Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Hum Mol Genet ; 28(13): 2212-2223, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31220269

ABSTRACT

Alström syndrome (OMIM #203800) is an autosomal recessive obesity ciliopathy caused by loss-of-function mutations in the ALMS1 gene. In addition to multi-organ dysfunction, such as cardiomyopathy, retinal degeneration and renal dysfunction, the disorder is characterized by high rates of obesity, insulin resistance and early-onset type 2 diabetes mellitus (T2DM). To investigate the underlying mechanisms of T2DM phenotypes, we generated a loss-of-function deletion of alms1 in the zebrafish. We demonstrate conservation of hallmark clinical characteristics alongside metabolic syndrome phenotypes, including a propensity for obesity and fatty livers, hyperinsulinemia and glucose response defects. Gene expression changes in ß-cells isolated from alms1-/- mutants revealed changes consistent with insulin hypersecretion and glucose sensing failure, which were corroborated in cultured murine ß-cells lacking Alms1. We also found evidence of defects in peripheral glucose uptake and concomitant hyperinsulinemia in the alms1-/- animals. We propose a model in which hyperinsulinemia is the primary and causative defect underlying generation of T2DM associated with alms1 deficiency. These observations support the alms1 loss-of-function zebrafish mutant as a monogenic model for mechanistic interrogation of T2DM phenotypes.


Subject(s)
Alstrom Syndrome/genetics , Diabetes Mellitus, Type 2/genetics , Insulin Resistance/genetics , Renal Insufficiency/genetics , Retinal Degeneration/genetics , Zebrafish/genetics , Alstrom Syndrome/physiopathology , Animals , Animals, Genetically Modified , Cell Line , Disease Models, Animal , Glucose Intolerance , Hyperinsulinism/genetics , Insulin-Secreting Cells/metabolism , Mice , Models, Biological , Obesity/genetics , Phenotype , Zebrafish/embryology
2.
Circulation ; 138(13): 1343-1355, 2018 09 25.
Article in English | MEDLINE | ID: mdl-29593015

ABSTRACT

BACKGROUND: Elevated levels of low-density lipoprotein cholesterol (LDL-C) are a major risk factor for cardiovascular disease via its contribution to the development and progression of atherosclerotic lesions. Although the genetic basis of LDL-C has been studied extensively, currently known genetic variants account for only ≈20% of the variation in LDL-C levels. METHODS: Through an array-based association analysis in 1102 Amish subjects, we identified a variant strongly associated with LDL-C levels. Using a combination of genetic analyses, zebrafish models, and in vitro experiments, we sought to identify the causal gene driving this association. RESULTS: We identified a founder haplotype associated with a 15 mg/dL increase in LDL-C on chromosome 5. After recombination mapping, the associated region contained 8 candidate genes. Using a zebrafish model to evaluate the relevance of these genes to cholesterol metabolism, we found that expression of the transcribed pseudogene, APOOP1, increased LDL-C and vascular plaque formation. CONCLUSIONS: Based on these data, we propose that APOOP1 regulates levels of LDL-C in humans, thus identifying a novel mechanism of lipid homeostasis.


Subject(s)
Amish/genetics , Atherosclerosis/genetics , Cholesterol, LDL/blood , Chromosomes, Human, Pair 5 , Dyslipidemias/genetics , Pseudogenes , Animals , Animals, Genetically Modified , Atherosclerosis/blood , Atherosclerosis/diagnosis , Atherosclerosis/ethnology , Dyslipidemias/blood , Dyslipidemias/diagnosis , Dyslipidemias/ethnology , Founder Effect , Genetic Association Studies , Genetic Predisposition to Disease , Haplotypes , Humans , Phenotype , Recombination, Genetic , Risk Factors , Zebrafish/genetics
3.
J Vis Exp ; (127)2017 09 20.
Article in English | MEDLINE | ID: mdl-28994750

ABSTRACT

Microtubules (MTs) are dynamic and fragile structures that are challenging to image in vivo, particularly in vertebrate embryos. Immunolabeling methods are described here to analyze distinct populations of MTs in the developing neural tube of the zebrafish embryo. While the focus is on neural tissue, this methodology is broadly applicable to other tissues. The procedures are optimized for early to mid-somitogenesis-stage embryos (1 somite to 12 somites), however they can be adapted to a range of other stages with relatively minor adjustments. The first protocol provides a method to assess the spatial distribution of stable and dynamic MTs and perform a quantitative analysis of these populations with image-processing software. This approach complements existing tools to image microtubule dynamics and distribution in real-time, using transgenic lines or transient expression of tagged constructs. Indeed, such tools are very useful, however they do not readily distinguish between dynamic and stable MTs. The ability to image and analyze these distinct microtubule populations has important implications for understanding mechanisms underlying cell polarization and morphogenesis. The second protocol outlines a technique to analyze nascent MTs specifically. This is accomplished by capturing the de novo growth properties of MTs over time, following microtubule depolymerization with the drug nocodazole and a recovery period after drug washout. This technique has not yet been applied to the study of MTs in zebrafish embryos, but is a valuable assay for investigating the in vivo function of proteins implicated in microtubule assembly.


Subject(s)
Immunohistochemistry/methods , Microtubules/metabolism , Zebrafish/embryology , Animals , Embryonic Development
4.
Hepatology ; 65(5): 1526-1542, 2017 05.
Article in English | MEDLINE | ID: mdl-28027591

ABSTRACT

The transmembrane 6 superfamily member 2 (TM6SF2) loss-of-function variant rs58542926 is a genetic risk factor for nonalcoholic fatty liver disease and progression to fibrosis but is paradoxically associated with lower levels of hepatically derived triglyceride-rich lipoproteins. TM6SF2 is expressed predominantly in liver and small intestine, sites for triglyceride-rich lipoprotein biogenesis and export. In light of this, we hypothesized that TM6SF2 may exhibit analogous effects on both liver and intestine lipid homeostasis. To test this, we genotyped rs58542926 in 983 bariatric surgery patients from the Geisinger Medical Center for Nutrition and Weight Management, Geisinger Health System, in Pennsylvania and from 3,556 study participants enrolled in the Amish Complex Disease Research Program. Although these two cohorts have different metabolic profiles, carriers in both cohorts had improved fasting lipid profiles. Importantly, following a high-fat challenge, carriers in the Amish Complex Disease Research Program cohort exhibited significantly lower postprandial serum triglycerides, suggestive of a role for TM6SF2 in the small intestine. To gain further insight into this putative role, effects of TM6SF2 deficiency were studied in a zebrafish model and in cultured human Caco-2 enterocytes. In both systems TM6SF2 deficiency resulted in defects in small intestine metabolism in response to dietary lipids, including significantly increased lipid accumulation, decreased lipid clearance, and increased endoplasmic reticulum stress. CONCLUSIONS: These data strongly support a role of TM6SF2 in the regulation of postprandial lipemia, potentially through a similar function for TM6SF2 in the lipidation and/or export of both hepatically and intestinally derived triglyceride-rich lipoproteins. (Hepatology 2017;65:1526-1542).


Subject(s)
Endoplasmic Reticulum Stress , Intestine, Small/metabolism , Lipid Metabolism/genetics , Liver/metabolism , Membrane Proteins/genetics , Animals , Base Sequence , Caco-2 Cells , Enterocytes/metabolism , Fatty Liver/genetics , Female , Hepatocytes/metabolism , Homeostasis , Humans , Intestine, Small/ultrastructure , Male , Membrane Proteins/metabolism , Mice , Middle Aged , Molecular Sequence Data , Polymorphism, Single Nucleotide , Postprandial Period , Triglycerides/biosynthesis , Triglycerides/blood , Tunicamycin , Zebrafish
5.
Neural Dev ; 11: 1, 2016 Jan 18.
Article in English | MEDLINE | ID: mdl-26782621

ABSTRACT

BACKGROUND: Shaping of the neural tube, the precursor of the brain and spinal cord, involves narrowing and elongation of the neural tissue, concomitantly with other morphogenetic changes that contribue to this process. In zebrafish, medial displacement of neural cells (neural convergence or NC), which drives the infolding and narrowing of the neural ectoderm, is mediated by polarized migration and cell elongation towards the dorsal midline. Failure to undergo proper NC results in severe neural tube defects, yet the molecular underpinnings of this process remain poorly understood. RESULTS: We investigated here the role of the microtubule (MT) cytoskeleton in mediating NC in zebrafish embryos using the MT destabilizing and hyperstabilizing drugs nocodazole and paclitaxel respectively. We found that MTs undergo major changes in organization and stability during neurulation and are required for the timely completion of NC by promoting cell elongation and polarity. We next examined the role of Microtubule-associated protein 1B (Map1b), previously shown to promote MT dynamicity in axons. map1b is expressed earlier than previously reported, in the developing neural tube and underlying mesoderm. Loss of Map1b function using morpholinos (MOs) or δMap1b (encoding a truncated Map1b protein product) resulted in delayed NC and duplication of the neural tube, a defect associated with impaired NC. We observed a loss of stable MTs in these embryos that is likely to contribute to the NC defect. Lastly, we found that Map1b mediates cell elongation in a cell autonomous manner and polarized protrusive activity, two cell behaviors that underlie NC and are MT-dependent. CONCLUSIONS: Together, these data highlight the importance of MTs in the early morphogenetic movements that shape the neural tube and reveal a novel role for the MT regulator Map1b in mediating cell elongation and polarized cell movement in neural progenitor cells.


Subject(s)
Microtubule-Associated Proteins/metabolism , Neural Tube/embryology , Neurulation , Zebrafish Proteins/metabolism , Animals , Cell Movement/drug effects , Cell Polarity/drug effects , Neural Tube/drug effects , Neural Tube/metabolism , Neurulation/drug effects , Nocodazole/administration & dosage , Paclitaxel/administration & dosage , Tubulin Modulators/administration & dosage , Zebrafish
6.
Int J Cell Biol ; 2014: 152645, 2014.
Article in English | MEDLINE | ID: mdl-24719618

ABSTRACT

A common assumption of excitotoxic mechanisms in the nervous system is that the ionic imbalance resulting from overstimulation of glutamate receptors and increased Na(+) and Ca(++) influx overwhelms cellular energy metabolic systems leading to cell death. The goal of this study was to examine how a chronic Na(+) channel leak current in developing Purkinje cells in the heterozygous Lurcher mutant (+/Lc) affects the expression and distribution of the α 3 subunit of the Na(+)/K(+) ATPase pump, a key component of the homeostasis system that maintains ionic equilibrium in neurons. The expression pattern of the catalytic α 3 Na(+)/K(+) ATPase subunit was analyzed by immunohistochemistry, histochemistry, and Western Blots in wild type (WT) and +/Lc cerebella at postnatal days P10, P15, and P25 to determine if there are changes in the distribution of active Na(+)/K(+) ATPase subunits in degenerating Purkinje cells. The results suggest that the expression of the catalytic α 3 subunit is altered in chronically depolarized +/Lc Purkinje cells, although the density of active Na(+)/K(+) ATPase pumps is not significantly altered compared with WT in the cerebellar cortex at P15, and then declines from P15 to P25 in the +/Lc cerebellum as the +/Lc Purkinje cells degenerate.

7.
Cerebellum ; 12(3): 377-89, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23136008

ABSTRACT

Recent studies using both dissociated and organotypic cell cultures have shown that heterozygous Lurcher (Lc/+) Purkinje cells (PCs) grown in vitro share many of the same survival and morphological characteristics as Lc/+ PCs in vivo. We have used this established tissue culture system as a valuable model for studying cell death mechanisms in a relatively simple system where neurodegeneration is induced by a constitutive cation leak mediated by the Lurcher mutation in the δ2 glutamate receptor (GluRδ2). In this study, Ca(++) imaging and immunocytochemistry studies indicate that intracellular levels of Ca(++) are chronically increased in Lc/+ PCs and the concentration and/or distribution of the conventional PKCγ isoform is altered in degenerating Lc/+ PCs. To begin to characterize the molecular mechanisms that regulate Lc/+ PC death, the contributions of conventional PKC pathways and of two MAP kinase family members, JNK and p38, were examined in slice cultures from wild-type and Lc/+ mutant mouse cerebellum. Cerebellar slice cultures from P0 pups were treated with either a conventional PKC inhibitor, a JNK inhibitor, or a p38 inhibitor either from 0 to 14 or 7 to 14 DIV. Treatment with either of the three inhibitors from 0 DIV significantly increased wild type and Lc/+ PC survival through 14 DIV, but only Lc/+ PC survival was significantly increased following treatments from 7 to 14 DIV. The results suggest that multiple PC death pathways are induced by the physical trauma of making organotypic slice cultures, naturally-occurring postnatal cell death, and the GluRδ2 (Lc) mutation.


Subject(s)
Cerebellum/cytology , Mitogen-Activated Protein Kinases/metabolism , Protein Kinase C/metabolism , Purkinje Cells/drug effects , Receptors, Glutamate/genetics , Signal Transduction/drug effects , Analysis of Variance , Animals , Animals, Newborn , Calbindins/metabolism , Calcium/metabolism , Caspase 3/metabolism , Cell Survival/drug effects , Cell Survival/genetics , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/genetics , Male , Mice , Mice, Inbred CBA , Mice, Transgenic , Organ Culture Techniques , Signal Transduction/genetics
8.
Eur J Neurosci ; 36(6): 2773-81, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22762562

ABSTRACT

Adult rats exposed to the DNA-methylating agent methylazoxymethanol on embryonic day 17 show a pattern of neurobiological deficits that model some of the neuropathological and behavioral changes observed in schizophrenia. Although it is generally assumed that these changes reflect targeted disruption of embryonic neurogenesis, it is unknown whether these effects generalise to other antimitotic agents administered at different stages of development. In the present study, neurochemical, behavioral and electrophysiological techniques were used to determine whether exposure to the antimitotic agent Ara-C later in development recapitulates some of the changes observed in methylazoxymethanol (MAM)-treated animals and in patients with schizophrenia. Male rats exposed to Ara-C (30 mg/kg/day) at embryonic days 19.5 and 20.5 show reduced cell numbers and heterotopias in hippocampal CA1 and CA2/3 regions, respectively, as well as cell loss in the superficial layers of the pre- and infralimbic cortex. Birth date labeling with bromodeoxyuridine reveals that the cytoarchitectural changes in CA2/3 are a consequence rather that a direct result of disrupted cortical neurogenesis. Ara-C-treated rats possess elevated levels of cortical dopamine and DOPAC (3,4-didyhydroxypheylacetic acid) but no change in norepinephrine or serotonin. Ara-C-treated rats are impaired in their ability to learn the Morris water maze task and showed diminished synaptic plasticity in the hippocampocortical pathway. These data indicate that disruption of neurogenesis at embryonic days 19.5 and 20.5 constitutes a useful model for the comparative study of deficits observed in other gestational models and their relationship to cognitive changes observed in schizophrenia.


Subject(s)
Endophenotypes , Hippocampus/physiopathology , Maze Learning/drug effects , Neuronal Plasticity , Schizophrenia/physiopathology , 3,4-Dihydroxyphenylacetic Acid/metabolism , Animals , Cytarabine , Disease Models, Animal , Dopamine/metabolism , Hippocampus/embryology , Hippocampus/pathology , Male , Neurogenesis/drug effects , Neuronal Plasticity/drug effects , Norepinephrine/metabolism , Rats , Rats, Sprague-Dawley , Schizophrenia/chemically induced , Schizophrenia/metabolism , Schizophrenia/pathology , Serotonin/metabolism
9.
Cerebellum ; 10(3): 504-14, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21104177

ABSTRACT

The Lurcher mutant mouse is characterized by its ataxic gait and loss of cerebellar Purkinje cells and their afferents, granule cells and olivary neurons, during the first weeks of postnatal development. For the 50 years since its discovery, the heterozygous Lurcher mutant has served as an important model system for studying neuron-target interactions in the developing cerebellum and cerebellar function. The identification of the Lurcher (Lc) gene over 10 years ago as a gain-of-function mutation in the δ2 glutamate receptor (GluRδ2) led to extensive studies of cell death mechanisms in the Lc/+ cerebellum. The advantage of this model system is that GluRδ2(+) receptors and GluRδ2(Lc) channels are expressed predominantly in Purkinje cells, making it possible to study the effects of a well-characterized leak current in a well-defined cell type during a critical phase of neuronal development. Yet there is still controversy surrounding the mechanisms of neuronal death in Lc/+ Purkinje cells with competing hypotheses for necrotic, apoptotic, and autophagic cell death pathways as a consequence of the excitotoxic stress caused by the GluRδ2(Lc) leak current. The goal of this review is to summarize recent studies that critically test the role of various cell death pathways in Lc/+ Purkinje cell degeneration with respect to evidence for the molecular heterogeneity of Purkinje cells. We propose that the expression of putative survival factors, such as heat shock proteins, in a subset of cerebellar Purkinje cells may affect cell death pathways and account for the pattern and diverse mechanisms of Lc/+ Purkinje degeneration.


Subject(s)
Apoptosis/genetics , Cerebellum/pathology , Mice, Neurologic Mutants , Purkinje Cells/pathology , Purkinje Cells/physiology , Animals , Animals, Newborn , Cell Survival/genetics , Cerebellum/growth & development , Heat-Shock Proteins/metabolism , Mice , Models, Neurological , Molecular Chaperones , Neoplasm Proteins/metabolism
10.
Dev Neurobiol ; 69(8): 505-17, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19294643

ABSTRACT

The differentiation and survival of heterozygous Lurcher (+/Lc) Purkinje cells in vitro was examined as a model system for studying how chronic ionic stress affects neuronal differentiation and survival. The Lurcher mutation in the delta2 glutamate receptor (GluRdelta2) converts an orphan receptor into a membrane channel that constitutively passes an inward cation current. In the GluRdelta2(+/Lc) mutant, Purkinje cell dendritic differentiation is disrupted and the cells degenerate following the first week of postnatal development. To determine if the GluRdelta2(+/Lc) Purkinje cell phenotype is recapitulated in vitro, +/+, and +/Lc Purkinje cells from postnatal Day 0 pups were grown in either isolated cell or cerebellar slice cultures. GluRdelta2(+/+) and GluRdelta2(+/Lc) Purkinje cells appeared to develop normally through the first 7 days in vitro (DIV), but by 11 DIV GluRdelta2(+/Lc) Purkinje cells exhibited a significantly higher cation leak current. By 14 DIV, GluRdelta2(+/Lc) Purkinje cell dendrites were stunted and the number of surviving GluRdelta2(+/Lc) Purkinje cells was reduced by 75% compared to controls. However, treatment of +/Lc cerebellar cultures with 1-naphthyl acetyl spermine increased +/Lc Purkinje cell survival to wild type levels. These results support the conclusion that the Lurcher mutation in GluRdelta2 induces cell autonomous defects in differentiation and survival. The establishment of a tissue culture system for studying cell injury and death mechanisms in a relatively simple system like GluRdelta2(+/Lc) Purkinje cells will provide a valuable model for studying how the induction of a chronic inward cation current in a single cell type affects neuronal differentiation and survival.


Subject(s)
Cerebellar Cortex/metabolism , Ion Channels/metabolism , Nerve Degeneration/metabolism , Purkinje Cells/metabolism , Receptors, Glutamate/genetics , Receptors, Glutamate/metabolism , Animals , Cations/metabolism , Cell Death/drug effects , Cell Death/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Survival/drug effects , Cell Survival/genetics , Cells, Cultured , Cerebellar Cortex/growth & development , Cerebellar Cortex/physiopathology , Disease Models, Animal , Female , Ion Channel Gating/genetics , Ion Channels/genetics , Male , Membrane Potentials/genetics , Mice , Mice, Neurologic Mutants , Mutation/genetics , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Organ Culture Techniques , Phenotype , Purkinje Cells/pathology , Spermine/analogs & derivatives , Spermine/pharmacology
11.
Dev Neurobiol ; 67(8): 1032-46, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17565706

ABSTRACT

Oxidative stress is postulated to play a role in cell death in many neurodegenerative diseases. As a model of neonatal neuronal cell death, we have examined the role of oxidative stress in Purkinje cell death in the heterozygous Lurcher mutant (+/Lc). Lurcher is a gain of function mutation in the delta2 glutamate receptor (GluRdelta2) that turns the receptor into a leaky membrane channel, resulting in chronic depolarization of +/Lc Purkinje cells starting around the first week of postnatal development. Virtually, all +/Lc Purkinje cells die by the end of the first postnatal month. To investigate the role of oxidative stress in +/Lc Purkinje cell death, we have examined nitric oxide synthase (NOS) activity and the expression of two markers for oxidative stress, nitrotyrosine and manganese super oxide dismutase (MnSOD), in wild type and +/Lc Purkinje cells at P10, P15, and P25. The results show that NOS activity and immunolabeling for nitrotyrosine and MnSOD are increased in +/Lc Purkinje cells. To determine whether peroxynitrite formation is a prerequisite for +/Lc Purkinje cell death, +/Lc mutants were crossed with an alpha-nNOS knockout mutant (nNOSalpha(-/-)) to reduce the production of NO. Analysis of the double mutants showed that blocking alpha-nNOS expression does not rescue +/Lc Purkinje cells. However, we present evidence for sustained NOS activity and nitrotyrosine formation in the GluRdelta2(+/Lc):nNOS(-/-) double mutant Purkinje cells, which suggests that the failure to rescue GluRdelta2(+/Lc):nNOS(-/-) Purkinje cells may be explained by the induction of alternative nNOS isoforms.


Subject(s)
Mice, Neurologic Mutants , Nitric Oxide Synthase Type I/metabolism , Nitric Oxide/physiology , Oxidative Stress/physiology , Purkinje Cells/physiology , Animals , Calbindins , Cell Death , Cerebellum/physiology , Cerebellum/physiopathology , Mice , Nitric Oxide Synthase/metabolism , Purkinje Cells/cytology , Purkinje Cells/enzymology , Purkinje Cells/pathology , S100 Calcium Binding Protein G/metabolism , Superoxide Dismutase/metabolism , Tyrosine/analogs & derivatives , Tyrosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...