Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Digit Health ; 10: 20552076241260507, 2024.
Article in English | MEDLINE | ID: mdl-38868368

ABSTRACT

Background: Wearable technology is used in healthcare to monitor the health of individuals. This study presents an updated systematic literature review of the use of wearable technology in promoting child and adolescent health, accompanied by recommendations for future research. Methods: This review focuses on studies involving children and adolescents aged between 2 and 18 years, regardless of their health condition or disabilities. Studies that were published from 2016 to 2024, and which met the inclusion criteria, were extracted from four academic databases (i.e. PubMed, Cochrane, Embase, and Web of Science) using the Preferred Reporting Items for Systematic Review and Meta-Analyses (PRISMA) protocol. Data on intervention purposes, interventions deployed, intervention duration, measurements, and the main outcomes of the studies were collected. Results: A total of 53 studies involving 14,852 participants were reviewed. They focused on various aspects, including the ownership and use of wearable devices (n = 3), the feasibility (n = 22), effectiveness (n = 4), and adherence (n = 2) of intervention strategies, or a combination of multiple aspects (n = 22). Among the interventions deployed, Fitbit was the most frequently used, featuring in 26 studies, followed by ActiGraph (n = 11). In intervention studies, the majority of studies focused on pre-morbidity prevention (n = 26) and the treatment of illnesses (n = 20), with limited attention given to postoperative monitoring (n = 4). Conclusions: The use of wearable technology by children and adolescents has proven to be both feasible and effective for health promotion. This systematic review summarizes existing research by exploring the use of wearable technology in promoting health across diverse youth populations, including healthy and unhealthy individuals. It examines health promotion at various stages of the disease continuum, including pre-disease prevention, in-disease treatment, and postoperative monitoring. Additionally, the review provides directions for future research.

2.
EMBO Mol Med ; 16(6): 1379-1403, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38684863

ABSTRACT

Polycystic kidney disease (PKD) is a genetic disorder characterized by bilateral cyst formation. We showed that PKD cells and kidneys display metabolic alterations, including the Warburg effect and glutaminolysis, sustained in vitro by the enzyme asparagine synthetase (ASNS). Here, we used antisense oligonucleotides (ASO) against Asns in orthologous and slowly progressive PKD murine models and show that treatment leads to a drastic reduction of total kidney volume (measured by MRI) and a prominent rescue of renal function in the mouse. Mechanistically, the upregulation of an ATF4-ASNS axis in PKD is driven by the amino acid response (AAR) branch of the integrated stress response (ISR). Metabolic profiling of PKD or control kidneys treated with Asns-ASO or Scr-ASO revealed major changes in the mutants, several of which are rescued by Asns silencing in vivo. Indeed, ASNS drives glutamine-dependent de novo pyrimidine synthesis and proliferation in cystic epithelia. Notably, while several metabolic pathways were completely corrected by Asns-ASO, glycolysis was only partially restored. Accordingly, combining the glycolytic inhibitor 2DG with Asns-ASO further improved efficacy. Our studies identify a new therapeutic target and novel metabolic vulnerabilities in PKD.


Subject(s)
Aspartate-Ammonia Ligase , Disease Models, Animal , Polycystic Kidney Diseases , Polycystic Kidney Diseases/metabolism , Polycystic Kidney Diseases/drug therapy , Polycystic Kidney Diseases/pathology , Polycystic Kidney Diseases/genetics , Animals , Aspartate-Ammonia Ligase/metabolism , Aspartate-Ammonia Ligase/genetics , Aspartate-Ammonia Ligase/antagonists & inhibitors , Mice , Oligonucleotides, Antisense/pharmacology , Oligonucleotides, Antisense/therapeutic use , Disease Progression , Humans , Kidney/pathology , Kidney/metabolism
3.
Nat Metab ; 5(3): 385-397, 2023 03.
Article in English | MEDLINE | ID: mdl-36879119

ABSTRACT

Depriving cells of nutrients triggers an energetic crisis, which is resolved by metabolic rewiring and organelle reorganization. Primary cilia are microtubule-based organelles at the cell surface, capable of integrating multiple metabolic and signalling cues, but their precise sensory function is not fully understood. Here we show that primary cilia respond to nutrient availability and adjust their length via glutamine-mediated anaplerosis facilitated by asparagine synthetase (ASNS). Nutrient deprivation causes cilia elongation, mediated by reduced mitochondrial function, ATP availability and AMPK activation independently of mTORC1. Of note, glutamine removal and replenishment is necessary and sufficient to induce ciliary elongation or retraction, respectively, under nutrient stress conditions both in vivo and in vitro by restoring mitochondrial anaplerosis via ASNS-dependent glutamate generation. Ift88-mutant cells lacking cilia show reduced glutamine-dependent mitochondrial anaplerosis during metabolic stress, due to reduced expression and activity of ASNS at the base of cilia. Our data indicate a role for cilia in responding to, and possibly sensing, cellular glutamine levels via ASNS during metabolic stress.


Subject(s)
Aspartate-Ammonia Ligase , Glutamine , Glutamine/metabolism , Aspartate-Ammonia Ligase/metabolism , Cilia/metabolism , Signal Transduction
4.
Microorganisms ; 11(2)2023 Feb 07.
Article in English | MEDLINE | ID: mdl-36838382

ABSTRACT

The tailoring of the skin microbiome is challenging and is a research hotspot in the pathogenesis of immune-mediated inflammatory skin diseases such as acne. Commonly encountered preservatives used as functional ingredients have an impact on the skin microbiota and are known to inhibit the survival of skin commensal bacteria. The selected species is Lactiplantibacillus plantarum, formulated with natural enhancers for topical use (SkinDuoTM). Ex vivo human skin models were used as a test system to assess the strain viability which was then validated on healthy volunteers. SkinDuoTM showed increased viability over time for in vitro skin models and a stable viability of over 50% on healthy skin. The strain was tested on human primary sebocytes obtained from sebaceous gland rich areas of facial skin and inoculated with the most abundant bacteria from the skin microbiota. Results on human ex vivo sebaceous gland models with the virulent phylotype of Cutibacterium acnes and Staphylococcus epidermidis present a significant reduction in viability, lipid production, and anti-inflammatory markers. We have developed an innovative anti-acne serum with L. plantarum that mimics the over-production of lipids, anti-inflammatory properties, and improves acne-disease skin models. Based on these results, we suggest that SkinDuoTM may be introduced as an acne-mitigating agent.

5.
Front Med (Lausanne) ; 8: 740087, 2021.
Article in English | MEDLINE | ID: mdl-34901057

ABSTRACT

Metabolic reprogramming is a key feature of Autosomal Dominant Polycystic Kidney Disease (ADPKD) characterized by changes in cellular pathways occurring in response to the pathological cell conditions. In ADPKD, a broad range of dysregulated pathways have been found. The studies supporting alterations in cell metabolism have shown that the metabolic preference for abnormal cystic growth is to utilize aerobic glycolysis, increasing glutamine uptake and reducing oxidative phosphorylation, consequently resulting in ADPKD cells shifting their energy to alternative energetic pathways. The mechanism behind the role of the polycystin proteins and how it leads to disease remains unclear, despite the identification of numerous signaling pathways. The integration of computational data analysis that accompanies experimental findings was pivotal in the identification of metabolic reprogramming in ADPKD. Here, we summarize the important results and argue that their exploitation may give further insights into the regulative mechanisms driving metabolic reprogramming in ADPKD. The aim of this review is to provide a comprehensive overview on metabolic focused studies and potential targets for treatment, and to propose that computational approaches could be instrumental in advancing this field of research.

6.
Cell Signal ; 67: 109495, 2020 03.
Article in English | MEDLINE | ID: mdl-31816397

ABSTRACT

Autosomal Dominant Polycystic Kidney Disease (ADPKD) is a slowly progressive disease characterized by the relentless growth of renal cysts throughout the life of affected individuals. Early evidence suggested that the epithelia lining the cysts share neoplastic features, leading to the definition of PKD as a "neoplasm in disguise". Recent work from our and other laboratories has identified a profound metabolic reprogramming in PKD, similar to the one reported in cancer and consistent with the reported increased proliferation. Multiple lines of evidence suggest that aerobic glycolysis (a Warburg-like effect) is present in the disease, along with other metabolic dysfunctions such as an increase in the pentose phosphate pathway, in glutamine anaplerosis and fatty acid biosynthesis, while fatty acid oxidation and oxidative phosphorylation (OXPHOS) are decreased. In addition to glutamine, other amino acid-related pathways appear altered, including asparagine and arginine. The precise origin of the metabolic alterations is not entirely clear, but two hypotheses can be formulated, not mutually exclusive. First, the polycystins have been recently shown to regulate directly mitochondrial function and structure either by regulating Ca2+ uptake in mitochondria at the Mitochondria Associated Membranes (MAMs) of the Endoplasmic Reticulum, or by a direct translocation of a small fragment of the protein into the matrix of mitochondria. One alternative possibility is that metabolic and mitochondrial dysfunctions in ADPKD are secondary to the de-regulation of proliferation, driven by the multiple signaling pathways identified in the disease, which include mTORC1 and AMPK among the most relevant. While the precise mechanisms underlying these novel alterations identified in ADPKD will need further investigation, it is evident that they offer a great opportunity for novel interventions in the disease.


Subject(s)
Mitochondria/metabolism , Polycystic Kidney Diseases/metabolism , Animals , Epigenesis, Genetic , Humans , Lipid Metabolism/genetics , Models, Biological , Polycystic Kidney Diseases/genetics , Polycystic Kidney Diseases/therapy , Signal Transduction/genetics
7.
Commun Biol ; 1: 194, 2018.
Article in English | MEDLINE | ID: mdl-30480096

ABSTRACT

Autosomal Dominant Polycystic Kidney Disease (ADPKD) is a genetic disorder caused by loss-of-function mutations in PKD1 or PKD2. Increased glycolysis is a prominent feature of the disease, but how it impacts on other metabolic pathways is unknown. Here, we present an analysis of mouse Pkd1 mutant cells and kidneys to investigate the metabolic reprogramming of this pathology. We show that loss of Pkd1 leads to profound metabolic changes that affect glycolysis, mitochondrial metabolism, and fatty acid synthesis (FAS). We find that Pkd1-mutant cells preferentially use glutamine to fuel the TCA cycle and to sustain FAS. Interfering with either glutamine uptake or FAS retards cell growth and survival. We also find that glutamine is diverted to asparagine via asparagine synthetase (ASNS). Transcriptional profiling of PKD1-mutant human kidneys confirmed these alterations. We find that silencing of Asns is lethal in Pkd1-mutant cells when combined with glucose deprivation, suggesting therapeutic approaches for ADPKD.

8.
Nat Rev Nephrol ; 14(11): 678-687, 2018 11.
Article in English | MEDLINE | ID: mdl-30120380

ABSTRACT

Autosomal dominant polycystic kidney disease (ADPKD) is one of the most common, potentially lethal, monogenic diseases and is caused predominantly by mutations in polycystic kidney disease 1 (PKD1) and PKD2, which encode polycystin 1 (PC1) and PC2, respectively. Over the decades-long course of the disease, patients develop large fluid-filled renal cysts that impair kidney function, leading to end-stage renal disease in ~50% of patients. Despite the identification of numerous dysregulated pathways in ADPKD, the molecular mechanisms underlying the renal dysfunction from mutations in PKD genes and the physiological functions of the polycystin proteins are still unclear. Alterations in cell metabolism have emerged in the past decade as a hallmark of ADPKD. ADPKD cells shift their mode of energy production from oxidative phosphorylation to alternative pathways, such as glycolysis. In addition, the polycystins seem to play regulatory roles in modulating mechanisms and machinery related to energy production and utilization, including AMPK, PPARα, PGC1α, calcium signalling at mitochondria-associated membranes, mTORC1, cAMP and CFTR-mediated ion transport as well as the expression of crucial components of the mitochondrial energy production apparatus. In this Review, we explore these metabolic changes and discuss in detail the relationship between energy metabolism and ADPKD pathogenesis and identify potential therapeutic targets.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Energy Metabolism , Mitochondria/physiology , Molecular Targeted Therapy , Polycystic Kidney, Autosomal Dominant/drug therapy , Polycystic Kidney, Autosomal Dominant/metabolism , Animals , Cyclic AMP/metabolism , Fatty Acids/metabolism , Glucose/metabolism , Glycolysis , Humans , Lipid Metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Oxidation-Reduction , Polycystic Kidney, Autosomal Dominant/genetics , Signal Transduction
9.
FASEB J ; 29(5): 1676-87, 2015 May.
Article in English | MEDLINE | ID: mdl-25526730

ABSTRACT

The importance of epigenetic changes in the development of hepatic steatosis is largely unknown. The histone variant macroH2A1 under alternative splicing gives rise to macroH2A1.1 and macroH2A1.2. In this study, we show that the macroH2A1 isoforms play an important role in the regulation of lipid accumulation in hepatocytes. Hepatoma cell line and immortalized human hepatocytes transiently transfected or knocked down with macroH2A1 isoforms were used as in vitro model of fat-induced steatosis. Gene expressions were analyzed by quantitative PCR array and Western blot. Chromatin immunoprecipitation analysis was performed to check the association of histone H3 lysine 27 trimethylation (H3K27me3) and histone H3 lysine 4 trimethylation (H3K4me3) with the promoter of lipogenic genes. Livers from knockout mice that are resistant to lipid deposition despite a high-fat diet were used for histopathology. We found that macroH2A1.2 is regulated by fat uptake and that its overexpression caused an increase in lipid uptake, triglycerides, and lipogenic genes compared with macroH2A1.1. This suggests that macroH2A1.2 is important for lipid uptake, whereas macroH2A1.1 was found to be protective. The result was supported by a high positivity for macroH2A1.1 in knockout mice for genes targeted by macroH2A1 (Atp5a1 and Fam73b), that under a high-fat diet presented minimal lipidosis. Moreover, macroH2A1 isoforms differentially regulate the expression of lipogenic genes by modulating the association of the active (H3K4me3) and repressive (H3K27me3) histone marks on their promoters. This study underlines the importance of the replacement of noncanonical histones in the regulation of genes involved in lipid metabolism in the progression of steatosis.


Subject(s)
Biomarkers/metabolism , Carcinoma, Hepatocellular/pathology , Diet, High-Fat/adverse effects , Epigenomics , Fatty Liver/metabolism , Fatty Liver/pathology , Hepatocytes/pathology , Histones/metabolism , Animals , Blotting, Western , Carcinoma, Hepatocellular/metabolism , Cells, Cultured , Chromatin Immunoprecipitation , Fatty Liver/etiology , Female , Fluorescent Antibody Technique , Gene Expression Profiling , Hepatocytes/metabolism , Histones/genetics , Humans , Immunoenzyme Techniques , Lipid Peroxidation , Liver/metabolism , Liver/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Proton-Translocating ATPases/physiology , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic/genetics , Protein Isoforms , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
10.
Endocrinology ; 155(3): 908-22, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24302625

ABSTRACT

Cushing's syndrome, which is characterized by excessive circulating glucocorticoid concentrations, may be due to ACTH-dependent or -independent causes that include anterior pituitary and adrenal cortical tumors, respectively. ACTH secretion is stimulated by CRH, and we report a mouse model for Cushing's syndrome due to an N-ethyl-N-nitrosourea (ENU) induced Crh mutation at -120 bp of the promoter region, which significantly increased luciferase reporter activity and was thus a gain-of-function mutation. Crh(-120/+) mice, when compared with wild-type littermates, had obesity, muscle wasting, thin skin, hair loss, and elevated plasma and urinary concentrations of corticosterone. In addition, Crh(-120/+) mice had hyperglycemia, hyperfructosaminemia, hyperinsulinemia, hypercholesterolemia, hypertriglyceridemia, and hyperleptinemia but normal adiponectin. Crh(-120/+) mice also had low bone mineral density, hypercalcemia, hypercalciuria, and decreased concentrations of plasma PTH and osteocalcin. Bone histomorphometry revealed Crh(-120/+) mice to have significant reductions in mineralizing surface area, mineral apposition, bone formation rates, osteoblast number, and the percentage of corticoendosteal bone covered by osteoblasts, which was accompanied by an increase in adipocytes in the bone marrow. Thus, a mouse model for Cushing's syndrome has been established, and this will help in further elucidating the pathophysiological effects of glucocorticoid excess and in evaluating treatments for corticosteroid-induced osteoporosis.


Subject(s)
Corticotropin-Releasing Hormone/genetics , Ethylnitrosourea/chemistry , Glucocorticoids/metabolism , Mutation , Promoter Regions, Genetic , Animals , Body Composition , Bone and Bones/metabolism , Calcium/metabolism , Cell Line , Chromosome Mapping , Corticosterone/metabolism , Cushing Syndrome/genetics , Disease Models, Animal , Female , Lipid Metabolism , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Osteoblasts/metabolism , Osteoporosis/metabolism
11.
Cell ; 154(2): 452-64, 2013 Jul 18.
Article in English | MEDLINE | ID: mdl-23870131

ABSTRACT

Mutations in whole organisms are powerful ways of interrogating gene function in a realistic context. We describe a program, the Sanger Institute Mouse Genetics Project, that provides a step toward the aim of knocking out all genes and screening each line for a broad range of traits. We found that hitherto unpublished genes were as likely to reveal phenotypes as known genes, suggesting that novel genes represent a rich resource for investigating the molecular basis of disease. We found many unexpected phenotypes detected only because we screened for them, emphasizing the value of screening all mutants for a wide range of traits. Haploinsufficiency and pleiotropy were both surprisingly common. Forty-two percent of genes were essential for viability, and these were less likely to have a paralog and more likely to contribute to a protein complex than other genes. Phenotypic data and more than 900 mutants are openly available for further analysis. PAPERCLIP:


Subject(s)
Genetic Techniques , Mice, Knockout , Phenotype , Animals , Disease/genetics , Disease Models, Animal , Female , Genes, Essential , Genome-Wide Association Study , Male , Mice
12.
Mamm Genome ; 24(5-6): 240-51, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23712496

ABSTRACT

C57BL/6N (B6N) is becoming the standard background for genetic manipulation of the mouse genome. The B6N, whose genome is very closely related to the reference C57BL/6J genome, is versatile in a wide range of phenotyping and experimental settings and large repositories of B6N ES cells have been developed. Here, we present a series of studies showing the baseline characteristics of B6N fed a high-fat diet (HFD) for up to 12 weeks. We show that HFD-fed B6N mice show increased weight gain, fat mass, and hypercholesterolemia compared to control diet-fed mice. In addition, HFD-fed B6N mice display a rapid onset of lipid accumulation in the liver with both macro- and microvacuolation, which became more severe with increasing duration of HFD. Our results suggest that the B6N mouse strain is a versatile background for studying diet-induced metabolic syndrome and may also represent a model for early nonalcoholic fatty liver disease.


Subject(s)
Disease Models, Animal , Lipid Metabolism , Mice/metabolism , Obesity/metabolism , Animals , Body Weight , Diet, High-Fat/adverse effects , Female , Humans , Liver/metabolism , Male , Mice/growth & development , Mice, Inbred C57BL , Obesity/etiology
13.
PLoS One ; 8(1): e54458, 2013.
Article in English | MEDLINE | ID: mdl-23372727

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) is one of the most common cancers worldwide. Prevention and risk reduction are important and the identification of specific biomarkers for early diagnosis of HCC represents an active field of research. Increasing evidence indicates that fat accumulation in the liver, defined as hepatosteatosis, is an independent and strong risk factor for developing an HCC. MacroH2A1, a histone protein generally associated with the repressed regions of chromosomes, is involved in hepatic lipid metabolism and is present in two alternative spliced isoforms, macroH2A1.1 and macroH2A1.2. These isoforms have been shown to predict lung and colon cancer recurrence but to our knowledge, their role in fatty-liver associated HCC has not been investigated previously. METHODS: We examined macroH2A1.1 and macroH2A1.2 protein expression levels in the liver of two murine models of fat-associated HCC, the high fat diet/diethylnistrosamine (DEN) and the phosphatase and tensin homolog (PTEN) liver specific knock-out (KO) mouse, and in human liver samples of subjects with steatosis or HCC, using immunoblotting and immunohistochemistry. RESULTS: Protein levels for both macroH2A1 isoforms were massively upregulated in HCC, whereas macroH2A1.2 was specifically upregulated in steatosis. In addition, examination of human liver samples showed a significant difference (p<0.01) in number of positive nuclei in HCC (100% of tumor cells positive for either macroH2A1.1 or macroH2A1.2), when compared to steatosis (<2% of hepatocytes positive for either isoform). The steatotic areas flanking the tumors were highly immunopositive for macroH2A1.1 and macroH2A1.2. CONCLUSIONS: These data obtained in mice and humans suggest that both macroH2A1 isoforms may play a role in HCC pathogenesis and moreover may be considered as novel diagnostic markers for human HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Fatty Liver/genetics , Histones/genetics , Liver Neoplasms/genetics , Animals , Biomarkers/metabolism , Carcinoma, Hepatocellular/etiology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cell Nucleus/pathology , Diet, High-Fat , Diethylnitrosamine , Fatty Liver/chemically induced , Fatty Liver/complications , Fatty Liver/metabolism , Gene Expression Regulation , Hepatocytes/metabolism , Hepatocytes/pathology , Histones/metabolism , Humans , Immunohistochemistry , Liver Neoplasms/etiology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice , Mice, Knockout , PTEN Phosphohydrolase/deficiency , PTEN Phosphohydrolase/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Tumor Microenvironment/genetics
14.
Curr Pharm Des ; 19(15): 2737-46, 2013.
Article in English | MEDLINE | ID: mdl-23092327

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD), an accumulation of intra-hepatic triglycerides that is often considered the hepatic manifestation of insulin resistance, is the most common cause of chronic liver disease in the Western countries with up to one third of the population affected. NAFLD is a spectrum of disturbances that encompasses various degrees of liver damage ranging from simple steatosis to non-alcoholic steatohepatitis (NASH). NASH is characterized by hepatocellular injury/inflammation with or without fibrosis. The individuals with NAFLD develop NASH in 10% of the cases, and are also at risk of developing hepatocellular carcinoma (HCC). Epigenetic mechanisms of nuclear chromatin remodeling, such as DNA methylation, post-translational modifications of histones, and incorporation of histone variants into the chromatin are increasingly recognized as crucial factors in the pathophysiology of NAFLD. NAFLD is often accompanied by oxidative stress: reactive oxygen species (ROS) are implicated in altered reduction/oxidation (redox) reactions that attack cellular macromolecules and are detected in the liver of patients and animal models of NAFLD. In this review, we summarize recent knowledge advancements in the hepatic epigenetic and redox mechanisms, and their possible links, involved in the pathogenesis and treatment of NAFLD.


Subject(s)
Epigenesis, Genetic , Fatty Liver/genetics , Fatty Liver/metabolism , Homeostasis , Cardiovascular Diseases/complications , Cardiovascular Diseases/genetics , Fatty Liver/complications , Humans , Non-alcoholic Fatty Liver Disease , Oxidation-Reduction , Oxidative Stress
15.
Mamm Genome ; 24(1-2): 44-53, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23160729

ABSTRACT

Large-scale N-ethyl-N-nitrosourea (ENU) mutagenesis has provided many rodent models for human disease. Here we describe the initial characterization and mapping of a recessive mutation that leads to degeneration of the incisors, failure of molars to erupt, a grey coat colour, and mild osteopetrosis. We mapped the omi mutation to chromosome 10 between D10Mit214 and D10Mit194. The Ostm1 gene is a likely candidate gene in this region and the grey-lethal allele, Ostm1 ( gl ), and omi mutations fail to complement each other. We show that om/om mice have reduced levels of Ostm1 protein. To date we have not been able to identify the causative mutation. We propose that omi is a novel hypomorphic mutation affecting Ostm1 expression, potentially in a regulatory element.


Subject(s)
Alleles , Chromosomes/genetics , Genes, Recessive , Membrane Proteins/genetics , Animals , Chromosome Mapping , Disease Models, Animal , Ethylnitrosourea , Female , Gene Expression Regulation , Genes, Lethal , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Mutagenesis , Mutation , Osteopetrosis/genetics , Phenotype
16.
Am J Hum Genet ; 91(6): 998-1010, 2012 Dec 07.
Article in English | MEDLINE | ID: mdl-23200864

ABSTRACT

Ubiquitination plays a crucial role in neurodevelopment as exemplified by Angelman syndrome, which is caused by genetic alterations of the ubiquitin ligase-encoding UBE3A gene. Although the function of UBE3A has been widely studied, little is known about its paralog UBE3B. By using exome and capillary sequencing, we here identify biallelic UBE3B mutations in four patients from three unrelated families presenting an autosomal-recessive blepharophimosis-ptosis-intellectual-disability syndrome characterized by developmental delay, growth retardation with a small head circumference, facial dysmorphisms, and low cholesterol levels. UBE3B encodes an uncharacterized E3 ubiquitin ligase. The identified UBE3B variants include one frameshift and two splice-site mutations as well as a missense substitution affecting the highly conserved HECT domain. Disruption of mouse Ube3b leads to reduced viability and recapitulates key aspects of the human disorder, such as reduced weight and brain size and a downregulation of cholesterol synthesis. We establish that the probable Caenorhabditis elegans ortholog of UBE3B, oxi-1, functions in the ubiquitin/proteasome system in vivo and is especially required under oxidative stress conditions. Our data reveal the pleiotropic effects of UBE3B deficiency and reinforce the physiological importance of ubiquitination in neuronal development and function in mammals.


Subject(s)
Blepharophimosis/genetics , Blepharoptosis/genetics , Intellectual Disability/genetics , Ubiquitin-Protein Ligases/genetics , Alleles , Amino Acid Sequence , Animals , Base Sequence , Blepharophimosis/diagnosis , Blepharoptosis/diagnosis , Brain/pathology , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Central Nervous System , Child , Child, Preschool , Exome , Facies , Female , Genotype , Humans , Infant , Intellectual Disability/diagnosis , Magnetic Resonance Imaging , Male , Mice , Mice, Knockout , Mutation , Oxidative Stress , Syndrome , Ubiquitin-Protein Ligases/deficiency
17.
Blood ; 119(6): 1370-9, 2012 Feb 09.
Article in English | MEDLINE | ID: mdl-22184403

ABSTRACT

Stem cell differentiation and lineage specification depend on coordinated programs of gene expression, but our knowledge of the chromatin-modifying factors regulating these events remains incomplete. Ubiquitination of histone H2A (H2A-K119u) is a common chromatin modification associated with gene silencing, and controlled by the ubiquitin-ligase polycomb repressor complex 1 (PRC1) and H2A-deubiquitinating enzymes (H2A-DUBs). The roles of H2A-DUBs in mammalian development, stem cells, and hematopoiesis have not been addressed. Here we characterized an H2A-DUB targeted mouse line Mysm1(tm1a/tm1a) and demonstrated defects in BM hematopoiesis, resulting in lymphopenia, anemia, and thrombocytosis. Development of lymphocytes was impaired from the earliest stages of their differentiation, and there was also a depletion of erythroid cells and a defect in erythroid progenitor function. These phenotypes resulted from a cell-intrinsic requirement for Mysm1 in the BM. Importantly, Mysm1(tm1a/tm1a) HSCs were functionally impaired, and this was associated with elevated levels of reactive oxygen species, γH2AX DNA damage marker, and p53 protein in the hematopoietic progenitors. Overall, these data establish a role for Mysm1 in the maintenance of BM stem cell function, in the control of oxidative stress and genetic stability in hematopoietic progenitors, and in the development of lymphoid and erythroid lineages.


Subject(s)
Cell Differentiation/genetics , Endopeptidases/genetics , Hematopoiesis/genetics , Lymphocytes/metabolism , Animals , Blood Cell Count , Blotting, Western , Endopeptidases/metabolism , Female , Flow Cytometry , Gene Expression Profiling , Genotype , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Histones/metabolism , Lymphocytes/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Phenotype , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Trans-Activators , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Specific Proteases
SELECTION OF CITATIONS
SEARCH DETAIL
...