Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Ageing Res Rev ; 96: 102281, 2024 04.
Article in English | MEDLINE | ID: mdl-38513771

ABSTRACT

Accumulation of the amyloid ß (Aß) peptide, derived from Aß precursor protein (APP), is a trait of Down syndrome (DS), as is early development of dementia that resembles Alzheimer's disease (AD). Treatments for this AD in DS simply do not. New drug therapies for AD, e.g., Lecanemab, are monoclonal antibodies designed to clear amyloid plaques composed of Aß. The increasingly real ability to target and dispose of Aß favors the use of these drugs in individuals with AD in DS, and, perhaps as earlier intervention for cognitive impairment. We present pertinent similarities between DS and AD in adult DS subjects, discuss challenges to target APP metabolites, and suggest that recently developed antibody treatments against Aß may be worth investigating to treat AD in DS.


Subject(s)
Alzheimer Disease , Down Syndrome , Humans , Down Syndrome/drug therapy , Down Syndrome/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Antibodies, Monoclonal , Phenotype
2.
Front Mol Neurosci ; 16: 1201744, 2023.
Article in English | MEDLINE | ID: mdl-37799731

ABSTRACT

Metabolites of the Amyloid-ß precursor protein (APP) proteolysis may underlie brain overgrowth in Autism Spectrum Disorder (ASD). We have found elevated APP metabolites (total APP, secreted (s) APPα, and α-secretase adamalysins in the plasma and brain tissue of children with ASD). In this review, we highlight several lines of evidence supporting APP metabolites' potential contribution to macrocephaly in ASD. First, APP appears early in corticogenesis, placing APP in a prime position to accelerate growth in neurons and glia. APP metabolites are upregulated in neuroinflammation, another potential contributor to excessive brain growth in ASD. APP metabolites appear to directly affect translational signaling pathways, which have been linked to single gene forms of syndromic ASD (Fragile X Syndrome, PTEN, Tuberous Sclerosis Complex). Finally, APP metabolites, and microRNA, which regulates APP expression, may contribute to ASD brain overgrowth, particularly increased white matter, through ERK receptor activation on the PI3K/Akt/mTOR/Rho GTPase pathway, favoring myelination.

3.
Front Mol Neurosci ; 16: 1201723, 2023.
Article in English | MEDLINE | ID: mdl-37808474

ABSTRACT

Recent studies promote new interest in the intersectionality between autism spectrum disorder (ASD) and Alzheimer's Disease. We have reported high levels of Amyloid-ß Precursor Protein (APP) and secreted APP-alpha (sAPPa) and low levels of amyloid-beta (Aß) peptides 1-40 and 1-42 (Aß40, Aß42) in plasma and brain tissue from children with ASD. A higher incidence of microcephaly (head circumference less than the 3rd percentile) associates with ASD compared to head size in individuals with typical development. The role of Aß peptides as contributors to acquired microcephaly in ASD is proposed. Aß may lead to microcephaly via disruption of neurogenesis, elongation of the G1/S cell cycle, and arrested cell cycle promoting apoptosis. As the APP gene exists on Chromosome 21, excess Aß peptides occur in Trisomy 21-T21 (Down's Syndrome). Microcephaly and some forms of ASD associate with T21, and therefore potential mechanisms underlying these associations will be examined in this review. Aß peptides' role in other neurodevelopmental disorders that feature ASD and acquired microcephaly are reviewed, including dup 15q11.2-q13, Angelman and Rett syndrome.

4.
5.
Radiol Case Rep ; 16(6): 1276-1279, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33854663

ABSTRACT

Pathogenic CACNA1A gene variants are associated with a spectrum of disorders including migraine with or without hemiplegia, ataxia, epilepsy, and developmental disability. We present a case of a pathogenic variant (c.4046G>A, p.R1349Q) in the CACNA1A gene associated with a clinical phenotype of global developmental delay, left hemiparesis, epilepsy, and stroke-like episodes. Longitudinal neuroimaging demonstrates hemispheric encephalomalacia with mismatched perfusion and angiographic imaging, in addition to progressive cerebellar atrophy.

7.
Sci Rep ; 10(1): 10781, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32612155

ABSTRACT

Fragile X mental retardation protein (FMRP) binds to and regulates the translation of amyloid-ß protein precursor (App) mRNA, but the detailed mechanism remains to be determined. Differential methylation of App mRNA could underlie FMRP binding, message localization and translation efficiency. We sought to determine the role of FMRP and N6-methyladeonsine (m6A) on nuclear export of App mRNA. We utilized the m6A dataset by Hsu and colleagues to identify m6A sites in App mRNA and to determine if the abundance of message in the cytoplasm relative to the nucleus is altered in Fmr1 knockout mouse brain cortex. Given that processing of APP to Aß and soluble APP alpha (sAPPα) contributes to disease phenotypes, we also investigated whether Fmr1KO associates with nuclear export of the mRNAs for APP protein processing enzymes, including ß-site amyloid cleaving enzyme (Bace1), A disintegrin and metalloproteinases (Adams), and presenilins (Psen). Fmr1KO did not alter the nuclear/cytoplasmic abundance of App mRNA. Of 36 validated FMRP targets, 35 messages contained m6A peaks but only Agap2 mRNA was selectively enriched in Fmr1KO nucleus. The abundance of the APP processing enzymes Adam9 and Psen1 mRNA, which code for a minor alpha-secretase and gamma-secretase, respectively, were selectively enriched in wild type cytoplasm.


Subject(s)
ADAM Proteins/metabolism , Adenosine/analogs & derivatives , Cell Nucleus/metabolism , Databases, Nucleic Acid , Fragile X Mental Retardation Protein/metabolism , Membrane Proteins/metabolism , Presenilin-1/metabolism , RNA, Messenger/metabolism , ADAM Proteins/genetics , Active Transport, Cell Nucleus/genetics , Adenosine/genetics , Adenosine/metabolism , Animals , Cell Nucleus/genetics , Fragile X Mental Retardation Protein/genetics , Membrane Proteins/genetics , Mice , Presenilin-1/genetics , RNA, Messenger/genetics
8.
Front Psychiatry ; 10: 165, 2019.
Article in English | MEDLINE | ID: mdl-31024350

ABSTRACT

The most replicated neuroanatomical finding in autism is the tendency toward brain overgrowth, especially in younger children. Research shows that both gray and white matter are enlarged. Proposed mechanisms underlying brain enlargement include abnormal inflammatory and neurotrophic signals that lead to excessive, aberrant dendritic connectivity via disrupted pruning and cell adhesion, and enlargement of white matter due to excessive gliogenesis and increased myelination. Amyloid-ß protein precursor (ßAPP) and its metabolites, more commonly associated with Alzheimer's disease (AD), are also dysregulated in autism plasma and brain tissue samples. This review highlights findings that demonstrate how one ßAPP metabolite, secreted APPα, and the ADAM family α-secretases, may lead to increased brain matter, with emphasis on increased white matter as seen in autism. sAPPα and the ADAM family α-secretases contribute to the anabolic, non-amyloidogenic pathway, which is in contrast to the amyloid (catabolic) pathway known to contribute to Alzheimer disease. The non-amyloidogenic pathway could produce brain enlargement via genetic mechanisms affecting mRNA translation and polygenic factors that converge on molecular pathways (mitogen-activated protein kinase/MAPK and mechanistic target of rapamycin/mTOR), promoting neuroinflammation. A novel mechanism linking the non-amyloidogenic pathway to white matter enlargement is proposed: α-secretase and/or sAPPα, activated by ERK receptor signaling activates P13K/AKt/mTOR and then Rho GTPases favoring myelination via oligodendrocyte progenitor cell (OPC) activation of cofilin. Applying known pathways in AD to autism should allow further understanding and provide options for new drug targets.

10.
Sci Rep ; 6: 26052, 2016 05 23.
Article in English | MEDLINE | ID: mdl-27212113

ABSTRACT

UNLABELLED: Autism spectrum disorder (ASD) and Fragile X syndrome (FXS) are developmental disorders. No validated blood-based biomarkers exist for either, which impedes bench-to-bedside approaches. Amyloid-ß (Aß) precursor protein (APP) and metabolites are usually associated with Alzheimer's disease (AD). APP cleavage by α-secretase produces potentially neurotrophic secreted APPα (sAPPα) and the P3 peptide fragment. ß-site APP cleaving enzyme (BACE1) cleavage produces secreted APPß (sAPPß) and intact Aß. Excess Aß is potentially neurotoxic and can lead to atrophy of brain regions such as amygdala in AD. By contrast, amygdala is enlarged in ASD but not FXS. We previously reported elevated levels of sAPPα in ASD and FXS vs. CONTROLS: We now report elevated plasma Aß and total APP levels in FXS compared to both ASD and typically developing controls, and elevated levels of sAPPα in ASD and FXS vs. CONTROLS: By contrast, plasma and brain sAPPß and Aß were lower in ASD vs. controls but elevated in FXS plasma vs. CONTROLS: We also detected age-dependent increase in an α-secretase in ASD brains. We report a novel mechanistic difference in APP pathways between ASD (processing) and FXS (expression) leading to distinct APP metabolite profiles in these two disorders. These novel, distinctive biochemical differences between ASD and FXS pave the way for blood-based biomarkers for ASD and FXS.


Subject(s)
Amygdala/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Aspartic Acid Endopeptidases/metabolism , Autism Spectrum Disorder/metabolism , Fragile X Syndrome/metabolism , Plasma/metabolism , Adolescent , Age Factors , Amygdala/pathology , Biomarkers/metabolism , Child , Child, Preschool , Humans , Organ Specificity , Protein Processing, Post-Translational , Young Adult
11.
J Psychiatr Res ; 59: 220-8, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25300441

ABSTRACT

BACKGROUND: Understanding of the pathophysiology of autism spectrum disorder (ASD) remains limited. Brain overgrowth has been hypothesized to be associated with the development of ASD. A derivative of amyloid-ß precursor protein (APP), secreted APPα (sAPPα), has neuroproliferative effects and has been shown to be elevated in the plasma of persons with ASD compared to control subjects. Reduction in sAPPα holds promise as a novel molecular target of treatment in ASD. Research into the neurochemistry of ASD has repeatedly implicated excessive glutamatergic and deficient GABAergic neurotransmission in the disorder. With this in mind, acamprosate, a novel modulator of glutamate and GABA function, has been studied in ASD. No data is available on the impact of glutamate or GABA modulation on sAPPα function. METHODS: Plasma APP derivative levels pre- and post-treatment with acamprosate were determined in two pilot studies involving youth with idiopathic and fragile X syndrome (FXS)-associated ASD. We additionally compared baseline APP derivative levels between youth with FXS-associated or idiopathic ASD. RESULTS: Acamprosate use was associated with a significant reduction in plasma sAPP(total) and sAPPα levels but no change occurred in Aß40 or Aß42 levels in 15 youth with ASD (mean age: 11.1 years). Youth with FXS-associated ASD (n = 12) showed increased sAPPα processing compared to age-, gender- and IQ-match youth with idiopathic ASD (n = 11). CONCLUSIONS: Plasma APP derivative analysis holds promise as a potential biomarker for use in ASD targeted treatment. Reduction in sAPP (total) and sAPPα may be a novel pharmacodynamic property of acamprosate. Future study is required to address limitations of the current study to determine if baseline APP derivative analysis may predict subgroups of persons with idiopathic or FXS-associated ASD who may respond best to acamprosate or to potentially other modulators of glutamate and/or GABA neurotransmission.


Subject(s)
Amyloid beta-Protein Precursor/blood , Child Development Disorders, Pervasive/blood , Child Development Disorders, Pervasive/drug therapy , Fragile X Syndrome/blood , Fragile X Syndrome/drug therapy , Taurine/analogs & derivatives , Acamprosate , Adolescent , Case-Control Studies , Child , Child, Preschool , Female , Humans , Male , Peptide Fragments/blood , Pilot Projects , Taurine/pharmacology , Taurine/therapeutic use
13.
Front Cell Neurosci ; 7: 94, 2013.
Article in English | MEDLINE | ID: mdl-23801940

ABSTRACT

Autism is a neurodevelopmental disorder marked by social skills and communication deficits and interfering repetitive behavior. Intellectual disability often accompanies autism. In addition to behavioral deficits, autism is characterized by neuropathology and brain overgrowth. Increased intracranial volume often accompanies this brain growth. We have found that the Alzheimer's disease (AD) associated amyloid-ß precursor protein (APP), especially its neuroprotective processing product, secreted APP α, is elevated in persons with autism. This has led to the "anabolic hypothesis" of autism etiology, in which neuronal overgrowth in the brain results in interneuronal misconnections that may underlie multiple autism symptoms. We review the contribution of research in brain volume and of APP to the anabolic hypothesis, and relate APP to other proteins and pathways that have already been directly associated with autism, such as fragile X mental retardation protein, Ras small GTPase/extracellular signal-regulated kinase, and phosphoinositide 3 kinase/protein kinase B/mammalian target of rapamycin. We also present additional evidence of magnetic resonance imaging intracranial measurements in favor of the anabolic hypothesis. Finally, since it appears that APP's involvement in autism is part of a multi-partner network, we extend this concept into the inherently interactive realm of epigenetics. We speculate that the underlying molecular abnormalities that influence APP's contribution to autism are epigenetic markers overlaid onto potentially vulnerable gene sequences due to environmental influence.

14.
Epilepsy Behav ; 22(3): 523-6, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21908237

ABSTRACT

Adults with periventricular nodular heterotopia (PNH) have epilepsy and dyslexia, but most have normal intelligence. It is not known whether PNH-related reading difficulty can be detected earlier in childhood or whether associated behavioral problems are present. We studied 10 children with PNH, 3 of whom did not have seizures, and 10 matched controls with neuropsychological testing and parental rating instruments at two time points separated by about 1 year. Children with PNH performed significantly worse than controls on a task related to reading fluency. In addition, those with PNH showed significantly worse adaptive skills, and a measure of conduct problems significantly worsened over time. Mood and behavioral problems were reported more commonly, though not significantly so, in children with PNH. These findings demonstrate that reading dysfluency can be evident in children with nodular heterotopia, even in the absence of epilepsy, but also highlight difficulties with behavior in this population.


Subject(s)
Child Behavior Disorders/etiology , Cognition Disorders/etiology , Periventricular Nodular Heterotopia/complications , Adolescent , Child , Child Behavior Disorders/diagnosis , Cognition Disorders/diagnosis , Executive Function , Female , Humans , Magnetic Resonance Imaging , Male , Neuropsychological Tests , Parents/psychology , Psychiatric Status Rating Scales , Statistics, Nonparametric
15.
PLoS One ; 6(6): e20405, 2011.
Article in English | MEDLINE | ID: mdl-21731612

ABSTRACT

Autism is a neurodevelopmental disorder characterized by deficits in verbal communication, social interactions, and the presence of repetitive, stereotyped and compulsive behaviors. Excessive early brain growth is found commonly in some patients and may contribute to disease phenotype. Reports of increased levels of brain-derived neurotrophic factor (BDNF) and other neurotrophic-like factors in autistic neonates suggest that enhanced anabolic activity in CNS mediates this overgrowth effect. We have shown previously that in a subset of patients with severe autism and aggression, plasma levels of the secreted amyloid-ß (Aß) precursor protein-alpha form (sAPPα) were significantly elevated relative to controls and patients with mild-to-moderate autism. Here we further tested the hypothesis that levels of sAPPα and sAPPß (proteolytic cleavage products of APP by α- and ß-secretase, respectively) are deranged in autism and may contribute to an anabolic environment leading to brain overgrowth. We measured plasma levels of sAPPα, sAPPß, Aß peptides and BDNF by corresponding ELISA in a well characterized set of subjects. We included for analysis 18 control, 6 mild-to-moderate, and 15 severely autistic patient plasma samples. We have observed that sAPPα levels are increased and BDNF levels decreased in the plasma of patients with severe autism as compared to controls. Further, we show that Aß1-40, Aß1-42, and sAPPß levels are significantly decreased in the plasma of patients with severe autism. These findings do not extend to patients with mild-to-moderate autism, providing a biochemical correlate of phenotypic severity. Taken together, this study provides evidence that sAPPα levels are generally elevated in severe autism and suggests that these patients may have aberrant non-amyloidogenic processing of APP.


Subject(s)
Amyloid beta-Protein Precursor/blood , Autistic Disorder/blood , Metabolic Networks and Pathways , Peptide Fragments/blood , Amyloid beta-Peptides/blood , Biomarkers/blood , Brain-Derived Neurotrophic Factor/blood , Case-Control Studies , Child , Demography , Humans , Male , Severity of Illness Index
16.
Pediatr Neurol ; 42(3): 213-4, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20159432

ABSTRACT

Anti-N-methyl-D-aspartate receptor antibody has been associated with a severe stereotypic form of subacute encephalitis, often found in women with ovarian teratoma. Reported here is the diagnosis of anti-N-methyl-D-aspartate receptor encephalitis in a 5-year-old girl who presented with subacute encephalopathy and movement disorder without evidence of malignancy. Early diagnosis and treatment with immune globulin and steroids resulted in near-complete recovery.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antibodies, Anti-Idiotypic/immunology , Antibodies, Anti-Idiotypic/metabolism , Autoimmune Diseases of the Nervous System , Encephalitis , Methylprednisolone/therapeutic use , Receptors, N-Methyl-D-Aspartate/immunology , Receptors, N-Methyl-D-Aspartate/metabolism , Autoimmune Diseases of the Nervous System/drug therapy , Autoimmune Diseases of the Nervous System/immunology , Autoimmune Diseases of the Nervous System/metabolism , Child, Preschool , Encephalitis/drug therapy , Encephalitis/immunology , Encephalitis/metabolism , Female , Humans
17.
Pediatr Neurol ; 40(4): 310-3, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19302947

ABSTRACT

A genetic defect causing autism and epilepsy involving the contactin associated protein-like 2 gene (CNTNAP2) has been discovered in a selected cohort of Amish children. These children were found to have focal seizures and autistic regression. Surgical biopsy of the anterior temporal lobe of two such children revealed cortical dysplasia and a single nucleotide polymorphism mutation of this gene. The present case is that of a related but geographically distant proband with a similar phenotype but a single-base-pair deletion in the CNTNAP2 gene. This patient exhibited the additional features of periventricular leukomalacia and hepatomegaly.


Subject(s)
Autistic Disorder/genetics , Epilepsy, Complex Partial/genetics , Hepatomegaly/genetics , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Adolescent , Autistic Disorder/complications , Epilepsy, Complex Partial/complications , Female , Hepatomegaly/complications , Hepatomegaly/pathology , Humans , Mutation/physiology , Pedigree , Polymorphism, Single Nucleotide , Splenomegaly/genetics , Splenomegaly/pathology
18.
Pediatr Neurol ; 39(4): 293-4, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18805373

ABSTRACT

A 12-year-old African American boy with mental retardation and Asperger's disorder presented with acute psychosis. Antiphospholipid antibody testing with enzyme-linked immunosorbent assay showed increased levels of immunoglobulin G anticardiolipin antibodies in the cerebrospinal fluid, but not in the serum. Although antiphospholipid antibodies have been reported in the serum of patients with thrombotic and neurologic disorders, there are only a few reports of these antibodies in cerebrospinal fluid. This finding is consistent with a recent report of antiphospholipid antibodies found in the cerebrospinal fluid of adults with acute psychosis.


Subject(s)
Antibodies, Antiphospholipid/cerebrospinal fluid , Immunoglobulin G/cerebrospinal fluid , Psychotic Disorders/cerebrospinal fluid , Antibodies, Antiphospholipid/immunology , Asperger Syndrome/cerebrospinal fluid , Asperger Syndrome/immunology , Child , Enzyme-Linked Immunosorbent Assay , Humans , Immunoglobulin G/immunology , Intellectual Disability/cerebrospinal fluid , Intellectual Disability/immunology , Male , Psychotic Disorders/immunology
19.
Pediatr Neurol ; 39(2): 137-8, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18639762

ABSTRACT

Absolute pitch is the ability to identify the pitch of an isolated tone. We report on a 4-year-old boy with autism and absolute pitch, one of the youngest reported in the literature. Absolute pitch is thought to be attributable to a single gene, transmitted in an autosomal-dominant fashion. The association of absolute pitch with autism raises the speculation that this talent could be linked to a genetically distinct subset of children with autism. Further, the identification of absolute pitch in even young children with autism may lead to a lifelong skill.


Subject(s)
Autistic Disorder/physiopathology , Pitch Discrimination/physiology , Child, Preschool , Humans , Male
20.
J Child Neurol ; 23(8): 950-3, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18660478

ABSTRACT

Periventricular nodular heterotopia is a common malformation of cortical development in which the migration of developing neurons destined for the cerebral cortex is abbreviated. Bilateral periventricular nodular heterotopia is most commonly an X-linked disorder that involves mutations in the filamin A (FLNA) gene, but an autosomal recessive form and sporadic forms have been identified. To our knowledge, autosomal dominant transmission of isolated periventricular nodular heterotopia has not been reported. Periventricular nodular heterotopia has a heterogeneous phenotype, associated commonly with seizure disorder, and more recently with reading deficits and visual-spatial deficits in some patients. We present a father and son with bilateral periventricular nodular heterotopia and similar visual-spatial learning deficits, consistent with nonverbal learning disability.


Subject(s)
Chromosome Aberrations , Developmental Disabilities/genetics , Genes, Dominant/genetics , Learning Disabilities/genetics , Periventricular Nodular Heterotopia/genetics , Adult , Cerebral Ventricles/pathology , Child , Concept Formation , Developmental Disabilities/diagnosis , Dominance, Cerebral/physiology , Frontal Lobe/pathology , Humans , Intelligence/genetics , Learning Disabilities/diagnosis , Magnetic Resonance Imaging , Male , Mathematics , Memory Disorders/diagnosis , Memory Disorders/genetics , Neuropsychological Tests , Orientation , Pattern Recognition, Visual , Periventricular Nodular Heterotopia/diagnosis , Problem Solving , Wechsler Scales
SELECTION OF CITATIONS
SEARCH DETAIL
...