Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Sci Adv ; 10(11): eadd9342, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38478609

ABSTRACT

Tumors represent ecosystems where subclones compete during tumor growth. While extensively investigated, a comprehensive picture of the interplay of clonal lineages during dissemination is still lacking. Using patient-derived pancreatic cancer cells, we created orthotopically implanted clonal replica tumors to trace clonal dynamics of unperturbed tumor expansion and dissemination. This model revealed the multifaceted nature of tumor growth, with rapid changes in clonal fitness leading to continuous reshuffling of tumor architecture and alternating clonal dominance as a distinct feature of cancer growth. Regarding dissemination, a large fraction of tumor lineages could be found at secondary sites each having distinctive organ growth patterns as well as numerous undescribed behaviors such as abortive colonization. Paired analysis of primary and secondary sites revealed fitness as major contributor to dissemination. From the analysis of pro- and nonmetastatic isogenic subclones, we identified a transcriptomic signature able to identify metastatic cells in human tumors and predict patients' survival.


Subject(s)
Ecosystem , Pancreatic Neoplasms , Humans , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Gene Expression Profiling , Transcriptome
2.
Sci Adv ; 10(13): eadk5386, 2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38536927

ABSTRACT

While pancreatic ductal adenocarcinomas (PDACs) are addicted to KRAS-activating mutations, inhibitors of downstream KRAS effectors, such as the MEK1/2 kinase inhibitor trametinib, are devoid of therapeutic effects. However, the extensive rewiring of regulatory circuits driven by the attenuation of the KRAS pathway may induce vulnerabilities of therapeutic relevance. An in-depth molecular analysis of the transcriptional and epigenomic alterations occurring in PDAC cells in the initial hours after MEK1/2 inhibition by trametinib unveiled the induction of endogenous retroviruses (ERVs) escaping epigenetic silencing, leading to the production of double-stranded RNAs and the increased expression of interferon (IFN) genes. We tracked ERV activation to the early induction of the transcription factor ELF3, which extensively bound and activated nonsilenced retroelements and synergized with IRF1 (interferon regulatory factor 1) in the activation of IFNs and IFN-stimulated genes. Trametinib-induced viral mimicry in PDAC may be exploited in the rational design of combination therapies in immuno-oncology.


Subject(s)
Carcinoma, Pancreatic Ductal , Endogenous Retroviruses , Pancreatic Neoplasms , Humans , Endogenous Retroviruses/genetics , Signal Transduction , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism
3.
Nat Commun ; 15(1): 1203, 2024 Feb 08.
Article in English | MEDLINE | ID: mdl-38331987

ABSTRACT

DNA damage resistance is a major barrier to effective DNA-damaging therapy in multiple myeloma (MM). To discover mechanisms through which MM cells overcome DNA damage, we investigate how MM cells become resistant to antisense oligonucleotide (ASO) therapy targeting Interleukin enhancer binding factor 2 (ILF2), a DNA damage regulator that is overexpressed in 70% of MM patients whose disease has progressed after standard therapies have failed. Here, we show that MM cells undergo adaptive metabolic rewiring to restore energy balance and promote survival in response to DNA damage activation. Using a CRISPR/Cas9 screening strategy, we identify the mitochondrial DNA repair protein DNA2, whose loss of function suppresses MM cells' ability to overcome ILF2 ASO-induced DNA damage, as being essential to counteracting oxidative DNA damage. Our study reveals a mechanism of vulnerability of MM cells that have an increased demand for mitochondrial metabolism upon DNA damage activation.


Subject(s)
Multiple Myeloma , Humans , Multiple Myeloma/genetics , DNA Helicases/metabolism , Metabolic Reprogramming , DNA Repair , DNA Damage
4.
J Inherit Metab Dis ; 46(2): 335-347, 2023 03.
Article in English | MEDLINE | ID: mdl-36433920

ABSTRACT

Multiple sulfatase deficiency (MSD) is an ultrarare lysosomal storage disorder due to deficiency of all known sulfatases. MSD is caused by mutations in the Sulfatase Modifying Factor 1 (SUMF1) gene encoding the enzyme responsible for the post-translational modification and activation of all sulfatases. Most MSD patients carry hypomorph SUMF1 variants resulting in variable degrees of residual sulfatase activities. In contrast, Sumf1 null mice with complete deficiency in all sulfatase enzyme activities, have very short lifespan with significant pre-wean lethality, owing to a challenging preclinical model. To overcome this limitation, we genetically engineered and characterized in mice two commonly identified patient-based SUMF1 pathogenic variants, namely p.Ser153Pro and p.Ala277Val. These pathogenic missense variants correspond to variants detected in patients with attenuated MSD presenting with partial-enzyme deficiency and relatively less severe disease. These novel MSD mouse models have a longer lifespan and show biochemical and pathological abnormalities observed in humans. In conclusion, mice harboring the p.Ser153Pro or the p.Ala277Val variant mimic the attenuated MSD and are attractive preclinical models for investigation of pathogenesis and treatments for MSD.


Subject(s)
Lysosomal Storage Diseases , Multiple Sulfatase Deficiency Disease , Humans , Animals , Mice , Multiple Sulfatase Deficiency Disease/genetics , Mutation , Sulfatases , Mutation, Missense , Oxidoreductases Acting on Sulfur Group Donors/genetics
5.
Nat Commun ; 13(1): 5212, 2022 09 05.
Article in English | MEDLINE | ID: mdl-36064721

ABSTRACT

Life-threatening hyperammonemia occurs in both inherited and acquired liver diseases affecting ureagenesis, the main pathway for detoxification of neurotoxic ammonia in mammals. Protein O-GlcNAcylation is a reversible and nutrient-sensitive post-translational modification using as substrate UDP-GlcNAc, the end-product of hexosamine biosynthesis pathway. Here we show that increased liver UDP-GlcNAc during hyperammonemia increases protein O-GlcNAcylation and enhances ureagenesis. Mechanistically, O-GlcNAcylation on specific threonine residues increased the catalytic efficiency for ammonia of carbamoyl phosphate synthetase 1 (CPS1), the rate-limiting enzyme in ureagenesis. Pharmacological inhibition of O-GlcNAcase, the enzyme removing O-GlcNAc from proteins, resulted in clinically relevant reductions of systemic ammonia in both genetic (hypomorphic mouse model of propionic acidemia) and acquired (thioacetamide-induced acute liver failure) mouse models of liver diseases. In conclusion, by fine-tuned control of ammonia entry into ureagenesis, hepatic O-GlcNAcylation of CPS1 increases ammonia detoxification and is a novel target for therapy of hyperammonemia in both genetic and acquired diseases.


Subject(s)
Ammonia , Carbamoyl-Phosphate Synthase (Ammonia) , Hyperammonemia , Urea , Uridine Diphosphate , Acetylglucosamine , Ammonia/metabolism , Animals , Biocatalysis , Carbamoyl-Phosphate Synthase (Ammonia)/genetics , Carbamoyl-Phosphate Synthase (Ammonia)/metabolism , Disease Models, Animal , Glycosylation , Humans , Hyperammonemia/genetics , Hyperammonemia/metabolism , Mammals/metabolism , Mice , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/metabolism , Propionic Acidemia/genetics , Propionic Acidemia/metabolism , Protein Processing, Post-Translational/genetics , Urea/metabolism , Uridine Diphosphate/genetics , Uridine Diphosphate/metabolism
6.
Proc Natl Acad Sci U S A ; 118(10)2021 03 09.
Article in English | MEDLINE | ID: mdl-33649241

ABSTRACT

α1-Antitrypsin (AAT) deficiency is a common genetic disease presenting with lung and liver diseases. AAT deficiency results from pathogenic variants in the SERPINA1 gene encoding AAT and the common mutant Z allele of SERPINA1 encodes for Z α1-antitrypsin (ATZ), a protein forming hepatotoxic polymers retained in the endoplasmic reticulum of hepatocytes. PiZ mice express the human ATZ and are a valuable model to investigate the human liver disease of AAT deficiency. In this study, we investigated differential expression of microRNAs (miRNAs) between PiZ and control mice and found that miR-34b/c was up-regulated and its levels correlated with intrahepatic ATZ. Furthermore, in PiZ mouse livers, we found that Forkhead Box O3 (FOXO3) driving microRNA-34b/c (miR-34b/c) expression was activated and miR-34b/c expression was dependent upon c-Jun N-terminal kinase (JNK) phosphorylation on Ser574 Deletion of miR-34b/c in PiZ mice resulted in early development of liver fibrosis and increased signaling of platelet-derived growth factor (PDGF), a target of miR-34b/c. Activation of FOXO3 and increased miR-34c were confirmed in livers of humans with AAT deficiency. In addition, JNK-activated FOXO3 and miR-34b/c up-regulation were detected in several mouse models of liver fibrosis. This study reveals a pathway involved in liver fibrosis and potentially implicated in both genetic and acquired causes of hepatic fibrosis.


Subject(s)
Forkhead Box Protein O3/metabolism , Liver Cirrhosis , MAP Kinase Kinase 4/metabolism , Up-Regulation , Animals , Disease Models, Animal , Forkhead Box Protein O3/genetics , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Liver Cirrhosis/prevention & control , MAP Kinase Kinase 4/genetics , Male , Mice , Mice, Knockout , MicroRNAs/biosynthesis , MicroRNAs/genetics , alpha 1-Antitrypsin/genetics , alpha 1-Antitrypsin/metabolism
7.
J Biol Chem ; 295(38): 13213-13223, 2020 09 18.
Article in English | MEDLINE | ID: mdl-32723872

ABSTRACT

α1-Antitrypsin (AAT) encoded by the SERPINA1 gene is an acute-phase protein synthesized in the liver and secreted into the circulation. Its primary role is to protect lung tissue by inhibiting neutrophil elastase. The Z allele of SERPINA1 encodes a mutant AAT, named ATZ, that changes the protein structure and leads to its misfolding and polymerization, which cause endoplasmic reticulum (ER) stress and liver disease through a gain-of-function toxic mechanism. Hepatic retention of ATZ results in deficiency of one of the most important circulating proteinase inhibitors and predisposes to early-onset emphysema through a loss-of-function mechanism. The pathogenetic mechanisms underlying the liver disease are not completely understood. C/EBP-homologous protein (CHOP), a transcription factor induced by ER stress, was found among the most up-regulated genes in livers of PiZ mice that express ATZ and in human livers of patients homozygous for the Z allele. Compared with controls, juvenile PiZ/Chop-/- mice showed reduced hepatic ATZ and a transcriptional response indicative of decreased ER stress by RNA-Seq analysis. Livers of PiZ/Chop-/- mice also showed reduced SERPINA1 mRNA levels. By chromatin immunoprecipitations and luciferase reporter-based transfection assays, CHOP was found to up-regulate SERPINA1 cooperating with c-JUN, which was previously shown to up-regulate SERPINA1, thus aggravating hepatic accumulation of ATZ. Increased CHOP levels were detected in diseased livers of children homozygous for the Z allele. In summary, CHOP and c-JUN up-regulate SERPINA1 transcription and play an important role in hepatic disease by increasing the burden of proteotoxic ATZ, particularly in the pediatric population.


Subject(s)
Liver Diseases/metabolism , Liver/metabolism , Mutation , Protein Aggregation, Pathological/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Transcription Factor CHOP/metabolism , alpha 1-Antitrypsin/biosynthesis , Alleles , Animals , Endoplasmic Reticulum Stress/genetics , Humans , Liver/pathology , Liver Diseases/genetics , Liver Diseases/pathology , Mice , Mice, Knockout , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/pathology , Protein Folding , Proto-Oncogene Proteins c-jun/genetics , Transcription Factor CHOP/genetics , Transcription, Genetic , Up-Regulation , alpha 1-Antitrypsin/genetics
8.
Eur J Hum Genet ; 27(9): 1475-1480, 2019 09.
Article in English | MEDLINE | ID: mdl-31152157

ABSTRACT

We identified a 14q21.2 microdeletion in a 16-year-old boy with autism spectrum disorder (ASD), IQ in the lower part of normal range but high-functioning memory skills. The deletion affects a gene desert, and the non-deleted gene closest to the microdeletion boundaries is LRFN5, which encodes a protein involved in synaptic plasticity and implicated in neuro-psychiatric disorders. LRFN5 expression was significantly decreased in the proband's skin fibroblasts. The deleted region includes the pseudogene chr14.232.a, which is transcribed into a long non-coding RNA (lncLRFN5-10), whose levels were also significantly reduced in the proband's fibroblasts compared to controls. Transfection of the patient's fibroblasts with a plasmid expressing chr14.232.a significantly increased LRFN5 expression, while siRNA targeting chr14.232.a-derived lncLRFN5-10 reduced LRFN5 levels. In summary, we report on an individual with ASD carrying a microdeletion encompassing the pseudogene chr14.232.a encoding for lncLRFN5-10, which was found to affect the expression levels of the nearby, non-deleted LRFN5. This case illustrates the potential role of long non-coding RNAs in regulating expression of neighbouring genes with a functional role in ASD pathogenesis.


Subject(s)
Autism Spectrum Disorder/diagnosis , Autism Spectrum Disorder/genetics , Chromosome Deletion , Chromosomes, Human, Pair 14 , Gene Expression , Membrane Glycoproteins/genetics , Pseudogenes , Adolescent , Fibroblasts/metabolism , Genetic Predisposition to Disease , Humans , Magnetic Resonance Imaging , Male , Skin/cytology , Exome Sequencing
9.
J Inherit Metab Dis ; 42(6): 1128-1135, 2019 11.
Article in English | MEDLINE | ID: mdl-30724386

ABSTRACT

The urea cycle and glutamine synthetase (GS) are the two main pathways for waste nitrogen removal and their deficiency results in hyperammonemia. Here, we investigated the efficacy of liver-specific GS overexpression for therapy of hyperammonemia. To achieve hepatic GS overexpression, we generated a helper-dependent adenoviral (HDAd) vector expressing the murine GS under the control of a liver-specific expression cassette (HDAd-GS). Compared to mice injected with a control vector expressing an unrelated reporter gene (HDAd-alpha-fetoprotein), wild-type mice with increased hepatic GS showed reduced blood ammonia levels and a concomitant increase of blood glutamine after intraperitoneal injections of ammonium chloride, whereas blood urea was unaffected. Moreover, injection of HDAd-GS reduced blood ammonia levels at baseline and protected against acute hyperammonemia following ammonia challenge in a mouse model with conditional hepatic deficiency of carbamoyl phosphate synthetase 1 (Cps1), the initial and rate-limiting step of ureagenesis. In summary, we found that upregulation of hepatic GS reduced hyperammonemia in wild-type and Cps1-deficient mice, thus confirming a key role of GS in ammonia detoxification. These results suggest that hepatic GS augmentation therapy has potential for treatment of both primary and secondary forms of hyperammonemia.


Subject(s)
Ammonia/metabolism , Genetic Therapy/methods , Glutamate-Ammonia Ligase/genetics , Hyperammonemia/genetics , Hyperammonemia/therapy , Liver/metabolism , Ammonia/toxicity , Animals , Carbamoyl-Phosphate Synthase (Ammonia)/genetics , Carbamoyl-Phosphate Synthase (Ammonia)/metabolism , Carbamoyl-Phosphate Synthase I Deficiency Disease/genetics , Carbamoyl-Phosphate Synthase I Deficiency Disease/metabolism , Carbamoyl-Phosphate Synthase I Deficiency Disease/therapy , Disease Models, Animal , Female , Gene Transfer Techniques , Glutamate-Ammonia Ligase/metabolism , Hyperammonemia/metabolism , Hyperammonemia/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organ Specificity/genetics
10.
Hepatology ; 66(2): 677-678, 2017 08.
Article in English | MEDLINE | ID: mdl-28437874
11.
Hepatology ; 66(1): 124-135, 2017 07.
Article in English | MEDLINE | ID: mdl-28295475

ABSTRACT

α1 -Antitrypsin (AAT) deficiency is one of the most common genetic disorders and the liver disease due to the Z mutant of AAT (ATZ) is a prototype of conformational disorder due to protein misfolding with consequent aberrant intermolecular protein aggregation. In the present study, we found that livers of PiZ transgenic mice expressing human ATZ have altered expression of a network of hepatocyte transcriptional factors, including hepatocyte nuclear factor-4α, that is early down-regulated and induces a transcriptional repression of ATZ expression. Reduced hepatocyte nuclear factor-4α was associated with activation of ß-catenin, which regulates liver zonation. Livers of PiZ mice and human patients with AAT deficiency were both found to have a severe perturbation of liver zonation. Functionally, PiZ mice showed a severe defect of ureagenesis, as shown by increased baseline ammonia, and reduced urea production and survival after an ammonia challenge. Down-regulation of hepatocyte nuclear factor-4α expression and defective zonation in livers have not been recognized so far as features of the liver disease caused by ATZ and are likely involved in metabolic disturbances and in the increased risk of hepatocellular carcinoma in patients with AAT deficiency. CONCLUSION: The findings of this study are consistent with the concept that abnormal AAT protein conformation and intrahepatic accumulation have broad effects on metabolic liver functions. (Hepatology 2017;66:124-135).


Subject(s)
Carcinoma, Hepatocellular/genetics , Gene Expression Regulation, Neoplastic , Hepatocyte Nuclear Factor 4/genetics , Liver Neoplasms/pathology , alpha 1-Antitrypsin Deficiency/genetics , Aging/genetics , Analysis of Variance , Animals , Carcinoma, Hepatocellular/pathology , Disease Models, Animal , Down-Regulation , Humans , Liver Neoplasms/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Random Allocation , Statistics, Nonparametric , alpha 1-Antitrypsin Deficiency/pathology
12.
Hepatology ; 65(6): 1865-1874, 2017 06.
Article in English | MEDLINE | ID: mdl-28073160

ABSTRACT

Alpha1-antitrypsin deficiency is a genetic disease that can affect both the lung and the liver. The vast majority of patients harbor a mutation in the serine protease inhibitor 1A (SERPINA1) gene leading to a single amino acid substitution that results in an unfolded protein that is prone to polymerization. Alpha1-antitrypsin defciency-related liver disease is therefore caused by a gain-of-function mechanism due to accumulation of the mutant Z alpha1-antitrypsin (ATZ) and is a key example of an disease mechanism induced by protein toxicity. Intracellular retention of ATZ triggers a complex injury cascade including apoptosis and other mechanisms, although several aspects of the disease pathogenesis are still unclear. We show that ATZ induces activation of c-Jun N-terminal kinase (JNK) and c-Jun and that genetic ablation of JNK1 or JNK2 decreased ATZ levels in vivo by reducing c-Jun-mediated SERPINA1 gene expression. JNK activation was confirmed in livers of patients homozygous for the Z allele, with severe liver disease requiring hepatic transplantation. Treatment of patient-derived induced pluripotent stem cell-hepatic cells with a JNK inhibitor reduced accumulation of ATZ. CONCLUSION: These data reveal that JNK is a key pathway in the disease pathogenesis and add new therapeutic entry points for liver disease caused by ATZ. (Hepatology 2017;65:1865-1874).


Subject(s)
JNK Mitogen-Activated Protein Kinases/metabolism , Signal Transduction/genetics , alpha 1-Antitrypsin/metabolism , Animals , Apoptosis/genetics , Blotting, Western , Cells, Cultured , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Hepatocytes/metabolism , Humans , Immunoprecipitation , Liver/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Sensitivity and Specificity , Up-Regulation , alpha 1-Antitrypsin/genetics
13.
Hum Mol Genet ; 26(1): 33-43, 2017 01 01.
Article in English | MEDLINE | ID: mdl-28013292

ABSTRACT

We performed whole exome sequencing in individuals from a family with autosomal dominant gastropathy resembling Ménétrier disease, a premalignant gastric disorder with epithelial hyperplasia and enhanced EGFR signalling. Ménétrier disease is believed to be an acquired disorder, but its aetiology is unknown. In affected members, we found a missense p.V742G variant in MIB2, a gene regulating NOTCH signalling that has not been previously linked to human diseases. The variant segregated with the disease in the pedigree, affected a highly conserved amino acid residue, and was predicted to be deleterious although it was found with a low frequency in control individuals. The purified protein carrying the p.V742G variant showed reduced ubiquitination activity in vitro and white blood cells from affected individuals exhibited significant reductions of HES1 and NOTCH3 expression reflecting alteration of NOTCH signalling. Because mutations of MIB1, the homolog of MIB2, have been found in patients with left ventricle non-compaction (LVNC), we investigated members of our family with Ménétrier-like disease for this cardiac abnormality. Asymptomatic left ventricular hypertrabeculation, the mildest end of the LVNC spectrum, was detected in two members carrying the MIB2 variant. Finally, we identified an additional MIB2 variant (p.V984L) affecting protein stability in an unrelated isolated case with LVNC. Expression of both MIB2 variants affected NOTCH signalling, proliferation and apoptosis in primary rat cardiomyocytes.In conclusion, we report the first example of left ventricular hypertrabeculation/LVNC with germline MIB2 variants resulting in altered NOTCH signalling that might be associated with a gastropathy clinically overlapping with Ménétrier disease.


Subject(s)
Cardiomyopathies/pathology , Gastritis, Hypertrophic/pathology , Mutation, Missense/genetics , Receptors, Notch/metabolism , Stomach Diseases/pathology , Ubiquitin-Protein Ligases/genetics , Ventricular Dysfunction, Left/pathology , Animals , Animals, Newborn , Cardiomyopathies/etiology , Cardiomyopathies/metabolism , Case-Control Studies , Cells, Cultured , Exome/genetics , Female , Gastritis, Hypertrophic/etiology , Gastritis, Hypertrophic/metabolism , Gene Expression Regulation , Humans , Male , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Pedigree , Phenotype , Rats , Receptors, Notch/genetics , Signal Transduction , Stomach Diseases/etiology , Stomach Diseases/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Ventricular Dysfunction, Left/etiology , Ventricular Dysfunction, Left/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...