Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 2269, 2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38480682

ABSTRACT

Primary familial brain calcification (PFBC) is characterized by calcium deposition in the brain, causing progressive movement disorders, psychiatric symptoms, and cognitive decline. PFBC is a heterogeneous disorder currently linked to variants in six different genes, but most patients remain genetically undiagnosed. Here, we identify biallelic NAA60 variants in ten individuals from seven families with autosomal recessive PFBC. The NAA60 variants lead to loss-of-function with lack of protein N-terminal (Nt)-acetylation activity. We show that the phosphate importer SLC20A2 is a substrate of NAA60 in vitro. In cells, loss of NAA60 caused reduced surface levels of SLC20A2 and a reduction in extracellular phosphate uptake. This study establishes NAA60 as a causal gene for PFBC, provides a possible biochemical explanation of its disease-causing mechanisms and underscores NAA60-mediated Nt-acetylation of transmembrane proteins as a fundamental process for healthy neurobiological functioning.


Subject(s)
Brain Diseases , Humans , Acetylation , Brain/diagnostic imaging , Brain/metabolism , Brain Diseases/genetics , Inheritance Patterns , Mutation , Phosphates/metabolism , Sodium-Phosphate Cotransporter Proteins, Type III/metabolism
2.
J Transl Med ; 21(1): 171, 2023 03 03.
Article in English | MEDLINE | ID: mdl-36869348

ABSTRACT

BACKGROUND: Type 2 diabetes (T2D) is a critical healthcare challenge and priority in Qatar which is listed amongst the top 10 countries in the world, with its prevalence presently at 17% double the global average. MicroRNAs (miRNAs) are implicated in the pathogenesis of (T2D) and long-term microvascular complications including diabetic retinopathy (DR). METHODS: In this study, a T2D cohort that accurately matches the characteristics of the general population was employed to find microRNA (miRNA) signatures that are correlated with glycemic and ß cell function measurements. Targeted miRNA profiling was performed in (471) T2D individuals with or without DR and (491) (non-diabetic) healthy controls from the Qatar Biobank. Discovery analysis identified 20 differentially expressed miRNAs in T2D compared to controls, of which miR-223-3p was significantly upregulated (fold change:5.16, p = 3.6e-02) and positively correlated with glucose and hemoglobin A1c (HbA1c) levels (p-value = 9.88e-04 and 1.64e-05, respectively), but did not show any significant associations with insulin or C-peptide. Accordingly, we performed functional validation using a miR-223-3p mimic (overexpression) under control and hyperglycemia-induced conditions in a zebrafish model. RESULTS: Over-expression of miR-223-3p alone was associated with significantly higher glucose (42.7 mg/dL, n = 75 vs 38.7 mg/dL, n = 75, p = 0.02) and degenerated retinal vasculature, and altered retinal morphology involving changes in the ganglion cell layer and inner and outer nuclear layers. Assessment of retinal angiogenesis revealed significant upregulation in the expression of vascular endothelial growth factor and its receptors, including kinase insert domain receptor. Further, the pancreatic markers, pancreatic and duodenal homeobox 1, and the insulin gene expressions were upregulated in the miR-223-3p group. CONCLUSION: Our zebrafish model validates a novel correlation between miR-223-3p and DR development. Targeting miR-223-3p in T2D patients may serve as a promising therapeutic strategy to control DR in at-risk individuals.


Subject(s)
Diabetes Mellitus, Type 2 , Diabetic Retinopathy , Hyperglycemia , MicroRNAs , Humans , Animals , Glycemic Control , Zebrafish , Vascular Endothelial Growth Factor A , Insulin , Glucose
3.
Blood Adv ; 7(5): 697-711, 2023 03 14.
Article in English | MEDLINE | ID: mdl-36477543

ABSTRACT

Emerging gene therapy clinical trials test the correction of hemophilia A (HA) by replacing factor VIII (FVIII) in autologous hematopoietic stem cells (HSCs). Although it is known that platelets, monocyte/macrophages, and mesenchymal stromal cells can secrete transgenic FVIII, a systematic examination of blood lineages as extrahepatic sources of FVIII, to our knowledge, has not yet been performed. In this study, we sought to provide a comprehensive map of native and lentivirus-based transgenic FVIII production from HSC stage to mature blood cells, through a flow cytometry analysis. In addition, we generated a model of transient HA in zebrafish based on antisense RNA, to assess the corrective potential of the FVIII-transduced HSCs. We discovered that FVIII production begins at the CD34+ progenitor stage after cytokine stimulation in culture. Among all mature white blood cells, monocytes are the largest producers of native FVIII and can maintain protein overexpression during differentiation from HSCs when transduced by a FVIII lentiviral vector. Moreover, the addition of the HSC self-renewal agonist UM171 to CD34+ cells during transduction expanded a subpopulation of CD14+/CD31+ monocytes with excellent ability to carry the FVIII transgene, allowing the correction of HA phenotype in zebrafish. Finally, the HA zebrafish model showed that f8 RNA is predominantly localized in the hematopoietic system at the larval stage, which indicates a potential contributory role of FVIII in hematopoiesis that warrants further investigation. We believe that this study may be of broad interest to hematologists and researchers striving to advance knowledge and permanent treatments for patients with HA.


Subject(s)
Hemophilia A , Hemostatics , Animals , Factor VIII/genetics , Hematopoietic Stem Cells/metabolism , Hemophilia A/therapy , Monocytes/metabolism , Zebrafish/metabolism , Humans
4.
Toxicol Res (Camb) ; 11(1): 77-87, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35237413

ABSTRACT

BACKGROUND: A sensitive method to investigate cellular stress and cytotoxicity is based on measuring mitochondrial membrane potential. Recently, JC-10, was developed to measure mitochondrial membrane potential in vitro and used as an indicator for cytotoxicity. Yet, JC-10 has never been used in vivo (whole organism). In normal cells, JC-10 concentrates in the mitochondrial matrix, where it forms red fluorescent aggregates. However, in apoptotic/necrotic cells, JC-10 diffuses out of the mitochondria, changes to monomeric form, and stains cells in green. Here, we aimed to develop and optimize a JC-10 assay to measure cytotoxicity in zebrafish embryo. We also investigated the effectiveness of JC-10 assay by comparing it to common cytotoxicity assays. METHODS: Zebrafish embryos were exposed to a toxic surfactant AEO-7 at no observed effect concentration (6.4 µg/L), and then cytotoxicity was measured using (i) JC-10 mitochondrial assay, (ii) acridine orange (AO), (iii) TUNEL assay, and (iv) measuring the level of Hsp70 by western blotting. RESULTS: As compared to the negative control, embryos treated with NOEC of AEO-7 did not show significant cytotoxicity when assessed by AO, TUNEL or western blotting. However, when JC-10 was used under the same experimental conditions, a significant increase of green:red fluorescent ratio signal was detected in the AEO-7 treated embryos, indicating mitochondrial damage and cellular cytotoxicity. Noteworthy, the observed green: red ratio increase was dose dependent, suggesting specificity of the JC-10 assay. CONCLUSION: JC-10 is a sensitive in vivo method, thus, can be used as surrogate assay to measure cytotoxicity in whole zebrafish embryos.

5.
Hum Mol Genet ; 31(16): 2796-2809, 2022 08 23.
Article in English | MEDLINE | ID: mdl-35348702

ABSTRACT

In order to report clinically actionable incidental findings in genetic testing, the American College of Medical Genetics and Genomics (ACMG) recommended the evaluation of variants in 59 genes associated with highly penetrant mutations. However, there is a lack of epidemiological data on medically actionable rare variants in these genes in Arab populations. We used whole genome sequencing data from 6045 participants from the Qatar Genome Programme and integrated it with phenotypic data collected by the Qatar Biobank. We identified novel putative pathogenic variants in the 59 ACMG genes by filtering previously unrecorded variants based on computational prediction of pathogenicity, variant rarity and segregation evidence. We assessed the phenotypic associations of candidate variants in genes linked to cardiovascular diseases. Finally, we used a zebrafish knockdown and synthetic human mRNA co-injection assay to functionally characterize two of these novel variants. We assessed the zebrafish cardiac function in terms of heart rate, rhythm and hemodynamics, as well as the heart structure. We identified 52 492 novel variants, which have not been reported in global and disease-specific databases. A total of 74 novel variants were selected with potentially pathogenic effect. We prioritized two novel cardiovascular variants, DSP c.1841A > G (p.Asp614Gly) and LMNA c.326 T > G (p.Val109Gly) for functional characterization. Our results showed that both variants resulted in abnormal zebrafish heart rate, rhythm and structure. This study highlights medically actionable variants that are specific to the Middle Eastern Qatari population.


Subject(s)
Desmoplakins/genetics , Incidental Findings , Lamin Type A , Animals , Genetic Predisposition to Disease , Genetic Testing/methods , Humans , Lamin Type A/genetics , Qatar , Zebrafish/genetics
6.
MethodsX ; 8: 101418, 2021.
Article in English | MEDLINE | ID: mdl-34430313

ABSTRACT

Microinjection technique is one of the essential methodologies that are used widely in zebrafish research. Microinjection is utilized to perform genetic manipulations within the developing zebrafish model. Further, this technique is used to study a wide range of genetic diseases and gene of interest role in early developmental processes. Thus, quality control for microinjection is an essential factor to ensure experimental reproducibility and consistency. In this technical note, in vitro transcribed synthetic mRNA encoding green fluorescence protein (eGFP), and red fluorescent protein (m-cherry) as well as fluorescein and rhodamine fluorescent dyes were injected into a single-cell zebrafish embryo for volume quality control. Given the importance of having quality control system and methodology to yield similar genetic manipulation within the zebrafish embryo:•We aimed to establish the unified delivery of injected material into zebrafish one cell stage embryo.•We aimed to establish consistency of the injected volume into mineral oil droplets that will serve as a quality control parameter to conforms a quality control practice to ensure the reproducibility of the microinjection technique.•The calibration of microinjection droplet size resulted in the visualization of fluorescent protein and dyes in the zebrafish embryo with precise volumes of delivered materials under the control of needle opening, injection pressure and time.

7.
Brain ; 144(2): 584-600, 2021 03 03.
Article in English | MEDLINE | ID: mdl-33559681

ABSTRACT

The extracellular matrix comprises a network of macromolecules such as collagens, proteoglycans and glycoproteins. VWA1 (von Willebrand factor A domain containing 1) encodes a component of the extracellular matrix that interacts with perlecan/collagen VI, appears to be involved in stabilizing extracellular matrix structures, and demonstrates high expression levels in tibial nerve. Vwa1-deficient mice manifest with abnormal peripheral nerve structure/function; however, VWA1 variants have not previously been associated with human disease. By interrogating the genome sequences of 74 180 individuals from the 100K Genomes Project in combination with international gene-matching efforts and targeted sequencing, we identified 17 individuals from 15 families with an autosomal-recessive, non-length dependent, hereditary motor neuropathy and rare biallelic variants in VWA1. A single disease-associated allele p.(G25Rfs*74), a 10-bp repeat expansion, was observed in 14/15 families and was homozygous in 10/15. Given an allele frequency in European populations approaching 1/1000, the seven unrelated homozygote individuals ascertained from the 100K Genomes Project represents a substantial enrichment above expected. Haplotype analysis identified a shared 220 kb region suggesting that this founder mutation arose >7000 years ago. A wide age-range of patients (6-83 years) helped delineate the clinical phenotype over time. The commonest disease presentation in the cohort was an early-onset (mean 2.0 ± 1.4 years) non-length-dependent axonal hereditary motor neuropathy, confirmed on electrophysiology, which will have to be differentiated from other predominantly or pure motor neuropathies and neuronopathies. Because of slow disease progression, ambulation was largely preserved. Neurophysiology, muscle histopathology, and muscle MRI findings typically revealed clear neurogenic changes with single isolated cases displaying additional myopathic process. We speculate that a few findings of myopathic changes might be secondary to chronic denervation rather than indicating an additional myopathic disease process. Duplex reverse transcription polymerase chain reaction and immunoblotting using patient fibroblasts revealed that the founder allele results in partial nonsense mediated decay and an absence of detectable protein. CRISPR and morpholino vwa1 modelling in zebrafish demonstrated reductions in motor neuron axonal growth, synaptic formation in the skeletal muscles and locomotive behaviour. In summary, we estimate that biallelic variants in VWA1 may be responsible for up to 1% of unexplained hereditary motor neuropathy cases in Europeans. The detailed clinical characterization provided here will facilitate targeted testing on suitable patient cohorts. This novel disease gene may have previously evaded detection because of high GC content, consequential low coverage and computational difficulties associated with robustly detecting repeat-expansions. Reviewing previously unsolved exomes using lower QC filters may generate further diagnoses.


Subject(s)
Extracellular Matrix Proteins/genetics , Hereditary Sensory and Motor Neuropathy/genetics , Adult , Aged , Animals , Behavior, Animal/physiology , Child , Female , Hereditary Sensory and Motor Neuropathy/pathology , Humans , Male , Middle Aged , Muscle, Skeletal/pathology , Mutation , Pedigree , Young Adult , Zebrafish
8.
Sci Total Environ ; 741: 140450, 2020 Nov 01.
Article in English | MEDLINE | ID: mdl-32886985

ABSTRACT

Surfactants are widely used in the industry of detergents, household products, and cosmetics. SAPDMA is a cationic surfactant that is used mostly in cosmetics, conditioning agents and has recently gained attention as a corrosion inhibitor in the sea pipelines industry. In this regard, literature concerning the ecotoxicological classification of SAPDMA on aquatic animals is lacking. This study aims to evaluate the potential ecotoxicity of SAPDMA using the aquatic zebrafish embryo model. The potential toxic effects of SAPDMA were assessed by different assays. This includes (i) mortality/survival assay to assess the median lethal concentration (LC50); (ii) teratogenicity assay to assess the no observed effect concentration (NOEC); (iii) organ-specific toxicity assays including cardiotoxicity, neurotoxicity (using locomotion assay), hematopoietic toxicity (hemoglobin synthesis using o-dianisidine staining), hepatotoxicity (liver steatosis and yolk retention using Oil Red O (ORO) stain); (iv) cellular cytotoxicity (mitochondrial membrane potential) by measuring the accumulation of JC-1 dye into mitochondria. Exposure of embryos to SAPDMA caused mortality in a dose-dependent manner with a calculated LC50 of 2.3 mg/L. Thus, based on the LC50 value and according to the Fish and Wildlife Service (FWS) Acute Toxicity Rating Scale, SAPDMA is classified as "moderately toxic". The No Observed Effect Concentration (NOEC) concerning a set of parameters including scoliosis, changes in body length, yolk, and eye sizes was 0.1 mg/L. At the same NOEC concentration (0.1 mg/L), no organ-specific toxicity was detected in fish treated with SAPDMA, except hepatomegaly with no associated liver dysfunctions. However, higher SAPDMA concentrations (0.8 mg/L) have dramatic effects on zebrafish organ development (eye, heart, and liver development). Our data recommend a re-evaluation of the SAPDMA employment in the industry setting and its strictly monitoring by environmental and public health agencies.


Subject(s)
Water Pollutants, Chemical , Zebrafish , Animals , Dimethylamines , Embryo, Nonmammalian , Lethal Dose 50 , Surface-Active Agents
9.
Cells ; 9(8)2020 07 27.
Article in English | MEDLINE | ID: mdl-32726939

ABSTRACT

Recessive mutations in Post-GPI attachment to proteins 3 (PGAP3) cause the rare neurological disorder hyperphosphatasia with mental retardation syndrome 4 type (HPMRS4). Here, we report a novel homozygous nonsense mutation in PGAP3 (c.265C>T-p.Gln89*), in a 3-year-old boy with unique novel clinical features. These include decreased intrauterine fetal movements, dysgenesis of the corpus callosum, olfactory bulb agenesis, dysmorphic features, cleft palate, left ear constriction, global developmental delay, and hypotonia. The zebrafish functional modeling of PGAP3 loss resulted in HPMRS4-like features, including structural brain abnormalities, dysmorphic cranial and facial features, hypotonia, and seizure-like behavior. Remarkably, morphants displayed defective neural tube formation during the early stages of nervous system development, affecting brain morphogenesis. The significant aberrant midbrain and hindbrain formation demonstrated by separation of the left and right tectal ventricles, defects in the cerebellar corpus, and caudal hindbrain formation disrupted oligodendrocytes expression leading to shorter motor neurons axons. Assessment of zebrafish neuromuscular responses revealed epileptic-like movements at early development, followed by seizure-like behavior, loss of touch response, and hypotonia, mimicking the clinical phenotype human patients. Altogether, we report a novel pathogenic PGAP3 variant associated with unique phenotypic hallmarks, which may be related to the gene's novel role in brain morphogenesis and neuronal wiring.


Subject(s)
Abnormalities, Multiple/genetics , Brain/pathology , Carboxylic Ester Hydrolases/adverse effects , Intellectual Disability/genetics , Phosphorus Metabolism Disorders/genetics , Abnormalities, Multiple/metabolism , Animals , Disease Models, Animal , Humans , Intellectual Disability/metabolism , Morphogenesis , Phosphorus Metabolism Disorders/metabolism , Receptors, Cell Surface , Zebrafish
10.
J Cell Physiol ; 235(11): 7870-7888, 2020 11.
Article in English | MEDLINE | ID: mdl-31943169

ABSTRACT

Myosin-binding protein C 3 (MYBPC3) variants are the most common cause of hypertrophic cardiomyopathy (HCM). HCM is a complex cardiac disorder due to its significant genetic and clinical heterogeneity. MYBPC3 variants genotype-phenotype associations remain poorly understood. We investigated the impact of two novel human MYBPC3 splice-site variants: V1: c.654+2_654+4dupTGG targeting exon 5 using morpholino MOe5i5; and V2: c.772+1G>A targeting exon 6 using MOe6i6; located within C1 domain of cMyBP-C protein, known to be critical in regulating sarcomere structure and contractility. Zebrafish MOe5i5 and MOe6i6 morphants recapitulated typical characteristics of human HCM with cardiac phenotypes of varying severity, including reduced cardiomyocyte count, thickened ventricular myocardial wall, a drastic reduction in heart rate, stroke volume, and cardiac output. Analysis of all cardiac morphological and functional parameters demonstrated that V2 cardiac phenotype was more severe than V1. Coinjection with synthetic human MYBPC3 messenger RNA (mRNA) partially rescued disparate cardiac phenotypes in each zebrafish morphant. While human MYBPC3 mRNA partially restored the decreased heart rate in V1 morphants and displayed increased percentages of ejection fraction, fractional shortening, and area change, it failed to revert the V1 ventricular myocardial thickness. These results suggest a possible V1 impact on cardiac contractility. In contrast, attempts to rescue V2 morphants only restored the ventricular myocardial wall hypertrophy phenotype but had no significant effect on impaired heart rate, suggesting a potential V2 impact on the cardiac structure. Our study provides evidence of an association between MYBPC3 exon-specific cardiac phenotypes in the zebrafish model providing important insights into how these genetic variants contribute to HCM disease.


Subject(s)
Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/pathology , Carrier Proteins/genetics , Animals , Disease Models, Animal , Exons/genetics , Humans , Phenotype , Protein Isoforms/genetics , Zebrafish
11.
Ann N Y Acad Sci ; 1448(1): 19-29, 2019 07.
Article in English | MEDLINE | ID: mdl-30937913

ABSTRACT

Calmodulin (CaM) is a universal calcium (Ca2+ )-binding messenger that regulates many vital cellular events. In cardiac muscle, CaM associates with ryanodine receptor 2 (RyR2) and regulates excitation-contraction coupling. Mutations in human genes CALM1, CALM2, and CALM3 have been associated with life-threatening heart disorders, such as long QT syndrome (LQTS) and catecholaminergic polymorphic ventricular tachycardia. A novel de novo LQTS-associated missense CaM mutation (E105A) was recently identified in a 6-year-old boy, who experienced an aborted first episode of cardiac arrest. Herein, we report the first molecular characterization of the CaM E105A mutation. Expression of the CaM E105A mutant in zebrafish embryos resulted in cardiac arrhythmia and increased heart rate, suggestive of ventricular tachycardia. In vitro biophysical and biochemical analysis revealed that E105A confers a deleterious effect on protein stability and a reduced Ca2+ -binding affinity due to loss of cooperativity. Finally, the CaM E105A mutation resulted in reduced CaM-RyR2 interaction and defective modulation of ryanodine binding. Our findings suggest that the CaM E105A mutation dysregulates normal cardiac function by a complex mechanism involving alterations in both CaM-Ca2+ and CaM-RyR2 interactions.


Subject(s)
Arrhythmias, Cardiac/genetics , Calmodulin/genetics , Calmodulin/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Tachycardia, Ventricular/genetics , Animals , Arrhythmias, Cardiac/pathology , Calcium/metabolism , Calcium Signaling/physiology , Child , Excitation Contraction Coupling/physiology , Heart Rate/genetics , Heart Rate/physiology , Humans , Male , Myocytes, Cardiac/metabolism , Tachycardia, Ventricular/physiopathology , Zebrafish
12.
Biochem J ; 475(24): 3933-3948, 2018 12 14.
Article in English | MEDLINE | ID: mdl-30446606

ABSTRACT

The most common inherited cardiac disorder, hypertrophic cardiomyopathy (HCM), is characterized by thickening of heart muscle, for which genetic mutations in cardiac myosin-binding protein C3 (c-MYBPC3) gene, is the leading cause. Notably, patients with HCM display a heterogeneous clinical presentation, onset and prognosis. Thus, delineating the molecular mechanisms that explain how disparate c-MYBPC3 variants lead to HCM is essential for correlating the impact of specific genotypes on clinical severity. Herein, five c-MYBPC3 missense variants clinically associated with HCM were investigated; namely V1 (R177H), V2 (A216T), V3 (E258K), V4 (E441K) and double mutation V5 (V3 + V4), all located within the C1 and C2 domains of MyBP-C, a region known to interact with sarcomeric protein, actin. Injection of the variant complementary RNAs in zebrafish embryos was observed to recapitulate phenotypic aspects of HCM in patients. Interestingly, V3- and V5-cRNA injection produced the most severe zebrafish cardiac phenotype, exhibiting increased diastolic/systolic myocardial thickness and significantly reduced heart rate compared with control zebrafish. Molecular analysis of recombinant C0-C2 protein fragments revealed that c-MYBPC3 variants alter the C0-C2 domain secondary structure, thermodynamic stability and importantly, result in a reduced binding affinity to cardiac actin. V5 (double mutant), displayed the greatest protein instability with concomitant loss of actin-binding function. Our study provides specific mechanistic insight into how c-MYBPC3 pathogenic variants alter both functional and structural characteristics of C0-C2 domains leading to impaired actin interaction and reduced contractility, which may provide a basis for elucidating the disease mechanism in HCM patients with c-MYBPC3 mutations.


Subject(s)
Actins/metabolism , Cardiomyopathy, Hypertrophic/metabolism , Carrier Proteins/metabolism , Genetic Variation/physiology , Mutation, Missense/physiology , Actins/genetics , Adult , Animals , Cardiomyopathy, Hypertrophic/genetics , Carrier Proteins/chemistry , Carrier Proteins/genetics , Humans , Protein Binding/physiology , Protein Structure, Secondary , Zebrafish
13.
Biomed Res Int ; 2018: 1642684, 2018.
Article in English | MEDLINE | ID: mdl-30363733

ABSTRACT

Over the last decade, the zebrafish (Danio rerio) has emerged as a model organism for cardiovascular research. Zebrafish have several advantages over mammalian models. For instance, the experimental cost of using zebrafish is comparatively low; the embryos are transparent, develop externally, and have high fecundity making them suitable for large-scale genetic screening. More recently, zebrafish embryos have been used for the screening of a variety of toxic agents, particularly for cardiotoxicity testing. Zebrafish has been shown to exhibit physiological responses that are similar to mammals after exposure to medicinal drugs including xenobiotics, hormones, cancer drugs, and also environmental pollutants, including pesticides and heavy metals. In this review, we provided a summary for recent studies that have used zebrafish to investigate the molecular mechanisms of drug-induced cardiotoxicity. More specifically, we focused on the techniques that were exploited by us and others for cardiovascular toxicity assessment and described several microscopic imaging and analysis protocols that are being used for the estimation of a variety of cardiac hemodynamic parameters.


Subject(s)
Cardiotoxicity/etiology , Drug-Related Side Effects and Adverse Reactions/etiology , Pharmaceutical Preparations/administration & dosage , Zebrafish/physiology , Animals , Hemodynamics/physiology , Humans
14.
J Cell Physiol ; 232(7): 1650-1659, 2017 Jul.
Article in English | MEDLINE | ID: mdl-27731493

ABSTRACT

Hypertrophic cardiomyopathy (HCM) is a common autosomal dominant genetic cardiovascular disorder marked by genetic and phenotypic heterogeneity. Mutations in the gene encodes the cardiac myosin-binding protein C, cMYBPC3 is amongst the various sarcomeric genes that are associated with HCM. These mutations produce mutated mRNAs and truncated cMyBP-C proteins. In this review, we will discuss the implications and molecular mechanisms involved in MYBPC3 different mutations. Further, we will highlight the novel targets that can be developed into potential therapeutics for the treatment of HMC. J. Cell. Physiol. 232: 1650-1659, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Cardiomyopathy, Hypertrophic/metabolism , Cardiomyopathy, Hypertrophic/therapy , Carrier Proteins/metabolism , Myocardium/metabolism , Animals , Carrier Proteins/genetics , Humans , Models, Molecular , Molecular Targeted Therapy , Mutation/genetics
15.
Methods Mol Biol ; 1220: 29-57, 2015.
Article in English | MEDLINE | ID: mdl-25388243

ABSTRACT

The many advantages of the zebrafish model provide a unique opportunity to integrate the tools of developmental embryology, transgenesis, and functional assays to elucidate the molecular pathways underlying hematopoiesis and for modeling human blood diseases. These methodologies have recently been applied to the zebrafish mast cell lineage and have resulted in a better understanding of vertebrate mast cell biology. By employing whole-mount in situ hybridization alone and in combination with co-localization approaches, fluorescence-activated cell sorting (FACS), and morpholino gene knockdown studies, new insights into early mast cell transcriptional regulation and ontogeny have been exposed in vivo. Transgenic strategies have permitted the modeling of human mast cell diseases, like systemic mastocytosis in zebrafish, which can subsequently be exploited for high-throughput chemical screens to identify potential therapies in these conditions. Mast cell functional assays have been adapted to zebrafish providing the opportunity to utilize this model for interrogating the cellular players in innate and adaptive immunity and as a live animal readout for drug responses in allergic and inflammatory reactions. These techniques are detailed in the following chapter.


Subject(s)
Mast Cells/cytology , Zebrafish/embryology , Animals , Animals, Genetically Modified , Antigens/immunology , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/immunology , Female , Flow Cytometry , Humans , Immunoglobulin E/immunology , Immunohistochemistry , In Situ Hybridization , Male , Mast Cells/immunology , Mast Cells/metabolism , Mast Cells/ultrastructure , Microscopy, Electron , RNA Probes/metabolism , Tissue Fixation , Tryptases/metabolism , Zebrafish/genetics
16.
Br J Haematol ; 167(1): 48-61, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24989799

ABSTRACT

Systemic mastocytosis (SM) is a rare myeloproliferative disease without curative therapy. Despite clinical variability, the majority of patients harbour a KIT-D816V mutation, but efforts to inhibit mutant KIT with tyrosine kinase inhibitors have been unsatisfactory, indicating a need for new preclinical approaches to identify alternative targets and novel therapies in this disease. Murine models to date have been limited and do not fully recapitulate the most aggressive forms of SM. We describe the generation of a transgenic zebrafish model expressing the human KIT-D816V mutation. Adult fish demonstrate a myeloproliferative disease phenotype, including features of aggressive SM in haematopoeitic tissues and high expression levels of endopeptidases, consistent with SM patients. Transgenic embryos demonstrate a cell-cycle phenotype with corresponding expression changes in genes associated with DNA maintenance and repair, such as reduced dnmt1. In addition, epcam was consistently downregulated in both transgenic adults and embryos. Decreased embryonic epcam expression was associated with reduced neuromast numbers, providing a robust in vivo phenotypic readout for chemical screening in KIT-D816V-induced disease. This study represents the first zebrafish model of a mast cell disease with an aggressive adult phenotype and embryonic markers that could be exploited to screen for novel agents in SM.


Subject(s)
Gene Expression , Mastocytosis, Systemic/genetics , Mutation , Proto-Oncogene Proteins c-kit/genetics , Animals , Animals, Genetically Modified , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Apoptosis/genetics , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Cycle/genetics , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , Disease Models, Animal , Embryo, Nonmammalian/metabolism , Epithelial Cell Adhesion Molecule , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Gene Order , Genetic Vectors , Hematopoiesis/genetics , Humans , Kidney/pathology , Mast Cells/enzymology , Mastocytosis , Peptide Hydrolases/genetics , Peptide Hydrolases/metabolism , Phenotype , Zebrafish , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
17.
Blood ; 119(15): 3585-94, 2012 Apr 12.
Article in English | MEDLINE | ID: mdl-22368273

ABSTRACT

We used the opportunities afforded by the zebrafish to determine upstream pathways regulating mast cell development in vivo and identify their cellular origin. Colocalization studies demonstrated zebrafish notch receptor expression in cells expressing carboxypeptidase A5 (cpa5), a zebrafish mast cell-specific marker. Inhibition of the Notch pathway resulted in decreased cpa5 expression in mindbomb mutants and wild-type embryos treated with the γ-secretase inhibitor, Compound E. A series of morpholino knockdown studies specifically identified notch1b and gata2 as the critical factors regulating mast cell fate. Moreover, hsp70::GAL4;UAS::nicd1a transgenic embryos overexpressing an activated form of notch1, nicd1a, displayed increased cpa5, gata2, and pu.1 expression. This increase in cpa5 expression could be reversed and reduced below baseline levels in a dose-dependent manner using Compound E. Finally, evidence that cpa5 expression colocalizes with lmo2 in the absence of hematopoietic stem cells revealed that definitive mast cells initially delineate from erythromyeloid progenitors. These studies identify a master role for Notch signaling in vertebrate mast cell development and establish developmental origins of this lineage. Moreover, these findings postulate targeting the Notch pathway as a therapeutic strategy in mast cell diseases.


Subject(s)
Cell Lineage/genetics , Homeodomain Proteins/physiology , Mast Cells/physiology , Nerve Tissue Proteins/physiology , Receptor, Notch1/physiology , Zebrafish Proteins/physiology , Zebrafish/immunology , Animals , Animals, Genetically Modified , Carboxypeptidases A/genetics , Carboxypeptidases A/metabolism , Carboxypeptidases A/physiology , Cell Differentiation/genetics , Embryo, Nonmammalian , Gene Expression Regulation, Developmental/drug effects , Homeodomain Proteins/antagonists & inhibitors , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mast Cells/metabolism , Morpholinos/pharmacology , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , Signal Transduction/physiology , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
18.
Br J Haematol ; 155(2): 167-81, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21810091

ABSTRACT

NUP98-HOXA9 [t(7;11) (p15;p15)] is associated with inferior prognosis in de novo and treatment-related acute myeloid leukaemia (AML) and contributes to blast crisis in chronic myeloid leukaemia (CML). We have engineered an inducible transgenic zebrafish harbouring human NUP98-HOXA9 under the zebrafish spi1(pu.1) promoter. NUP98-HOXA9 perturbed zebrafish embryonic haematopoiesis, with upregulated spi1 expression at the expense of gata1a. Markers associated with more differentiated myeloid cells, lcp1, lyz, and mpx were also elevated, but to a lesser extent than spi1, suggesting differentiation of early myeloid progenitors may be impaired by NUP98-HOXA9. Following irradiation, NUP98-HOXA9-expressing embryos showed increased numbers of cells in G2-M transition compared to controls and absence of a normal apoptotic response, which may result from an upregulation of bcl2. These data suggest NUP98-HOXA9-induced oncogenesis may result from a combination of defects in haematopoiesis and an aberrant response to DNA damage. Importantly, 23% of adult NUP98-HOXA9-transgenic fish developed a myeloproliferative neoplasm (MPN) at 19-23 months of age. In summary, we have identified an embryonic haematopoietic phenotype in a transgenic zebrafish line that subsequently develops MPN. This tool provides a unique opportunity for high-throughput in vivo chemical modifier screens to identify novel therapeutic agents in high risk AML.


Subject(s)
Cell Transformation, Neoplastic/genetics , Homeodomain Proteins/genetics , Leukemia, Experimental/genetics , Myeloid Cells/pathology , Myeloproliferative Disorders/genetics , Nuclear Pore Complex Proteins/genetics , Oncogene Proteins, Fusion/genetics , Animals , Animals, Genetically Modified , Apoptosis , Cell Cycle , Cell Lineage , DNA Damage , GATA1 Transcription Factor/physiology , Gene Expression Regulation, Developmental , Gene Expression Regulation, Leukemic , Genes, Reporter , Hematopoiesis/genetics , Homeodomain Proteins/physiology , Humans , Leukemia, Experimental/pathology , Leukemia, Radiation-Induced/genetics , Leukemia, Radiation-Induced/pathology , Myeloid Cells/radiation effects , Myeloproliferative Disorders/pathology , Nuclear Pore Complex Proteins/physiology , Oncogene Proteins, Fusion/physiology , Phenotype , Promoter Regions, Genetic , Proto-Oncogene Proteins/genetics , Recombinant Fusion Proteins/physiology , Trans-Activators/genetics , Transgenes , Zebrafish/embryology , Zebrafish Proteins/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...