Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Cell Rep ; 42(7): 112706, 2023 07 25.
Article in English | MEDLINE | ID: mdl-37389991

ABSTRACT

The E3 ubiquitin ligase Ube3a is biallelically expressed in neural progenitors and glial cells, suggesting that UBE3A gain-of-function mutations might cause neurodevelopmental disorders irrespective of parent of origin. Here, we engineered a mouse line that harbors an autism-linked UBE3AT485A (T503A in mouse) gain-of-function mutation and evaluated phenotypes in animals that inherited the mutant allele paternally, maternally, or from both parents. We find that paternally and maternally expressed UBE3AT503A results in elevated UBE3A activity in neural progenitors and glial cells. Expression of UBE3AT503A from the maternal allele, but not the paternal one, leads to a persistent elevation of UBE3A activity in neurons. Mutant mice display behavioral phenotypes that differ by parent of origin. Expression of UBE3AT503A, irrespective of its parent of origin, promotes transient embryonic expansion of Zcchc12 lineage interneurons. Phenotypes of Ube3aT503A mice are distinct from Angelman syndrome model mice. Our study has clinical implications for a growing number of disease-linked UBE3A gain-of-function mutations.


Subject(s)
Angelman Syndrome , Autistic Disorder , Animals , Mice , Autistic Disorder/genetics , Disease Models, Animal , Gain of Function Mutation , Interneurons/metabolism , Maternal Inheritance , Phenotype , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
2.
Elife ; 102021 11 15.
Article in English | MEDLINE | ID: mdl-34779768

ABSTRACT

Triple-negative breast cancer (TNBC) is an aggressive and highly lethal disease, which warrants the critical need to identify new therapeutic targets. We show that Zinc Fingers and Homeoboxes 2 (ZHX2) is amplified or overexpressed in TNBC cell lines and patients. Functionally, depletion of ZHX2 inhibited TNBC cell growth and invasion in vitro, orthotopic tumor growth, and spontaneous lung metastasis in vivo. Mechanistically, ZHX2 bound with hypoxia-inducible factor (HIF) family members and positively regulated HIF1α activity in TNBC. Integrated ChIP-seq and gene expression profiling demonstrated that ZHX2 co-occupied with HIF1α on transcriptionally active promoters marked by H3K4me3 and H3K27ac, thereby promoting gene expression. Among the identified ZHX2 and HIF1α coregulated genes, overexpression of AP2B1, COX20, KDM3A, or PTGES3L could partially rescue TNBC cell growth defect by ZHX2 depletion, suggested that these downstream targets contribute to the oncogenic role of ZHX2 in an accumulative fashion. Furthermore, multiple residues (R491, R581, and R674) on ZHX2 are important in regulating its phenotype, which correspond with their roles on controlling ZHX2 transcriptional activity in TNBC cells. These studies establish that ZHX2 activates oncogenic HIF1α signaling, therefore serving as a potential therapeutic target for TNBC.


Subject(s)
Carcinogenesis/genetics , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Transcription Factors/genetics , Triple Negative Breast Neoplasms/genetics , Cell Line, Tumor , Gene Expression Profiling , Homeodomain Proteins/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Transcription Factors/metabolism
3.
Dis Model Mech ; 14(6)2021 06 01.
Article in English | MEDLINE | ID: mdl-34137816

ABSTRACT

Genetics are a known contributor to differences in alcohol sensitivity in humans with fetal alcohol spectrum disorders (FASDs) and in animal models. Our study profiled gene expression in gastrulation-stage embryos from two commonly used, genetically similar mouse substrains, C57BL/6J (6J) and C57BL/6NHsd (6N), that differ in alcohol sensitivity. First, we established normal gene expression patterns at three finely resolved time points during gastrulation and developed a web-based interactive tool. Baseline transcriptional differences across strains were associated with immune signaling. Second, we examined the gene networks impacted by alcohol in each strain. Alcohol caused a more pronounced transcriptional effect in the 6J versus 6N mice, matching the increased susceptibility of the 6J mice. The 6J strain exhibited dysregulation of pathways related to cell death, proliferation, morphogenic signaling and craniofacial defects, while the 6N strain showed enrichment of hypoxia and cellular metabolism pathways. These datasets provide insight into the changing transcriptional landscape across mouse gastrulation, establish a valuable resource that enables the discovery of candidate genes that may modify alcohol susceptibility that can be validated in humans, and identify novel pathogenic mechanisms of alcohol. This article has an associated First Person interview with the first author of the paper.


Subject(s)
Embryo, Mammalian/metabolism , Ethanol/toxicity , Gastrulation , Gene Expression Profiling , Animals , Embryo, Mammalian/drug effects , Mice
5.
Cell Rep ; 35(1): 108946, 2021 04 06.
Article in English | MEDLINE | ID: mdl-33826889

ABSTRACT

Although embryonic brain development and neurodegeneration have received considerable attention, the events that govern postnatal brain maturation are less understood. Here, we identify the miR-29 family to be strikingly induced during the late stages of brain maturation. Brain maturation is associated with a transient, postnatal period of de novo non-CG (CH) DNA methylation mediated by DNMT3A. We examine whether an important function of miR-29 during brain maturation is to restrict the period of CH methylation via its targeting of Dnmt3a. Deletion of miR-29 in the brain, or knockin mutations preventing miR-29 to specifically target Dnmt3a, result in increased DNMT3A expression, higher CH methylation, and repression of genes associated with neuronal activity and neuropsychiatric disorders. These mouse models also develop neurological deficits and premature lethality. Our results identify an essential role for miR-29 in restricting CH methylation in the brain and illustrate the importance of CH methylation regulation for normal brain maturation.


Subject(s)
Brain/growth & development , Brain/metabolism , DNA Methylation/genetics , MicroRNAs/metabolism , 3' Untranslated Regions/genetics , Animals , Animals, Newborn , Base Sequence , Behavior, Animal , DNA (Cytosine-5-)-Methyltransferases/metabolism , Down-Regulation/genetics , Gene Expression Regulation, Developmental , Mice, Inbred C57BL , MicroRNAs/genetics , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/pathology , Neurons/metabolism , Neurons/pathology , Seizures/genetics , Seizures/pathology , Signal Transduction , Synapses/metabolism , Up-Regulation/genetics
6.
Mol Autism ; 11(1): 74, 2020 10 05.
Article in English | MEDLINE | ID: mdl-33023670

ABSTRACT

BACKGROUND: Chromodomain helicase DNA-binding protein 8 (Chd8) is a high-confidence risk gene for autism spectrum disorder (ASD). However, how Chd8 haploinsufficiency impairs gene expression in the brain and impacts behavior at different stages of life is unknown. METHODS: We generated a mutant mouse line with an ASD-linked loss-of-function mutation in Chd8 (V986*; stop codon mutation). We examined the behavior of Chd8 mutant mice along with transcriptional changes in the cerebral cortex as a function of age, with a focus on one embryonic (E14.5) and three postnatal ages (1, 6, and 12 months). RESULTS: Chd8V986*/+ mutant mice displayed macrocephaly, reduced rearing responses and reduced center time in the open field, and enhanced social novelty preference. Behavioral phenotypes were more evident in Chd8V986*/+ mutant mice at 1 year of age. Pup survival was reduced in wild-type x Chd8V986*/+ crosses when the mutant parent was female. Transcriptomic analyses indicated that pathways associated with synaptic and neuronal projections and sodium channel activity were reduced in the cortex of embryonic Chd8V986*/+ mice and then equalized relative to wild-type mice in the postnatal period. At 12 months of age, expression of genes associated with endoplasmic reticulum (ER) stress, chaperone-mediated protein folding, and the unfolded protein response (UPR) were reduced in Chd8V986*/+ mice, whereas genes associated with the c-MET signaling pathway were increased in expression. LIMITATIONS: It is unclear whether the transcriptional changes observed with age in Chd8V986*/+ mice reflect a direct effect of CHD8-regulated gene expression, or if CHD8 indirectly affects the expression of UPR/ER stress genes in adult mice as a consequence of neurodevelopmental abnormalities. CONCLUSIONS: Collectively, these data suggest that UPR/ER stress pathways are reduced in the cerebral cortex of aged Chd8V986*/+ mice. Our study uncovers neurodevelopmental and age-related phenotypes in Chd8V986*/+ mice and highlights the importance of controlling for age when studying Chd8 haploinsufficient mice.


Subject(s)
Brain/embryology , Brain/metabolism , DNA-Binding Proteins/genetics , Haploinsufficiency/genetics , Proteostasis/genetics , Animals , Anxiety/physiopathology , Behavior, Animal , Gene Expression Profiling , Mice, Inbred C57BL , Organ Size , Phenotype , Phosphorylation , Reflex, Startle , Ribosomal Protein S6/metabolism , Social Interaction , Survival Analysis , Time Factors
7.
Cancer Discov ; 10(11): 1706-1721, 2020 11.
Article in English | MEDLINE | ID: mdl-32690540

ABSTRACT

Triple-negative breast cancer (TNBC) is an aggressive and highly lethal disease. Because of its heterogeneity and lack of hormone receptors or HER2 expression, targeted therapy is limited. Here, by performing a functional siRNA screening for 2-OG-dependent enzymes, we identified gamma-butyrobetaine hydroxylase 1 (BBOX1) as an essential gene for TNBC tumorigenesis. BBOX1 depletion inhibits TNBC cell growth while not affecting normal breast cells. Mechanistically, BBOX1 binds with the calcium channel inositol-1,4,5-trisphosphate receptor type 3 (IP3R3) in an enzymatic-dependent manner and prevents its ubiquitination and proteasomal degradation. BBOX1 depletion suppresses IP3R3-mediated endoplasmic reticulum calcium release, therefore impairing calcium-dependent energy-generating processes including mitochondrial respiration and mTORC1-mediated glycolysis, which leads to apoptosis and impaired cell-cycle progression in TNBC cells. Therapeutically, genetic depletion or pharmacologic inhibition of BBOX1 inhibits TNBC tumor growth in vitro and in vivo. Our study highlights the importance of targeting the previously uncharacterized BBOX1-IP3R3-calcium oncogenic signaling axis in TNBC. SIGNIFICANCE: We provide evidence from unbiased screens that BBOX1 is a potential therapeutic target in TNBC and that genetic knockdown or pharmacologic inhibition of BBOX1 leads to decreased TNBC cell fitness. This study lays the foundation for developing effective BBOX1 inhibitors for treatment of this lethal disease.This article is highlighted in the In This Issue feature, p. 1611.


Subject(s)
gamma-Butyrobetaine Dioxygenase/metabolism , Cell Proliferation , Female , Humans , Signal Transduction
8.
J Pathol ; 252(2): 125-137, 2020 10.
Article in English | MEDLINE | ID: mdl-32619021

ABSTRACT

Activation of the nuclear factor (erythroid-derived 2)-like 2 (NFE2L2 or NRF2) transcription factor is a critical and evolutionarily conserved cellular response to oxidative stress, metabolic stress, and xenobiotic insult. Deficiency of NRF2 results in hypersensitivity to a variety of stressors, whereas its aberrant activation contributes to several cancer types, most commonly squamous cell carcinomas of the esophagus, oral cavity, bladder, and lung. Between 10% and 35% of patients with squamous cell carcinomas display hyperactive NRF2 signaling, harboring activating mutations and copy number amplifications of the NFE2L2 oncogene or inactivating mutations or deletions of KEAP1 or CUL3, the proteins of which co-complex to ubiquitylate and degrade NRF2 protein. To better understand the role of NRF2 in tumorigenesis and more broadly in development, we engineered the endogenous Nfe2l2 genomic locus to create a conditional mutant LSL-Nrf2E79Q mouse model. The E79Q mutation, one of the most commonly observed NRF2-activating mutations in human squamous cancers, codes for a mutant protein that does not undergo KEAP1/CUL3-dependent degradation, resulting in its constitutive activity. Expression of NRF2 E79Q protein in keratin 14 (KRT14)-positive murine tissues resulted in hyperplasia of squamous cell tissues of the tongue, forestomach, and esophagus, a stunted body axis, decreased weight, and decreased visceral adipose depots. RNA-seq profiling and follow-up validation studies of cultured NRF2E79Q murine esophageal epithelial cells revealed known and novel NRF2-regulated transcriptional programs, including genes associated with squamous cell carcinoma (e.g. Myc), lipid and cellular metabolism (Hk2, Ppard), and growth factors (Areg, Bmp6, Vegfa). These data suggest that in addition to decreasing adipogenesis, KRT14-restricted NRF2 activation drives hyperplasia of the esophagus, forestomach, and tongue, but not formation of squamous cell carcinoma. © 2020 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Adipose Tissue, White/pathology , Carcinogenesis/genetics , Disease Models, Animal , NF-E2-Related Factor 2/genetics , Precancerous Conditions/genetics , Upper Gastrointestinal Tract/pathology , Animals , Carcinoma, Squamous Cell/genetics , Esophagus/pathology , Humans , Hyperplasia/genetics , Mice , Mutation , Tongue/pathology
9.
Article in English | MEDLINE | ID: mdl-32490353

ABSTRACT

BACKGROUND: Increasing evidence suggests a causal relationship between the gut microbiome and psychiatric illnesses. In particular, autism spectrum disorder is associated with gastrointestinal symptoms and alterations in the gut microbiome. Administration of probiotics is a commonly used strategy by caregivers of people with neurodevelopmental illness. However, evidence for successful improvement in gut microbiome and (behavioral) symptoms has been lacking. RESULTS: Here, we use a novel ferret model of maternal immune activation to show that high-dose probiotic administration in a placebo-controlled study design causes changes in the gut microbiome in the form of a transient increase in the administered bacterial species. In contrast, we found no differences in baseline microbiome composition or changes induced by probiotic administration between animals exposed in utero to maternal immune activation and control animals. However, the relative presence of several bacterial species correlated with an increased preference for novelty (object and conspecific). Intriguingly, several of the hits in this screen are species that have previously emerged in the literature as being associated with autism and anxiety. CONCLUSIONS: Together, our results suggest that high-dose probiotic interventions may be beneficial for the adjunct treatment of psychiatric illnesses. Placebo-controlled clinical trials in humans are urgently needed.

10.
Cell Rep ; 31(10): 107751, 2020 06 09.
Article in English | MEDLINE | ID: mdl-32521276

ABSTRACT

Set2 co-transcriptionally methylates lysine 36 of histone H3 (H3K36), producing mono-, di-, and trimethylation (H3K36me1/2/3). These modifications recruit or repel chromatin effector proteins important for transcriptional fidelity, mRNA splicing, and DNA repair. However, it was not known whether the different methylation states of H3K36 have distinct biological functions. Here, we use engineered forms of Set2 that produce different lysine methylation states to identify unique and shared functions for H3K36 modifications. Although H3K36me1/2 and H3K36me3 are functionally redundant in many SET2 deletion phenotypes, we found that H3K36me3 has a unique function related to Bur1 kinase activity and FACT (facilitates chromatin transcription) complex function. Further, during nutrient stress, either H3K36me1/2 or H3K36me3 represses high levels of histone acetylation and cryptic transcription that arises from within genes. Our findings uncover the potential for the regulation of diverse chromatin functions by different H3K36 methylation states.


Subject(s)
Histones/metabolism , Protein Processing, Post-Translational/genetics , Transcription, Genetic/genetics , Humans , Methylation
11.
Alcohol Clin Exp Res ; 44(8): 1540-1550, 2020 08.
Article in English | MEDLINE | ID: mdl-32557641

ABSTRACT

BACKGROUND: Early gestational alcohol exposure is associated with severe craniofacial and CNS dysmorphologies and behavioral abnormalities during adolescence and adulthood. Alcohol exposure during the formation of the neural tube (gestational day [GD] 8 to 10 in mice; equivalent to4th week of human pregnancy) disrupts development of ventral midline brain structures such as the pituitary, septum, and ventricles. This study identifies transcriptomic changes in the rostroventral neural tube (RVNT), the region of the neural tube that gives rise to the midline structures sensitive to alcohol exposure during neurulation. METHODS: Female C57BL/6J mice were administered 2 doses of alcohol (2.9 g/kg) or vehicle 4 hours apart on GD 9.0. The RVNTs of embryos were collected 6 or 24 hours after the first dose and processed for RNA-seq. RESULTS: Six hours following GD 9.0 alcohol exposure (GD 9.25), over 2,300 genes in the RVNT were determined to be differentially regulated by alcohol. Enrichment analysis determined that PAE affected pathways related to cell proliferation, p53 signaling, ribosome biogenesis, and immune activation. In addition, over 100 genes involved in primary cilia formation and function and regulation of morphogenic pathways were altered 6 hours after alcohol exposure. The changes to gene expression were largely transient, as only 91 genes identified as differentially regulated by prenatal alcohol at GD 10 (24 hours postexposure). Functionally, the differentially regulated genes at GD 10 were related to organogenesis and cell migration. CONCLUSIONS: These data give a comprehensive view of the changing landscape of the embryonic transcriptome networks in regions of the neural tube that give rise to brain structures impacted by a neurulation-stage alcohol exposure. Identification of gene networks dysregulated by alcohol will help elucidate the pathogenic mechanisms of alcohol's actions.


Subject(s)
Central Nervous System Depressants/pharmacology , Embryo, Mammalian/drug effects , Ethanol/pharmacology , Gene Expression Regulation, Developmental/drug effects , Neural Tube/drug effects , Neurulation/drug effects , Animals , Cell Proliferation/genetics , Cilia/genetics , Embryo, Mammalian/metabolism , Female , Gene Expression Profiling , Mice , Neural Tube/metabolism , Neurulation/genetics , Organelle Biogenesis , Pregnancy , RNA-Seq , Ribosomes/genetics , Tumor Suppressor Protein p53
12.
Proc Natl Acad Sci U S A ; 117(23): 13023-13032, 2020 06 09.
Article in English | MEDLINE | ID: mdl-32461361

ABSTRACT

Clear cell renal cell carcinoma (ccRCC) is characterized by loss of tumor suppressor Von Hippel Lindau (VHL) function, which leads to accumulation of hypoxia inducible factor α (including HIF1α and HIF2α). HIF2α was previously reported to be one of the major oncogenic drivers in ccRCC, however, its therapeutic targets remain challenging. Here we performed a deubiquitinase (DUB) complementary DNA (cDNA) library binding screen and discovered that ubiquitin-specific peptidase 37 (USP37) is a DUB that binds HIF2α and promotes HIF2α deubiquitination. As a result, USP37 promotes HIF2α protein stability in an enzymatically dependent manner, and depletion of USP37 leads to HIF2α down-regulation in ccRCC. Functionally, USP37 depletion causes decreased cell proliferation measured by MTS, two-dimensional (2D) colony formation as well as three-dimensional (3D) anchorage- independent growth. USP37 is also essential for maintaining kidney tumorigenesis in an orthotopic xenograft model and its depletion leads to both decreased primary kidney tumorigenesis and spontaneous lung metastasis. Our results suggest that USP37 is a potential therapeutic target in ccRCC.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinoma, Renal Cell/pathology , Endopeptidases/metabolism , Kidney Neoplasms/pathology , Animals , Carcinogenesis , Carcinoma, Renal Cell/genetics , Cell Line, Tumor , Chromatin Immunoprecipitation Sequencing , Down-Regulation , Endopeptidases/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Kidney Neoplasms/genetics , Mice , Protein Stability , RNA, Small Interfering/metabolism , RNA-Seq , Ubiquitination , Xenograft Model Antitumor Assays
13.
Mol Cell ; 77(6): 1294-1306.e5, 2020 03 19.
Article in English | MEDLINE | ID: mdl-32023483

ABSTRACT

von Hippel-Lindau (VHL) is a critical tumor suppressor in clear cell renal cell carcinomas (ccRCCs). It is important to identify additional therapeutic targets in ccRCC downstream of VHL loss besides hypoxia-inducible factor 2α (HIF2α). By performing a genome-wide screen, we identified Scm-like with four malignant brain tumor domains 1 (SFMBT1) as a candidate pVHL target. SFMBT1 was considered to be a transcriptional repressor but its role in cancer remains unclear. ccRCC patients with VHL loss-of-function mutations displayed elevated SFMBT1 protein levels. SFMBT1 hydroxylation on Proline residue 651 by EglN1 mediated its ubiquitination and degradation governed by pVHL. Depletion of SFMBT1 abolished ccRCC cell proliferation in vitro and inhibited orthotopic tumor growth in vivo. Integrated analyses of ChIP-seq, RNA-seq, and patient prognosis identified sphingosine kinase 1 (SPHK1) as a key SFMBT1 target gene contributing to its oncogenic phenotype. Therefore, the pVHL-SFMBT1-SPHK1 axis serves as a potential therapeutic avenue for ccRCC.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Renal Cell/pathology , Gene Expression Regulation, Neoplastic , Kidney Neoplasms/pathology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Repressor Proteins/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Cell Cycle , Cell Movement , Cell Proliferation , Genome-Wide Association Study , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Phosphotransferases (Alcohol Group Acceptor)/genetics , Prognosis , Prolyl Hydroxylases/genetics , Prolyl Hydroxylases/metabolism , Repressor Proteins/genetics , Tumor Cells, Cultured , Ubiquitination , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Xenograft Model Antitumor Assays
14.
Nat Commun ; 10(1): 5177, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31729379

ABSTRACT

Protein hydroxylation affects protein stability, activity, and interactome, therefore contributing to various diseases including cancers. However, the transiency of the hydroxylation reaction hinders the identification of hydroxylase substrates. By developing an enzyme-substrate trapping strategy coupled with TAP-TAG or orthogonal GST- purification followed by mass spectrometry, we identify adenylosuccinate lyase (ADSL) as an EglN2 hydroxylase substrate in triple negative breast cancer (TNBC). ADSL expression is higher in TNBC than other breast cancer subtypes or normal breast tissues. ADSL knockout impairs TNBC cell proliferation and invasiveness in vitro and in vivo. An integrated transcriptomics and metabolomics analysis reveals that ADSL activates the oncogenic cMYC pathway by regulating cMYC protein level via a mechanism requiring ADSL proline 24 hydroxylation. Hydroxylation-proficient ADSL, by affecting adenosine levels, represses the expression of the long non-coding RNA MIR22HG, thus upregulating cMYC protein level. Our findings highlight the role of ADSL hydroxylation in controlling cMYC and TNBC tumorigenesis.


Subject(s)
Adenylosuccinate Lyase/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Triple Negative Breast Neoplasms/enzymology , Adenosine/metabolism , Adenylosuccinate Lyase/genetics , Carcinogenesis , Cell Line, Tumor , Cell Proliferation , Female , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/physiopathology
15.
eNeuro ; 5(5)2018.
Article in English | MEDLINE | ID: mdl-30406186

ABSTRACT

Maternal immune activation (MIA) has been identified as a causal factor in psychiatric disorders by epidemiological studies in humans and mechanistic studies in rodent models. Addressing this gap in species between mice and human will accelerate the understanding of the role of MIA in the etiology of psychiatric disorders. Here, we provide the first study of MIA in the ferret (Mustela putorius furo), an animal model with a rich history of developmental investigations due to the similarities in developmental programs and cortical organization with primates. We found that after MIA by injection of PolyIC in the pregnant mother animal, the adult offspring exhibited reduced social behavior, less eye contact with humans, decreased recognition memory, a sex-specific increase in amphetamine-induced hyperlocomotion, and altered gut microbiome. We also studied the neurophysiological properties of the MIA ferrets in development by in-vivo recordings of the local field potential (LFP) from visual cortex in five- to six-week-old animals, and found that the spontaneous and sensory-evoked LFP had decreased power, especially in the gamma frequency band. Overall, our results provide the first evidence for the detrimental effect of MIA in ferrets and support the use of the ferret as an intermediate model species for the study of disorders with neurodevelopmental origin.


Subject(s)
Behavior, Animal/physiology , Brain/immunology , Gastrointestinal Microbiome/physiology , Mental Disorders/physiopathology , Aging , Animals , Brain/physiology , Disease Models, Animal , Female , Ferrets , Pregnancy , Prenatal Exposure Delayed Effects , Social Behavior , Visual Cortex/physiology
16.
Support Care Cancer ; 25(5): 1563-1570, 2017 05.
Article in English | MEDLINE | ID: mdl-28064384

ABSTRACT

PURPOSE: In this proof-of-concept pilot study, our purpose was to determine correlations between gut microbiota composition and alterations in cardiorespiratory fitness and psychosocial outcomes among post-primary treatment breast cancer survivors (BCS). METHODS: Composition of the gut microbiota in BCS (n = 12) was assessed at baseline (M0) and at the end of 3 months (M3) using Illumina MiSeq DNA Sequencing of the 16S rRNA gene. Gut microbiota composition was analyzed using the QIIME bioinformatics software and represented through diversity metrics and taxa analyses. Cardiorespiratory fitness, fatigue, anxiety, depression, and sleep dysfunction were assessed at M0 and M3 via the submaximal treadmill test, Fatigue Symptom Inventory, Hospital Anxiety and Depression Scale, and Pittsburgh Sleep Quality Index, respectively. RESULTS: Increased fatigue interference in BCS was associated with increased mean within-sample Shannon diversity (organism richness and evenness) (p = 0.009). Weighted UniFrac analysis (shifts in taxa relative abundance) revealed significant differences in between-sample (beta) diversity for changes in fatigue interference (p = 0.01) and anxiety (p = 0.022), with a trend observed for fatigue intensity and sleep dysfunction (p < 0.1). Unweighted UniFrac analysis (shifts in taxa types) found significant beta diversity differences for cardiorespiratory fitness (p = 0.026). Prior to false discovery correction (FDR), changes in fitness, fatigue, anxiety, and sleep dysfunction were associated with the frequency of certain gut bacteria genera (e.g., Faecalibacterium, Prevotella, Bacteroides) (p < 0.05). CONCLUSIONS: Correlations may exist between alterations in gut microbiota composition and longitudinal changes in cardiorespiratory fitness, fatigue, and anxiety in BCS. Further research examining the role of the microbiota-gut-brain axis in exercise-induced effects on psychosocial outcomes in BCS is warranted.


Subject(s)
Breast Neoplasms/psychology , Cardiorespiratory Fitness/physiology , Exercise/physiology , Gastrointestinal Microbiome/immunology , Adolescent , Adult , Aged , Breast Neoplasms/mortality , Female , Humans , Middle Aged , Survival Rate , Survivors/psychology , Young Adult
17.
Arthritis Rheumatol ; 66(5): 1291-9, 2014 May.
Article in English | MEDLINE | ID: mdl-24782186

ABSTRACT

OBJECTIVE: To investigate whether the Fcγ receptor IIIa-66L/R/H (FcγRIIIa-66L/R/H) polymorphism influences net effective receptor function and to assess if the FCGR3A combined genotypes formed by FcγRIIIa-66L/R/H and FcγRIIIa-176F/V, as well as copy number variation (CNV), confer risk of developing systemic lupus erythematosus (SLE) and lupus nephritis. METHODS: FcγRIIIa variants, expressed on A20 IIA1.6 cells, were used in flow cytometry-based human IgG-binding assays. Using Pyrosequencing methodology, FCGR3A single-nucleotide polymorphism and CNV genotypes were determined in a cohort of 1,728 SLE patients and 2,404 healthy controls. RESULTS: The FcγRIIIa-66L/R/H (rs10127939) polymorphism influenced ligand binding capacity in the presence of the FcγRIIIa-176V (rs396991) allele. There was a trend toward an association of the low-binding FcγRIIIa-176F allele with lupus nephritis among African Americans (P = 0.0609) but not among European Americans (P > 0.10). Nephritis among African American patients with SLE was associated with FcγRIIIa low-binding haplotypes containing the 66L/R/H and 176F variants (P = 0.03) and with low-binding genotype combinations (P = 0.002). No association was observed among European American patients with SLE. The distribution of FCGR3A CNV was not significantly different among controls and SLE patients with or without nephritis. CONCLUSION: FcγRIIIa-66L/R/H influences ligand binding. The low-binding haplotypes formed by 66L/R/H and 176F confer enhanced risk of lupus nephritis in African Americans. FCGR3A CNVs are not associated with SLE or lupus nephritis in either African Americans or European Americans.


Subject(s)
Black or African American/genetics , Immunoglobulin G/metabolism , Lupus Nephritis/ethnology , Lupus Nephritis/genetics , Receptors, IgG/genetics , Alleles , Case-Control Studies , Genetic Predisposition to Disease/ethnology , Genetic Predisposition to Disease/genetics , Genotype , Haplotypes/genetics , Humans , Lupus Nephritis/epidemiology , Phenotype , Polymorphism, Single Nucleotide/genetics , Protein Binding , Risk Factors , White People/genetics
18.
Pharmacogenet Genomics ; 23(9): 455-62, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23778324

ABSTRACT

OBJECTIVE: Kawasaki disease (KD), response to intravenous immunoglobulin (IVIG) therapy, and associated coronary artery disease progression have been associated with genetic polymorphisms in Fc gamma receptor (FcγR) genes. However, it is not known whether the existing gene copy number (GCN) variability relates to KD treatment response, susceptibility, or associated sequelae. METHODS: The copy number of individuals with KD (n=510) and their family members (n=808) for three variable FcγRs was assessed using pyrosequencing. We performed the transmission disequilibrium test to examine the association of GCN for FcγRs (FcγR2C, FcγR3A, and FcγR3B) with susceptibility and used logistic regression models to determine its association with IVIG treatment outcomes. RESULTS: FcγR2C and FcγR3B GCN were significantly associated with KD susceptibility. IVIG response was associated with GCN variations of FcγR3B in Whites and FcγR2C in Hispanics, and gene risk score based on single nucleotide polymorphism and GCN in FcγRs were significantly different between IVIG responders and nonresponders among Whites. We found no significant associations between coronary artery disease and any of the FcγR copy numbers. CONCLUSION: GCN of FcγR2C and FcγR3B influences IVIG treatment response and predisposes individuals to KD, providing potential insights into understanding the mechanism of the FcγR gene family in the IVIG pathway.


Subject(s)
Gene Dosage , Immunoglobulins, Intravenous/therapeutic use , Mucocutaneous Lymph Node Syndrome/genetics , Mucocutaneous Lymph Node Syndrome/therapy , Receptors, IgG/genetics , Adult , Asian People/genetics , Base Sequence , Child , Cohort Studies , Coronary Artery Disease/complications , Coronary Artery Disease/epidemiology , Coronary Artery Disease/ethnology , Coronary Artery Disease/genetics , Genetic Predisposition to Disease , Genetic Variation , Hispanic or Latino/genetics , Humans , Linkage Disequilibrium , Molecular Sequence Data , Mucocutaneous Lymph Node Syndrome/complications , Mucocutaneous Lymph Node Syndrome/ethnology , Polymorphism, Single Nucleotide , Receptors, IgG/immunology , Treatment Outcome , White People/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...