Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Mol Genet Metab ; 125(4): 345-350, 2018 12.
Article in English | MEDLINE | ID: mdl-30392741

ABSTRACT

Niemann-Pick disease, type C1 (NPC1) is an inborn error of metabolism that results in endolysosomal accumulation of unesterified cholesterol. Clinically, NPC1 manifests as cholestatic liver disease in the newborn or as a progressive neurogenerative condition characterized by cerebellar ataxia and cognitive decline. Currently there are no FDA approved therapies for NPC1. Thus, understanding the pathological processes that contribute to neurodegeneration will be important in both developing and testing potential therapeutic interventions. Neuroinflammation and necroptosis contribute to the NPC1 pathological cascade. Receptor Interacting Protein Kinase 1 and 3 (RIPK1 and RIPK3), are protein kinases that play a central role in mediating neuronal necroptosis. Our prior work suggested that pharmacological inhibition of RIPK1 had a significant but modest beneficial effect; however, the inhibitors used in that study had suboptimal pharmacokinetic properties. In this work we evaluated both pharmacological and genetic inhibition of RIPK1 kinase activity. Lifespan in both Npc1-/- mice treated with GSK'547, a RIPK1 inhibitor with better pharmacokinetic properties, and Npc1-/-:Ripk1kd/kd double mutant mice was significantly increased. In both cases the increase in lifespan was modest, suggesting that the therapeutic potential of RIPK1 inhibition, as a monotherapy, is limited. We thus investigated the potential of combining RIPK1 inhibition with 2-hydroxypropyl-ß-cyclodextrin (HPßCD) therapy HPßCD has been shown to slow neurological disease progression in NPC1 mice, cats and patients. HPßCD appeared to have an additive positive effect on the pathology and survival of Npc1-/-:Ripk1kd/kd mice. RIPK1 and RIPK3 are both critical components of the necrosome, thus we were surprised to observe no increase survival in Npc1-/-;Ripk3-/- mice compared to Npc1-/- mice. These data suggest that although necroptosis is occurring in NPC1, the observed effects of RIPK1 inhibition may be related to its RIPK3-independent role in neuroinflammation and cytokine production.


Subject(s)
2-Hydroxypropyl-beta-cyclodextrin/pharmacology , Apoptosis , Necrosis , Niemann-Pick Disease, Type C/therapy , Proteins/physiology , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Animals , Combined Modality Therapy , Disease Models, Animal , Excipients/pharmacology , Intracellular Signaling Peptides and Proteins , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Niemann-Pick C1 Protein , Niemann-Pick Disease, Type C/genetics , Niemann-Pick Disease, Type C/pathology , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors
2.
Cell Death Dis ; 7: e2147, 2016 Mar 17.
Article in English | MEDLINE | ID: mdl-26986514

ABSTRACT

Niemann-Pick disease, type C1 (NPC1) is a neurodegenerative, lysosomal storage disorder due to mutation of the NPC1 gene. The NPC1 phenotype is characterized by progressive neuronal dysfunction, including cerebellar ataxia and dementia. There is histological evidence of neuroinflammation and progressive neuronal loss, with cerebellar Purkinje cells particularly vulnerable to loss of NPC1 function. Necroptosis was evaluated as a mechanism of neuronal loss. Receptor-interacting protein kinase 1 (RIP1) and RIP3 are key components of the necrosomal complex that regulates necroptotic cell death. We report increased expression of RIP1 and RIP3 in NPC1 fibroblasts, NPC1 iPS cell-derived neuronal precursors, and in cerebellar tissue from both NPC1 mice and patients. Our data suggest a positive correlation between NPC1 neurological disease severity and assembly of the necrosome complex. Furthermore, we demonstrate that pharmacological inhibition of RIP1 decreases cell death both in vitro and in vivo. Treatment of Npc1-mutant mice with necrostatin-1, an allosteric inhibitor of RIP1, significantly delayed cerebellar Purkinje cell loss, progression of neurological symptoms, and death. Collectively, our data identified necroptosis as a key component of the molecular network that contributes to neuronal loss in NPC1 and establish that inhibition of necroptosis is a potential therapeutic intervention.


Subject(s)
Neural Stem Cells/metabolism , Niemann-Pick Disease, Type C/metabolism , Niemann-Pick Disease, Type C/therapy , Purkinje Cells/metabolism , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Mutant Strains , Necrosis , Neural Stem Cells/pathology , Niemann-Pick C1 Protein , Niemann-Pick Disease, Type C/genetics , Niemann-Pick Disease, Type C/pathology , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/metabolism , Proteins/genetics , Proteins/metabolism , Purkinje Cells/pathology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
3.
Clin Genet ; 87(6): 570-5, 2015 Jun.
Article in English | MEDLINE | ID: mdl-24813812

ABSTRACT

Data from massively parallel sequencing or 'Next Generation Sequencing' of the human exome has reached a critical mass in both public and private databases, in that these collections now allow researchers to critically evaluate population genetics in a manner that was not feasible a decade ago. The ability to determine pathogenic allele frequencies by evaluation of the full coding sequences and not merely a single nucleotide polymorphism (SNP) or series of SNPs will lead to more accurate estimations of incidence. For demonstrative purposes, we analyzed the causative gene for the disorder Smith-Lemli-Opitz Syndrome (SLOS), the 7-dehydrocholesterol reductase (DHCR7) gene and determined both the carrier frequency for DHCR7 mutations, and predicted an expected incidence of the disorder. Estimations of the incidence of SLOS have ranged widely from 1:10,000 to 1:70,000 while the carrier frequency has been reported as high as 1 in 30. Using four exome data sets with a total of 17,836 chromosomes, we ascertained a carrier frequency of pathogenic DHRC7 mutations of 1.01%, and predict a SLOS disease incidence of 1/39,215 conceptions. This approach highlights yet another valuable aspect of the exome sequencing databases, to inform clinical and health policy decisions related to genetic counseling, prenatal testing and newborn screening.


Subject(s)
Gene Frequency , Mutation , Oxidoreductases Acting on CH-CH Group Donors/genetics , Smith-Lemli-Opitz Syndrome/epidemiology , Smith-Lemli-Opitz Syndrome/genetics , Alleles , Datasets as Topic , Genotype , High-Throughput Nucleotide Sequencing , Humans , Incidence
4.
J Inherit Metab Dis ; 37(3): 415-20, 2014 May.
Article in English | MEDLINE | ID: mdl-24500076

ABSTRACT

Smith-Lemli-Opitz syndrome (SLOS) is an autosomal recessive, multiple congenital anomaly syndrome with cognitive impairment and a distinct behavioral phenotype that includes autistic features. SLOS is caused by a defect in 3ß-hydroxysterol Δ(7)-reductase which leads to decreased cholesterol levels and elevated cholesterol precursors, specifically 7- and 8-dehydrocholesterol. However, the pathological processes contributing to the neurological abnormalities in SLOS have not been defined. In view of prior data suggesting defects in SLOS in vesicular release and given the association of altered serotonin metabolism with autism, we were interested in measuring neurotransmitter metabolite levels in SLOS to assess their potential to be used as biomarkers in therapeutic trials. We measured cerebral spinal fluid levels of serotonin and dopamine metabolites, 5-hydroxyindoleacetic acid (5HIAA) and homovanillic acid (HVA) respectively, in 21 SLOS subjects. Results were correlated with the SLOS anatomical severity score, Aberrant Behavior Checklist scores and concurrent sterol biochemistry. Cerebral spinal fluid (CSF) levels of both 5HIAA and HVA were significantly reduced in SLOS subjects. In individual patients, the levels of both 5HIAA and HVA were reduced to a similar degree. CSF neurotransmitter metabolite levels did not correlate with either CSF sterols or behavioral measures. This is the first study demonstrating decreased levels of CSF neurotransmitter metabolites in SLOS. We propose that decreased levels of neurotransmitters in SLOS are caused by a sterol-related defect in synaptic vesicle formation and that CSF 5HIAA and HVA will be useful biomarkers in development of future therapeutic trials.


Subject(s)
Homovanillic Acid/cerebrospinal fluid , Hydroxyindoleacetic Acid/cerebrospinal fluid , Smith-Lemli-Opitz Syndrome/cerebrospinal fluid , Adolescent , Child , Child, Preschool , Cholesterol/administration & dosage , Cholesterol/cerebrospinal fluid , Cross-Over Studies , Female , Humans , Male , Smith-Lemli-Opitz Syndrome/physiopathology , Synaptic Vesicles/physiology
6.
Neurology ; 76(4): 366-72, 2011 Jan 25.
Article in English | MEDLINE | ID: mdl-21205675

ABSTRACT

OBJECTIVE: Niemann-Pick disease type C (NPC) is an inherited disorder characterized by intracellular accumulation of lipids such as cholesterol and glycosphingolipids in endosomes and lysosomes. This accumulation induces progressive degeneration of the nervous system. NPC shows some intriguing similarities with Alzheimer disease (AD), including neurofibrillary tangles, but patients with NPC generally lack amyloid-ß (Aß) plaques. Lipids affect γ-secretase-dependent amyloid precursor protein (APP) metabolism that generates Aß in vitro, but this has been difficult to prove in vivo. Our aim was to assess the effect of altered lipid constituents in neuronal membranes on amyloidogenic APP processing in humans. METHODS: We examined Aß in CSF from patients with NPC (n = 38) and controls (n = 14). CSF was analyzed for Aß(38), Aß(40), Aß(42), α-cleaved soluble APP, ß-cleaved soluble APP, total-tau, and phospho-tau. RESULTS: Aß release was markedly increased in NPC, with a shift toward the Aß(42) isoform. Levels of α- and ß-cleaved soluble APP were similar in patients and controls. Patients with NPC had increased total-tau. Patients on treatment with miglustat (n = 18), a glucosylceramide synthase blocker, had lower Aß(42) and total-tau than untreated patients. CONCLUSION: Increased CSF levels of Aß(38), Aß(40), and Aß(42) and unaltered levels of ß-cleaved soluble APP are consistent with increased γ-secretase-dependent Aß release in the brains of patients with NPC. These results provide the first in vivo evidence that neuronal lipid accumulation facilitates γ-secretase-dependent Aß production in humans and may be of relevance to AD pathogenesis.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Niemann-Pick Diseases/enzymology , 1-Deoxynojirimycin/analogs & derivatives , 1-Deoxynojirimycin/therapeutic use , Adolescent , Amyloid beta-Peptides/cerebrospinal fluid , Amyloid beta-Protein Precursor/cerebrospinal fluid , Apolipoproteins E/genetics , Axons/pathology , Biomarkers/cerebrospinal fluid , Child , Child, Preschool , Cross-Sectional Studies , Female , Genotype , Glucosyltransferases/antagonists & inhibitors , Humans , Infant , Male , Niemann-Pick Diseases/cerebrospinal fluid , Niemann-Pick Diseases/drug therapy , Peptide Fragments/cerebrospinal fluid , Phosphoproteins/cerebrospinal fluid , Young Adult , tau Proteins/cerebrospinal fluid
7.
Neuroscience ; 170(3): 858-64, 2010 Oct 27.
Article in English | MEDLINE | ID: mdl-20670678

ABSTRACT

Cholesterol is an essential component of cellular membranes that is required for normal lipid organization and cell signaling. While the mechanisms associated with maintaining cholesterol homeostasis in the plasma and peripheral tissues have been well studied, the role and regulation of cholesterol biosynthesis in normal brain function and development have proven much more challenging to investigate. Smith-Lemli-Opitz syndrome (SLOS) is a disorder of cholesterol synthesis characterized by mutations of 7-dehydrocholesterol reductase (DHCR7) that impair the reduction of 7-dehydrocholesterol (7DHC) to cholesterol and lead to neurocognitive deficits, including cerebellar hypoplasia and austism behaviors. Here we have used a novel mass spectrometry-based imaging technique called cation-enhanced nanostructure-initiator mass spectrometry (NIMS) for the in situ detection of intact cholesterol molecules from biological tissues. We provide the first images of brain sterol localization in a mouse model for SLOS (Dhcr7(-/-)). In SLOS mice, there is a striking localization of both 7DHC and residual cholesterol in the abnormally developing cerebellum and brainstem. In contrast, the distribution of cholesterol in 1-day old healthy pups was diffuse throughout the cerebrum and comparable to that of adult mice. This study represents the first application of NIMS to localize perturbations in metabolism within pathological tissues and demonstrates that abnormal cholesterol biosynthesis may be particularly important for the development of these brain regions.


Subject(s)
Cholesterol/metabolism , Mass Spectrometry/methods , Smith-Lemli-Opitz Syndrome/metabolism , Animals , Brain/embryology , Brain/metabolism , Disease Models, Animal , Mice , Mice, Mutant Strains , Nanotechnology/methods
10.
Am J Med Genet ; 103(3): 223-5, 2001 Oct 15.
Article in English | MEDLINE | ID: mdl-11745994

ABSTRACT

Smith-Lemli-Opitz syndrome (SLOS) is an autosomal recessive disorder caused by mutations in the 7-dehydrocholesterol reductase gene, DHCR7. The diagnosis is based on the biochemical findings of elevated plasma 7-dehydrocholesterol (7DHC) levels. Adrenal insufficiency with hyponatremia has been reported in 3 patients with severe SLOS; in those cases it was thought to be caused by aldosterone deficiency because it responded to mineralocorticoid replacement. We present a fourth patient with a severe form of SLOS and adrenal insufficiency who had unexplained persistent hypertension, a combination of signs that has not been reported previously in SLOS.


Subject(s)
Adrenal Insufficiency/congenital , Hypertension/congenital , Smith-Lemli-Opitz Syndrome/diagnosis , Cholesterol/blood , Dehydrocholesterols/blood , Humans , Infant, Newborn , Male
11.
Neuron ; 32(4): 591-604, 2001 Nov 20.
Article in English | MEDLINE | ID: mdl-11719201

ABSTRACT

The organizing centers and molecules that pattern the cerebral cortex have been elusive. Here we show that cortical patterning involves regulation of the Lhx2 homeobox gene by the roof plate. Roof plate ablation results in reduced cortical size and Lhx2 expression defects that implicate roof plate signals in the bimodal regulation of Lhx2 in vivo. Bimodal Lhx2 regulation can be recapitulated in explants using two roof plate-derived signaling molecules, Bmp4 and Bmp2. Loss of Lhx2 function results in profound losses of cortical progenitors and neurons, but Lhx2 mutants continue to generate cortical neurons from dorsal sources that may include the roof plate region itself. These findings provide evidence for the roof plate as an organizing center of the developing cortex and for a roof plate-Lhx2 pathway in cortical patterning.


Subject(s)
Cerebral Cortex/embryology , Cerebral Cortex/physiology , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transforming Growth Factor beta , Animals , Bone Morphogenetic Protein 2 , Bone Morphogenetic Protein 4 , Bone Morphogenetic Proteins/pharmacology , Cell Lineage/physiology , Cells, Cultured , Cerebral Cortex/cytology , Female , Growth Differentiation Factors , Growth Substances/genetics , LIM-Homeodomain Proteins , Male , Mice , Mice, Knockout , Neurons/cytology , Neurons/physiology , Pregnancy
12.
Mol Genet Metab ; 74(1-2): 105-19, 2001.
Article in English | MEDLINE | ID: mdl-11592808

ABSTRACT

Over the past few years, the number of identified inborn errors of cholesterol biosynthesis has increased significantly. The first inborn error of cholesterol biosynthesis to be characterized, in the mid 1980s, was mevalonic aciduria. In 1993, Irons et al. ( 1 ) (M. Irons, E. R. Elias, G. Salen, G. S. Tint, and A. K. Batta, Lancet 341:1414, 1993) reported that Smith-Lemli-Opitz syndrome, a classic autosomal recessive malformation syndrome, was due to an inborn error of cholesterol biosynthesis. This was the first inborn error of postsqualene cholesterol biosynthesis to be identified, and subsequently additional inborn errors of postsqualene cholesterol biosynthesis have been characterized to various extent. To date, eight inborn errors of cholesterol metabolism have been described in human patients or in mutant mice. The enzymatic steps impaired in these inborn errors of metabolism include mevolonate kinase (mevalonic aciduria as well as hyperimmunoglobulinemia D and periodic fever syndrome), squalene synthase (Ss-/- mouse), 3beta-hydroxysteroid Delta14-reductase (hydrops-ectopic calcification-moth-eaten skeletal dysplasia), 3beta-hydroxysteroid dehydrogenase (CHILD syndrome, bare patches mouse, and striated mouse), 3beta-hydroxysteroid Delta8,Delta7-isomerase (X-linked dominant chondrodysplasia punctata type 2, CHILD syndrome, and tattered mouse), 3beta-hydroxysteroid Delta24-reductase (desmosterolosis) and 3beta-hydroxysteroid Delta7-reductase (RSH/Smith-Lemli-Opitz syndrome and Dhcr7-/- mouse). Identification of the genetic and biochemical defects which give rise to these syndromes has provided the first step in understanding the pathophysiological processes which underlie these malformation syndromes.


Subject(s)
Cholesterol/biosynthesis , Lipid Metabolism, Inborn Errors/genetics , Animals , Cholesterol/metabolism , Disease Models, Animal , Humans , Lipid Metabolism, Inborn Errors/enzymology , Lipid Metabolism, Inborn Errors/urine , Mevalonic Acid/urine
13.
Am J Med Genet ; 103(1): 75-80, 2001 Sep 15.
Article in English | MEDLINE | ID: mdl-11562938

ABSTRACT

Smith-Lemli-Opitz syndrome (RHS) (SLOS, OMIM 270400) is an autosomal recessive disorder of cholesterol biosynthesis caused by mutations of the 3beta-hydroxysterol Delta(7)-Delta(8)-reductase gene, DHCR7. We report a fetus with holoprosencephaly and multiple congenital anomalies who was homozygous for the IVS8-1G-->C mutation. Following termination of pregnancy, both the elevated amniotic fluid 7-dehydrocholesterol level and the DHCR7 mutations were demonstrated. Two other newborn infants with IVS8-1G-->C/IVS8-1G-->C genotype are described. This report illustrates a severe phenotypic extreme of SLOS associated with a null genotype, underscores the complex relationship between SLOS and holoprosencephaly, and discusses the possible pathogenetic mechanisms of the development of holoprosencephaly in SLOS.


Subject(s)
Oxidoreductases Acting on CH-CH Group Donors , Smith-Lemli-Opitz Syndrome/genetics , Base Sequence , Fatal Outcome , Fetal Death , Fetus , Genotype , Holoprosencephaly/pathology , Homozygote , Humans , Infant, Newborn , Male , Oxidoreductases/genetics , Point Mutation , Smith-Lemli-Opitz Syndrome/pathology
14.
Am J Med Genet ; 102(4): 383-6, 2001 Sep 01.
Article in English | MEDLINE | ID: mdl-11503168

ABSTRACT

Smith-Lemli-Opitz syndrome (SLOS) is an inherited multiple malformation syndrome caused by enzymatic deficiency of 3beta-hydroxysterol-Delta(7)-reductase (DHCR7). SLOS is thought to be most common among European Caucasians, with an incidence of 1 in 20,000 to 1 in 30,000 births. To define the carrier rate and ethnic distribution of SLOS, we screened DNA samples from 2,978 unrelated individuals for the most common SLOS mutation (IVS8-1G-->C). Twenty-four heterozygotes of the IVS8-1G-->C mutation were detected in 2,978 individuals of European Caucasian and Black backgrounds. For European Caucasians, the carrier rate for SLOS may be as high as 1 in 30, suggesting an incidence of 1 in 1,700 to 1 in 13,400. This high number is supported by the recent observation of newborn and prenatal incidence of 1 in 22,000 in the Caucasian population. Ours is the first report of the IVS8-1G-->C mutation in persons of African ancestry. Published 2001 Wiley-Liss, Inc.


Subject(s)
Gene Frequency/genetics , Mutation/genetics , Oxidoreductases Acting on CH-CH Group Donors , Oxidoreductases/genetics , Smith-Lemli-Opitz Syndrome/enzymology , Smith-Lemli-Opitz Syndrome/genetics , Europe/epidemiology , Europe/ethnology , Genetic Carrier Screening , Genetic Testing , Humans , Smith-Lemli-Opitz Syndrome/ethnology
15.
Am J Med Genet ; 102(1): 18-20, 2001 Jul 22.
Article in English | MEDLINE | ID: mdl-11471166

ABSTRACT

Smith-Lemli-Opitz syndrome (OMIM 270400) (SLOS) is caused by inherited enzymatic deficiency of 3beta-hydroxysterol-Delta7-reductase (7-dehydrocholesterol-Delta7-reductase, DHCR7). SLOS is diagnosed clinically by the demonstration of elevated levels of 7-dehydrocholesterol (7DHC) in body fluids or tissues. SLOS is associated with mental retardation of variable degree and severe behavior abnormalities. The physical abnormalities range from minor facial anomalies to lethal malformations of the central nervous system, heart, kidneys, and other organs. The exact incidence of SLOS is not known. Although there exist estimates of the incidence of SLOS ranging from 1 in 20,000 to 1 in 60,000, no prospective studies of the incidence of SLOS, based on the clinical data and biochemical diagnosis of SLOS, have been performed. Five unrelated cases of SLOS were diagnosed in Ontario during a 12-month period. The diagnoses were made based on the demonstration of elevated 7DHC in plasma or amniotic fluid. The birth rate for Ontario for that period was 132,000 births. The incidence of SLOS in Ontario was at least 1 in 26,500 pregnancies in 1999-2000. Given that 86% of the population of Ontario is of European origin, the incidence of SLOS in the Ontario population of European origin was at least 1 in 22,700. As infants with mild forms of SLOS born during this period may remain undiagnosed, these numbers likely are underestimates. This observation has implications for prenatal and newborn screening for this potentially treatable inherited disorder.


Subject(s)
Oxidoreductases Acting on CH-CH Group Donors , Oxidoreductases/genetics , Smith-Lemli-Opitz Syndrome/genetics , Dehydrocholesterols/blood , Humans , Incidence , Mutation , Ontario/epidemiology , Oxidoreductases/deficiency , Severity of Illness Index , Smith-Lemli-Opitz Syndrome/enzymology , Smith-Lemli-Opitz Syndrome/epidemiology
17.
Hum Mol Genet ; 10(6): 555-64, 2001 Mar 15.
Article in English | MEDLINE | ID: mdl-11230174

ABSTRACT

The RSH/Smith--Lemli--Opitz syndrome (RSH/SLOS) is a human autosomal recessive syndrome characterized by multiple malformations, a distinct behavioral phenotype with autistic features and mental retardation. RSH/SLOS is due to an inborn error of cholesterol biosynthesis caused by mutation of the 3 beta-hydroxysterol Delta(7)-reductase gene. To further our understanding of the developmental and neurological processes that underlie the pathophysiology of this disorder, we have developed a mouse model of RSH/SLOS by disruption of the 3 beta-hydroxysterol Delta(7)-reductase gene. Here we provide the biochemical, phenotypic and neurophysiological characterization of this genetic mouse model. As in human patients, the RSH/SLOS mouse has a marked reduction of serum and tissue cholesterol levels and a marked increase of serum and tissue 7-dehydrocholesterol levels. Phenotypic similarities between this mouse model and the human syndrome include intra-uterine growth retardation, variable craniofacial anomalies including cleft palate, poor feeding with an uncoordinated suck, hypotonia and decreased movement. Neurophysiological studies showed that although the response of frontal cortex neurons to the neurotransmitter gamma-amino-n-butyric acid was normal, the response of these same neurons to glutamate was significantly impaired. This finding provides insight into potential mechanisms underlying the neurological dysfunction seen in this human mental retardation syndrome and suggests that this mouse model will allow the testing of potential therapeutic interventions.


Subject(s)
Oxidoreductases Acting on CH-CH Group Donors , Oxidoreductases/genetics , Smith-Lemli-Opitz Syndrome/genetics , Animals , Disease Models, Animal , Mice , Mice, Inbred C57BL , Oxidoreductases/deficiency , Oxidoreductases/metabolism , Phenotype , Smith-Lemli-Opitz Syndrome/metabolism , Smith-Lemli-Opitz Syndrome/pathology
18.
Am J Med Genet ; 98(2): 191-200, 2001 Jan 15.
Article in English | MEDLINE | ID: mdl-11223857

ABSTRACT

The behavior phenotype of Smith-Lemli-Opitz syndrome (SLOS) was studied by assessing behavior, social, and communication abilities, sensory hyperreactivity, and the deficits associated with autistic disorder. Fifty-six SLOS subjects, age 0.3 to 32.3 years, were evaluated by multiple age-dependent questionnaires and telephone interviews. Of the 56 subjects, 50 (89%) had a history of repeated self-injury: 30 (54%) bit themselves; 27 (48%) head-banged; and 30 (54%) threw themselves backward in a highly characteristic upper body movement ("opisthokinesis"). Forty-seven of these subjects were also evaluated by direct observation and by direct interview of the parent or caregiver. Of 11 subjects 10 years or older, three (27%) had a stereotypic stretching motion of the upper body accompanied by hand flicking. Additional measures showed sensory hyperreactivity, temperament dysregulation, sleep disturbance, and social and communication deficits. Nine of 17 subjects (53%) met the diagnostic criteria for autistic disorder by the Autism Diagnostic Interview-Revised (ADI-R) algorithm questions [Lord et al., 1993, 1994]. Thus, SLOS is a metabolic disorder that can be associated with autism and other behavioral characteristics that define a distinctive and diagnostically important behavioral disorder.


Subject(s)
Behavior , Smith-Lemli-Opitz Syndrome/psychology , Adolescent , Adult , Algorithms , Autistic Disorder/complications , Autistic Disorder/diagnosis , Child , Child, Preschool , Cholesterol/therapeutic use , Female , Humans , Infant , Interpersonal Relations , Interview, Psychological , Male , Motor Activity , Phenotype , Sensation Disorders/complications , Smith-Lemli-Opitz Syndrome/genetics , Smith-Lemli-Opitz Syndrome/physiopathology
19.
Mech Dev ; 100(2): 165-75, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11165475

ABSTRACT

We are interested in the early mechanisms that initiate regional patterning in the dorsal telencephalon, which gives rise to cerebral cortex. Members of the LIM-homeodomain (LIM-HD) family of transcription factors are implicated in patterning and cell fate specification in several systems including the mammalian forebrain. Mice in which Lhx2 is disrupted were reported to have reduced telencephalic development, and the hippocampal primordium appeared to be missing, by morphological observation. We hypothesized that this may be due to a defect in the cortical hem, a Wnt- and Bmp-rich putative signaling center in the medial telencephalon, a source of regulatory signals for hippocampal development. We asked if the expression of any known hem-specific signaling molecule is deficient in Lhx2-/- mice. Our results reveal, unexpectedly, that at embryonic day (E)12.5, what appears to be some spared 'lateral' cortex is instead an expanded cortical hem. Normally restricted to the extreme medial edge of the telencephalon, the hem covers almost the entire dorsal telencephalon in the Lhx2-/- mice. This indicates a role for Lhx2 in the regulation of the extent of the cortical hem. In spite of an expanded, mislocated hem in the Lhx2-/- telencephalon, a potential source of ectopic dorsalizing cues, no hippocampal differentiation is detected in tissue adjacent to the mutant hem, nor does the overall dorsoventral patterning appear perturbed. We propose that Lhx2 is involved at a crucial early step in patterning the telencephalon, where the neuroepithelium is first divided into presumptive cortical tissue, and the cortical hem. The defect in the Lhx2-/- telencephalon appears to be at this step.


Subject(s)
Cerebral Cortex/embryology , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Homeodomain Proteins/physiology , Telencephalon/metabolism , Transcription Factors/metabolism , Transcription Factors/physiology , Zebrafish Proteins , Animals , Cell Lineage , Choroid Plexus/metabolism , Genotype , Hippocampus/embryology , Hippocampus/metabolism , Homeodomain Proteins/genetics , Homozygote , Immunohistochemistry , LIM-Homeodomain Proteins , Mice , Multigene Family , Protein Structure, Tertiary , Proto-Oncogene Proteins/biosynthesis , Signal Transduction , Time Factors , Transcription Factors/genetics , Wnt Proteins
20.
Mol Genet Metab ; 71(1-2): 163-74, 2000.
Article in English | MEDLINE | ID: mdl-11001807

ABSTRACT

The RSH/Smith-Lemli-Opitz syndrome (RSH/SLOS) is an autosomal recessive multiple congenital anomaly/mental retardation syndrome caused by an inborn error of cholesterol biosynthesis. The RSH/SLOS phenotypic spectrum is broad; however, typical features include microcephaly, ptosis, a small upturned nose, micrognathia, postaxial polydactaly, second and third toe syndactaly, genital anomalies, growth failure, and mental retardation. RSH/SLOS is due to a deficiency of the 3beta-hydroxysterol Delta(7)-reductase, which catalyzes the reduction of 7-dehydrocholesterol (7-DHC) to cholesterol. This inborn error of cholesterol biosynthesis results in elevated serum and tissue 7-DHC levels. The 3beta-hydroxysterol Delta(7)-reductase gene (DHCR7) maps to chromosome 11q12-13, and to date 66 different mutations of this gene have been identified in RSH/SLOS patients. Identification of the biochemical basis of RSH/SLOS has led to development of therapeutic regimens based on dietary cholesterol supplementation and has increased our understanding of the role cholesterol plays during embryonic development.


Subject(s)
Cholesterol/biosynthesis , Oxidoreductases Acting on CH-CH Group Donors , Smith-Lemli-Opitz Syndrome/metabolism , Child , Child, Preschool , Female , Holoprosencephaly/genetics , Holoprosencephaly/metabolism , Humans , Infant , Male , Molecular Biology , Mutation , Oxidoreductases/deficiency , Oxidoreductases/genetics , Phenotype , Smith-Lemli-Opitz Syndrome/genetics , Smith-Lemli-Opitz Syndrome/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...