428 results on '"BROWN WT"'
Search Results
2. Professional Engineers and Agricultural Extension Services
- Author
-
National Engineering Conference (1985 : Melbourne, Vic.), Brown, WT, and Foster, MP
- Published
- 1985
3. Engineering Australian Agriculture - a Biased Historical Perspective
- Author
-
Engineering Conference (1984 : Brisbane, Qld.), Brown, WT, and O'Shea, JA
- Published
- 1984
4. Genetic analysis of first-trimester miscarriages with a combination of cytogenetic karyotyping, microsatellite genotyping and arrayCGH
- Author
-
Zhang Yx, Jenkins Ec, Shupin Li, Gu Y, Ju W, Xie Js, Wu Bl, Zhang Yp, Brown Wt, Nanbert Zhong, Yiping Shen, and Guan Fj
- Subjects
medicine.medical_specialty ,Genotype ,Genetic counseling ,Biology ,Gene dosage ,Genetic analysis ,Cytogenetics ,Pregnancy ,Prenatal Diagnosis ,Genetics ,medicine ,Humans ,Genotyping ,Genetics (clinical) ,Comparative Genomic Hybridization ,Karyotype ,medicine.disease ,Abortion, Spontaneous ,Pregnancy Trimester, First ,Karyotyping ,Chromosome abnormality ,Female ,Microsatellite Repeats ,Comparative genomic hybridization - Abstract
Miscarriage is the spontaneous loss of an embryo or fetus before the 20th week of pregnancy. Most miscarriages occur before the end of the first trimester (
- Published
- 2009
- Full Text
- View/download PDF
5. Premature Aging Syndromes
- Author
-
Brown Wt
- Subjects
Premature aging ,Progeria ,Down syndrome ,business.industry ,media_common.quotation_subject ,Longevity ,Physiology ,medicine.disease ,Bioinformatics ,Cockayne syndrome ,medicine ,business ,Werner syndrome ,media_common - Published
- 2015
- Full Text
- View/download PDF
6. Perspectives and Molecular Diagnosis of the Fragile X Syndrome
- Author
-
Brown Wt
- Subjects
Genetics ,Triplet repeat ,Biochemistry (medical) ,Clinical Biochemistry ,RNA-binding protein ,Disease ,Biology ,medicine.disease ,law.invention ,Fragile X syndrome ,law ,Mutation (genetic algorithm) ,medicine ,Gene ,Polymerase chain reaction ,Southern blot - Abstract
The fragile X syndrome is the most common mendelianly inherited form of mental retardation. The underlying mutation is usually a triplet repeat (CGG) that is variable in length and undergoes a tremendous length amplification in affected individuals. The mutation leads to absence expression of a gene, which apparently functions as an RNA binding protein. Molecular diagnostic testing for the mutation is conducted using direct genomic Southern blot analysis and polymerase chain reaction. Because the mutation is so common and a single type of mutation accounts for most individuals with the disease, widespread genetic screening can be considered.
- Published
- 1995
- Full Text
- View/download PDF
7. Mapping autism risk loci using genetic linkage and chromosomal rearrangements
- Author
-
Szatmari, P, Paterson, AD, Zwaigenbaum, L, Roberts, W, Brian, J, Liu, XQ, Vincent, JB, Skaug, JL, Thompson, AP, Senman, L, Feuk, L, Qian, C, Bryson, SE, Jones, MB, Marshall, CR, Scherer, SW, Vieland, VJ, Bartlett, C, Mangin, LV, Goedken, R, Segre, A, Pericak-Vance, MA, Cuccaro, ML, Gilbert, JR, Wright, HH, Abramson, RK, Betancur, C, Bourgeron, T, Gillberg, C, Leboyer, M, Buxbaum, JD, Davis, KL, Hollander, E, Silverman, JM, Hallmayer, J, Lotspeich, L, Sutcliffe, JS, Haines, JL, Folstein, SE, Piven, J, Wassink, TH, Sheffield, V, Geschwind, DH, Bucan, M, Brown, WT, Cantor, RM, Constantino, JN, Gilliam, TC, Herbert, M, LaJonchere, C, Ledbetter, DH, Lese-Martin, C, Miller, J, Nelson, S, Samango-Sprouse, CA, Spence, S, State, M, Tanzi, RE, Coon, H, Dawson, G, Devlin, B, Estes, A, Flodman, P, Klei, L, McMahon, WM, Minshew, N, Munson, J, Korvatska, E, Rodier, PM, Schellenberg, GD, Smith, M, Spence, MA, Stodgell, C, Tepper, PG, Wijsman, EM, Yu, CE, Rogé, B, Mantoulan, C, Wittemeyer, K, Poustka, A, Felder, B, Klauck, SM, Schuster, C, Poustka, F, Bölte, S, Feineis-Matthews, S, Herbrecht, E, Schmötzer, G, Tsiantis, J, Papanikolaou, K, Maestrini, E, and Bacchelli, E
- Subjects
mental disorders - Abstract
Autism spectrum disorders (ASDs) are common, heritable neurodevelopmental conditions. The genetic architecture of ASDs is complex, requiring large samples to overcome heterogeneity. Here we broaden coverage and sample size relative to other studies of ASDs by using Affymetrix 10K SNP arrays and 1,168 families with at least two affected individuals, performing the largest linkage scan to date while also analyzing copy number variation in these families. Linkage and copy number variation analyses implicate chromosome 11p12-p13 and neurexins, respectively, among other candidate loci. Neurexins team with previously implicated neuroligins for glutamatergic synaptogenesis, highlighting glutamate-related genes as promising candidates for contributing to ASDs. © 2007 Nature Publishing Group.
- Published
- 2007
- Full Text
- View/download PDF
8. 5-HTTLPR variants not associated with autistic spectrum disorders
- Author
-
Brown Wt, Ju W, Zhong N, Ye L, Tsiouris J, and Cohen I
- Subjects
Serotonin ,Genotype ,Nerve Tissue Proteins ,Autistic spectrum ,Cellular and Molecular Neuroscience ,Polymorphism (computer science) ,Reference Values ,Germany ,mental disorders ,Genetics ,Ethnicity ,Medicine ,Humans ,Genetic Predisposition to Disease ,Allele ,Autistic Disorder ,Child ,Genetics (clinical) ,Serotonin transporter ,Alleles ,Serotonin Plasma Membrane Transport Proteins ,Membrane Glycoproteins ,Polymorphism, Genetic ,biology ,business.industry ,Racial Groups ,Genetic Variation ,Membrane Transport Proteins ,medicine.disease ,Human genetics ,United States ,Fragile X syndrome ,5-HTTLPR ,Fragile X Syndrome ,behavior and behavior mechanisms ,biology.protein ,business ,Carrier Proteins ,psychological phenomena and processes - Abstract
To determine whether there is an association of polymorphic variants of the serotonin transporter (5-HTT) gene-linked polymorphic region (5-HTTLPR) and autistic spectrum disorders, we analyzed the 5-HTTLPR genotypes of 72 autistic subjects, 11 fragile X syndrome patients with autistic behavior, 43 normal subjects, and 49 fragile X syndrome non-autistic subjects. The distribution frequency of 5-HTTLPR long allele (L) and the short allele (S) variants showed no differences between subjects. Our findings do not support the hypothesis that polymorphic 5-HTTLPR variants are a susceptibility factor for autistic disorders.
- Published
- 2001
9. CLN-encoded proteins do not interact with each other
- Author
-
Ju W, Moroziewicz Dn, Wisniewski Ke, Zhong Na, Jurkiewicz A, and Brown Wt
- Subjects
Two-hybrid screening ,engineering.material ,Biology ,medicine.disease_cause ,Protein–protein interaction ,Cellular and Molecular Neuroscience ,Open Reading Frames ,Neuronal Ceroid-Lipofuscinoses ,Genetics ,medicine ,Humans ,Cloning, Molecular ,Gene ,Genetics (clinical) ,Mutation ,Tripeptidyl-Peptidase 1 ,Reverse Transcriptase Polymerase Chain Reaction ,Chromosome Mapping ,Membrane Proteins ,medicine.disease ,Phenotype ,Batten ,CLN3 ,engineering ,Neuronal ceroid lipofuscinosis ,Lysosomes - Abstract
The lysosomal storage of lipofuscins is the common pathological feature that characterizes the infantile, late-infantile, juvenile (Batten's disease), and Finnish-variant neuronal ceroid lipofuscinosis (INCL, LINCL, JNCL and FNCL), which are due to mutations in the genes CLN1 , CLN2 , CLN3 , and CLN5 , respectively. The CLN1 and CLN2 genes encode lysosomal enzymes, but the CLN3 and CLN5 genes encode membrane-spanning proteins. Why deficiencies of lysosomal enzymes and membrane-spanning proteins produce similar clinical phenotypes and pathological changes is still unanswered. We hypothesize that CLN -encoded proteins may comprise a functional pathogenic pathway, in which protein associations may play important roles. To test this hypothesis, we studied protein-protein interactions among the CLN1 -, CLN2 -, and CLN3 -encoded proteins using a yeast two-hybrid system. Our results provided no evidence that CLN -encoded proteins interact with each other. This suggests there may be unidentified components in NCL pathogenesis.
- Published
- 2000
10. New X-linked mental retardation (XLMR) syndrome with distinct facial appearance and growth retardation
- Author
-
Brown Wt, Krystyna E. Wisniewski, and Susan Sklower Brooks
- Subjects
Adult ,Male ,medicine.medical_specialty ,X Chromosome ,Adolescent ,Genetic Linkage ,Genes, Recessive ,Nystagmus ,Cerebral Ventricles ,Atrophy ,Internal medicine ,Intellectual Disability ,Spastic diplegia ,medicine ,Humans ,Abnormalities, Multiple ,Global developmental delay ,Genetics (clinical) ,Growth Disorders ,Sex Chromosome Aberrations ,Philtrum ,business.industry ,Infant ,Anatomy ,Syndrome ,medicine.disease ,Blepharophimosis ,Pedigree ,medicine.anatomical_structure ,Endocrinology ,Face ,Small for gestational age ,medicine.symptom ,Agenesis of Corpus Callosum ,business ,Esotropia - Abstract
We report on 2 brothers and their nephew with an apparently new X-linked mental retardation (XLMR) syndrome characterized by a distinct facial appearance, growth retardation, and severe mental retardation. The facial traits included triangular shape; bifrontal narrowness; malar flatness; blepharophimosis; very deeply set eyes; epicanthus inversus; bulbous nose; low hairline; low-set, deeply cupped, and protruding ears; short ill-defined philtrum; and thin tented upper lip. These facial anomalies are particularly striking and recognizable even at birth. The boys were small for gestational age and remained below −2 SD in growth parameters. With age, large joint contractures developed. Pectus excavatum was apparent at birth but became more obvious with age. Global developmental delay was evident in infancy. The brothers were nonverbal while their nephew spoke simple words. Optic atrophy, esotropia, nystagmus, and spastic diplegia were evident. They were self-abusive, hyperactive, and poorly coordinated. CT scans demonstrated atrophic hydrocephalus. No EEG abnormalities were detected. Karyotypes were 46,XY and fragile X negative. Routine chemistries; amino, organic, and uronic acids; oligosaccharides; lysosomal enzymes; and very long chain fatty acids were normal. Remarkable phenotypic similarity between these brothers and their nephew and lack of manifestations in their mothers makes X-linked recessive inheritance likely. This syndrome, which does not appear to have been reported previously, adds to the delineation of XLMR. © 1994 Wiley-Liss, Inc.
- Published
- 1994
11. COLLABORATIVE PROSPECTIVE-STUDY OF THE FRAGILE-X SYNDROME - ONE-YEAR PROGRESS REPORT
- Author
-
Sherman, Sl, Barbi, G, Brondumnielsen, K, Brown, Wt, Carpenter, Nj, Chudley, Ae, Ferraz, Op, Ferreira, P, Gustavson, Kh, Halliday, J, Hockey, A, Howardpeebles, Pn, Jenkins, E, Kennerknecht, I, Kahkonen, M, Ladaique, P, Leisti, J, Maddalena, A, Mazurczak, T, Mattei, Jf, Mattina, Teresa, Mckinley, Mj, Murphy, P, Pellissier, Mc, Purvissmith, S, Robinson, H, Scapagnini, U, Schaap, T, Shapiro, Lr, Smits, Apt, Steinbach, P, Turner, G, Uchida, Ia, Vanoost, Ba, Voelckel, Ma, Weaver, Dd, Webb, T., and University of Groningen
- Subjects
PARENTAL ORIGIN ,FRAGILE-X SYNDROME ,MALES ,RECURRENCE RISKS ,X-LINKED MENTAL RETARDATION ,ISOLATED CASE - Abstract
A prospective study of the fragile X syndrome [fra(X)] was initiated one year ago to refine the estimates of recurrence risks based on the phenotype of the mother and the family history of the syndrome. The basic unit of data consists of the description of the conceptus of women known to carry the fra(X) gene or of mothers of an isolated case. To date, information on 261 women and their conceptuses was ascertained primarily through prenatal diagnosis; these data are summarized here. Although tests of significance were limited due to small sample sizes in subgroups, the following trends were observed: 1) the penetrance of fra(X) site expression was 80% in both male and female conceptuses suggesting that fra(X) site expression is equally penetrant early in development; 2) the sex ratio at the time of prenatal diagnosis did not differ from one, indicating that selection against fra(X) fetuses, if any, does not differ among sexes; 3) the recurrence risk among offspring of borderline/mildly retarded mothers was higher than that among offspring of intellectually normal mothers; 4) the recurrence risk in offspring did not differ based on the mother's fra(X) site expression; and 5) the recurrence risk in offspring of mothers with isolated cases was slightly less (34%) than that of obligate carrier mothers (41%) although this was not significant. The potential use of these prospective data on the fra(X) syndrome is emphasized.
- Published
- 1992
12. Association of autism severity with a monoamine oxidase A functional polymorphism
- Author
-
Cohen, IL, primary, Liu, X, additional, Schutz, C, additional, White, BN, additional, Jenkins, EC, additional, Brown, WT, additional, and Holden, JJA, additional
- Published
- 2003
- Full Text
- View/download PDF
13. Progeria: a human-disease model of accelerated aging
- Author
-
Brown, WT, primary
- Published
- 1992
- Full Text
- View/download PDF
14. Treatment of early non-small cell lung cancer, stage IA, by image-guided robotic stereotactic radioablation -- cyberknife.
- Author
-
Brown WT, Wu X, Amendola B, Perman M, Han H, Fayad F, Garcia S, Lewin A, Abitbol A, de la Zerda A, and Schwade JG
- Abstract
OBJECTIVE:: To evaluate the efficacy of using image-guided robotic stereotactic radioablation as an alternative treatment modality for patients with surgically resectable, but medically inoperable, T1 N0 M0, stage IA non-small cell lung cancer. METHODS:: Between January 2004 and May 2006, 19 patients, 11 women and 8 men ranging in age from 52 to 88 years, with stage IA non-small cell lung cancer were treated. Tumor volume ranged from 1.7 to 13 mL. Total doses ranged from 24 to 60 Gy delivered in 3 fractions. Eleven patients received 60 Gy. Real-time target localization was accomplished by radiographic detection of fiducial marker(s) implanted within the tumor combined with respiratory motion tracking. RESULTS:: All patients tolerated radioablation well with fatigue as the main side effect. Fourteen patients are alive from 1 to 25 months posttreatment. Four patients died: 2 of comorbid disease and 2 of cancer progression (status post 60 and 55.5 Gy). Three patients developed grade I radiation pneumonitis. Two patients have stable disease. In 3 patients, cancer recurred in the planning treatment volume: in 2 patients after treatment with 60 Gy and in 1 patient after treatment with 55.5 Gy. One patient had local control in the target volume but developed metastasis to the ipsilateral hilum. Nine patients had a complete response and show no evidence of disease. CONCLUSIONS:: In our early experience, stereotactic radioablation using the CyberKnife system appears to be a safe, minimally invasive, and effective modality for treating early stage lung cancer in patients with medically inoperable disease. Dose escalation and/or increasing the treatment volumes, with the aid of the high conformality of this technique, may help to achieve further improvements in these promising results. [ABSTRACT FROM AUTHOR]
- Published
- 2007
- Full Text
- View/download PDF
15. Association of autism severity with a monoamine oxidase A functional polymorphism.
- Author
-
IL Cohen, M.D., X Liu, Schutz, C, White, BN, Jenkins, EC, Brown, WT, and Holdend, JJA
- Subjects
AUTISM ,MONOAMINE oxidase ,GENETIC polymorphisms ,SEROTONINERGIC mechanisms - Abstract
A functional polymorphism (the upstream variable-number tandem repeat region, or uVNTR) in the monoamine oxidase A (MAOA) promoter region has been reported to be associated with behavioral abnormalities as well as increased serotonergic responsivity. We examined the relation between MAOA-uVNTR alleles and the phenotypic expression of autism in 41 males younger than 12.6 years of age. Children with the low-activity MAOA allele had both lower intelligence quotients (IQ) and more severe autistic behavior than children with the high-activity allele. In follow-up testing of 34 of the males at the 1-year time-point, those with the low-activity allele showed a worsening in IQ but no change in the severity of their autistic behavior. We conclude that functional MAOA-uVNTR alleles may act as a genetic modifier of the severity of autism in males. [ABSTRACT FROM AUTHOR]
- Published
- 2003
- Full Text
- View/download PDF
16. Cystathionine Disappearance with Neuronal Loss: A Possible Neuronal Marker
- Author
-
Brown Wt, Krystyna E. Wisniewski, Wisniewski Hm, Sturman Ja, Devine E, and R. Rudelli
- Subjects
medicine.medical_specialty ,Cerebellum ,Adolescent ,Transsulfuration pathway ,Cystathionine ,Neuronal Ceroid-Lipofuscinoses ,Cortex (anatomy) ,Internal medicine ,medicine ,Humans ,Brain Chemistry ,Cerebral Cortex ,biology ,business.industry ,Brain ,General Medicine ,Spinal cord ,Cystathionine beta synthase ,medicine.anatomical_structure ,Endocrinology ,Spinal Cord ,Cerebral cortex ,Cerebellar cortex ,Pediatrics, Perinatology and Child Health ,biology.protein ,Female ,Neurology (clinical) ,Neuron ,business - Abstract
Cystathionine is an important intermediate in the transsulfuration pathway of methionine catabolism and is normally present in high concentration in the human CNS. We have measured the concentration of cystathionine, other amino acids, and brain proteins in the cerebral cortex, cerebellar cortex and spinal cord of two cases with ceroid lipofuscinosis. Neuropathological and biochemical studies of Case 1, at an advanced stage, Case 2, at an early stage, and five controls were correlated with clinical and neurological findings. The concentration of an unidentified 54,000 Dalton protein was greatly increased in Case 1 as observed by 2-D gel electrophoresis. Neurons and cystathionine were almost totally absent from the cortex and cerebellum of Case 1, while they were slightly reduced in Case 2, in comparison to control brains. These studies suggest that cystathionine may be specifically located within neurons. We present for the first time the observation that there was a strikingly low brain concentration of cystathionine, a potential neuronal marker, in an advanced stage of a neurodegenerative process.
- Published
- 1985
- Full Text
- View/download PDF
17. Assignment of proteins to human chromosome 21 using two-dimensional gel electrophoresis and somatic cell genetics: An approach to the study of Down syndrome
- Author
-
E. A. Devine-Gage, Brown Wt, Dutkowski R, Sammons D, and Jenkins Ec
- Subjects
Genetics ,Premature aging ,Gel electrophoresis ,Two-dimensional gel electrophoresis ,Chromosome ,Aneuploidy ,Biology ,medicine.disease ,Molecular biology ,Cellular and Molecular Neuroscience ,medicine ,Chromosome abnormality ,Trisomy ,Chromosome 21 - Abstract
A mouse hybrid cell line (WA17d) was derived which contained multiple copies of human chromosome 21 and no other human chromosome. Cell extracts of this line were prepared and subjected to two-dimensional gel electrophoresis. Several proteins were identified whose synthesis was altered by the presence of chromosome 21 and 6 proteins were identified as being specific to this human chromosome. These gene products might be involved in the pathogenesis of Down syndrome and be related to the neurologic defects and premature aging seen in this common chromosome abnormality.
- Published
- 1987
- Full Text
- View/download PDF
18. Autism is associated with the fragile-X syndrome
- Author
-
S. Raguthu, E. Friedman, Brown Wt, Jenkins Ec, J.H. French, Brooks J, and Krystyna E. Wisniewski
- Subjects
Male ,medicine.medical_specialty ,Adolescent ,Research methodology ,Public health ,medicine.disease ,Fragile X syndrome ,Fragile X Syndrome ,Clinical diagnosis ,Testis ,Developmental and Educational Psychology ,medicine ,Etiology ,Humans ,Autism ,Autistic Disorder ,Child ,Psychiatry ,Psychology ,Sex Chromosome Aberrations ,Clinical psychology - Published
- 1982
- Full Text
- View/download PDF
19. A Comparison of Adult and Childhood Progerias: Werner Syndrome and Hutchinson-Gilford Progeria Syndrome
- Author
-
Kieras Fj, Houck Ge, Dutkowski R, Brown Wt, and Jenkins Ec
- Subjects
Genetics ,Senescence ,congenital, hereditary, and neonatal diseases and abnormalities ,Mutation ,Progeria ,integumentary system ,nutritional and metabolic diseases ,Crouzon syndrome ,Consanguinity ,Disease ,Biology ,medicine.disease ,medicine.disease_cause ,Phenotype ,medicine ,Werner syndrome - Abstract
The Werner syndrome, also known as progeria of the adult, and the childhood Hutchinson-Gilford Progeria Syndrome (hereafter Progeria), both serve as genetic disease models of human aging (Brown, 1979). A comparison of their similarities and differences may be useful in order to gain insight into the nature of the genetic mutations underlying these conditions. Their modes of inheritance indicate the involvement of a single gene. This implies that some specific genes may lead to a phenotype of greatly accelerated senescence and that such genes may have direct effects on the rate of aging. Determining the basic mechanisms involved in producing their phenotypes may point the way to an understanding of important pathogenetic aspects underlying the aging process.
- Published
- 1985
- Full Text
- View/download PDF
20. Fragile X expression in short-term whole blood cultures is affected by cell density
- Author
-
M. S. Krawczun, Brown Wt, Edmund C. Jenkins, and Wayne Silverman
- Subjects
Genetics ,Male ,Fragile x ,Heterozygote ,Blood Cells ,Chromosomal fragile site ,Cell Count ,Biology ,Standard methods ,Molecular biology ,Genetic Techniques ,Fragile X Syndrome ,Cell density ,Martin-bell syndrome ,Humans ,Female ,Floxuridine ,Genetics (clinical) ,Sex Chromosome Aberrations ,Whole blood - Abstract
The effect of cell density on expression of the fragile site at (X)(q27.3) in short-term whole-blood cultures from patients with fragile X [fra(X)] or Martin-Bell syndrome was studied. A significant increase in fra(X) frequency was observed in 7 of 8 samples when cell density was decreased. Higher fra(X) frequency was not always noted at below-routine density, but in some cases fra(X) expression was depressed at above-routine density. We conclude that decay of the FUdR effect explains the fact that fra(X) expression is affected by culture density. It is significant that a relationship exists between the two; it suggests that in order to maximize fra(X) expression in cases with low-percentage fra(X) with standard methods, cell density may have to be adjusted. It is possible that in individuals who are normally nonexpressing, such as some obligate female carriers and nonpenetrant males, fra(X) expression may be sensitive to cell density effects.
- Published
- 1988
21. Megavitamin and orthomolecular therapy of schizophrenia
- Author
-
Brown Wt
- Subjects
Psychiatry ,medicine.medical_specialty ,business.industry ,Attitude of Health Personnel ,Schizophrenia (object-oriented programming) ,MEDLINE ,General Medicine ,Vitamins ,Orthomolecular therapy ,medicine ,Schizophrenia ,Humans ,business - Published
- 1975
22. A PvuII RFLP detected in the human prion protein (PrP) gene
- Author
-
Ye Wu, P. Merz, H. M. Wisniewski, Evelyn A. Devine-Gage, Brown Wt, Nikolaos K. Robakis, and C. Dobkin
- Subjects
Genetics ,Male ,Polymorphism, Genetic ,biology ,Chromosomes, Human, Pair 20 ,DNA Restriction Enzymes ,Molecular biology ,Restriction fragment ,Pedigree ,Genes ,biology.protein ,Prnp gene ,Humans ,Female ,Restriction fragment length polymorphism ,Prion protein ,Deoxyribonucleases, Type II Site-Specific ,Gene ,Polymorphism, Restriction Fragment Length - Published
- 1987
23. RFLP analysis in 5 Sicilian families with the fragile X syndrome
- Author
-
Romano, V., Mascali, G., Chiavetta, V., Ragusa, Rm, Barletta, C., Corrado Romano, Mollica, F., Mattina, Teresa, Gross, A., and Brown, Wt
24. An anonymous single copy chromosome 21 probe, DS21D2, associated with a frequent RFLP
- Author
-
Evelyn A. Devine-Gage, Brown Wt, G.E. Houck, and C.B. Chan
- Subjects
Genetics ,Polymorphism, Genetic ,Chromosome Mapping ,DNA Restriction Enzymes ,Biology ,Single copy ,Deoxyribonuclease EcoRI ,Gene Frequency ,Chromosomes, Human, 21-22 and Y ,Humans ,Allele ,Restriction fragment length polymorphism ,Chromosome 21 ,Allele frequency ,Alleles - Published
- 1985
- Full Text
- View/download PDF
25. Video-assisted thoracic surgery: The Miami experience
- Author
-
Brown, WT
- Published
- 1998
26. Mapping autism risk loci using genetic linkage and chromosomal rearrangements
- Author
-
Peter, Szatmari, Andrew D, Paterson, Lonnie, Zwaigenbaum, Wendy, Roberts, Jessica, Brian, Xiao-Qing, Liu, John B, Vincent, Jennifer L, Skaug, Ann P, Thompson, Lili, Senman, Lars, Feuk, Cheng, Qian, Susan E, Bryson, Marshall B, Jones, Christian R, Marshall, Stephen W, Scherer, Veronica J, Vieland, Christopher, Bartlett, La Vonne, Mangin, Rhinda, Goedken, Alberto, Segre, Margaret A, Pericak-Vance, Michael L, Cuccaro, John R, Gilbert, Harry H, Wright, Ruth K, Abramson, Catalina, Betancur, Thomas, Bourgeron, Christopher, Gillberg, Marion, Leboyer, Joseph D, Buxbaum, Kenneth L, Davis, Eric, Hollander, Jeremy M, Silverman, Joachim, Hallmayer, Linda, Lotspeich, James S, Sutcliffe, Jonathan L, Haines, Susan E, Folstein, Joseph, Piven, Thomas H, Wassink, Val, Sheffield, Daniel H, Geschwind, Maja, Bucan, W Ted, Brown, Rita M, Cantor, John N, Constantino, T Conrad, Gilliam, Martha, Herbert, Clara, Lajonchere, David H, Ledbetter, Christa, Lese-Martin, Janet, Miller, Stan, Nelson, Carol A, Samango-Sprouse, Sarah, Spence, Matthew, State, Rudolph E, Tanzi, Hilary, Coon, Geraldine, Dawson, Bernie, Devlin, Annette, Estes, Pamela, Flodman, Lambertus, Klei, William M, McMahon, Nancy, Minshew, Jeff, Munson, Elena, Korvatska, Patricia M, Rodier, Gerard D, Schellenberg, Moyra, Smith, M Anne, Spence, Chris, Stodgell, Ping Guo, Tepper, Ellen M, Wijsman, Chang-En, Yu, Bernadette, Rogé, Carine, Mantoulan, Kerstin, Wittemeyer, Annemarie, Poustka, Bärbel, Felder, Sabine M, Klauck, Claudia, Schuster, Fritz, Poustka, Sven, Bölte, Sabine, Feineis-Matthews, Evelyn, Herbrecht, Gabi, Schmötzer, John, Tsiantis, Katerina, Papanikolaou, Elena, Maestrini, Elena, Bacchelli, Francesca, Blasi, Simona, Carone, Claudio, Toma, Herman, Van Engeland, Maretha, de Jonge, Chantal, Kemner, Frederieke, Koop, Frederike, Koop, Marjolein, Langemeijer, Marjolijn, Langemeijer, Channa, Hijmans, Channa, Hijimans, Wouter G, Staal, Gillian, Baird, Patrick F, Bolton, Michael L, Rutter, Emma, Weisblatt, Jonathan, Green, Catherine, Aldred, Julie-Anne, Wilkinson, Andrew, Pickles, Ann, Le Couteur, Tom, Berney, Helen, McConachie, Anthony J, Bailey, Kostas, Francis, Gemma, Honeyman, Aislinn, Hutchinson, Jeremy R, Parr, Simon, Wallace, Anthony P, Monaco, Gabrielle, Barnby, Kazuhiro, Kobayashi, Janine A, Lamb, Ines, Sousa, Nuala, Sykes, Edwin H, Cook, Stephen J, Guter, Bennett L, Leventhal, Jeff, Salt, Catherine, Lord, Christina, Corsello, Vanessa, Hus, Daniel E, Weeks, Fred, Volkmar, Maïté, Tauber, Eric, Fombonne, Andy, Shih, Kacie J, Meyer, Department of Psychiatry and Behavioural Neurosciences, McMaster University [Hamilton, Ontario]-Offord Centre for Child Studies, The Centre for Applied Genomics, Toronto, University of Toronto-The Hospital for sick children [Toronto] (SickKids)-Department of Molecular Genetics-McLaughlin Centre, Department of Pediatrics, University of Alberta, Autism Research Unit, The Hospital for sick children [Toronto] (SickKids)-University of Toronto, Department of Psychiatry, University of Toronto, Departments of Pediatrics and Psychology, Dalhousie University [Halifax], Department of Neural and Behavioral Sciences, Pennsylvania State University (Penn State), Penn State System-Penn State System, Department of Molecular Genetics [Toronto], Battelle Center for Mathematical Medicine, Ohio State University [Columbus] (OSU)-Nationwide Children's Hospital, Department of Pathology and Laboratory Medicine, University of North Carolina [Chapel Hill] (UNC), University of North Carolina System (UNC)-University of North Carolina System (UNC), Department of Computer Science, University of Iowa [Iowa City], John P. Hussman Institute for Human Genomics, University of Miami [Coral Gables], W.S. Hall Psychiatric Institute, University of South Carolina [Columbia], Physiopathologie des Maladies du Système Nerveux Central, Université Pierre et Marie Curie - Paris 6 (UPMC)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Génétique Humaine et Fonctions Cognitives, Institut Pasteur [Paris]-Centre National de la Recherche Scientifique (CNRS), Gillberg Neuropsychiatry Centre [Göteborg, Sueden], Institute of Neuroscience and Physiology [Göteborg]-University of Gothenburg (GU), Institute of Child Health, University College of London [London] (UCL), Institut Mondor de Recherche Biomédicale (IMRB), Institut National de la Santé et de la Recherche Médicale (INSERM)-IFR10-Université Paris-Est Créteil Val-de-Marne - Paris 12 (UPEC UP12), Friedman Brain Institute, Mount Sinai, Icahn School of Medicine at Mount Sinai [New York] (MSSM), Department of Neuroscience, PennState Meteorology Department, Department of Psychiatry [Pittsburgh], University of Pittsburgh School of Medicine, Pennsylvania Commonwealth System of Higher Education (PCSHE)-Pennsylvania Commonwealth System of Higher Education (PCSHE), Stanford School of Medicine [Stanford], Stanford Medicine, Stanford University-Stanford University, Department of Psychiatry and Behavioral Sciences [Stanford], Vanderbilt Brain Institute, Vanderbilt University School of Medicine [Nashville], Department of Molecular Physiology & Biophysics and Psychiatry, Vanderbilt University [Nashville]-Centers for Human Genetics Research and Molecular Neuroscience, Johns Hopkins University (JHU), Carolina Institute for Developmental Disabilities, Carver College of Medicine [Iowa City], University of Iowa [Iowa City]-University of Iowa [Iowa City], University of Iowa [Iowa City]-Howard Hughes Medical-Institute Carver College of Medicine, Department of Neurology, UCLA School of Medicine, Department of Genetics, University of Pennsylvania [Philadelphia]-School of Medicine, N.Y.S. Institute for Basic Research in Developmental Disabilities, Department of Human Genetics, UCLA, University of California [Los Angeles] (UCLA), University of California-University of California-Semel Institute, Washington University in Saint Louis (WUSTL), University of Chicago, Harvard Medical School [Boston] (HMS), Autism Genetic Resource Exchange, Autism Speaks, Emory University [Atlanta, GA], Developmental Brain and Behaviour Unit, University of Southampton, Cure Autism Now, Institute of Human Genetics, Rheinische Friedrich-Wilhelms-Universität Bonn, Children's National Medical Center, The George Washington University (GW), Massachusetts General Hospital, Massachusetts General Hospital [Boston], Department of Developmental Neuroscience, IRCCS Fondazione Stella Maris [Pisa], Autism Speaks and the Department of Psychiatry, Department of Speech and Hearing Sciences [Washington], University of Washington [Seattle], University of California [Irvine] (UCI), University of California-University of California, Department of Psychiatry and Behavioral Sciences, Department of OB/GYN, University of Rochester Medical Center, Pathology and Laboratory Medicine, University of Pennsylvania [Philadelphia], Department of Epidemiology, University of Pittsburgh (PITT), Departments of Biostatistics and Medicine, Department of Medicine, Octogone Unité de Recherche Interdisciplinaire (Octogone), Université Toulouse - Jean Jaurès (UT2J), Centre de Référence du Syndrome de Prader-Willi, CHU Toulouse [Toulouse], University of Oxford [Oxford]-Warneford Hospital, Division of Molecular Genome Analysis, German Cancer Research Center - Deutsches Krebsforschungszentrum [Heidelberg] (DKFZ), Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Goethe-Universität Frankfurt am Main, University Department of Child Psychiatry, National and Kapodistrian University of Athens (NKUA), Department of Pharmacy and Biotechnology, Alma Mater Studiorum Università di Bologna [Bologna] (UNIBO), Medical Genetics Laboratory, Policlinico S. Orsola-Malpighi, University Medical Center [Utrecht]-Brain Center Rudolf Magnus, Department of Neurocognition, Maastricht University [Maastricht], Newcomen Centre, Guy's Hospital [London], Department of Child and Adolescent Psychiatry, Institute of psychiatry, MRC Social, Genetic and Developmental Psychiatry Centre (SGDP), The Institute of Psychiatry-King‘s College London, University of Cambridge Clinical School, University of Cambridge [UK] (CAM), Manchester Academic Health Sciences Centre, Department of Medicine, Manchester, University of Manchester [Manchester]-School of Epidemiology and Health Science, Newcastle University [Newcastle]-Institute of Health & Society (Child & Adolescent Psychiatry), Child and Adolescent Mental Health, Newcastle University [Newcastle], Institutes of Neuroscience and Health and Society, The Wellcome Trust Centre for Human Genetics [Oxford], University of Oxford [Oxford], Centre for Integrated Genomic Medical Research, Manchester, University of Manchester [Manchester], Institute for Juvenile Research-University of Illinois [Chicago] (UIC), University of Illinois System-University of Illinois System, Institute for Juvenile Research, University of Illinois [Chicago] (UIC), Department of Disability and Human Development, New York University [New York] (NYU), NYU System (NYU)-NYU System (NYU), Autism and Communicative Disorders Centre, University of Michigan [Ann Arbor], University of Michigan System-University of Michigan System, Human Genetics Department, SFU Discrete Mathematics Group (SFU-DMG), Simon Fraser University (SFU.ca), Child Study Centre, Yale University School of Medicine, Centre d'Endocrinologie, Maladies Osseuses, Génétique et Gynécologie Médicale, Hôpital des Enfants, CHU Toulouse [Toulouse]-CHU Toulouse [Toulouse], Department of Child Psychiatry, McGill University = Université McGill [Montréal, Canada]-Montreal Children's Hospital, McGill University Health Center [Montreal] (MUHC)-McGill University Health Center [Montreal] (MUHC), Scientific Affairs, Autism Genome Project Consortium, RS: FPN CN II, Cognitive Neuroscience, MUMC+: HZC Klinische Neurofysiologie (5), The Hospital for sick children [Toronto] (SickKids)-University of Toronto-Department of Molecular Genetics-McLaughlin Centre, University of California (UC)-University of California (UC)-Semel Institute, University of California [Irvine] (UC Irvine), University of California (UC)-University of California (UC), King‘s College London-The Institute of Psychiatry, Yale School of Medicine [New Haven, Connecticut] (YSM), Betancur, Catalina, Institut Pasteur [Paris] (IP)-Centre National de la Recherche Scientifique (CNRS), University of Pennsylvania-School of Medicine, University of Pennsylvania, Pôle Enfants [CHU Toulouse], Centre Hospitalier Universitaire de Toulouse (CHU Toulouse)-Centre Hospitalier Universitaire de Toulouse (CHU Toulouse), University of Oxford-Warneford Hospital, University of Oxford, Centre Hospitalier Universitaire de Toulouse (CHU Toulouse), Université de Toulouse (UT)-Université de Toulouse (UT), Szatmari P, Paterson AD, Zwaigenbaum L, Roberts W, Brian J, Liu XQ, Vincent JB, Skaug JL, Thompson AP, Senman L, Feuk L, Qian C, Bryson SE, Jones MB, Marshall CR, Scherer SW, Vieland VJ, Bartlett C, Mangin LV, Goedken R, Segre A, Pericak-Vance MA, Cuccaro ML, Gilbert JR, Wright HH, Abramson RK, Betancur C, Bourgeron T, Gillberg C, Leboyer M, Buxbaum JD, Davis KL, Hollander E, Silverman JM, Hallmayer J, Lotspeich L, Sutcliffe JS, Haines JL, Folstein SE, Piven J, Wassink TH, Sheffield V, Geschwind DH, Bucan M, Brown WT, Cantor RM, Constantino JN, Gilliam TC, Herbert M, Lajonchere C, Ledbetter DH, Lese-Martin C, Miller J, Nelson S, Samango-Sprouse CA, Spence S, State M, Tanzi RE, Coon H, Dawson G, Devlin B, Estes A, Flodman P, Klei L, McMahon WM, Minshew N, Munson J, Korvatska E, Rodier PM, Schellenberg GD, Smith M, Spence MA, Stodgell C, Tepper PG, Wijsman EM, Yu CE, Roge B, Mantoulan C, Wittemeyer K, Poustka A, Felder B, Klauck SM, Schuster C, Poustka F, Bolte S, Feineis-Matthews S, Herbrecht E, Schmotzer G, Tsiantis J, Papanikolaou K, Maestrini E, Bacchelli E, Blasi F, Carone S, Toma C, Van Engeland H, de Jonge M, Kemner C, Koop F, Langemeijer M, Hijimans C, Staal WG, Baird G, Bolton PF, Rutter ML, Weisblatt E, Green J, Aldred C, Wilkinson JA, Pickles A, Le Couteur A, Berney T, McConachie H, Bailey AJ, Francis K, Honeyman G, Hutchinson A, Parr JR, Wallace S, Monaco AP, Barnby G, Kobayashi K, Lamb JA, Sousa I, Sykes N, Cook EH, Guter SJ, Leventhal BL, Salt J, Lord C, Corsello C, Hus V, Weeks DE, Volkmar F, Tauber M, Fombonne E, and Shih A.
- Subjects
Male ,genetic structures ,Genetic Linkage ,Neurexin ,[SDV.GEN] Life Sciences [q-bio]/Genetics ,0302 clinical medicine ,Risk Factors ,MESH: Risk Factors ,Heritability of autism ,Copy-number variation ,MESH: Genetic Variation ,Genetics ,0303 health sciences ,medicine.diagnostic_test ,MESH: Genetic Testing ,MESH: Genetic Predisposition to Disease ,Chromosome Mapping ,3. Good health ,Female ,MESH: Genetic Linkage ,MESH: Autistic Disorder ,Epigenetics of autism ,Biology ,Article ,03 medical and health sciences ,Genetic linkage ,mental disorders ,medicine ,Humans ,MESH: Chromosome Aberrations ,Family ,Genetic Predisposition to Disease ,Genetic Testing ,Autistic Disorder ,MESH: Family ,030304 developmental biology ,Genetic testing ,Chromosome Aberrations ,[SDV.GEN]Life Sciences [q-bio]/Genetics ,MESH: Humans ,Genetic Variation ,medicine.disease ,Genetic architecture ,MESH: Male ,MESH: Lod Score ,Autism ,Lod Score ,MESH: Chromosome Mapping ,MESH: Female ,030217 neurology & neurosurgery - Abstract
International audience; Autism spectrum disorders (ASDs) are common, heritable neurodevelopmental conditions. The genetic architecture of ASDs is complex, requiring large samples to overcome heterogeneity. Here we broaden coverage and sample size relative to other studies of ASDs by using Affymetrix 10K SNP arrays and 1,181 [corrected] families with at least two affected individuals, performing the largest linkage scan to date while also analyzing copy number variation in these families. Linkage and copy number variation analyses implicate chromosome 11p12-p13 and neurexins, respectively, among other candidate loci. Neurexins team with previously implicated neuroligins for glutamatergic synaptogenesis, highlighting glutamate-related genes as promising candidates for contributing to ASDs.
- Published
- 2007
- Full Text
- View/download PDF
27. Isolation and characterization of a highly polymorphic human locus (DXS455) in proximal Xq28
- Author
-
W. Ted Brown, Nelson B. Freimer, Carol L. Stayton, Ponmani Goonewardena, T. Conrad Gilliam, G. Giacomo Consalez, Stephen T. Warren, Consalez, GIAN GIACOMO, Stayton, Cl, Freimer, Nb, Goonewardena, P, Brown, Wt, Gilliam, Tc, and Warren, St
- Subjects
Genetic Markers ,Male ,Genetics ,Heterozygote ,congenital, hereditary, and neonatal diseases and abnormalities ,Linkage disequilibrium ,Polymorphism, Genetic ,X Chromosome ,Chromosome Mapping ,Locus (genetics) ,Biology ,Cosmids ,Genetic analysis ,Pedigree ,Loss of heterozygosity ,Genetic marker ,Fragile X Syndrome ,Cosmid ,Humans ,Female ,Restriction fragment length polymorphism ,Allele ,Alleles - Abstract
Human Xq28 is highly gene dense with over 27 loci. Because most of these genes have been mapped by linkage to polymorphic loci, only one of which (DXS52) is informative in most families, a search was conducted for new, highly polymorphic Xq28 markers. From a cosmid library constructed using a somatic cell hybrid containing human Xq27.3----qter as the sole human DNA, a human-insert cosmid (c346) was identified and found to reveal variation on Southern blot analyses with female DNA digested with any of several different restriction endonucleases. Two subclones of c346, p346.8 and p346.T, that respectively identify a multiallelic VNTR locus and a frequent two-allele TaqI polymorphism were isolated. Examination of 21 unrelated females showed heterozygosity of 76 and 57%, respectively. These two markers appeared to be in linkage equilibrium, and a combined analysis revealed heterozygosity in 91% of unrelated females. Families segregating the fragile X syndrome with key Xq28 crossovers position this locus (designated DXS455) between the proximal Xq28 locus DXS296 (VK21) and the more distal locus DXS374 (1A1), which is proximal to DXS52. DXS455 is therefore the most polymorphic locus identified in Xq28 and will be useful in the genetic analysis of this gene dense region, including the diagnosis of nearby genetic disease loci by linkage.
- Published
- 1992
28. Expansion of the phenotypic spectrum of KARS1-related disorders to include arthrogryposis multiplex congenita and summary of experiences with lysine supplementation.
- Author
-
Bejma TA, Beidler WS, VanSickle EA, Prokop JW, Brown WT, Scheurer-Monaghan A, and Rossetti LZ
- Subjects
- Child, Child, Preschool, Female, Humans, Infant, Male, Dietary Supplements, Mutation genetics, Phenotype, Arthrogryposis genetics, Arthrogryposis pathology, Lysine genetics, Lysine-tRNA Ligase genetics
- Abstract
There are currently multiple disorders of aminoacyl-tRNA synthetases described, including KARS1-related disorder resulting from dysfunctional lysyl-tRNA synthetases. In this report, we describe four novel KARS1 variants in three affected individuals, two of whom displayed arthrogryposis-like phenotypes, suggestive of phenotypic expansion. We also highlight subjective clinical improvement in one subject following lysine supplementation in conjunction with a protein-fortified diet, suggesting its potential as a novel treatment modality for KARS1-related disorders. This report offers additional insight into the etiology and management of KARS1-related disorders and expands our ability to provide guidance to affected individuals and their families., (© 2024 The Author(s). American Journal of Medical Genetics Part A published by Wiley Periodicals LLC.)
- Published
- 2024
- Full Text
- View/download PDF
29. Interactive effects of mindfulness and negative urgency on intimate partner aggression perpetration.
- Author
-
Brown WT, Martelli AM, and Chester DS
- Subjects
- Humans, Aggression, Interpersonal Relations, Sexual Behavior, Sexual Partners, Mindfulness, Intimate Partner Violence
- Abstract
Intimate partner aggression (IPA) is a costly and incompletely understood phenomenon. Negative urgency, the tendency to act impulsively in response to negative affect, is predictive of IPA perpetration. Mindfulness, by virtue of its emphasis on nonreactivity to negative affect, is an opposing force to urgent tendencies that may mitigate the negative urgency-IPA link. Yet, no research to date investigates the interactive effects of negative urgency and mindfulness on IPA perpetration. Two studies were conducted that measured and manipulated multiple facets of mindfulness alongside measures of negative urgency and tendencies of IPA perpetration (combined N = 508 undergraduate students in monogamous intimate relationships). Counter to our preregistered predictions, we found that negative urgency's association with greater IPA perpetration increased at higher levels of mindfulness. These findings suggest that mindfulness may not be a protective factor against IPA perpetration for individuals higher in negative urgency, but rather may serve as a risk factor., (© 2023 Wiley Periodicals LLC.)
- Published
- 2024
- Full Text
- View/download PDF
30. Involvement of Type 10 17β-Hydroxysteroid Dehydrogenase in the Pathogenesis of Infantile Neurodegeneration and Alzheimer's Disease.
- Author
-
He XY, Frackowiak J, Dobkin C, Brown WT, and Yang SY
- Subjects
- Animals, Humans, Mice, Alcohol Dehydrogenase metabolism, Brain metabolism, 17-Hydroxysteroid Dehydrogenases genetics, 17-Hydroxysteroid Dehydrogenases metabolism, Alzheimer Disease metabolism
- Abstract
Type 10 17β-hydroxysteroid dehydrogenase (17β-HSD10) is the HSD17B10 gene product playing an appreciable role in cognitive functions. It is the main hub of exercise-upregulated mitochondrial proteins and is involved in a variety of metabolic pathways including neurosteroid metabolism to regulate allopregnanolone homeostasis. Deacetylation of 17β-HSD10 by sirtuins helps regulate its catalytic activities. 17β-HSD10 may also play a critical role in the control of mitochondrial structure, morphology and dynamics by acting as a member of the Parkin/PINK1 pathway, and by binding to cyclophilin D to open mitochondrial permeability pore. 17β-HSD10 also serves as a component of RNase P necessary for mitochondrial tRNA maturation. This dehydrogenase can bind with the Aβ peptide thereby enhancing neurotoxicity to brain cells. Even in the absence of Aβ, its quantitative and qualitative variations can result in neurodegeneration. Since elevated levels of 17β-HSD10 were found in brain cells of Alzheimer's disease (AD) patients and mouse AD models, it is considered to be a key factor in AD pathogenesis. Since data underlying Aβ-binding-alcohol dehydrogenase (ABAD) were not secured from reported experiments, ABAD appears to be a fabricated alternative term for the HSD17B10 gene product. Results of this study would encourage researchers to solve the question why elevated levels of 17β-HSD10 are present in brains of AD patients and mouse AD models. Searching specific inhibitors of 17β-HSD10 may find candidates to reduce senile neurodegeneration and open new approaches for the treatment of AD.
- Published
- 2023
- Full Text
- View/download PDF
31. Editorial for the Fragile X Syndrome Genetics Special Issue: May 2023.
- Author
-
Godler DE and Brown WT
- Subjects
- Humans, Fragile X Syndrome genetics, Intellectual Disability genetics, Autistic Disorder genetics
- Abstract
Fragile X syndrome (FXS) is the leading single-gene cause of inherited intellectual disability and autism [...].
- Published
- 2023
- Full Text
- View/download PDF
32. Infantile Neurodegeneration Results from Mutants of 17β-Hydroxysteroid Dehydrogenase Type 10 Rather Than Aβ-Binding Alcohol Dehydrogenase.
- Author
-
He XY, Dobkin C, Brown WT, and Yang SY
- Subjects
- Humans, Mutation, Missense, Alcohol Dehydrogenase genetics, Alzheimer Disease genetics, 3-Hydroxyacyl CoA Dehydrogenases
- Abstract
Type 10 17β-hydroxysteroid dehydrogenase (17β-HSD10), a homo-tetrameric multifunctional protein with 1044 residues encoded by the HSD17B10 gene, is necessary for brain cognitive function. Missense mutations result in infantile neurodegeneration, an inborn error in isoleucine metabolism. A 5-methylcytosine hotspot underlying a 388-T transition leads to the HSD10 (p.R130C) mutant to be responsible for approximately half of all cases suffering with this mitochondrial disease. Fewer females suffer with this disease due to X-inactivation. The binding capability of this dehydrogenase to Aβ-peptide may play a role in Alzheimer's disease, but it appears unrelated to infantile neurodegeneration. Research on this enzyme was complicated by reports of a purported Aβ-peptide-binding alcohol dehydrogenase (ABAD), formerly referred to as endoplasmic-reticulum-associated Aβ-binding protein (ERAB). Reports concerning both ABAD and ERAB in the literature reflect features inconsistent with the known functions of 17β-HSD10. It is clarified here that ERAB is reportedly a longer subunit of 17β-HSD10 (262 residues). 17β-HSD10 exhibits L-3-hydroxyacyl-CoA dehydrogenase activity and is thus also referred to in the literature as short-chain 3-hydorxyacyl-CoA dehydrogenase or type II 3-hydorxyacyl-CoA dehydrogenase. However, 17β-HSD10 is not involved in ketone body metabolism, as reported in the literature for ABAD. Reports in the literature referring to ABAD (i.e., 17β-HSD10) as a generalized alcohol dehydrogenase, relying on data underlying ABAD's activities, were found to be unreproducible. Furthermore, the rediscovery of ABAD/ERAB's mitochondrial localization did not cite any published research on 17β-HSD10. Clarification of the purported ABAD/ERAB function derived from these reports on ABAD/ERAB may invigorate this research field and encourage new approaches to the understanding and treatment of HSD17B10 -gene-related disorders. We establish here that infantile neurodegeneration is caused by mutants of 17β-HSD10 but not ABAD, and so we conclude that ABAD represents a misnomer employed in high-impact journals.
- Published
- 2023
- Full Text
- View/download PDF
33. 3-Hydroxyacyl-CoA and Alcohol Dehydrogenase Activities of Mitochondrial Type 10 17β-Hydroxysteroid Dehydrogenase in Neurodegeneration Study.
- Author
-
He XY, Dobkin C, Brown WT, and Yang SY
- Subjects
- 17-Hydroxysteroid Dehydrogenases, Alcohol Dehydrogenase, Amyloid beta-Peptides metabolism, Coenzyme A, Humans, 3-Hydroxyacyl CoA Dehydrogenases metabolism, Alzheimer Disease metabolism
- Abstract
Background: Mitochondrial 17β-hydroxysteroid dehydrogenase type 10 (17β-HSD10) is necessary for brain cognitive function, but its studies were confounded by reports of Aβ-peptide binding alcohol dehydrogenase (ABAD), formerly endoplasmic reticulum-associated Aβ-peptide binding protein (ERAB), for two decades so long as ABAD serves as the alternative term of 17β-HSD10., Objective: To determine whether those ABAD reports are true or false, even if they were published in prestigious journals., Methods: 6xHis-tagged 17β-HSD10 was prepared and characterized by well-established experimental procedures., Results: The N-terminal 6xHis tag did not significantly interfere with the dehydrogenase activities of 17β-HSD10, but the kinetic constants of its 3-hydroxyacyl-CoA dehydrogenase activity are drastically distinct from those of ABAD, and it was not involved in ketone body metabolism as previously reported for ABAD. Furthermore, it was impossible to measure its generalized alcohol dehydrogenase activities underlying the concept of ABAD because the experimental procedures described in ABAD reports violated basic chemical and/or biochemical principles. More incredibly, both authors and journals had not yet agreed to make any corrigenda of ABAD reports., Conclusion: Brain 17β-HSD10 plays a key role in neurosteroid metabolism and further studies in this area may lead to potential treatments of neurodegeneration including AD.
- Published
- 2022
- Full Text
- View/download PDF
34. Daily Living Skills in Adolescent and Young Adult Males With Fragile X Syndrome.
- Author
-
Thurman AJ, Swinehart SS, Klusek J, Roberts JE, Bullard L, Marzan JCB, Brown WT, and Abbeduto L
- Subjects
- Activities of Daily Living, Adolescent, Adult, Humans, Language, Language Tests, Male, Young Adult, Autistic Disorder, Fragile X Syndrome
- Abstract
By adulthood, most males with fragile X syndrome (FXS) require support to navigate day-to-day settings. The present study cross-sectionally: (1) characterized the profile of daily living skills in males with FXS and (2) examined associated participant characteristics (i.e., fragile X mental retardation protein [FMRP] expression, nonverbal cognition, language, autism symptomatology, and anxiety symptomatology) using the Waisman-Activities of Daily Living questionnaire. Males with FXS (n = 57, ages 15-23 years) needed more help/support in the areas of domestic and community daily livings skills, than in the area of personal daily living skills. Significant associations were observed between reduced daily living skills and lower nonverbal cognition, receptive language, expressive language, and increased autism symptomatology. Receptive language emerged as the strongest unique predictor of daily living skill performance., (©AAIDD.)
- Published
- 2022
- Full Text
- View/download PDF
35. Development of a Quantitative FMRP Assay for Mouse Tissue Applications.
- Author
-
Adayev T, LaFauci G, Xu W, Dobkin C, Kascsak R, Brown WT, and Goodman JH
- Subjects
- Animals, Brain growth & development, Brain metabolism, Dried Blood Spot Testing standards, Female, Fragile X Mental Retardation Protein genetics, Fragile X Syndrome genetics, Immunoassay methods, Immunoassay standards, Male, Mice, Mice, Inbred C57BL, Organ Specificity, Sensitivity and Specificity, Dried Blood Spot Testing methods, Fragile X Mental Retardation Protein metabolism, Fragile X Syndrome metabolism
- Abstract
Fragile X syndrome results from the absence of the FMR1 gene product-Fragile X Mental Retardation Protein (FMRP). Fragile X animal research has lacked a reliable method to quantify FMRP. We report the development of an array of FMRP-specific monoclonal antibodies and their application for quantitative assessment of FMRP (qFMRPm) in mouse tissue. To characterize the assay, we determined the normal variability of FMRP expression in four brain structures of six different mouse strains at seven weeks of age. There was a hierarchy of FMRP expression: neocortex > hippocampus > cerebellum > brainstem. The expression of FMRP was highest and least variable in the neocortex, whereas it was most variable in the hippocampus. Male C57Bl/6J and FVB mice were selected to determine FMRP developmental differences in the brain at 3, 7, 10, and 14 weeks of age. We examined the four structures and found a developmental decline in FMRP expression with age, except for the brainstem where it remained stable. qFMRPm assay of blood had highest values in 3 week old animals and dropped by 2.5-fold with age. Sex differences were not significant. The results establish qFMRPm as a valuable tool due to its ease of methodology, cost effectiveness, and accuracy.
- Published
- 2021
- Full Text
- View/download PDF
36. A Genotype-Phenotype Study of High-Resolution FMR1 Nucleic Acid and Protein Analyses in Fragile X Patients with Neurobehavioral Assessments.
- Author
-
Budimirovic DB, Schlageter A, Filipovic-Sadic S, Protic DD, Bram E, Mahone EM, Nicholson K, Culp K, Javanmardi K, Kemppainen J, Hadd A, Sharp K, Adayev T, LaFauci G, Dobkin C, Zhou L, Brown WT, Berry-Kravis E, Kaufmann WE, and Latham GJ
- Abstract
Fragile X syndrome (FXS) is caused by silencing of the FMR1 gene, which encodes a protein with a critical role in synaptic plasticity. The molecular abnormality underlying FMR1 silencing, CGG repeat expansion, is well characterized; however, delineation of the pathway from DNA to RNA to protein using biosamples from well characterized patients with FXS is limited. Since FXS is a common and prototypical genetic disorder associated with intellectual disability (ID) and autism spectrum disorder (ASD), a comprehensive assessment of the FMR1 DNA-RNA-protein pathway and its correlations with the neurobehavioral phenotype is a priority. We applied nine sensitive and quantitative assays evaluating FMR1 DNA, RNA, and FMRP parameters to a reference set of cell lines representing the range of FMR1 expansions. We then used the most informative of these assays on blood and buccal specimens from cohorts of patients with different FMR1 expansions, with emphasis on those with FXS (N = 42 total, N = 31 with FMRP measurements). The group with FMRP data was also evaluated comprehensively in terms of its neurobehavioral profile, which allowed molecular-neurobehavioral correlations. FMR1 CGG repeat expansions, methylation levels, and FMRP levels, in both cell lines and blood samples, were consistent with findings of previous FMR1 genomic and protein studies. They also demonstrated a high level of agreement between blood and buccal specimens. These assays further corroborated previous reports of the relatively high prevalence of methylation mosaicism (slightly over 50% of the samples). Molecular-neurobehavioral correlations confirmed the inverse relationship between overall severity of the FXS phenotype and decrease in FMRP levels (N = 26 males, mean 4.2 ± 3.3 pg FMRP/ng genomic DNA). Other intriguing findings included a significant relationship between the diagnosis of FXS with ASD and two-fold lower levels of FMRP (mean 2.8 ± 1.3 pg FMRP/ng genomic DNA, p = 0.04), in particular observed in younger age- and IQ-adjusted males (mean age 6.9 ± 0.9 years with mean 3.2 ± 1.2 pg FMRP/ng genomic DNA, 57% with severe ASD), compared to FXS without ASD. Those with severe ID had even lower FMRP levels independent of ASD status in the male-only subset. The results underscore the link between FMR1 expansion, gene methylation, and FMRP deficit. The association between FMRP deficiency and overall severity of the neurobehavioral phenotype invites follow up studies in larger patient cohorts. They would be valuable to confirm and potentially extend our initial findings of the relationship between ASD and other neurobehavioral features and the magnitude of FMRP deficit. Molecular profiling of individuals with FXS may have important implications in research and clinical practice.
- Published
- 2020
- Full Text
- View/download PDF
37. The role of reduced expression of fragile X mental retardation protein in neurons and increased expression in astrocytes in idiopathic and syndromic autism (duplications 15q11.2-q13).
- Author
-
Wegiel J, Brown WT, La Fauci G, Adayev T, Kascsak R, Kascsak R, Flory M, Kaczmarski W, Kuchna I, Nowicki K, Martinez-Cerdeno V, Wisniewski T, and Wegiel J
- Subjects
- Adolescent, Adult, Animals, Brain metabolism, Cerebral Cortex pathology, Child, Child, Preschool, Female, Humans, Immunohistochemistry, Male, Middle Aged, Young Adult, Astrocytes metabolism, Autistic Disorder genetics, Autistic Disorder metabolism, Fragile X Mental Retardation Protein genetics, Fragile X Mental Retardation Protein metabolism, Neurons metabolism
- Abstract
Fragile X syndrome (FXS), caused by lack of fragile X mental retardation protein (FMRP), is associated with a high prevalence of autism. The deficit of FMRP reported in idiopathic autism suggests a mechanistic overlap between FXS and autism. The overall goal of this study is to detect neuropathological commonalities of FMRP deficits in the brains of people with idiopathic autism and with syndromic autism caused by dup15q11.2-q13 (dup15). This study tests the hypothesis based on our preliminary data that both idiopathic and syndromic autism are associated with brain region-specific deficits of neuronal FMRP and structural changes of the affected neurons. This immunocytochemical study revealed neuronal FMRP deficits and shrinkage of deficient neurons in the cerebral cortex, subcortical structures, and cerebellum in subjects with idiopathic and dup(15)/autism. Neuronal FMRP deficit coexists with surprising infiltration of the brains of autistic children and adults with FMRP-positive astrocytes known to be typical only for the fetal and short postnatal periods. In the examined autistic subjects, these astrocytes selectively infiltrate the border between white and gray matter in the cerebral and cerebellar cortex, the molecular layer of the cortex, part of the amygdala and thalamus, central cerebellar white matter, and dentate nucleus. Astrocyte pathology results in an additional local loss of FMRP in neurons and their shrinkage. Neuronal deficit of FMRP and shrinkage of affected neurons in structures free of FMRP-positive astrocytes and regions infiltrated with FMRP-expressing astrocytes appear to reflect mechanistic, neuropathological, and functional commonalities of FMRP abnormalities in FXS and autism spectrum disorder. Autism Res 2018, 11: 1316-1331. © 2018 International Society for Autism Research, Wiley Periodicals, Inc. LAY SUMMARY: Immunocytochemistry reveals a deficit of fragile X mental retardation protein (FMRP) in neurons of cortical and subcortical brain structures but increased FMRP expression in astrocytes infiltrating gray and white matter. The detected shrinkage of FMRP-deficient neurons may provide a mechanistic explanation of reported neuronal structural and functional changes in autism. This study contributes to growing evidence of mechanistic commonalities between fragile X syndrome and autism spectrum disorder., (© 2018 International Society for Autism Research, Wiley Periodicals, Inc.)
- Published
- 2018
- Full Text
- View/download PDF
38. Folate receptor autoantibodies are prevalent in children diagnosed with autism spectrum disorder, their normal siblings and parents.
- Author
-
Quadros EV, Sequeira JM, Brown WT, Mevs C, Marchi E, Flory M, Jenkins EC, Velinov MT, and Cohen IL
- Subjects
- Adult, Autism Spectrum Disorder diagnosis, Child, Child, Preschool, Female, Humans, Male, Autism Spectrum Disorder immunology, Autoantibodies immunology, Folate Receptor 1 immunology, Parents, Siblings
- Abstract
Folate deficiency can affect fetal and neonatal brain development Considering the reported association of Folate receptor alpha (FRα) autoantibodies (Abs) with autism and developmental disorders, we sought to confirm this in families of 82 children with ASD, 53 unaffected siblings, 65 fathers, and 70 mothers, along with 52 unrelated normal controls. Overall, 76% of the affected children, 75% of the unaffected siblings, 69% of fathers and 59% of mothers were positive for either blocking or binding Ab, whereas the prevalence of this Ab in the normal controls was 29%. The Ab was highly prevalent in affected families including unaffected siblings. The appearance of these antibodies may have a familial origin but the risk of developing ASD is likely influenced by other mitigating factors since some siblings who had the antibodies were not affected. The antibody response appears heritable with the blocking autoantibody in the parents and affected child increasing the risk of ASD. Autism Res 2018, 11: 707-712. © 2018 International Society for Autism Research, Wiley Periodicals, Inc., Lay Summary: Folate is an essential nutrient during fetal and infant development. Autoantibodies against the folate receptor alpha can block folate transport from the mother to the fetus and to the brain in infants. Children diagnosed with autism and their immediate family members were evaluated for the prevalence of folate receptor autoantibodies. The autoantibody was highly prevalent in affected families with similar distribution in parents, normal siblings and affected children. The presence of these antibodies appears to have a familial origin and may contribute to developmental deficits when combined with other factors., (© 2018 International Society for Autism Research, Wiley Periodicals, Inc.)
- Published
- 2018
- Full Text
- View/download PDF
39. Changes in Cerebral Oxygenation in Preterm Infants With Progressive Posthemorrhagic Ventricular Dilatation.
- Author
-
Kochan M, McPadden J, Bass WT, Shah T, Brown WT, Tye GW, and Vazifedan T
- Subjects
- Carbon Dioxide analysis, Cerebral Hemorrhage diagnostic imaging, Dilatation, Pathologic, Disease Progression, Female, Gestational Age, Humans, Hydrocephalus diagnostic imaging, Infant, Infant, Newborn, Infant, Premature, Diseases pathology, Infant, Premature, Diseases physiopathology, Male, Oximetry, Spectroscopy, Near-Infrared, Cerebral Hemorrhage etiology, Cerebral Ventricles pathology, Cerebrovascular Circulation physiology, Hydrocephalus complications, Infant, Premature, Oxygen analysis
- Abstract
Background: Optimal timing of intervention in neonatal progressive posthemorrhagic hydrocephalus is often a difficult decision. Unchecked hydrocephalus can lead to irreversible brain injury through impaired perfusion, while placement of a shunt is not without long-term morbidity. The purpose of this study was to assess the use of near-infrared spectroscopy to measure changes in regional cerebral oxygen saturation as an indicator of cerebral perfusion in infants with progressive posthemorrhagic ventricular dilatation., Methods: Near-infrared spectroscopy was used to measure regional cerebral oxygen saturation for more than a one-hour period in infants within 24 hours of cranial ultrasound. Simultaneous pulse oximetry was recorded and oxygen extraction was calculated. Ventricular size was measured by ultrasound using the frontal-occipital horn ratio and compared with average oxygen saturation and oxygen extraction. Statistical analysis was done using the Spearman rank test and analysis of variance., Results: Ventricular measurements were made in 20 very low birth weight premature infants with periventricular-intraventricular hemorrhage and 12 infants with normal ultrasound scans. Ventricular dilatation was associated with lower cerebral oxygen saturation and higher oxygen extraction (P < 0.001). Progressive ventricular dilatation was inversely related to changes in cerebral oxygen saturation (P < 0.001)., Conclusions: Progressive posthemorrhagic ventricular dilatation is associated with a significant decrease in cerebral oxygenation and increase in oxygen extraction suggesting a decrease in cerebral perfusion. Near-infrared spectroscopy could potentially provide additional clinical information to assist in determining optimal timing of surgical intervention in preterm infants with progressive ventricular enlargement., (Copyright © 2017 Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
40. Fragile X targeted pharmacotherapy: lessons learned and future directions.
- Author
-
Erickson CA, Davenport MH, Schaefer TL, Wink LK, Pedapati EV, Sweeney JA, Fitzpatrick SE, Brown WT, Budimirovic D, Hagerman RJ, Hessl D, Kaufmann WE, and Berry-Kravis E
- Abstract
Our understanding of fragile X syndrome (FXS) pathophysiology continues to improve and numerous potential drug targets have been identified. Yet, current prescribing practices are only symptom-based in order to manage difficult behaviors, as no drug to date is approved for the treatment of FXS. Drugs impacting a diversity of targets in the brain have been studied in recent FXS-specific clinical trials. While many drugs have focused on regulation of enhanced glutamatergic or deficient GABAergic neurotransmission, compounds studied have not been limited to these mechanisms. As a single-gene disorder, it was thought that FXS would have consistent drug targets that could be modulated with pharmacotherapy and lead to significant improvement. Unfortunately, despite promising results in FXS animal models, translational drug treatment development in FXS has largely failed. Future success in this field will depend on learning from past challenges to improve clinical trial design, choose appropriate outcome measures and age range choices, and find readily modulated drug targets. Even with many negative placebo-controlled study results, the field continues to move forward exploring both the new mechanistic drug approaches combined with ways to improve trial execution. This review summarizes the known phenotype and pathophysiology of FXS and past clinical trial rationale and results, and discusses current challenges facing the field and lessons from which to learn for future treatment development efforts.
- Published
- 2017
- Full Text
- View/download PDF
41. Ubiquitin-Proteasome-Collagen (CUP) Pathway in Preterm Premature Rupture of Fetal Membranes.
- Author
-
Zhao X, Dong X, Luo X, Pan J, Ju W, Zhang M, Wang P, Zhong M, Yu Y, Brown WT, and Zhong N
- Abstract
Spontaneous preterm birth (sPTB) occurs before 37 gestational weeks, with preterm premature rupture of the membranes (PPROM) and spontaneous preterm labor (sPTL) as the predominant adverse outcomes. Previously, we identified altered expression of long non-coding RNAs (lncRNAs) and message RNAs (mRNAs) related to the ubiquitin proteasome system (UPS) in human placentas following pregnancy loss and PTB. We therefore hypothesized that similar mechanisms might underlie PPROM. In the current study, nine pairs of ubiquitin-proteasome-collagen (CUP) pathway-related mRNAs and associated lncRNAs were found to be differentially expressed in PPROM and sPTL. Pathway analysis showed that the functions of their protein products were inter-connected by ring finger protein. Twenty variants including five mutations were identified in CUP-related genes in sPTL samples. Copy number variations were found in COL19A1, COL28A1, COL5A1, and UBAP2 of sPTL samples. The results reinforced our previous findings and indicated the association of the CUP pathway with the development of sPTL and PPROM. This association was due not only to the genetic variation, but also to the epigenetic regulatory function of lncRNAs. Furthermore, the findings suggested that the loss of collagen content in PPROM could result from degradation and/or suppressed expression of collagens.
- Published
- 2017
- Full Text
- View/download PDF
42. FORWARD: A Registry and Longitudinal Clinical Database to Study Fragile X Syndrome.
- Author
-
Sherman SL, Kidd SA, Riley C, Berry-Kravis E, Andrews HF, Miller RM, Lincoln S, Swanson M, Kaufmann WE, and Brown WT
- Subjects
- Activities of Daily Living psychology, Adolescent, Adult, Caregivers psychology, Child, Child, Preschool, Female, Fragile X Syndrome psychology, Fragile X Syndrome therapy, Humans, Infant, Infant, Newborn, Longitudinal Studies, Male, United States, Young Adult, Databases as Topic, Fragile X Syndrome diagnosis, Fragile X Syndrome genetics, Registries
- Abstract
Background and Objective: Advances in the care of patients with fragile X syndrome (FXS) have been hampered by lack of data. This deficiency has produced fragmentary knowledge regarding the natural history of this condition, healthcare needs, and the effects of the disease on caregivers. To remedy this deficiency, the Fragile X Clinic and Research Consortium was established to facilitate research. Through a collective effort, the Fragile X Clinic and Research Consortium developed the Fragile X Online Registry With Accessible Research Database (FORWARD) to facilitate multisite data collection. This report describes FORWARD and the way it can be used to improve health and quality of life of FXS patients and their relatives and caregivers., Methods: FORWARD collects demographic information on individuals with FXS and their family members (affected and unaffected) through a 1-time registry form. The longitudinal database collects clinician- and parent-reported data on individuals diagnosed with FXS, focused on those who are 0 to 24 years of age, although individuals of any age can participate., Results: The registry includes >2300 registrants (data collected September 7, 2009 to August 31, 2014). The longitudinal database includes data on 713 individuals diagnosed with FXS (data collected September 7, 2012 to August 31, 2014). Longitudinal data continue to be collected on enrolled patients along with baseline data on new patients., Conclusions: FORWARD represents the largest resource of clinical and demographic data for the FXS population in the United States. These data can be used to advance our understanding of FXS: the impact of cooccurring conditions, the impact on the day-to-day lives of individuals living with FXS and their families, and short-term and long-term outcomes., Competing Interests: POTENTIAL CONFLICT OF INTEREST: Dr Kaufmann is a consultant to Neuren, Edison, Newron, EryDel, Marinus, Anavex, and GW Pharmaceuticals; he has received research support from Novartis, Ipsen and Eloxx; EBK has received funding from Seaside Therapeutics, Novartis, Roche, Alcobra, Neuren, Cydan, Fulcrum and Neurotrope Pharmaceuticals to consult on trial design or development strategies and/or conduct clinical trials in FXS, Rett syndrome or Down syndrome, from Vtesse to conduct clinical trials in Niemann-Pick disease, type C, and from Asuragen Inc to develop testing standards for FMR1 testing. The other authors have indicated they have no potential conflicts of interest to disclose., (Copyright © 2017 by the American Academy of Pediatrics.)
- Published
- 2017
- Full Text
- View/download PDF
43. Autism Spectrum Disorder in Fragile X Syndrome: Cooccurring Conditions and Current Treatment.
- Author
-
Kaufmann WE, Kidd SA, Andrews HF, Budimirovic DB, Esler A, Haas-Givler B, Stackhouse T, Riley C, Peacock G, Sherman SL, Brown WT, and Berry-Kravis E
- Subjects
- Adolescent, Adrenergic alpha-Agonists therapeutic use, Adult, Antipsychotic Agents therapeutic use, Applied Behavior Analysis, Autism Spectrum Disorder psychology, Child, Child Behavior Disorders diagnosis, Child Behavior Disorders psychology, Child Behavior Disorders therapy, Child, Preschool, Combined Modality Therapy, Comorbidity, Diagnostic and Statistical Manual of Mental Disorders, Female, Fragile X Syndrome psychology, Humans, Infant, Longitudinal Studies, Male, Registries, Autism Spectrum Disorder diagnosis, Autism Spectrum Disorder therapy, Fragile X Syndrome diagnosis, Fragile X Syndrome therapy
- Abstract
Background and Objective: Individuals with fragile X syndrome (FXS) are frequently codiagnosed with autism spectrum disorder (ASD). Most of our current knowledge about ASD in FXS comes from family surveys and small studies. The objective of this study was to examine the impact of the ASD diagnosis in a large clinic-based FXS population to better inform the care of people with FXS., Methods: The study employed a data set populated by data from individuals with FXS seen at specialty clinics across the country. The data were collected by clinicians at the patient visit and by parent report for nonclinical and behavioral outcomes from September 7, 2012 through August 31, 2014. Data analyses were performed by using χ
2 tests for association, t tests, and multiple logistic regression to examine the association between clinical and other factors with ASD status., Results: Half of the males and nearly 20% of females met Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition criteria for current ASD. Relative to the FXS-only group, the FXS with ASD (FXS+ASD) group had a higher prevalence of seizures (20.7% vs 7.6%, P < .001), persistence of sleep problems later in childhood, increased behavior problems, especially aggressive/disruptive behavior, and higher use of α-agonists and antipsychotics. Behavioral services, including applied behavior analysis, appeared to be underused in children with FXS+ASD (only 26% and 16% in prekindergarten and school-age periods, respectively) relative to other populations with idiopathic ASD., Conclusions: These findings confirm among individuals with FXS an association of an ASD diagnosis with important cooccurring conditions and identify gaps between expected and observed treatments among individuals with FXS+ASD., Competing Interests: POTENTIAL CONFLICT OF INTEREST: The authors have indicated they have no potential conflicts of interest to disclose., (Copyright © 2017 by the American Academy of Pediatrics.)- Published
- 2017
- Full Text
- View/download PDF
44. Reduced vagal tone in women with the FMR1 premutation is associated with FMR1 mRNA but not depression or anxiety.
- Author
-
Klusek J, LaFauci G, Adayev T, Brown WT, Tassone F, and Roberts JE
- Abstract
Background: Autonomic dysfunction is implicated in a range of psychological conditions, including depression and anxiety. The fragile X mental retardation-1 ( FMR1 ) premutation is a common genetic mutation that affects ~1:150 women and is associated with psychological vulnerability. This study examined cardiac indicators of autonomic function among women with the FMR1 premutation and control women as potential biomarkers for psychological risk that may be linked to FMR1 ., Methods: Baseline inter-beat interval and respiratory sinus arrhythmia (a measure of parasympathetic vagal tone) were measured in 35 women with the FMR1 premutation and 28 controls. The women completed anxiety and depression questionnaires. FMR1 genetic indices (i.e., CGG repeat, quantitative FMRP, FMR1 mRNA, activation ratio) were obtained for the premutation group., Results: Respiratory sinus arrhythmia was reduced in the FMR1 premutation group relative to controls. While depression symptoms were associated with reduced respiratory sinus arrhythmia among control women, these variables were unrelated in the FMR1 premutation. Elevated FMR1 mRNA was associated with higher respiratory sinus arrhythmia., Conclusions: Women with the FMR1 premutation demonstrated autonomic dysregulation characterized by reduced vagal tone. Unlike patterns observed in the general population and in study controls, vagal activity and depression symptoms were decoupled in women with the FMR1 premutation, suggesting independence between autonomic regulation and psychopathological symptoms that is atypical and potentially specific to the FMR1 premutation. The association between vagal tone and mRNA suggests that molecular variation associated with FMR1 plays a role in autonomic regulation.
- Published
- 2017
- Full Text
- View/download PDF
45. Partial Agenesis and Hypoplasia of the Corpus Callosum in Idiopathic Autism.
- Author
-
Wegiel J, Flory M, Kaczmarski W, Brown WT, Chadman K, Wisniewski T, Nowicki K, Kuchna I, Ma SY, and Wegiel J
- Subjects
- Adolescent, Adult, Agenesis of Corpus Callosum complications, Autistic Disorder complications, Child, Child, Preschool, Corpus Callosum diagnostic imaging, Corpus Callosum pathology, Female, Humans, Male, Middle Aged, Young Adult, Agenesis of Corpus Callosum diagnostic imaging, Agenesis of Corpus Callosum pathology, Autistic Disorder diagnostic imaging, Autistic Disorder pathology
- Abstract
To test the hypothesis that developmental anomalies of the corpus callosum (CC), contribute to the pathogenesis of autism, we characterized the type, topography, and severity of CC pathology corresponding to reduced CC areas that are detected by magnetic resonance imaging in the brains of 11 individuals with autism and 11 controls. In the brains of 3 autistic subjects, partial CC agenesis resulted in complete or partial lack of interhemispheric axonal connections in CC segments III-V. In these cases, a combination of focal agenesis and uniform axonal deficit caused reduction of CC areas by 37%, of axon numbers by 62%, and of the numerical density of axons by 39%. In the CC of 8 autistic subjects without agenesis, there was an 18% deficit of the midsagittal CC area, 48.4% deficit of axon numbers, and 37% reduction of the numerical density of axons. The significantly thinner CC, reduced CC area, and uniform axonal deficit in all autistic subjects were classified as CC hypoplasia. Thus, the byproduct of partial CC agenesis and hypoplasia is reduction of axonal connections between cortical areas known to be involved in behavioral alterations observed in people with autism., (2017 American Association of Neuropathologists, Inc. This work is written by US Government employees and is in the public domain in the US.)
- Published
- 2017
- Full Text
- View/download PDF
46. Is Taurine a Biomarker in Autistic Spectrum Disorder?
- Author
-
Park E, Cohen I, Gonzalez M, Castellano MR, Flory M, Jenkins EC, Brown WT, and Schuller-Levis G
- Subjects
- Adolescent, Child, Child, Preschool, Female, Humans, Male, Autism Spectrum Disorder blood, Biomarkers blood, Taurine blood
- Abstract
Taurine is a sulfur-containing amino acid which is not incorporated into protein. However, taurine has various critical physiological functions including development of the eye and brain, reproduction, osmoregulation, and immune functions including anti-inflammatory as well as anti-oxidant activity. The causes of autistic spectrum disorder (ASD) are not clear but a high heritability implicates an important role for genetic factors. Reports also implicate oxidative stress and inflammation in the etiology of ASD. Thus, taurine, a well-known antioxidant and regulator of inflammation, was investigated here using the sera from both girls and boys with ASD as well as their siblings and parents. Previous reports regarding taurine serum concentrations in ASD from various laboratories have been controversial. To address the potential role of taurine in ASD, we collected sera from 66 children with ASD (males: 45; females: 21, age 1.5-11.5 years, average age 5.2 ± 1.6) as well as their unaffected siblings (brothers: 24; sisters: 32, age 1.5-17 years, average age 7.0 ± 2.0) as controls of the children with ASD along with parents (fathers: 49; mothers: 54, age 28-45 years). The sera from normal adult controls (males: 47; females: 51, age 28-48 years) were used as controls for the parents. Taurine concentrations in all sera samples were measured using high performance liquid chromatography (HPLC) using a phenylisothiocyanate labeling technique. Taurine concentrations from female and male children with ASD were 123.8 ± 15.2 and 145.8 ± 8.1 μM, respectively, and those from their unaffected brothers and sisters were 142.6 ± 10.4 and 150.8 ± 8.4 μM, respectively. There was no significant difference in taurine concentration between autistic children and their unaffected siblings. Taurine concentrations in children with ASD were also not significantly different from their parents (mothers: 139.6 ± 7.7 μM, fathers: 147.4 ± 7.5 μM). No significant difference was observed between adult controls and parents of ASD children (control females: 164.8 ± 4.8 μM, control males: 163.0 ± 7.0 μM). However, 21 out of 66 children with ASD had low taurine concentrations (<106 μM). Since taurine has anti-oxidant activity, children with ASD with low taurine concentrations will be examined for abnormal mitochondrial function. Our data imply that taurine may be a valid biomarker in a subgroup of ASD.
- Published
- 2017
- Full Text
- View/download PDF
47. Detection and Quantification of the Fragile X Mental Retardation Protein 1 (FMRP).
- Author
-
LaFauci G, Adayev T, Kascsak R, and Brown WT
- Abstract
The final product of FMR1 gene transcription, Fragile X Mental Retardation Protein 1 (FMRP), is an RNA binding protein that acts as a repressor of translation. FMRP is expressed in several tissues and plays important roles in neurogenesis, synaptic plasticity, and ovarian functions and has been implicated in a number of neuropsychological disorders. The loss of FMRP causes Fragile X Syndrome (FXS). In most cases, FXS is due to large expansions of a CGG repeat in FMR1 -normally containing 6-54 repeats-to over 200 CGGs and identified as full mutation (FM). Hypermethylation of the repeat induces FMR1 silencing and lack of FMRP expression in FM male. Mosaic FM males express low levels of FMRP and present a less severe phenotype that inversely correlates with FMRP levels. Carriers of pre-mutations (55-200 CGG) show increased mRNA, and normal to reduced FMRP levels. Alternative splicing of FMR1 mRNA results in 24 FMRP predicted isoforms whose expression are tissues and developmentally regulated. Here, we summarize the approaches used by several laboratories including our own to (a) detect and estimate the amount of FMRP in different tissues, developmental stages and various pathologies; and (b) to accurately quantifying FMRP for a direct diagnosis of FXS in adults and newborns., Competing Interests: On 14 January 2014, the patent “System and Method for Quantifying Fragile X Mental 1 Protein in tissue and blood samples” (United States Patent # 8628934) was issued. The assignee for the patent is the Research Foundation for Mental Hygiene, Inc. The inventors are Giuseppe LaFauci, Richard Kascsak and W. Ted Brown. G.L., R.K., and W.T.B. declare no conflict of interest.
- Published
- 2016
- Full Text
- View/download PDF
48. Retraction Note: Alteration of astrocytes and Wnt/β-catenin signaling in the frontal cortex of autistic subjects.
- Author
-
Cao F, Yin A, Wen G, Sheikh AM, Tauqeer Z, Malik M, Nagori A, Schirripa M, Schirripa F, Merz G, Feng S, Brown WT, and Li X
- Published
- 2016
- Full Text
- View/download PDF
49. DNA Methylation Profiling at Single-Base Resolution Reveals Gestational Folic Acid Supplementation Influences the Epigenome of Mouse Offspring Cerebellum.
- Author
-
Barua S, Kuizon S, Brown WT, and Junaid MA
- Abstract
It is becoming increasingly more evident that lifestyle, environmental factors, and maternal nutrition during gestation can influence the epigenome of the developing fetus and thus modulate the physiological outcome. Variations in the intake of maternal nutrients affecting one-carbon metabolism may influence brain development and exert long-term effects on the health of the progeny. In this study, we investigated whether supplementation with high maternal folic acid during gestation alters DNA methylation and gene expression in the cerebellum of mouse offspring. We used reduced representation bisulfite sequencing to analyze the DNA methylation profile at the single-base resolution level. The genome-wide DNA methylation analysis revealed that supplementation with higher maternal folic acid resulted in distinct methylation patterns (P < 0.05) of CpG and non-CpG sites in the cerebellum of offspring. Such variations of methylation and gene expression in the cerebellum of offspring were highly sex-specific, including several genes of the neuronal pathways. These findings demonstrate that alterations in the level of maternal folic acid during gestation can influence methylation and gene expression in the cerebellum of offspring. Such changes in the offspring epigenome may alter neurodevelopment and influence the functional outcome of neurologic and psychiatric diseases.
- Published
- 2016
- Full Text
- View/download PDF
50. High Gestational Folic Acid Supplementation Alters Expression of Imprinted and Candidate Autism Susceptibility Genes in a sex-Specific Manner in Mouse Offspring.
- Author
-
Barua S, Kuizon S, Brown WT, and Junaid MA
- Subjects
- Animals, Cerebral Cortex embryology, Cerebral Cortex metabolism, DNA Methylation, Dietary Supplements, Female, Folic Acid administration & dosage, Genetic Predisposition to Disease, Male, Mice, Mice, Inbred C57BL, Pregnancy, Sex Factors, Vitamin B Complex administration & dosage, Autistic Disorder genetics, Cerebral Cortex drug effects, Folic Acid pharmacology, Genomic Imprinting, Prenatal Exposure Delayed Effects genetics, Prenatal Nutritional Physiological Phenomena genetics, Vitamin B Complex pharmacology
- Abstract
Maternal nutrients play critical roles in modulating epigenetic events and exert long-term influences on the progeny's health. Folic acid (FA) supplementation during pregnancy has decreased the incidence of neural tube defects in newborns, but the influence of high doses of maternal FA supplementation on infants' brain development is unclear. The present study was aimed at investigating the effects of a high dose of gestational FA on the expression of genes in the cerebral hemispheres (CHs) of 1-day-old pups. One week prior to mating and throughout the entire period of gestation, female C57BL/6J mice were fed a diet, containing FA at either 2 mg/kg (control diet (CD)) or 20 mg/kg (high maternal folic acid (HMFA)). At postnatal day 1, pups from different dams were sacrificed and CH tissues were collected. Quantitative RT-PCR and Western blot analysis confirmed sex-specific alterations in the expression of several genes that modulate various cellular functions (P < 0.05) in pups from the HMFA group. Genomic DNA methylation analysis showed no difference in the level of overall methylation in pups from the HMFA group. These findings demonstrate that HMFA supplementation alters offsprings' CH gene expression in a sex-specific manner. These changes may influence infants' brain development.
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.