of these iHeps. More interestingly, these iHeps are expandable in vitro and can reconstitute the damaged hepatic tissues of the fumarylacetoacetate hydrolase-deficient (Fah-/-) mice. Our study provides a strategy to generate functional hepatocyte-like cells by using a single TF plus a chemical cocktail and is one step closer to generate the full-chemical iHeps.


Subject(s)
Cell Transdifferentiation/genetics , Cellular Reprogramming/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Hepatocytes/cytology , Hepatocytes/metabolism , Transcription Factors/genetics , Animals , Biomarkers , Cell Lineage , Cell Transplantation , Cells, Cultured , Fluorescent Antibody Technique , Gene Expression , Hydrolases/deficiency , Immunophenotyping , Liver/metabolism , Liver/pathology , Mice , Mice, Knockout , Regenerative Medicine , Transcription Factors/metabolism
18.
Adv Exp Med Biol ; 959: 215-230, 2017.
Article in English | MEDLINE | ID: mdl-28755199

ABSTRACT

Several animal models of Fah deficiency have been developed, including mice, pigs and most recently rats. Initially, the murine models were developed with the intent to mirror the human disease for pathophysiologic and therapeutic studies. However, it soon became apparent that Fah-positive hepatocytes have a potent selective growth advantage in mutant liver and can extensively repopulate the diseased organ. For this reason, Fah mutant mice have become a workhorse for liver biology and are widely used in liver stem cell and hepatic gene therapy research. Immune deficient Fah-knockout mice can be repopulated with human hepatocytes, creating "mice with human livers". These chimeric animals have become an important preclinical model for infectious diseases, metabolism and gene therapy. The potent expansion of human hepatocytes in Fah knockout mice has given rise to the concept of using Fah mutants as living bioreactors to produce large quantities of fully mature hepatocytes. As a consequence, larger animal models of Fah deficiency have recently been developed.


Subject(s)
Hydrolases/deficiency , Liver/pathology , Animals , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Hydrolases/genetics , Hydrolases/metabolism , Liver/drug effects , Liver/metabolism , Models, Animal
19.
Mol Ther ; 25(5): 1155-1162, 2017 05 03.
Article in English | MEDLINE | ID: mdl-28389320

ABSTRACT

Lysosomal storage disorders (LSDs) are a broad class of monogenic diseases with an overall incidence of 1:7,000 newborns, due to the defective activity of one or more lysosomal hydrolases or related proteins resulting in storage of un-degraded substrates in the lysosomes. The over 40 different known LSDs share a life-threatening nature, but they are present with extremely variable clinical manifestations, determined by the characteristics and tissue distribution of the material accumulating due to the lysosomal dysfunction. The majority of LSDs lack a curative treatment. This is particularly true for LSDs severely affecting the CNS. Based on current preclinical and clinical evidences, among other treatment modalities, hematopoietic stem cell gene therapy could potentially result in robust therapeutic benefit for LSD patients, with particular indication for those characterized by severe brain damage. Optimization of current approaches and technology, as well as implementation of clinical trials for novel indications, and prolonged and more extensive follow-up of the already treated patients will allow translating this promise into new medicinal products.


Subject(s)
Brain Damage, Chronic/therapy , Genetic Therapy/methods , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Hydrolases/genetics , Lysosomal Storage Diseases/therapy , Animals , Brain Damage, Chronic/enzymology , Brain Damage, Chronic/genetics , Brain Damage, Chronic/pathology , Disease Models, Animal , Enzyme Replacement Therapy/methods , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Hematopoietic Stem Cells/cytology , Humans , Hydrolases/deficiency , Lentivirus/genetics , Lentivirus/metabolism , Lysosomal Storage Diseases/enzymology , Lysosomal Storage Diseases/genetics , Lysosomal Storage Diseases/pathology , Lysosomes/metabolism , Transplantation, Homologous
SELECTION OF CITATIONS
SEARCH DETAIL