Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 101
Filter
1.
Mol Cancer ; 23(1): 105, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38755661

ABSTRACT

BACKGROUND: The main drawback of BRAF/MEK inhibitors (BRAF/MEKi)-based targeted therapy in the management of BRAF-mutated cutaneous metastatic melanoma (MM) is the development of therapeutic resistance. We aimed to assess in this context the role of mTORC2, a signaling complex defined by the presence of the essential RICTOR subunit, regarded as an oncogenic driver in several tumor types, including MM. METHODS: After analyzing The Cancer Genome Atlas MM patients' database to explore both overall survival and molecular signatures as a function of intra-tumor RICTOR levels, we investigated the effects of RICTOR downregulation in BRAFV600E MM cell lines on their response to BRAF/MEKi. We performed proteomic screening to identify proteins modulated by changes in RICTOR expression, and Seahorse analysis to evaluate the effects of RICTOR depletion on mitochondrial respiration. The combination of BRAFi with drugs targeting proteins and processes emerged in the proteomic screening was carried out on RICTOR-deficient cells in vitro and in a xenograft setting in vivo. RESULTS: Low RICTOR levels in BRAF-mutated MM correlate with a worse clinical outcome. Gene Set Enrichment Analysis of low-RICTOR tumors display gene signatures suggestive of activation of the mitochondrial Electron Transport Chain (ETC) energy production. RICTOR-deficient BRAFV600E cells are intrinsically tolerant to BRAF/MEKi and anticipate the onset of resistance to BRAFi upon prolonged drug exposure. Moreover, in drug-naïve cells we observed a decline in RICTOR expression shortly after BRAFi exposure. In RICTOR-depleted cells, both mitochondrial respiration and expression of nicotinamide phosphoribosyltransferase (NAMPT) are enhanced, and their pharmacological inhibition restores sensitivity to BRAFi. CONCLUSIONS: Our work unveils an unforeseen tumor-suppressing role for mTORC2 in the early adaptation phase of BRAFV600E melanoma cells to targeted therapy and identifies the NAMPT-ETC axis as a potential therapeutic vulnerability of low RICTOR tumors. Importantly, our findings indicate that the evaluation of intra-tumor RICTOR levels has a prognostic value in metastatic melanoma and may help to guide therapeutic strategies in a personalized manner.


Subject(s)
Drug Resistance, Neoplasm , Mechanistic Target of Rapamycin Complex 2 , Melanoma , Protein Kinase Inhibitors , Proto-Oncogene Proteins B-raf , Rapamycin-Insensitive Companion of mTOR Protein , Animals , Humans , Mice , Cell Line, Tumor , Down-Regulation , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Mechanistic Target of Rapamycin Complex 2/metabolism , Mechanistic Target of Rapamycin Complex 2/genetics , Melanoma/genetics , Melanoma/drug therapy , Melanoma/metabolism , Melanoma/pathology , Mutation , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proteomics/methods , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Rapamycin-Insensitive Companion of mTOR Protein/metabolism , Rapamycin-Insensitive Companion of mTOR Protein/genetics , Xenograft Model Antitumor Assays , MAP Kinase Kinase Kinases/antagonists & inhibitors
2.
Int J Mol Sci ; 24(16)2023 08 17.
Article in English | MEDLINE | ID: mdl-37629051

ABSTRACT

Obesity is a growing public health problem associated with increased risk of type 2 diabetes, cardiovascular disease, nonalcoholic fatty liver disease (NAFLD) and cancer. Here, we identify microRNA-22 (miR-22) as an essential rheostat involved in the control of lipid and energy homeostasis as well as the onset and maintenance of obesity. We demonstrate through knockout and transgenic mouse models that miR-22 loss-of-function protects against obesity and hepatic steatosis, while its overexpression promotes both phenotypes even when mice are fed a regular chow diet. Mechanistically, we show that miR-22 controls multiple pathways related to lipid biogenesis and differentiation. Importantly, genetic ablation of miR-22 favors metabolic rewiring towards higher energy expenditure and browning of white adipose tissue, suggesting that modulation of miR-22 could represent a viable therapeutic strategy for treatment of obesity and other metabolic disorders.


Subject(s)
Diabetes Mellitus, Type 2 , MicroRNAs , Non-alcoholic Fatty Liver Disease , Animals , Mice , Homeostasis , Mice, Transgenic , Non-alcoholic Fatty Liver Disease/genetics , Obesity/genetics , MicroRNAs/genetics , Lipids
3.
Int J Mol Sci ; 21(2)2020 Jan 16.
Article in English | MEDLINE | ID: mdl-31963158

ABSTRACT

The RNA-binding protein, Epithelial Splicing Regulatory Protein 1 (ESRP1) can promote or suppress tumorigenesis depending on the cell type and disease context. In colorectal cancer, we have previously shown that aberrantly high ESRP1 expression can drive tumor progression. In order to unveil the mechanisms by which ESRP1 can modulate cancer traits, we searched for proteins affected by modulation of Esrp1 in two human colorectal cancer cell lines, HCA24 and COLO320DM, by proteomics analysis. Proteins hosted by endogenous ESRP1 ribonucleoprotein complex in HCA24 cells were also analyzed following RNA-immunoprecipitation. Proteomics data were complemented with bioinformatics approach to exploit publicly available data on protein-protein interaction (PPI). Gene Ontology was analysed to identify a common molecular signature possibly explaining the pro-tumorigenic role of ESRP1. Interestingly, proteins identified herein support a role for ESRP1 in response to external stimulus, regulation of cell cycle and hypoxia. Our data provide further insights into factors affected by and entwined with ESRP1 in colorectal cancer.


Subject(s)
Colorectal Neoplasms/metabolism , Proteomics/methods , RNA-Binding Proteins/metabolism , Cell Cycle/genetics , Cell Cycle/physiology , Cell Line, Tumor , Colorectal Neoplasms/genetics , Computational Biology/methods , Gene Expression Regulation, Neoplastic/genetics , Humans , Protein Binding , RNA-Binding Proteins/genetics
4.
Neurobiol Dis ; 124: 14-28, 2019 04.
Article in English | MEDLINE | ID: mdl-30389403

ABSTRACT

Spinocerebellar ataxia 28 is an autosomal dominant neurodegenerative disorder caused by missense mutations affecting the proteolytic domain of AFG3L2, a major component of the mitochondrial m-AAA protease. However, little is known of the underlying pathogenetic mechanisms or how to treat patients with SCA28. Currently available Afg3l2 mutant mice harbour deletions that lead to severe, early-onset neurological phenotypes that do not faithfully reproduce the late-onset and slowly progressing SCA28 phenotype. Here we describe production and detailed analysis of a new knock-in murine model harbouring an Afg3l2 allele carrying the p.Met665Arg patient-derived mutation. Heterozygous mutant mice developed normally but adult mice showed signs of cerebellar ataxia detectable by beam test. Although cerebellar pathology was negative, electrophysiological analysis showed a trend towards increased spontaneous firing in Purkinje cells from heterozygous mutants with respect to wild-type controls. As homozygous mutants died perinatally with evidence of cardiac atrophy, for each genotype we generated mouse embryonic fibroblasts (MEFs) to investigate mitochondrial function. MEFs from mutant mice showed altered mitochondrial bioenergetics, with decreased basal oxygen consumption rate, ATP synthesis and mitochondrial membrane potential. Mitochondrial network formation and morphology was altered, with greatly reduced expression of fusogenic Opa1 isoforms. Mitochondrial alterations were also detected in cerebella of 18-month-old heterozygous mutants and may be a hallmark of disease. Pharmacological inhibition of de novo mitochondrial protein translation with chloramphenicol caused reversal of mitochondrial morphology in homozygous mutant MEFs, supporting the relevance of mitochondrial proteotoxicity for SCA28 pathogenesis and therapy development.


Subject(s)
ATP-Dependent Proteases/genetics , ATPases Associated with Diverse Cellular Activities/genetics , Disease Models, Animal , Mitochondria/metabolism , Spinocerebellar Ataxias/congenital , Animals , Female , Gene Knock-In Techniques , Membrane Potential, Mitochondrial , Mice, Inbred C57BL , Mitochondrial Proteins/metabolism , Mutation, Missense , Purkinje Cells/physiology , Purkinje Cells/ultrastructure , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/metabolism , Spinocerebellar Ataxias/pathology
5.
Mol Cell ; 42(1): 84-95, 2011 Apr 08.
Article in English | MEDLINE | ID: mdl-21474070

ABSTRACT

Adrenergic stimulation of the heart engages cAMP and phosphoinositide second messenger signaling cascades. Cardiac phosphoinositide 3-kinase p110γ participates in these processes by sustaining ß-adrenergic receptor internalization through its catalytic function and by controlling phosphodiesterase 3B (PDE3B) activity via an unknown kinase-independent mechanism. We have discovered that p110γ anchors protein kinase A (PKA) through a site in its N-terminal region. Anchored PKA activates PDE3B to enhance cAMP degradation and phosphorylates p110γ to inhibit PIP(3) production. This provides local feedback control of PIP(3) and cAMP signaling events. In congestive heart failure, p110γ is upregulated and escapes PKA-mediated inhibition, contributing to a reduction in ß-adrenergic receptor density. Pharmacological inhibition of p110γ normalizes ß-adrenergic receptor density and improves contractility in failing hearts.


Subject(s)
A Kinase Anchor Proteins/metabolism , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Myocytes, Cardiac/metabolism , Phosphatidylinositol Phosphates/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Class Ib Phosphatidylinositol 3-Kinase/chemistry , Class Ib Phosphatidylinositol 3-Kinase/deficiency , Class Ib Phosphatidylinositol 3-Kinase/genetics , Cyclic AMP-Dependent Protein Kinase RIIalpha Subunit/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , DNA/genetics , Enzyme Activation , Enzyme Inhibitors/pharmacology , Heart Failure/drug therapy , Heart Failure/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Protein Interaction Mapping , Quinoxalines/pharmacology , Receptors, Adrenergic, beta/metabolism , Second Messenger Systems , Sequence Homology, Amino Acid , Thiazolidinediones/pharmacology
6.
PLoS Genet ; 12(12): e1006461, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27923065

ABSTRACT

Pain is necessary to alert us to actual or potential tissue damage. Specialized nerve cells in the body periphery, so called nociceptors, are fundamental to mediate pain perception and humans without pain perception are at permanent risk for injuries, burns and mutilations. Pain insensitivity can be caused by sensory neurodegeneration which is a hallmark of hereditary sensory and autonomic neuropathies (HSANs). Although mutations in several genes were previously associated with sensory neurodegeneration, the etiology of many cases remains unknown. Using next generation sequencing in patients with congenital loss of pain perception, we here identify bi-allelic mutations in the FLVCR1 (Feline Leukemia Virus subgroup C Receptor 1) gene, which encodes a broadly expressed heme exporter. Different FLVCR1 isoforms control the size of the cytosolic heme pool required to sustain metabolic activity of different cell types. Mutations in FLVCR1 have previously been linked to vision impairment and posterior column ataxia in humans, but not to HSAN. Using fibroblasts and lymphoblastoid cell lines from patients with sensory neurodegeneration, we here show that the FLVCR1-mutations reduce heme export activity, enhance oxidative stress and increase sensitivity to programmed cell death. Our data link heme metabolism to sensory neuron maintenance and suggest that intracellular heme overload causes early-onset degeneration of pain-sensing neurons in humans.


Subject(s)
Membrane Transport Proteins/genetics , Nerve Degeneration/genetics , Oxidative Stress/genetics , Pain/genetics , Receptors, Virus/genetics , Apoptosis/genetics , Cell Line , Exome/genetics , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Frameshift Mutation/genetics , Heme/genetics , Humans , Immunoprecipitation , Male , Nerve Degeneration/pathology , Nociceptors/metabolism , Nociceptors/pathology , Pain/pathology , Primary Cell Culture , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/pathology
7.
Nucleic Acids Res ; 43(2): 862-74, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25567987

ABSTRACT

p63 is a crucial regulator of epidermal development, but its transcriptional control has remained elusive. Here, we report the identification of a long-range enhancer (p63LRE) that is composed of two evolutionary conserved modules (C38 and C40), acting in concert to control tissue- and layer-specific expression of the p63 gene. Both modules are in an open and active chromatin state in human and mouse keratinocytes and in embryonic epidermis, and are strongly bound by p63. p63LRE activity is dependent on p63 expression in embryonic skin, and also in the commitment of human induced pluripotent stem cells toward an epithelial cell fate. A search for other transcription factors involved in p63LRE regulation revealed that the CAAT enhancer binding proteins Cebpa and Cebpb and the POU domain-containing protein Pou3f1 repress p63 expression during keratinocyte differentiation by binding the p63LRE enhancer. Collectively, our data indicate that p63LRE is composed of additive and partly redundant enhancer modules that act to direct robust p63 expression selectively in the basal layer of the epidermis.


Subject(s)
Enhancer Elements, Genetic , Epidermis/embryology , Epidermis/metabolism , Gene Expression Regulation, Developmental , Keratinocytes/metabolism , Phosphoproteins/genetics , Trans-Activators/genetics , Animals , Cell Differentiation/genetics , Cells, Cultured , Humans , Keratinocytes/cytology , Mice, Inbred C57BL , Morphogenesis/genetics , Transcription Factors/metabolism , Transcriptional Activation
8.
Eur J Haematol ; 96(4): 367-74, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26058344

ABSTRACT

Diamond-Blackfan anemia (DBA) is a congenital pure red cell aplasia often associated with skeletal malformations. Mutations in ribosomal protein coding genes, mainly in RPS19, account for the majority of DBA cases. The molecular mechanisms underlying DBA pathogenesis are still not completely understood. Alternative spliced isoforms of FLVCR1 (feline leukemia virus subgroup C receptor 1) transcript coding for non-functional proteins have been reported in some DBA patients. Consistently, a phenotype very close to DBA has been described in animal models of FLVCR1 deficiency. FLVCR1 gene codes for two proteins: the plasma membrane heme exporter FLVCR1a and the mitochondrial heme exporter FLVCR1b. The coordinated expression of both FLVCR1 isoforms regulates an intracellular heme pool, necessary for proper expansion and differentiation of erythroid precursors. Here, we investigate the role of FLVCR1 isoforms in a cellular model of DBA. RPS19-downregulated TF1 cells show reduced FLVCR1a and FLVCR1b mRNA levels associated with heme overload. The downregulation of FLVCR1 isoforms affects cell cycle progression and apoptosis in differentiating K562 cells, a phenotype similar to DBA. Taken together, these data suggest that alteration of heme metabolism could play a role in the pathogenesis of DBA.


Subject(s)
Gene Expression Regulation, Leukemic , Heme/biosynthesis , Membrane Transport Proteins/genetics , RNA, Messenger/genetics , Receptors, Virus/genetics , Ribosomal Proteins/genetics , Alternative Splicing , Anemia, Diamond-Blackfan/genetics , Anemia, Diamond-Blackfan/metabolism , Anemia, Diamond-Blackfan/pathology , Apoptosis , Cell Cycle , Cell Line, Tumor , Cell Membrane/metabolism , Heme/agonists , Heme/antagonists & inhibitors , Humans , K562 Cells , Membrane Transport Proteins/metabolism , Mitochondria/metabolism , Models, Biological , Mutation , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Virus/antagonists & inhibitors , Receptors, Virus/metabolism , Ribosomal Proteins/antagonists & inhibitors , Ribosomal Proteins/metabolism
9.
Cereb Cortex ; 25(9): 2729-40, 2015 Sep.
Article in English | MEDLINE | ID: mdl-24735673

ABSTRACT

Alternative splicing in the brain is dynamic and instrumental to adaptive changes in response to stimuli. Lysine-specific demethylase 1 (LSD1/KDM1A) is a ubiquitously expressed histone H3Lys4 demethylase that acts as a transcriptional co-repressor in complex with its molecular partners CoREST and HDAC1/2. In mammalian brain, alternative splicing of LSD1 mini-exon E8a gives rise to neuroLSD1, a neurospecific isoform that, upon phosphorylation, acts as a dominant-negative causing disassembly of the co-repressor complex and de-repression of target genes. Here we show that the LSD1/neuroLSD1 ratio changes in response to neuronal activation and such effect is mediated by neurospecific splicing factors NOVA1 and nSR100/SRRM4 together with a novel cis-silencer. Indeed, we found that, in response to epileptogenic stimuli, downregulation of NOVA1 reduces exon E8a splicing and expression of neuroLSD1. Using behavioral and EEG analyses we observed that neuroLSD1-specific null mice are hypoexcitable and display decreased seizure susceptibility. Conversely, in a mouse model of Rett syndrome characterized by hyperexcitability, we measured higher levels of NOVA1 protein and upregulation of neuroLSD1. In conclusion, we propose that, in the brain, correct ratio between LSD1 and neuroLSD1 contributes to excitability and, when altered, could represent a pathogenic event associated with neurological disorders involving altered E/I.


Subject(s)
Alternative Splicing/genetics , Brain/pathology , Down-Regulation/genetics , Epilepsy/genetics , Histone Demethylases/metabolism , Neurons/physiology , Analysis of Variance , Animals , Antigens, Neoplasm/metabolism , Brain/physiopathology , Cell Line, Tumor , Chromatin Immunoprecipitation , Disease Models, Animal , Electroencephalography , Histone Demethylases/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Neuro-Oncological Ventral Antigen , Neuroblastoma/pathology , RNA-Binding Proteins/metabolism , Transfection
10.
Biochim Biophys Acta ; 1839(4): 259-64, 2014.
Article in English | MEDLINE | ID: mdl-24576667

ABSTRACT

The tissue-specific gene expression changes mediated by the hypoxia inducible factors (HIFs) allow the adaptation of cells to low oxygen tension and control several processes including erythropoiesis, angiogenesis and vasculogenesis. The Feline Leukemia Virus, subgroup C, Receptor 1 (Flvcr1) gene encodes for two isoforms, Flvcr1a and 1b, involved in the export of heme out of the cell and of mitochondria respectively. Studies in mouse models demonstrated a crucial role of Flvcr1 isoforms in erythropoiesis and during embryo development. Here, we showed the modulation of Flvcr1 gene expression in different tissues and cell lines in response to hypoxia. Chromatin immunoprecipitation analysis demonstrated that HIF2α and HIF-dependent transcription factor ETS1 (v-ets avian erythroblastosis virus E26 oncogene homolog 1) bind at the region -318/+39 of the Flvcr1 promoter. Analysis of Caco2 cells in which HIF2α or ETS1 were silenced or overexpressed demonstrated that, both HIF2α and ETS1 are involved in the transcriptional regulation of Flvcr1a and that HIF2α is absolutely required for Flvcr1a induction upon hypoxia. The inclusion of the Flvcr1 gene in the group of HIF2α-responsive genes strengthens its role in hypoxia-stimulated processes like erythropoiesis, vasculogenesis and heme absorption.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Hypoxia/genetics , Membrane Transport Proteins/genetics , Proto-Oncogene Protein c-ets-1/genetics , Receptors, Virus/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Caco-2 Cells , Embryonic Development , Erythropoiesis/genetics , Gene Expression Regulation, Developmental , Humans , Membrane Transport Proteins/metabolism , Mice , Neovascularization, Physiologic/genetics , Organ Specificity , Proto-Oncogene Protein c-ets-1/metabolism , Receptors, Virus/metabolism
11.
Gastroenterology ; 146(5): 1325-38, 2014 May.
Article in English | MEDLINE | ID: mdl-24486949

ABSTRACT

BACKGROUND & AIMS: The liver has one of the highest rates of heme synthesis of any organ. More than 50% of the heme synthesized in the liver is used for synthesis of P450 enzymes, which metabolize exogenous and endogenous compounds that include natural products, hormones, drugs, and carcinogens. Feline leukemia virus subgroup C cellular receptor 1a (FLVCR1a) is plasma membrane heme exporter that is ubiquitously expressed and controls intracellular heme content in hematopoietic lineages. We investigated the role of Flvcr1a in liver function in mice. METHODS: We created mice with conditional disruption of Mfsd7b, which encodes Flvcr1a, in hepatocytes (Flvcr1a(fl/fl);alb-cre mice). Mice were analyzed under basal conditions, after phenylhydrazine-induced hemolysis, and after induction of cytochromes P450 synthesis. Livers were collected and analyzed by histologic, quantitative real-time polymerase chain reaction, and immunoblot analyses. Hepatic P450 enzymatic activities were measured. RESULTS: Flvcr1a(fl/fl);alb-cre mice accumulated heme and iron in liver despite up-regulation of heme oxygenase 1, ferroportin, and ferritins. Hepatic heme export activity of Flvcr1a was closely associated with heme biosynthesis, which is required to sustain cytochrome induction. Upon cytochromes P450 stimulation, Flvcr1a(fl/fl);alb-cre mice had reduced cytochrome activity, associated with accumulation of heme in hepatocytes. The expansion of the cytosolic heme pool in these mice was likely responsible for the early inhibition of heme synthesis and increased degradation of heme, which reduced expression and activity of cytochromes P450. CONCLUSIONS: In livers of mice, Flvcr1a maintains a free heme pool that regulates heme synthesis and degradation as well as cytochromes P450 expression and activity. These findings have important implications for drug metabolism.


Subject(s)
Cytochrome P-450 Enzyme System/biosynthesis , Heme/metabolism , Hepatocytes/enzymology , Membrane Transport Proteins/metabolism , Receptors, Virus/metabolism , Animals , Benzo(a)pyrene/pharmacology , Cation Transport Proteins/metabolism , Cytochrome P-450 Enzyme System/genetics , Dexamethasone/pharmacology , Enzyme Induction , Ferritins/metabolism , Heme/biosynthesis , Heme Oxygenase-1/metabolism , Hemolysis , Hepatocytes/drug effects , Homeostasis , Imidazoles/pharmacology , Membrane Proteins/metabolism , Membrane Transport Proteins/genetics , Mice , Mice, Knockout , Phenylhydrazines/pharmacology , RNA, Messenger/metabolism , Receptors, Virus/genetics
12.
Haematologica ; 100(6): 720-9, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25795718

ABSTRACT

Feline leukemia virus subgroup C receptor 1 (Flvcr1) encodes two heme exporters: FLVCR1a, which localizes to the plasma membrane, and FLVCR1b, which localizes to mitochondria. Here, we investigated the role of the two Flvcr1 isoforms during erythropoiesis. We showed that, in mice and zebrafish, Flvcr1a is required for the expansion of committed erythroid progenitors but cannot drive their terminal differentiation, while Flvcr1b contributes to the expansion phase and is required for differentiation. FLVCR1a-down-regulated K562 cells have defective proliferation, enhanced differentiation, and heme loading in the cytosol, while FLVCR1a/1b-deficient K562 cells show impairment in both proliferation and differentiation, and accumulate heme in mitochondria. These data support a model in which the coordinated expression of Flvcr1a and Flvcr1b contributes to control the size of the cytosolic heme pool required to sustain metabolic activity during the expansion of erythroid progenitors and to allow hemoglobinization during their terminal maturation. Consistently, reduction or increase of the cytosolic heme rescued the erythroid defects in zebrafish deficient in Flvcr1a or Flvcr1b, respectively. Thus, heme export represents a tightly regulated process that controls erythropoiesis.


Subject(s)
Cell Differentiation/physiology , Erythropoiesis/physiology , Heme/metabolism , Intracellular Fluid/metabolism , Membrane Transport Proteins/physiology , Receptors, Virus/physiology , Amino Acid Sequence , Animals , Humans , K562 Cells , Mice , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Zebrafish
13.
J Immunol ; 191(11): 5451-9, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24154625

ABSTRACT

Hemopexin (Hx) is an acute-phase protein synthesized by hepatocytes in response to the proinflammatory cytokines IL-6, IL-1ß, and TNF-α. Hx is the plasma protein with the highest binding affinity to heme and controls heme-iron availability in tissues and also in T lymphocytes, where it modulates their responsiveness to IFN-γ. Recent data have questioned regarding an anti-inflammatory role of Hx, a role that may be both heme-binding dependent and independent. The aim of this study was to investigate the role of Hx in the development of a T cell-mediated inflammatory autoimmune response. During experimental autoimmune encephalomyelitis (EAE), the mouse model of multiple sclerosis, Hx content in serum increased and remained high. When EAE was induced in Hx knockout (Hx(-/-)) mice, they developed a clinically earlier and exacerbated EAE compared with wild-type mice, associated to a higher amount of CD4(+)-infiltrating T cells. The severe EAE developed by Hx(-/-) mice could be ascribed to an enhanced expansion of Th17 cells accounting for both a higher disposition of naive T cells to differentiate toward the Th17 lineage and a higher production of Th17 differentiating cytokines IL-6 and IL-23 by APCs. When purified human Hx was injected in Hx(-/-) mice before EAE induction, Th17 expansion, as well as disease severity, were comparable with those of wild-type mice. Taken together, these data indicate that Hx has a negative regulatory role in Th17-mediated inflammation and prospect its pharmacological use to limit the expansion of this cell subset in inflammatory and autoimmune disease.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Hemopexin/metabolism , Hepatocytes/immunology , Multiple Sclerosis/immunology , Th17 Cells/immunology , Animals , Cells, Cultured , Cytokines/immunology , Disease Models, Animal , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/genetics , Hemopexin/genetics , Hemopexin/immunology , Humans , Immunity, Cellular/genetics , Immunosuppression Therapy , Inflammation Mediators/immunology , Male , Mice , Mice, 129 Strain , Mice, Knockout , Multiple Sclerosis/genetics
14.
J Am Soc Nephrol ; 25(2): 316-28, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24136918

ABSTRACT

Spermatogonial stem cells reside in specific niches within seminiferous tubules and continuously generate differentiating daughter cells for production of spermatozoa. Although spermatogonial stem cells are unipotent, these cells are able to spontaneously convert to germline cell-derived pluripotent stem cells (GPSCs) in vitro. GPSCs have many properties of embryonic stem cells and are highly plastic, but their therapeutic potential in tissue regeneration has not been fully explored. Using a novel renal epithelial differentiation protocol, we obtained GPSC-derived tubular-like cells (GTCs) that were functional in vitro, as demonstrated through transepithelial electrical resistance analysis. In mice, GTCs injected after ischemic renal injury homed to the renal parenchyma, and GTC-treated mice showed reduced renal oxidative stress, tubular apoptosis, and cortical damage and upregulated tubular expression of the antioxidant enzyme hemeoxygenase-1. Six weeks after ischemic injury, kidneys of GTC-treated mice had less fibrosis and inflammatory infiltrate than kidneys of vehicle-treated mice. In conclusion, we show that GPSCs can be differentiated into functionally active renal tubular-like cells that therapeutically prevent chronic ischemic damage in vivo, introducing the potential utility of GPSCs in regenerative cell therapy.


Subject(s)
Acute Kidney Injury/surgery , Adult Stem Cells/transplantation , Kidney Tubules/cytology , Reperfusion Injury/surgery , Spermatogonia/cytology , Stem Cell Transplantation , Acute Kidney Injury/pathology , Acute Kidney Injury/prevention & control , Animals , Apoptosis , Biomarkers , Cell Differentiation , Cell Movement , Cells, Cultured , Collagen Type IV/pharmacology , Electric Impedance , Embryoid Bodies , Female , Fibrosis , Gene Expression Profiling , Heme Oxygenase-1/analysis , Kidney Failure, Chronic/prevention & control , Male , Membrane Proteins/analysis , Mice , Oxidative Stress , Postoperative Complications/etiology , Regenerative Medicine/methods , Reperfusion Injury/pathology , Seminiferous Tubules/cytology , Stem Cell Transplantation/adverse effects , Teratoma/etiology
15.
Circulation ; 127(12): 1317-29, 2013 Mar 26.
Article in English | MEDLINE | ID: mdl-23446829

ABSTRACT

BACKGROUND: Hemolytic diseases are characterized by enhanced intravascular hemolysis resulting in heme-catalyzed reactive oxygen species generation, which leads to endothelial dysfunction and oxidative damage. Hemopexin (Hx) is a plasma heme scavenger able to prevent endothelial damage and tissue congestion in a model of heme overload. Here, we tested whether Hx could be used as a therapeutic tool to counteract heme toxic effects on the cardiovascular system in hemolytic diseases. METHODS AND RESULTS: By using a model of heme overload in Hx-null mice, we demonstrated that heme excess in plasma, if not bound to Hx, promoted the production of reactive oxygen species and the induction of adhesion molecules and caused the reduction of nitric oxide availability. Then, we used ß-thalassemia and sickle cell disease mice as models of hemolytic diseases to evaluate the efficacy of an Hx-based therapy in the treatment of vascular dysfunction related to heme overload. Our data demonstrated that Hx prevented heme-iron loading in the cardiovascular system, thus limiting the production of reactive oxygen species, the induction of adhesion molecules, and the oxidative inactivation of nitric oxide synthase/nitric oxide, and promoted heme recovery and detoxification by the liver mainly through the induction of heme oxygenase activity. Moreover, we showed that in sickle cell disease mice, endothelial activation and oxidation were associated with increased blood pressure and altered cardiac function, and the administration of exogenous Hx was found to almost completely normalize these parameters. CONCLUSIONS: Hemopexin treatment is a promising novel therapy to protect against heme-induced cardiovascular dysfunction in hemolytic disorders.


Subject(s)
Anemia, Sickle Cell/drug therapy , Cardiovascular System/physiopathology , Endothelium, Vascular/physiopathology , Heme/adverse effects , Hemopexin/therapeutic use , beta-Thalassemia/drug therapy , Anemia, Sickle Cell/metabolism , Anemia, Sickle Cell/physiopathology , Animals , Cardiovascular System/drug effects , Disease Models, Animal , Endothelium, Vascular/drug effects , Heme/metabolism , Hemopexin/genetics , Hemopexin/pharmacology , Mice , Mice, Knockout , Mice, SCID , Nitric Oxide/metabolism , Reactive Oxygen Species/metabolism , Treatment Outcome , beta-Thalassemia/metabolism , beta-Thalassemia/physiopathology
16.
Biomolecules ; 14(2)2024 Jan 26.
Article in English | MEDLINE | ID: mdl-38397386

ABSTRACT

Feline leukemia virus C receptor 1a (FLVCR1a), initially identified as a retroviral receptor and localized on the plasma membrane, has emerged as a crucial regulator of heme homeostasis. Functioning as a positive regulator of δ-aminolevulinic acid synthase 1 (ALAS1), the rate-limiting enzyme in the heme biosynthetic pathway, FLVCR1a influences TCA cycle cataplerosis, thus impacting TCA flux and interconnected metabolic pathways. This study reveals an unexplored link between FLVCR1a, heme synthesis, and cholesterol production in endothelial cells. Using cellular models with manipulated FLVCR1a expression and inducible endothelial-specific Flvcr1a-null mice, we demonstrate that FLVCR1a-mediated control of heme synthesis regulates citrate availability for cholesterol synthesis, thereby influencing cellular cholesterol levels. Moreover, alterations in FLVCR1a expression affect membrane cholesterol content and fluidity, supporting a role for FLVCR1a in the intricate regulation of processes crucial for vascular development and endothelial function. Our results underscore FLVCR1a as a positive regulator of heme synthesis, emphasizing its integration with metabolic pathways involved in cellular energy metabolism. Furthermore, this study suggests that the dysregulation of heme metabolism may have implications for modulating lipid metabolism. We discuss these findings in the context of FLVCR1a's potential heme-independent function as a choline importer, introducing additional complexity to the interplay between heme and lipid metabolism.


Subject(s)
Citric Acid Cycle , Endothelial Cells , Mice , Animals , Endothelial Cells/metabolism , Membrane Transport Proteins/metabolism , Cell Membrane/metabolism , Mice, Knockout , Heme/metabolism
17.
Biomolecules ; 14(3)2024 Feb 26.
Article in English | MEDLINE | ID: mdl-38540698

ABSTRACT

In recent years, EVs have emerged as promising vehicles for coding and non-coding RNAs (ncRNAs), which have demonstrated remarkable potential as biomarkers for various diseases, including chronic liver diseases (CLDs). EVs are small, membrane-bound particles released by cells, carrying an arsenal of ncRNAs, including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and other ncRNA species, such as piRNAs, circRNAs, and tsRNAs. These ncRNAs act as key regulators of gene expression, splicing, and translation, providing a comprehensive molecular snapshot of the cells of origin. The non-invasive nature of EV sampling, typically via blood or serum collection, makes them highly attractive candidates for clinical biomarker applications. Moreover, EV-encapsulated ncRNAs offer unique advantages over traditional cell-free ncRNAs due to their enhanced stability within the EVs, hence allowing for their detection in circulation for extended periods and enabling more sensitive and reliable biomarker measurements. Numerous studies have investigated the potential of EV-enclosed ncRNAs as biomarkers for CLD. MiRNAs, in particular, have gained significant attention due to their ability to rapidly respond to changes in cellular stress and inflammation, hallmarks of CLD pathogenesis. Elevated levels of specific miRNAs have been consistently associated with various CLD subtypes, including metabolic dysfunction-associated steatotic liver disease (MASLD), metabolic dysfunction-associated steatohepatitis (MASH), and chronic hepatitis B and C. LncRNAs have also emerged as promising biomarkers for CLD. These transcripts are involved in a wide range of cellular processes, including liver regeneration, fibrosis, and cancer progression. Studies have shown that lncRNA expression profiles can distinguish between different CLD subtypes, providing valuable insights into disease progression and therapeutic response. Promising EV-enclosed ncRNA biomarkers for CLD included miR-122 (elevated levels of miR-122 are associated with MASLD progression and liver fibrosis), miR-21 (increased expression of miR-21 is linked to liver inflammation and fibrosis in CLD patients), miR-192 (elevated levels of miR-192 are associated with more advanced stages of CLD, including cirrhosis and HCC), LncRNA HOTAIR (increased HOTAIR expression is associated with MASLD progression and MASH development), and LncRNA H19 (dysregulation of H19 expression is linked to liver fibrosis and HCC progression). In the present review, we focus on the EV-enclosed ncRNAs as promising tools for the diagnosis and monitoring of CLD of various etiologies.


Subject(s)
Carcinoma, Hepatocellular , Extracellular Vesicles , Fatty Liver , Liver Neoplasms , MicroRNAs , RNA, Long Noncoding , Humans , RNA, Long Noncoding/genetics , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , RNA, Untranslated/physiology , MicroRNAs/genetics , Biomarkers/metabolism , Liver Cirrhosis/diagnosis , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Extracellular Vesicles/metabolism , Fatty Liver/pathology
18.
Antioxidants (Basel) ; 13(9)2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39334706

ABSTRACT

The relationship between serum 25-hydroxyvitamin D (25(OH)D) levels, genomic response to vitamin D (Vit.D), and positivity to SARS-CoV-2 remains understudied. In this pilot study, during the follow-up of patients with Inflammatory Bowel Disease (IBD) and COVID-19, we investigated this issue by analyzing the molecular contents of serum extracellular vesicles (EVs) from six groups of IBD patients (n = 32), classified according to anti-SARS-CoV-2 status, 25(OH)D level, and Vit.D supplementation, by small RNA-seq. This analysis revealed differentially expressed miRNAs, PIWI-RNA, transfer RNA, small nucleolar RNAs, and protein-coding RNAs in the EVs obtained from these cohorts of IBD patients. Experimental validation evidenced a statistically significant increase in miR30d-5p, miR150-5p, Let-7f-5p, and Let-7a-5p in the anti-SARS-CoV-2-positive and low 25(OH)D and Vit.D supplemented groups with respect to the non-Vit.D supplemented group, indicating their responsiveness to Vit.D treatment. Bioinformatics analysis highlighted the regulation of these validated miRNAs by oxidative stress and inflammation, hallmarks of IBD and COVID-19. Our study reports an unprecedented panel of circulating EV-enclosed inflammation- and oxidative stress-related miRNAs, the potentiality of which, as biomarkers for Vit.D responsivity in IBD patients, needs to be explored in future studies on larger cohorts in order to allow clinicians to optimize current treatment strategies upon viral infection.

19.
EMBO Mol Med ; 2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39271959

ABSTRACT

Defining the molecular mechanisms underlying cardiac resilience is crucial to find effective approaches to protect the heart. A physiologic level of ROS is produced in the heart by fatty acid oxidation, but stressful events can boost ROS and cause mitochondrial dysfunction and cardiac functional impairment. Melusin is a muscle specific chaperone required for myocardial compensatory remodeling during stress. Here we report that Melusin localizes in mitochondria where it binds the mitochondrial trifunctional protein, a key enzyme in fatty acid oxidation, and decreases it activity. Studying both mice and human induced pluripotent stem cell-derived cardiomyocytes, we found that Melusin reduces lipid oxidation in the myocardium and limits ROS generation in steady state and during pressure overload and doxorubicin treatment, preventing mitochondrial dysfunction. Accordingly, the treatment with the lipid oxidation inhibitor Trimetazidine concomitantly with stressful stimuli limits ROS accumulation and prevents long-term heart dysfunction. These findings disclose a physiologic mechanism of metabolic regulation in the heart and demonstrate that a timely restriction of lipid metabolism represents a potential therapeutic strategy to improve cardiac resilience to stress.

20.
Cell Rep Med ; 5(7): 101647, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39019006

ABSTRACT

Congenital hydrocephalus (CH), occurring in approximately 1/1,000 live births, represents an important clinical challenge due to the limited knowledge of underlying molecular mechanisms. The discovery of novel CH genes is thus essential to shed light on the intricate processes responsible for ventricular dilatation in CH. Here, we identify FLVCR1 (feline leukemia virus subgroup C receptor 1) as a gene responsible for a severe form of CH in humans and mice. Mechanistically, our data reveal that the full-length isoform encoded by the FLVCR1 gene, FLVCR1a, interacts with the IP3R3-VDAC complex located on mitochondria-associated membranes (MAMs) that controls mitochondrial calcium handling. Loss of Flvcr1a in mouse neural progenitor cells (NPCs) affects mitochondrial calcium levels and energy metabolism, leading to defective cortical neurogenesis and brain ventricle enlargement. These data point to defective NPCs calcium handling and metabolic activity as one of the pathogenetic mechanisms driving CH.


Subject(s)
Calcium , Hydrocephalus , Membrane Transport Proteins , Mitochondria , Neural Stem Cells , Receptors, Virus , Animals , Humans , Mice , Calcium/metabolism , Hydrocephalus/metabolism , Hydrocephalus/genetics , Hydrocephalus/pathology , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Inositol 1,4,5-Trisphosphate Receptors/genetics , Membrane Transport Proteins/metabolism , Membrane Transport Proteins/genetics , Mitochondria/metabolism , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Neurogenesis/genetics , Receptors, Virus/metabolism , Receptors, Virus/genetics
SELECTION OF CITATIONS
SEARCH DETAIL