Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
NPJ Precis Oncol ; 8(1): 29, 2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38448512

ABSTRACT

ALK and ROS1 fusions are effectively targeted by tyrosine kinase inhibitors (TKIs), however patients inevitably relapse after an initial response, often due to kinase domain mutations. We investigated circulating DNA from TKI-relapsed NSCLC patients by deep-sequencing. New EML4::ALK substitutions, L1198R, C1237Y and L1196P, were identified in the plasma of NSCLC ALK patients and characterized in a Ba/F3 cell model. Variants C1237Y and L1196P demonstrated pan-inhibitor resistance across 5 clinical and 2 investigational TKIs.

2.
Int J Mol Sci ; 24(16)2023 Aug 13.
Article in English | MEDLINE | ID: mdl-37628929

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD, including nonalcoholic fatty liver (NAFL) and nonalcoholic steatohepatitis (NASH)) is a high-prevalence disorder, affecting about 1 billion people, which can evolve to more severe conditions like cirrhosis or hepatocellular carcinoma. NAFLD is often concomitant with conditions of the metabolic syndrome, such as central obesity and insulin-resistance, but a specific drug able to revert NAFL and prevent its evolution towards NASH is still lacking. With the liver being a key organ in metabolic processes, the potential therapeutic strategies are many, and range from directly targeting the lipid metabolism to the prevention of tissue inflammation. However, side effects have been reported for the drugs tested up to now. In this review, different approaches to the treatment of NAFLD are presented, including newer therapies and ongoing clinical trials. Particular focus is placed on the reverse cholesterol transport system and on the agonists for nuclear factors like PPAR and FXR, but also drugs initially developed for other conditions such as incretins and thyromimetics along with validated natural compounds that have anti-inflammatory potential. This work provides an overview of the different therapeutic strategies currently being tested for NAFLD, other than, or along with, the recommendation of weight loss.


Subject(s)
Liver Neoplasms , Non-alcoholic Fatty Liver Disease , Humans , Non-alcoholic Fatty Liver Disease/drug therapy , Lipid Metabolism , Biological Transport
3.
Cancers (Basel) ; 15(14)2023 Jul 14.
Article in English | MEDLINE | ID: mdl-37509285

ABSTRACT

Infant acute myeloid leukemia (AML) is a heterogeneous disease, genetically distinct from its adult counterpart. Chromosomal translocations involving the KMT2A gene (MLL) are especially common in affected infants of less than 1 year of age, and are associated with a dismal prognosis. While these rearrangements are likely to arise in utero, the cell of origin has not been conclusively identified. This knowledge could lead to a better understanding of the biology of the disease and support the identification of new therapeutic vulnerabilities. Over the last few years, important progress in understanding the dynamics of fetal hematopoiesis has been made. Several reports have highlighted how hematopoietic stem cells (HSC) provide little contribution to fetal hematopoiesis, which is instead largely sustained by HSC-independent progenitors. Here, we used conditional Cre-Lox transgenic mouse models to engineer the Mll-Af9 translocation in defined subsets of embryonic hematopoietic progenitors. We show that embryonic hematopoiesis is generally permissive for Mll-Af9-induced leukemic transformation. Surprisingly, the selective introduction of Mll-Af9 in HSC-independent progenitors generated a transplantable myeloid leukemia, whereas it did not when introduced in embryonic HSC-derived cells. Ex vivo engineering of the Mll-Af9 rearrangement in HSC-independent progenitors using a CRISPR/Cas9-based approach resulted in the activation of an aberrant myeloid-biased self-renewal program. Overall, our results demonstrate that HSC-independent hematopoietic progenitors represent a permissive environment for Mll-Af9-induced leukemic transformation, and can likely act as cells of origin of infant AML.

4.
Int J Mol Sci ; 23(8)2022 Apr 14.
Article in English | MEDLINE | ID: mdl-35457155

ABSTRACT

The use of nanoparticles (NPs) has surely grown in recent years due to their versatility, with a spectrum of applications that range from nanomedicine to the food industry. Recent research focuses on the development of NPs for the oral administration route rather than the intravenous one, placing the interactions between NPs and the intestine at the centre of the attention. This allows the NPs functionalization to exploit the different characteristics of the digestive tract, such as the different pH, the intestinal mucus layer, or the intestinal absorption capacity. On the other hand, these same characteristics can represent a problem for their complexity, also considering the potential interactions with the food matrix or the microbiota. This review intends to give a comprehensive look into three main branches of NPs delivery through the oral route: the functionalization of NPs drug carriers for systemic targets, with the case of insulin carriers as an example; NPs for the delivery of drugs locally active in the intestine, for the treatment of inflammatory bowel diseases and colon cancer; finally, the potential concerns and side effects of the accidental and uncontrolled exposure to NPs employed as food additives, with focus on E171 (titanium dioxide) and E174 (silver NPs).


Subject(s)
Metal Nanoparticles , Nanoparticles , Administration, Oral , Food Additives , Gastrointestinal Tract , Intestinal Absorption , Intestines
5.
Cells ; 11(6)2022 03 21.
Article in English | MEDLINE | ID: mdl-35326511

ABSTRACT

Our knowledge of the complexity of the developing hematopoietic system has dramatically expanded over the course of the last few decades. We now know that, while hematopoietic stem cells (HSCs) firmly reside at the top of the adult hematopoietic hierarchy, multiple HSC-independent progenitor populations play variegated and fundamental roles during fetal life, which reflect on adult physiology and can lead to disease if subject to perturbations. The importance of obtaining a high-resolution picture of the mechanisms by which the developing embryo establishes a functional hematopoietic system is demonstrated by many recent indications showing that ontogeny is a primary determinant of function of multiple critical cell types. This review will specifically focus on exploring the diversity of hematopoietic stem and progenitor cells unique to embryonic and fetal life. We will initially examine the evidence demonstrating heterogeneity within the hemogenic endothelium, precursor to all definitive hematopoietic cells. Next, we will summarize the dynamics and characteristics of the so-called "hematopoietic waves" taking place during vertebrate development. For each of these waves, we will define the cellular identities of their components, the extent and relevance of their respective contributions as well as potential drivers of heterogeneity.


Subject(s)
Hematopoiesis , Hematopoietic Stem Cells , Embryo, Mammalian , Hematopoietic Stem Cells/metabolism
6.
World J Gastroenterol ; 28(4): 449-463, 2022 Jan 28.
Article in English | MEDLINE | ID: mdl-35125829

ABSTRACT

Celiac disease (CeD) is a multifactorial autoimmune disorder spread worldwide. The exposure to gluten, a protein found in cereals like wheat, barley and rye, is the main environmental factor involved in its pathogenesis. Even if the genetic predisposition represented by HLA-DQ2 or HLA-DQ8 haplotypes is widely recognised as mandatory for CeD development, it is not enough to explain the total predisposition for the disease. Furthermore, the onset of CeD comprehend a wide spectrum of symptoms, that often leads to a delay in CeD diagnosis. To overcome this deficiency and help detecting people with increased risk for CeD, also clarifying CeD traits linked to disease familiarity, different studies have tried to make light on other predisposing elements. These were in many cases genetic variants shared with other autoimmune diseases. Since inherited traits can be regulated by epigenetic modifications, also induced by environmental factors, the most recent studies focused on the potential involvement of epigenetics in CeD. Epigenetic factors can in fact modulate gene expression with many mechanisms, generating more or less stable changes in gene expression without affecting the DNA sequence. Here we analyze the different epigenetic modifications in CeD, in particular DNA methylation, histone modifications, non-coding RNAs and RNA methylation. Special attention is dedicated to the additional predispositions to CeD, the involvement of epigenetics in developing CeD complications, the pathogenic pathways modulated by epigenetic factors such as microRNAs and the potential use of epigenetic profiling as biomarker to discriminate different classes of patients.


Subject(s)
Celiac Disease , MicroRNAs , Celiac Disease/diagnosis , Celiac Disease/genetics , Epigenesis, Genetic , Genetic Predisposition to Disease , Glutens , Humans , MicroRNAs/genetics
7.
Int J Mol Sci ; 22(3)2021 Jan 28.
Article in English | MEDLINE | ID: mdl-33525473

ABSTRACT

Celiac disease (CD) is an autoimmune enteropathy arising in genetically predisposed subjects exposed to gluten, which activates both innate and adaptive immunity. Although the pathogenesis is common to all patients, the clinical spectrum is quite variable, and differences could be explained by gene expression variations. Among the factors able to affect gene expression, there are lncRNAs. We evaluated the expression profile of 87 lncRNAs in CD vs. healthy control (HC) intestinal biopsies by RT-qPCR array. Nuclear enriched abundant transcript 1 (NEAT1) and taurine upregulated gene 1 (TUG1) were detected as downregulated in CD patients at diagnosis, but their expression increased in biopsies of patients on a gluten-free diet (GFD) exposed to gluten. The increase in NEAT1 expression after gluten exposure was mediated by IL-15 and STAT3 activation and binding to the NEAT1 promoter, as demonstrated by gel shift assay. NEAT1 is localized in the nucleus and can regulate gene expression by sequestering transcription factors, and it has been implicated in immune regulation and control of cell proliferation. The demonstration of its regulation by gluten thus also supports the role of lncRNAs in CD and prompts further research on these RNAs as gene expression regulators.


Subject(s)
Celiac Disease/genetics , Down-Regulation , Duodenum/chemistry , Gliadin/adverse effects , RNA, Long Noncoding/genetics , Adult , Case-Control Studies , Celiac Disease/immunology , Cell Proliferation , Cells, Cultured , Child , Down-Regulation/drug effects , Duodenum/immunology , Female , Gene Expression Regulation , Humans , Immunity, Innate , Interleukin-15/genetics , Intestinal Mucosa/chemistry , Intestinal Mucosa/immunology , Male , STAT3 Transcription Factor/genetics
8.
Front Cell Dev Biol ; 8: 618164, 2020.
Article in English | MEDLINE | ID: mdl-33511126

ABSTRACT

Several lines of evidence suggest that childhood leukemia, the most common cancer in young age, originates during in utero development. However, our knowledge of the cellular origin of this large and heterogeneous group of malignancies is still incomplete. The identification and characterization of their cell of origin is of crucial importance in order to define the processes that initiate and sustain disease progression, to refine faithful animal models and to identify novel therapeutic approaches. During embryogenesis, hematopoiesis takes place at different anatomical sites in sequential waves, and occurs in both a hematopoietic stem cell (HSC)-dependent and a HSC-independent fashion. Despite the recently described relevance and complexity of HSC-independent hematopoiesis, few studies have so far investigated its potential involvement in leukemogenesis. Here, we review the current knowledge on prenatal origin of leukemias in the context of recent insights in developmental hematopoiesis.

9.
J Immunol ; 203(1): 247-258, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31127033

ABSTRACT

The signals that control endothelial plasticity in inflamed tissues have only been partially characterized. For example, it has been shown that inadequate vasculogenesis in systemic sclerosis (SSc) has been associated with an endothelial defect. We used a genetic lineage tracing model to investigate whether endothelial cells die or change phenotypically after fibrosis induction and whether signals released by cells of the innate immune system and in the blood of patients influence their commitment. We observed that in the lineage-tracing transgenic mice Cdh5-CreERT2::R26R-EYFP, endothelial-derived cells (EdCs) underwent fibrosis after treatment with bleomycin, and EdCs retrieved from the lung showed expression of endothelial-to-mesenchymal transition (EndoMT) markers. Liposome-encapsulated clodronate was used to assess macrophage impact on EdCs. Clodronate treatment affected the number of alternatively activated macrophages in the lung, with upregulated expression of EndoMT markers in lung EdCs. Endothelial fate and function were investigated in vitro upon challenge with serum signals from SSc patients or released by activated macrophages. Sera of SSc patients with anti-Scl70 Abs, at higher risk of visceral organ fibrosis, induced EndoMT and jeopardized endothelial function. In conclusion, EdCs in SSc might be defective because of commitment to a mesenchymal fate, which is sustained by soluble signals in the patient's blood. Macrophages contribute to preserve the endothelial identity of precursor cells. Altered macrophage-dependent plasticity of EdCs could contribute to link vasculopathy with fibrosis.


Subject(s)
Endothelium/physiology , Inflammation/immunology , Lung/pathology , Macrophages/physiology , Mesenchymal Stem Cells/physiology , Scleroderma, Systemic/immunology , Animals , Autoantibodies/metabolism , Cell Differentiation , Cell Lineage , Cell Plasticity , Cells, Cultured , Clodronic Acid , DNA Topoisomerases, Type I , Fibrosis , Humans , Immunity, Innate , Mice , Mice, Transgenic , Neovascularization, Pathologic , Nuclear Proteins/immunology
10.
Genes Nutr ; 10(5): 482, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26233308

ABSTRACT

Celiac disease is an intestinal disease which shows different symptoms and clinical manifestations among pediatric and adult patients. These variations could be imputable to age-related changes in gut architecture and intestinal immune system, which could be characterized by gene expression differences possibly regulated by miRNAs. We analyzed a panel of miRNAs and their target genes in duodenal biopsies of Marsh 3AB and 3C pediatric celiac patients, compared to controls. Moreover, to assess variation of expression in plasma samples, we evaluated circulating miRNA levels in controls and patients at diagnosis or on gluten-free diet. We detected a decreased miR-192-5p expression in celiac patients, but no variations in NOD2 and CXCL2, targets previously identified in adults. Conversely, we detected a significant increase in mRNA and protein levels of another target, MAD2L1, protein related to cell cycle control. miR-31-5p and miR-338-3p were down-regulated and their respective targets, FOXP3 and RUNX1, involved in Treg function, resulted up-regulated in celiac patients. Finally, we detected, in celiac patients, an increased expression of miR-21-5p, possibly caused by a regulatory loop with its putative target STAT3, which showed an increased activation in Marsh 3C patients. The analysis of plasma revealed a trend similar to that observed in biopsies, but in presence of gluten-free diet we could not detect circulating miRNAs values comparable to controls. miRNAs and their gene targets showed an altered expression in duodenal mucosa and plasma of celiac disease pediatric patients, and these alterations could be different from adult ones.

11.
PLoS One ; 9(10): e108411, 2014.
Article in English | MEDLINE | ID: mdl-25285664

ABSTRACT

Emerging evidence has demonstrated that miRNA sequences can regulate skeletal myogenesis by controlling the process of myoblast proliferation and differentiation. However, at present a deep analysis of miRNA expression in control and FSHD myoblasts during differentiation has not yet been derived. To close this gap, we used a next-generation sequencing (NGS) approach applied to in vitro myogenesis. Furthermore, to minimize sample genetic heterogeneity and muscle-type specific patterns of gene expression, miRNA profiling from NGS data was filtered with FC ≥ 4 (log(2)FC ≥ 2) and p-value<0.05, and its validation was derived by qRT-PCR on myoblasts from seven muscle districts. In particular, control myogenesis showed the modulation of 38 miRNAs, the majority of which (34 out 38) were up-regulated, including myomiRs (miR-1, -133a, -133b and -206). Approximately one third of the modulated miRNAs were not previously reported to be involved in muscle differentiation, and interestingly some of these (i.e. miR-874, -1290, -95 and -146a) were previously shown to regulate cell proliferation and differentiation. FSHD myogenesis evidenced a reduced number of modulated miRNAs than healthy muscle cells. The two processes shared nine miRNAs, including myomiRs, although with FC values lower in FSHD than in control cells. In addition, FSHD cells showed the modulation of six miRNAs (miR-1268, -1268b, -1908, 4258, -4508- and -4516) not evidenced in control cells and that therefore could be considered FSHD-specific, likewise three novel miRNAs that seem to be specifically expressed in FSHD myotubes. These data further clarify the impact of miRNA regulation during control myogenesis and strongly suggest that a complex dysregulation of miRNA expression characterizes FSHD, impairing two important features of myogenesis: cell cycle and muscle development. The derived miRNA profiling could represent a novel molecular signature for FSHD that includes diagnostic biomarkers and possibly therapeutic targets.


Subject(s)
Gene Expression Regulation , High-Throughput Nucleotide Sequencing/methods , MicroRNAs/genetics , Muscle Development/genetics , Muscular Dystrophy, Facioscapulohumeral/genetics , Case-Control Studies , Cell Differentiation/genetics , Cell Line , Humans , MicroRNAs/metabolism , Muscular Dystrophy, Facioscapulohumeral/pathology , Myoblasts/pathology , Reproducibility of Results
12.
Am J Gastroenterol ; 109(10): 1662-74, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25070052

ABSTRACT

OBJECTIVES: microRNAs (miRNAs) are short RNAs that regulate gene expression in various processes, including immune response. Altered immune response is a pivotal event in the pathogenesis of celiac disease (CD), and miRNAs could have a role in modulating both innate and adaptive response to gluten in celiac patients. METHODS: We compared miRNA profiles in duodenal biopsies of controls and CD patients by miRNA array. Differentially expressed miRNAs were validated in controls, Marsh 3A-B, and Marsh 3C patients by quantitative PCR (qPCR). Target gene expression was assessed by qPCR, western blotting, and immunohistochemistry, and the effect of gliadin was evaluated by in vitro stimulation experiments on duodenal biopsies. RESULTS: Seven miRNAs were identified as significantly downregulated in the duodenum of adult CD patients as compared with controls. qPCR validated the decreased expression of miR-192-5p, miR-31-5p, miR-338-3p, and miR-197, in particular in patients with more severe histological lesions (Marsh 3C). In silico analysis of possible miRNA targets identified several genes involved in innate and adaptive immunity. Among these, chemokine C-X-C motif ligand 2 (CXCL2) and NOD2 showed significantly increased mRNA and protein level in Marsh 3C patients and a significant inverse correlation with the regulatory miR-192-5p. In addition, forkhead box P3 (FOXP3), Run-related transcription factor 1, and interleukin-18 (targets of miR-31-5p, miR-338-3p, and miR-197, respectively) showed upregulation in CD patients. Furthermore, alterations in CXCL2 and NOD2, FOXP3, miR-192-5p, and miR-31-5p expression were triggered by gliadin exposure in CD patients. CONCLUSIONS: miRNA expression is significantly altered in duodenal mucosa of CD patients, and this alteration can increase the expression of molecules involved in immune response.


Subject(s)
Adaptive Immunity/physiology , Celiac Disease/genetics , Celiac Disease/immunology , Immunity, Innate/physiology , MicroRNAs/metabolism , Adult , Case-Control Studies , Celiac Disease/metabolism , Cohort Studies , Duodenum/metabolism , Duodenum/pathology , Female , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , Microarray Analysis , Middle Aged
13.
PLoS One ; 7(8): e43335, 2012.
Article in English | MEDLINE | ID: mdl-22905258

ABSTRACT

Regeneration of muscle fibers, lost during pathological muscle degeneration or after injuries, is sustained by the production of new myofibers by means of the satellite cells. Survival of the satellite cells is a critical requirement for efficient muscle reconstitution. Necdin, a member of the MAGE proteins family, is expressed in satellite cell-derived myogenic precursors during perinatal growth and in the adult upon activation during muscle regeneration, where it plays an important role both in myoblast differentiation and survival. We show here that necdin exerts its pro-survival activity by counteracting the action of the pro-apoptotic protein Cell Cycle Apoptosis Regulatory Protein (CCAR1/CARP1) that we have identified as a new molecular interactor of necdin by two-hybrid screening. Necdin is responsible for the maintenance of CCAR1 protein levels, by implementing its ubiquitination and degradation through the proteasome. Taken together, these data shed new light on the molecular mechanism of necdin anti-apoptotic activity in myogenesis.


Subject(s)
Apoptosis Regulatory Proteins/biosynthesis , Apoptosis , Cell Cycle Proteins/biosynthesis , Gene Expression Regulation , Myoblasts/cytology , Nerve Tissue Proteins/biosynthesis , Nuclear Proteins/biosynthesis , Ubiquitin-Protein Ligases/biosynthesis , Animals , Cell Differentiation , Cell Survival , Gene Library , Humans , Mice , Mice, Inbred C57BL , Muscle Development , Muscles/metabolism , Proteasome Endopeptidase Complex/metabolism , Satellite Cells, Skeletal Muscle/cytology , Two-Hybrid System Techniques
14.
Genomics ; 100(5): 289-96, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22824653

ABSTRACT

We performed a detailed genomic investigation of the chimpanzee locus syntenic to human chromosome 4q35.2, associated to the facioscapulohumeral dystrophy. Two contigs of approximately 150 kb and 200 kb were derived from PTR chromosomes 4q35 and 3p12, respectively: both regions showed a very similar sequence organization, including D4Z4 and Beta satellite linked clusters. Starting from these findings, we derived a hypothetical evolutionary history of human 4q35, 10q26 and 3p12 chromosome regions focusing on the D4Z4-Beta satellite linked organization. The D4Z4 unit showed an open reading frame (DUX4) at both PTR 4q35 and 3p12 regions; furthermore some subregions of the Beta satellite unit showed a high degree of conservation between chimpanzee and humans. In conclusion, this paper provides evidence that at the 4q subtelomere the linkage between D4Z4 and Beta satellite arrays is a feature that appeared late during evolution and is conserved between chimpanzee and humans.


Subject(s)
Chromosomes, Human, Pair 10/genetics , Chromosomes, Human, Pair 3/genetics , Chromosomes, Human, Pair 4/genetics , Evolution, Molecular , Muscular Dystrophy, Facioscapulohumeral/genetics , Pan troglodytes/genetics , Animals , Base Sequence , Blotting, Southern , Contig Mapping , Genetic Linkage , Genomic Library , Homeodomain Proteins/genetics , Humans , In Situ Hybridization, Fluorescence , Molecular Sequence Data , Multigene Family/genetics , Sequence Analysis, DNA , Species Specificity , Synteny/genetics
15.
PLoS One ; 6(6): e20966, 2011.
Article in English | MEDLINE | ID: mdl-21695143

ABSTRACT

BACKGROUND: Determine global gene dysregulation affecting 4q-linked (FSHD-1) and non 4q-linked (FSHD-2) cells during early stages of myogenic differentiation. This approach has been never applied to FSHD pathogenesis. METHODOLOGY/PRINCIPAL FINDINGS: By in vitro differentiation of FSHD-1 and FSHD-2 myoblasts and gene chip analysis we derived that gene expression profile is altered only in FSHD-1 myoblasts and FSHD-2 myotubes. The changes seen in FSHD-1 regarded a general defect in cell cycle progression, probably due to the upregulation of myogenic markers PAX3 and MYOD1, and a deficit of factors (SUV39H1 and HMGB2) involved in D4Z4 chromatin conformation. On the other hand, FSHD-2 mytubes were characterized by a general defect in RNA metabolism, protein synthesis and degradation and, to a lesser extent, in cell cycle. Common dysregulations regarded genes involved in response to oxidative stress and in sterol biosynthetic process. Interestingly, our results also suggest that miRNAs might be implied in both FSHD-1 and FSHD-2 gene dysregulation. Finally, in both cell differentiation systems, we did not observe a gradient of altered gene expression throughout the 4q35 chromosome. CONCLUSIONS/SIGNIFICANCE: FSHD-1 and FSHD-2 cells showed, in different steps of myogenic differentiation, a global deregulation of gene expression rather than an alteration of expression of 4q35 specific genes. In general, FSHD-1 and FSHD-2 global gene deregulation interested common and distinctive biological processes. In this regard, defects of cell cycle progression (FSHD-1 and to a lesser extent FSHD-2), protein synthesis and degradation (FSHD-2), response to oxidative stress (FSHD-1 and FSHD-2), and cholesterol homeostasis (FSHD-1 and FSHD-2) may in general impair a correct myogenesis. Taken together our results recapitulate previously reported defects of FSHD-1, and add new insights into the gene deregulation characterizing both FSHD-1 and FSHD-2, in which miRNAs may play a role.


Subject(s)
Cell Differentiation/genetics , Gene Expression Profiling , Muscle Development/genetics , Nuclear Proteins/genetics , Adolescent , Adult , Aged , Case-Control Studies , Cell Line , Child , Child, Preschool , Female , Humans , Male , MicroRNAs/genetics , Microfilament Proteins , Middle Aged , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscular Dystrophy, Facioscapulohumeral/genetics , Muscular Dystrophy, Facioscapulohumeral/pathology , Myoblasts/cytology , Myoblasts/metabolism , Myoblasts/pathology , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , RNA-Binding Proteins , Reproducibility of Results , Young Adult
16.
BMC Biol ; 7: 41, 2009 Jul 16.
Article in English | MEDLINE | ID: mdl-19607661

ABSTRACT

BACKGROUND: Facioscapulohumeral muscular dystrophy (FSHD) is an autosomal dominant neuromuscular disorder associated with the partial deletion of integral numbers of 3.3 kb D4Z4 DNA repeats within the subtelomere of chromosome 4q. A number of candidate FSHD genes, adenine nucleotide translocator 1 gene (ANT1), FSHD-related gene 1 (FRG1), FRG2 and DUX4c, upstream of the D4Z4 array (FSHD locus), and double homeobox chromosome 4 (DUX4) within the repeat itself, are upregulated in some patients, thus suggesting an underlying perturbation of the chromatin structure. Furthermore, a mouse model overexpressing FRG1 has been generated, displaying skeletal muscle defects. RESULTS: In the context of myogenic differentiation, we compared the chromatin structure and tridimensional interaction of the D4Z4 array and FRG1 gene promoter, and FRG1 expression, in control and FSHD cells. The FRG1 gene was prematurely expressed during FSHD myoblast differentiation, thus suggesting that the number of D4Z4 repeats in the array may affect the correct timing of FRG1 expression. Using chromosome conformation capture (3C) technology, we revealed that the FRG1 promoter and D4Z4 array physically interacted. Furthermore, this chromatin structure underwent dynamic changes during myogenic differentiation that led to the loosening of the FRG1/4q-D4Z4 array loop in myotubes. The FRG1 promoter in both normal and FSHD myoblasts was characterized by H3K27 trimethylation and Polycomb repressor complex binding, but these repression signs were replaced by H3K4 trimethylation during differentiation. The D4Z4 sequences behaved similarly, with H3K27 trimethylation and Polycomb binding being lost upon myogenic differentiation. CONCLUSION: We propose a model in which the D4Z4 array may play a critical chromatin function as an orchestrator of in cis chromatin loops, thus suggesting that this repeat may play a role in coordinating gene expression.


Subject(s)
Chromatin Assembly and Disassembly/physiology , Muscle Development/genetics , Muscular Dystrophy, Facioscapulohumeral/genetics , Myoblasts, Skeletal/metabolism , Nuclear Proteins/metabolism , Cell Differentiation/genetics , Cells, Cultured , Chromatin/chemistry , Chromatin/metabolism , Chromosomes, Human, Pair 4/genetics , DNA-Binding Proteins/metabolism , Female , Gene Expression Regulation , Humans , Jumonji Domain-Containing Histone Demethylases , Male , Microfilament Proteins , Muscle Fibers, Skeletal/metabolism , Muscular Dystrophy, Facioscapulohumeral/metabolism , Myoblasts, Skeletal/ultrastructure , Nuclear Proteins/genetics , Oxidoreductases, N-Demethylating/metabolism , Polycomb-Group Proteins , Promoter Regions, Genetic , RNA-Binding Proteins , Repressor Proteins/metabolism , Tandem Repeat Sequences/physiology
17.
Mol Biol Cell ; 20(16): 3620-7, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19553472

ABSTRACT

Regeneration of muscle fibers, lost during pathological muscle degeneration or after injuries, is mediated by the production of new myofibres. This process, sustained by the resident stem cells of the muscle, the satellite cells, is finely regulated by local cues, in particular by cytokines and growth factors. Evidence in the literature suggests that nerve growth factor (NGF) is involved in muscle fiber regeneration; however, its role and mechanism of action were unclear. We have investigated this issue in in vivo mouse models of muscle regeneration and in primary myogenic cells. Our results demonstrate that NGF acts through its low-affinity receptor p75(NTR) in a developmentally regulated signaling pathway necessary to myogenic differentiation and muscle repair in vivo. We also demonstrate that this action of NGF is mediated by the down-regulation of RhoA-GTP signaling in myogenic cells.


Subject(s)
Muscle, Skeletal/metabolism , Muscle, Skeletal/physiology , Nerve Growth Factor/metabolism , Receptors, Nerve Growth Factor/metabolism , Regeneration/physiology , Satellite Cells, Skeletal Muscle/physiology , rhoA GTP-Binding Protein/metabolism , Animals , Cell Differentiation/physiology , Cell Fusion , Cells, Cultured , Cytoskeleton/metabolism , Humans , Mice , Muscle Fibers, Skeletal/physiology , Muscle Fibers, Skeletal/ultrastructure , Muscle, Skeletal/cytology , Muscle, Skeletal/pathology , Satellite Cells, Skeletal Muscle/cytology , Signal Transduction/physiology
18.
J Cell Sci ; 122(Pt 8): 1119-25, 2009 Apr 15.
Article in English | MEDLINE | ID: mdl-19339547

ABSTRACT

Skeletal muscles of subjects with advanced cancer undergo progressive wasting, referred to as cachexia. Cachexia is an important area for medical research because strategies proposed until now have yielded little benefit. We have recently identified necdin as a key player in fetal and postnatal physiological myogenesis and in muscle regeneration. Here we show that necdin is selectively expressed in muscles of cachetic mice and prove that its expression is causally linked to a protective response of the tissue against tumor-induced wasting, inhibition of myogenic differentiation and fiber regeneration. Necdin carries out this role mainly via interference with TNFalpha signaling at various levels, including regulation of expression of TNFR1 and p53, and regulation of the activity of caspase 3 and caspase 9. These data suggest that inhibition of muscle wasting using necdin is a feasible approach to treat cachexia in neoplastic patients.


Subject(s)
Adenocarcinoma/metabolism , Cachexia/prevention & control , Colonic Neoplasms/metabolism , Muscle, Skeletal/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Adenocarcinoma/complications , Adenocarcinoma/pathology , Animals , Cachexia/etiology , Cachexia/metabolism , Cachexia/pathology , Caspase 3/metabolism , Caspase 9/metabolism , Cell Differentiation , Cell Line, Tumor , Colonic Neoplasms/complications , Colonic Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Muscle, Skeletal/enzymology , Muscle, Skeletal/pathology , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Regeneration , Signal Transduction , Time Factors , Tumor Necrosis Factor-alpha/metabolism , Tumor Suppressor Protein p53/metabolism
19.
J Hepatol ; 49(1): 123-33, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18462824

ABSTRACT

BACKGROUND/AIMS: Many patients with hepatic iron overload do not have identifiable mutations and often present with metabolic disorders and hepatic steatosis. Since the pathophysiology of Dysmetabolic Hepatic Iron Overload (DHIO) is still obscure, the aim of this study was to evaluate, in these patients, possible alterations in iron-related molecule expression. METHODS: Iron-related gene mRNA levels were determined by quantitative-PCR in liver biopsies of subjects with NAFLD without iron overload and patients with HFE-hemochromatosis, beta-thalassemia major and DHIO. Urinary hepcidin was measured by immunoblotting. RESULTS: No alterations in mRNA expression of either iron transporters or exporters were found in DHIO. mRNA and urinary hepcidin levels normalized for the amount of iron overload showed a significantly lower ratio than in controls, although not as low as in hemochromatosis or beta-thalassemia. Differently from what observed in hemochromatosis, hepcidin mRNA did not correlate with urinary hepcidin. CONCLUSIONS: Patients with DHIO show appropriate regulation of mRNAs encoding proteins involved in iron uptake and efflux but dysregulation of hepcidin production. The relatively elevated urinary hepcidin can explain the iron phenotype in DHIO (more macrophage iron retention and low/normal transferrin saturation).


Subject(s)
Antimicrobial Cationic Peptides/genetics , Iron Overload/genetics , Iron/metabolism , Liver Diseases/genetics , Adult , Aged , Antigens, CD/genetics , Antimicrobial Cationic Peptides/urine , Biopsy , Cation Transport Proteins/genetics , Ceruloplasmin/genetics , Female , Genotype , Hemochromatosis/genetics , Hemochromatosis/urine , Hepcidins , Humans , Iron Overload/urine , Liver Diseases/pathology , Liver Diseases/urine , Male , Membrane Proteins/genetics , Middle Aged , RNA, Messenger/metabolism , Receptors, Transferrin/genetics
20.
J Neurochem ; 104(6): 1577-87, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17988240

ABSTRACT

GN11 and GT1-7 are immortalized gonadotropin-releasing hormone-positive murine cell lines exhibiting the features of immature olfactory neurons and differentiated hypothalamic neurons, respectively. Using electron microscopy and biochemical assays (RT-PCR and immunoblotting) we determined the presence of numerous caveolae invaginations and of caveolin-1 and -2 mRNAs and proteins in GN11 cells, and their absence in GT1-7 cells. The lack of caveolins in GT1-7 cells might be due to the silencing of gene transcription caused by estrogen receptor alpha whose inhibitory activity in GN11 cells could be counter-balanced by co-expression of caveolin-permissive estrogen receptor beta. To test whether the unique expression of caveolins in GN11 cells is related to their immature state, we treated GN11 cells for 24-72 h with retinoic acid or phorbol ester. Both treatments led to neuronal differentiation of GN11 cells, as shown by emission of long neuritic processes, increased expression of growth cone-associated protein-43 and appearance of voltage-gated K+ and C2+ channel currents. Concurrently, caveolins 1 and 2, and estrogen receptor beta were down-regulated in differentiated GN11, whereas estrogen receptor alpha was unaffected by differentiation. We conclude that caveolin expression in GN11 neurons is down-regulated upon differentiation and up-regulated by estrogen receptor beta.


Subject(s)
Antineoplastic Agents/pharmacology , Caveolin 1/genetics , Caveolin 2/genetics , Neurons/physiology , Phorbol Esters/pharmacology , Tretinoin/pharmacology , Animals , Brain Neoplasms , Calcium Channels/physiology , Cell Differentiation/drug effects , Cell Line, Tumor , Down-Regulation/drug effects , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/genetics , Gene Expression/drug effects , Gonadotropin-Releasing Hormone/metabolism , Membrane Potentials/drug effects , Mice , Neurons/cytology , Neurons/drug effects , Patch-Clamp Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...