Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Alzheimers Dis ; 97(4): 1793-1806, 2024.
Article in English | MEDLINE | ID: mdl-38306050

ABSTRACT

Background: Some epidemiologic studies associate traumatic brain injury (TBI) with Alzheimer's disease (AD). Objective: To test whether a TBI-induced acceleration of age-related mitochondrial change could potentially mediate the reported TBI-AD association. Methods: We administered unilateral controlled cortical impact (CCI) or sham injuries to 5-month-old C57BL/6J and tau transgenic rTg4510 mice. In the non-transgenics, we assessed behavior (1-5 days, 1 month, and 15 months), lesion size (1 and 15 months), respiratory chain enzymes (1 and 15 months), and mitochondrial DNA copy number (mtDNAcn) (1 and 15 months) after CCI/sham. In the transgenics we quantified post-injury mtDNAcn and tangle burden. Results: In the non-transgenics CCI caused acute behavioral deficits that improved or resolved by 1-month post-injury. Protein-normalized complex I and cytochrome oxidase activities were not significantly altered at 1 or 15 months, although complex I activity in the CCI ipsilesional cortex declined during that period. Hippocampal mtDNAcn was not altered by injury at 1 month, increased with age, and rose to the greatest extent in the CCI contralesional hippocampus. In the injured then aged transgenics, the ipsilesional hippocampus contained less mtDNA and fewer tangles than the contralesional hippocampus; mtDNAcn and tangle counts did not correlate. Conclusions: As mice age their brains increase mtDNAcn as part of a compensatory response that preserves mitochondrial function, and TBI enhances this response. TBI may, therefore, increase the amount of compensation required to preserve late-life mitochondrial function. If TBI does modify AD risk, altering the trajectory or biology of aging-related mitochondrial changes could mediate the effect.


Subject(s)
Alzheimer Disease , Brain Injuries, Traumatic , Mice , Animals , Mice, Inbred C57BL , Brain Injuries, Traumatic/pathology , Brain/pathology , Mitochondria/pathology , DNA, Mitochondrial/genetics , Mice, Transgenic , Disease Models, Animal
2.
Sci Transl Med ; 15(688): eabf4077, 2023 03 22.
Article in English | MEDLINE | ID: mdl-36947593

ABSTRACT

Angelman syndrome is a devastating neurogenetic disorder for which there is currently no effective treatment. It is caused by mutations or epimutations affecting the expression or function of the maternally inherited allele of the ubiquitin-protein ligase E3A (UBE3A) gene. The paternal UBE3A allele is imprinted in neurons of the central nervous system (CNS) by the UBE3A antisense (UBE3A-AS) transcript, which represents the distal end of the small nucleolar host gene 14 (SNHG14) transcription unit. Reactivating the expression of the paternal UBE3A allele in the CNS has long been pursued as a therapeutic option for Angelman syndrome. Here, we described the development of an antisense oligonucleotide (ASO) therapy for Angelman syndrome that targets an evolutionarily conserved region demarcating the start of the UBE3A-AS transcript. We designed and chemically optimized gapmer ASOs targeting specific sequences at the start of the human UBE3A-AS transcript. We showed that ASOs targeting this region precisely and efficiently repress the transcription of UBE3A-AS, reactivating the expression of the paternal UBE3A allele in neurotypical and Angelman syndrome induced pluripotent stem cell-derived neurons. We further showed that human-targeted ASOs administered to the CNS of cynomolgus macaques by lumbar intrathecal injection repress UBE3A-AS and reactivate the expression of the paternal UBE3A allele throughout the CNS. These findings support the advancement of this investigational molecular therapy for Angelman syndrome into clinical development (ClinicalTrials.gov, NCT04259281).


Subject(s)
Angelman Syndrome , Humans , Angelman Syndrome/therapy , Angelman Syndrome/drug therapy , Alleles , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
3.
mSystems ; 8(1): e0060822, 2023 02 23.
Article in English | MEDLINE | ID: mdl-36598241

ABSTRACT

A large subset of patients with Angelman syndrome (AS) suffer from concurrent gastrointestinal (GI) issues, including constipation, poor feeding, and reflux. AS is caused by the loss of ubiquitin ligase E3A (UBE3A) gene expression in the brain. Clinical features of AS, which include developmental delays, intellectual disability, microcephaly, and seizures, are primarily due to the deficient expression or function of the maternally inherited UBE3A allele. The association between neurodevelopmental delay and GI disorders is part of the increasing evidence suggesting a link between the brain and the gut microbiome via the microbiota-gut-brain axis. To investigate the associations between colonization of the gut microbiota in AS, we characterized the fecal microbiome in three animal models of AS involving maternal deletions of Ube3A, including mouse, rat, and pig, using 16S rRNA amplicon sequencing. Overall, we identified changes in bacterial abundance across all three animal models of AS. Specific bacterial groups were significantly increased across all animal models, including Lachnospiraceae Incertae sedis, Desulfovibrios sp., and Odoribacter, which have been correlated with neuropsychiatric disorders. Taken together, these findings suggest that specific changes to the local environment in the gut are driven by a Ube3a maternal deletion, unaffected by varying housing conditions, and are prominent and detectable across multiple small and large animal model species. These findings begin to uncover the underlying mechanistic causes of GI disorders in AS patients and provide future therapeutic options for AS patients. IMPORTANCE Angelman syndrome (AS)-associated gastrointestinal (GI) symptoms significantly impact quality of life in patients. In AS models in mouse, rat, and pig, AS animals showed impaired colonization of the gut microbiota compared to wild-type (healthy) control animals. Common changes in AS microbiomes across all three animal models may play a causal effect for GI symptoms and may help to identify ways to treat these comorbidities in patients in the future.


Subject(s)
Angelman Syndrome , Gastrointestinal Diseases , Gastrointestinal Microbiome , Mice , Rats , Animals , Swine , Angelman Syndrome/genetics , Gastrointestinal Microbiome/genetics , RNA, Ribosomal, 16S/genetics , Quality of Life , Disease Models, Animal , Ubiquitin-Protein Ligases/genetics
4.
Brain Res ; 1789: 147945, 2022 08 15.
Article in English | MEDLINE | ID: mdl-35595066

ABSTRACT

Traumatic brain injury (TBI) is one of the leading causes of death and disability worldwide. Cerebral edema following TBI is known to play a critical role in injury severity and prognosis. In the current study we used multimodal magnetic resonance imaging (MRI) to assess cerebral edema 24 h after unilateral contusive TBI in male and female rats. We then directly quantified brain water content in the same subjectsex vivo.We found that both males and females had similarly elevated T2 values after TBI compared with sham controls. Apparent diffusion coefficient (ADC) was more variable than T2 and did not show significant injury effects in males or females. Brain water was elevated in male TBI rats compared with sham controls, but there was no difference between female TBI and sham groups. Notably, MRI biomarkers of edema were more closely correlated with brain water in male rats; female rats did not show any relationship between brain water and T2 or ADC. These observations raise questions about the interpretation of radiological findings traditionally interpreted as edema in female TBI patients. A better understanding of sex differences and similarities in the pathophysiology of post-traumatic edema is needed to help improve patient management and the development of effective treatment strategies for men and women.


Subject(s)
Brain Edema , Brain Injuries, Traumatic , Brain Injuries , Animals , Biomarkers , Brain Edema/diagnostic imaging , Brain Edema/etiology , Brain Edema/pathology , Brain Injuries/pathology , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/diagnostic imaging , Edema/complications , Female , Humans , Male , Rats , Rats, Sprague-Dawley , Water
5.
PeerJ ; 8: e8956, 2020.
Article in English | MEDLINE | ID: mdl-32341898

ABSTRACT

Synthetic molecules that mimic the function of natural enzymes or molecules have untapped potential for use in the next generation of drugs. Cyclic compounds that contain aromatic rings are macrocyclic cyclophanes, and when they coordinate iron ions are of particular interest due to their antioxidant and biomimetic properties. However, little is known about the molecular responses at the cellular level. This study aims to evaluate the changes in immune gene expression in human cells exposed to the cyclophanes Fe2PO and Fe2PC. Confluent human embryonic kidney cells were exposed to either the cyclophane Fe2PO or Fe2PC before extraction of RNA. The expression of a panel of innate and adaptive immune genes was analyzed by quantitative real-time PCR. Evidence was found for an inflammatory response elicited by the cyclophane exposures. After 8 h of exposure, the cells increased the relative expression of inflammatory mediators such as interleukin 1; IRAK, which transduces signals between interleukin 1 receptors and the NFκB pathway; and the LPS pattern recognition receptor CD14. After 24 h of exposure, regulatory genes begin to counter the inflammation, as some genes involved in oxidative stress, apoptosis and non-inflammatory immune responses come into play. Both Fe2PO and Fe2PC induced similar immunogenetic changes in transcription profiles, but equal molar doses of Fe2PC resulted in more robust responses. These data suggest that further work in whole animal models may provide more insights into the extent of systemic physiological changes induced by these cyclophanes.

6.
Brain Imaging Behav ; 13(2): 461-471, 2019 Apr.
Article in English | MEDLINE | ID: mdl-29656312

ABSTRACT

Despite higher rates of hospitalization and mortality following traumatic brain injury (TBI) in patients over 65 years old, older patients remain underrepresented in drug development studies. Worse outcomes in older individuals compared to younger adults could be attributed to exacerbated injury mechanisms including oxidative stress, inflammation, blood-brain barrier disruption, and bioenergetic dysfunction. Accordingly, pleiotropic treatments are attractive candidates for neuroprotection. Taurine, an endogenous amino acid with antioxidant, anti-inflammatory, anti-apoptotic, osmolytic, and neuromodulator effects, is neuroprotective in adult rats with TBI. However, its effects in the aged brain have not been evaluated. We subjected aged male rats to a unilateral controlled cortical impact injury to the sensorimotor cortex, and randomized them into four treatment groups: saline or 25 mg/kg, 50 mg/kg, or 200 mg/kg i.p. taurine. Treatments were administered 20 min post-injury and daily for 7 days. We assessed sensorimotor function on post-TBI days 1-14 and tissue loss on day 14 using T2-weighted magnetic resonance imaging. Experimenters were blinded to the treatment group for the duration of the study. We did not observe neuroprotective effects of taurine on functional impairment or tissue loss in aged rats after TBI. These findings in aged rats are in contrast to previous reports of taurine neuroprotection in younger animals. Advanced age is an important variable for drug development studies in TBI, and further research is required to better understand how aging may influence mechanisms of taurine neuroprotection.


Subject(s)
Brain Injuries, Traumatic/pathology , Disease Models, Animal , Neuroprotection/drug effects , Taurine/administration & dosage , Animals , Humans , Magnetic Resonance Imaging , Male , Neuroprotective Agents/pharmacology , Rats , Recovery of Function/drug effects
7.
Hum Mol Genet ; 26(20): 3995-4010, 2017 10 15.
Article in English | MEDLINE | ID: mdl-29016856

ABSTRACT

Maternally derived copy number gains of human chromosome 15q11.2-q13.3 (Dup15q syndrome or Dup15q) cause intellectual disability, epilepsy, developmental delay, hypotonia, speech impairments, and minor dysmorphic features. Dup15q syndrome is one of the most common and penetrant chromosomal abnormalities observed in individuals with autism spectrum disorder (ASD). Although ∼40 genes are located in the 15q11.2-q13.3 region, overexpression of the ubiquitin-protein E3A ligase (UBE3A) gene is thought to be the predominant molecular cause of the phenotypes observed in Dup15q syndrome. The UBE3A gene demonstrates maternal-specific expression in neurons and loss of maternal UBE3A causes Angelman syndrome, a neurodevelopmental disorder with some overlapping neurological features to Dup15q. To directly test the hypothesis that overexpression of UBE3A is an important underlying molecular cause of neurodevelopmental dysfunction, we developed and characterized a mouse overexpressing Ube3a isoform 2 in excitatory neurons. Ube3a isoform 2 is conserved between mouse and human and known to play key roles in neuronal function. Transgenic mice overexpressing Ube3a isoform 2 in excitatory forebrain neurons exhibited increased anxiety-like behaviors, learning impairments, and reduced seizure thresholds. However, these transgenic mice displayed normal social approach, social interactions, and repetitive motor stereotypies that are relevant to ASD. Reduced forebrain, hippocampus, striatum, amygdala, and cortical volume were also observed. Altogether, these findings show neuronal overexpression of Ube3a isoform 2 causes phenotypes translatable to neurodevelopmental disorders.


Subject(s)
Intellectual Disability/enzymology , Neurons/enzymology , Ubiquitin-Protein Ligases/biosynthesis , Animals , Antisocial Personality Disorder/genetics , Antisocial Personality Disorder/metabolism , Anxiety/genetics , Anxiety/metabolism , Chromosome Aberrations , Chromosomes, Human, Pair 15/enzymology , Chromosomes, Human, Pair 15/genetics , Chromosomes, Human, Pair 15/metabolism , Disease Models, Animal , Female , Gene Expression , Humans , Intellectual Disability/genetics , Intellectual Disability/metabolism , Intellectual Disability/pathology , Male , Mice , Mice, Transgenic , Neurons/metabolism , Neurons/pathology , Phenotype , Seizures/genetics , Seizures/metabolism , Ubiquitin-Protein Ligases/genetics
8.
Epigenetics Chromatin ; 10: 27, 2017.
Article in English | MEDLINE | ID: mdl-28515788

ABSTRACT

BACKGROUND: The ubiquitin protein E3A ligase gene (UBE3A) gene is imprinted with maternal-specific expression in neurons and biallelically expressed in all other cell types. Both loss-of-function and gain-of-function mutations affecting the dosage of UBE3A are associated with several neurodevelopmental syndromes and psychological conditions, suggesting that UBE3A is dosage-sensitive in the brain. The observation that loss of imprinting increases the dosage of UBE3A in brain further suggests that inactivation of the paternal UBE3A allele evolved as a dosage-regulating mechanism. To test this hypothesis, we examined UBE3A transcript and protein levels among cells, tissues, and species with different imprinting states of UBE3A. RESULTS: Overall, we found no correlation between the imprinting status and dosage of UBE3A. Importantly, we found that maternal Ube3a protein levels increase in step with decreasing paternal Ube3a protein levels during neurogenesis in mouse, fully compensating for loss of expression of the paternal Ube3a allele in neurons. CONCLUSIONS: Based on our findings, we propose that imprinting of UBE3A does not function to reduce the dosage of UBE3A in neurons but rather to regulate some other, as yet unknown, aspect of gene expression or protein function.


Subject(s)
Gene Dosage/genetics , Genomic Imprinting , Maternal Inheritance/genetics , Ubiquitin-Protein Ligases/genetics , Alleles , Animals , Female , Gene Expression Regulation , Mice , Neurogenesis/genetics , Neurons/metabolism , RNA, Antisense/genetics , Ubiquitin-Protein Ligases/biosynthesis
9.
J Med Chem ; 57(16): 6965-72, 2014 Aug 28.
Article in English | MEDLINE | ID: mdl-25119815

ABSTRACT

The cytochrome P450 eicosanoid 14,15-epoxyeicosa-5,8,11-trienoic acid (14,15-EET) is a powerful endogenous autacoid that has been ascribed an impressive array of physiologic functions including regulation of blood pressure. Because 14,15-EET is chemically and metabolically labile, structurally related surrogates containing epoxide bioisosteres were introduced and have become useful in vitro pharmacologic tools but are not suitable for in vivo applications. A new generation of EET mimics incorporating modifications to the carboxylate were prepared and evaluated for vasorelaxation and inhibition of soluble epoxide hydrolase (sEH). Tetrazole 19 (ED50 0.18 µM) and oxadiazole-5-thione 25 (ED50 0.36 µM) were 12- and 6-fold more potent, respectively, than 14,15-EET as vasorelaxants; on the other hand, their ability to block sEH differed substantially, i.e., 11 vs >500 nM. These data will expedite the development of potent and specific in vivo drug candidates.


Subject(s)
8,11,14-Eicosatrienoic Acid/analogs & derivatives , Vasodilator Agents/chemistry , Vasodilator Agents/pharmacology , 8,11,14-Eicosatrienoic Acid/chemistry , Animals , Cattle , Chemistry Techniques, Synthetic , Coronary Vessels/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Epoxide Hydrolases/antagonists & inhibitors , Humans , In Vitro Techniques , Molecular Mimicry , Oxadiazoles/chemistry , Oxadiazoles/pharmacology , Oxamic Acid/analogs & derivatives , Oxamic Acid/chemistry , Oxamic Acid/pharmacology , Tetrazoles/chemistry , Tetrazoles/pharmacology , Vasodilation/drug effects , Vasodilator Agents/chemical synthesis
10.
J Marital Fam Ther ; 38(1): 281-304, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22283391

ABSTRACT

This meta-analysis summarizes results from k = 24 studies comparing either Brief Strategic Family Therapy, Functional Family Therapy, Multidimensional Family Therapy, or Multisystemic Therapy to either treatment-as-usual, an alternative therapy, or a control group in the treatment of adolescent substance abuse and delinquency. Additionally, the authors reviewed and applied three advanced meta-analysis methods including influence analysis, multivariate meta-analysis, and publication bias analyses. The results suggested that as a group the four family therapies had statistically significant, but modest effects as compared to treatment-as-usual (d = 0.21; k = 11) and as compared to alternative therapies (d = 0.26; k = 11). The effect of family therapy compared to control was larger (d = 0.70; k = 4) but was not statistically significant probably because of low power. There was insufficient evidence to determine whether the various models differed in their effectiveness relative to each other. Influence analyses suggested that three studies had a large effect on aggregate effect sizes and heterogeneity statistics. Moderator and multivariate analyses were largely underpowered but will be useful as this literature grows.


Subject(s)
Adolescent Behavior , Family Therapy/methods , Juvenile Delinquency/rehabilitation , Substance-Related Disorders/therapy , Adolescent , Adolescent Health Services/organization & administration , Cognitive Behavioral Therapy/methods , Combined Modality Therapy/methods , Comorbidity , Evidence-Based Medicine , Humans , Juvenile Delinquency/statistics & numerical data , Psychotherapy, Group/methods , Randomized Controlled Trials as Topic , Substance-Related Disorders/epidemiology
SELECTION OF CITATIONS
SEARCH DETAIL