90 results on '"Fullston, T."'
Search Results
2. Influence of fibroids on cell‐free DNA screening accuracy
- Author
-
Scott, F., primary, Menezes, M., additional, Smet, M. E., additional, Carey, K., additional, Hardy, T., additional, Fullston, T., additional, Rolnik, D. L., additional, and McLennan, A., additional
- Published
- 2022
- Full Text
- View/download PDF
3. Diet-induced paternal obesity in the absence of diabetes diminishes the reproductive health of two subsequent generations of mice
- Author
-
Fullston, T., Palmer, N.O., Owens, J.A., Mitchell, M., Bakos, H.W., and Lane, M.
- Published
- 2012
- Full Text
- View/download PDF
4. Screening and cell-based assessment of mutations in the Aristaless-related homeobox (ARX) gene
- Author
-
Fullston, T, Finnis, M, Hackett, A, Hodgson, B, Brueton, L, Baynam, G, Norman, A, Reish, O, Shoubridge, C, and Gecz, J
- Published
- 2011
- Full Text
- View/download PDF
5. X-Linked Lissencephaly With Absent Corpus Callosum and Abnormal Genitalia: An Evolving Multisystem Syndrome With Severe Congenital Intestinal Diarrhea Disease.
- Author
-
Shoubridge C., Leventer R., Wong F., Nazaretian S., Simpson I., Gecz J., McGillivray G., Fullston T., Coman D., Shoubridge C., Leventer R., Wong F., Nazaretian S., Simpson I., Gecz J., McGillivray G., Fullston T., and Coman D.
- Abstract
X-linked lissencephaly with abnormal genitalia is a rare and devastating syndrome. The authors present an infant with a multisystem phenotype where the intestinal manifestations were as life limiting as the central nervous system features. Severe chronic diarrhea resulted in failure to thrive, dehydration, electrolyte derangements, long-term hospitalization, and prompted transition to palliative care. Other multisystem manifestations included megacolon, colitis, pancreatic insufficiency hypothalamic dysfunction, hypothyroidism, and hypophosphatasia. A novel aristaless-related homeobox gene mutation, c.1136G>T/p.R379L, was identified. This case contributes to the clinical, histological, and molecular understanding of the multisystem nature of this disorder, especially the role of ARX in the development of the enteroendocrine system.Copyright © The Author(s) 2017.
- Published
- 2018
6. Defining the limits of detection for chromosome rearrangements in the preimplantation embryo using next generation sequencing
- Author
-
Cuman, C, primary, Beyer, C E, additional, Brodie, D, additional, Fullston, T, additional, Lin, J I, additional, Willats, E, additional, Zander-Fox, D, additional, and Mullen, J, additional
- Published
- 2018
- Full Text
- View/download PDF
7. X-Linked Lissencephaly With Absent Corpus Callosum and Abnormal Genitalia: An Evolving Multisystem Syndrome With Severe Congenital Intestinal Diarrhea Disease.
- Author
-
Coman, D, Fullston, T, Shoubridge, C, Leventer, R, Wong, F, Nazaretian, S, Simpson, I, Gecz, J, McGillivray, G, Coman, D, Fullston, T, Shoubridge, C, Leventer, R, Wong, F, Nazaretian, S, Simpson, I, Gecz, J, and McGillivray, G
- Abstract
X-linked lissencephaly with abnormal genitalia is a rare and devastating syndrome. The authors present an infant with a multisystem phenotype where the intestinal manifestations were as life limiting as the central nervous system features. Severe chronic diarrhea resulted in failure to thrive, dehydration, electrolyte derangements, long-term hospitalization, and prompted transition to palliative care. Other multisystem manifestations included megacolon, colitis, pancreatic insufficiency hypothalamic dysfunction, hypothyroidism, and hypophosphatasia. A novel aristaless-related homeobox gene mutation, c.1136G>T/p.R379L, was identified. This case contributes to the clinical, histological, and molecular understanding of the multisystem nature of this disorder, especially the role of ARX in the development of the enteroendocrine system.
- Published
- 2017
8. The current state of reproductive biology research in Australia and New Zealand: core themes from the Society for Reproductive Biology Annual Meeting, 2016
- Author
-
Akison, L. K., primary, Andraweera, P. H., additional, Bertoldo, M. J., additional, Brown, H. M., additional, Cuffe, J. S. M., additional, Fullston, T., additional, Holland, O., additional, and Schjenken, J. E., additional
- Published
- 2017
- Full Text
- View/download PDF
9. A review of fundamental principles for animal models of DOHaD research: an Australian perspective
- Author
-
Dickinson, H., primary, Moss, T. J., additional, Gatford, K. L., additional, Moritz, K. M., additional, Akison, L., additional, Fullston, T., additional, Hryciw, D. H., additional, Maloney, C. A., additional, Morris, M. J., additional, Wooldridge, A. L., additional, Schjenken, J. E., additional, Robertson, S. A., additional, Waddell, B. J., additional, Mark, P. J., additional, Wyrwoll, C. S., additional, Ellery, S. J., additional, Thornburg, K. L., additional, Muhlhausler, B. S., additional, and Morrison, J. L., additional
- Published
- 2016
- Full Text
- View/download PDF
10. When two obese parents are worse than one! Impacts on embryo and fetal development
- Author
-
McPherson, N. O., primary, Bell, V. G., additional, Zander-Fox, D. L., additional, Fullston, T., additional, Wu, L. L., additional, Robker, R. L., additional, and Lane, M., additional
- Published
- 2015
- Full Text
- View/download PDF
11. Screening and cell-based assessment of mutations in the Aristaless-related homeobox (ARX) gene
- Author
-
Fullston, T., Finnis, M., Hackett, A., Hodgson, B., Brueton, L., Baynam, G., Norman, A., Reish, O., Shoubridge, C., Gecz, J., Fullston, T., Finnis, M., Hackett, A., Hodgson, B., Brueton, L., Baynam, G., Norman, A., Reish, O., Shoubridge, C., and Gecz, J.
- Abstract
ARX mutations cause a diverse spectrum of human disorders, ranging from severe brain and genital malformations to non-syndromic intellectual disability (ID). ARX is a transcription factor with multiple domains that include four polyalanine (pA) tracts, the first two of which are frequently expanded by mutations. We progressively screened DNA samples from 613 individuals with ID initially for the most frequent ARX mutations (c.304ins(GCG)7‘expansion’ of pA1 and c.429_452dup ‘dup24bp’ of pA2). Five hundred samples without pA1 or pA2 mutations had the entire ARX ORF screened by single stranded polymorphism conformation (SSCP) and/or denaturing high pressure liquid chromatography (dHPLC) analysis. Overall, eight families with six mutations in ARX were identified (1.31%): five duplication mutations in pA2 (0.82%) with three new clinical reports of families with the dup24bp and two duplications larger than the dup24bp mutation discovered (dup27bp, dup33bp); and three point mutations (0.6%), including one novel mutation in the homeodomain (c.1074G>T). Four ultraconserved regions distal to ARX (uc466–469) were also screened in a subset of 94 patients, with three unique nucleotide changes identified in two (uc466, uc467). The subcellular localization of full length ARX proteins was assessed for 11 variants. Protein mislocalization increased as a function of pA2 tract length and phenotypic severity, as has been previously suggested for pA1. Similarly, protein mislocalization of the homeodomain mutations also correlated with clinical severity, suggesting an emerging genotype vs cellular phenotype correlation.
- Published
- 2011
12. Mutations in the nuclear localization sequence of the Aristaless related homeobox; sequestration of mutant ARX with IPO13 disrupts normal subcellular distribution of the transcription factor and retards cell division.
- Author
-
Shoubridge, C., Tan, M.H., Fullston, T., Cloosterman, D., Coman, D., McGillivray, G., Mancini, G.M.S., Kleefstra, T., Gecz, J., Shoubridge, C., Tan, M.H., Fullston, T., Cloosterman, D., Coman, D., McGillivray, G., Mancini, G.M.S., Kleefstra, T., and Gecz, J.
- Abstract
Contains fulltext : 89419.pdf (publisher's version ) (Open Access), BACKGROUND: Aristaless related homeobox (ARX) is a paired-type homeobox gene. ARX function is frequently affected by naturally occurring mutations. Nonsense mutations, polyalanine tract expansions and missense mutations in ARX cause a range of intellectual disability and epilepsy phenotypes with or without additional features including hand dystonia, lissencephaly, autism or dysarthria. Severe malformation phenotypes, such as X-linked lissencephaly with ambiguous genitalia (XLAG), are frequently observed in individuals with protein truncating or missense mutations clustered in the highly conserved paired-type homeodomain. RESULTS: We have identified two novel point mutations in the R379 residue of the ARX homeodomain; c.1135C>A, p.R379S in a patient with infantile spasms and intellectual disability and c.1136G>T, p.R379L in a patient with XLAG. We investigated these and other missense mutations (R332P, R332H, R332C, T333N: associated with XLAG and Proud syndrome) predicted to affect the nuclear localisation sequences (NLS) flanking either end of the ARX homeodomain. The NLS regions are required for correct nuclear import facilitated by Importin 13 (IPO13). We demonstrate that missense mutations in either the N- or C-terminal NLS regions of the homeodomain cause significant disruption to nuclear localisation of the ARX protein in vitro. Surprisingly, none of these mutations abolished the binding of ARX to IPO13. This was confirmed by co-immunoprecipitation and immmuno fluorescence studies. Instead, tagged and endogenous IPO13 remained bound to the mutant ARX proteins, even in the RanGTP rich nuclear environment. We also identify the microtubule protein TUBA1A as a novel interacting protein for ARX and show cells expressing mutant ARX protein accumulate in mitosis, indicating normal cell division may be disrupted. CONCLUSIONS: We show that the most likely, common pathogenic mechanism of the missense mutations in NLS regions of the ARX homeodomain is inadequate accumul
- Published
- 2010
13. Mutations in the nuclear localization sequence of the Aristaless related homeobox; Sequestration of mutant ARX with IPO13 disrupts normal subcellular distribution of the transcription factor and retards cell division
- Author
-
Shoubridge, C. (Cheryl), Tan, M. (May), Fullston, T. (Tod), Cloosterman, D. (Desiree), Coman, D. (David), McGillivray, G. (George), Mancini, G.M.S. (Grazia), Kleefstra, T. (Tjitske), Gecz, J. (Jozef), Shoubridge, C. (Cheryl), Tan, M. (May), Fullston, T. (Tod), Cloosterman, D. (Desiree), Coman, D. (David), McGillivray, G. (George), Mancini, G.M.S. (Grazia), Kleefstra, T. (Tjitske), and Gecz, J. (Jozef)
- Abstract
Background. Aristaless related homeobox (ARX) is a paired-type homeobox gene. ARX function is frequently affected by naturally occurring mutations. Nonsense mutations, polyalanine tract expansions and missense mutations in ARX cause a range of intellectual disability and epilepsy phenotypes with or without additional features including hand dystonia, lissencephaly, autism or dysarthria. Severe malformation phenotypes, such as X-linked lissencephaly with ambiguous genitalia (XLAG), are frequently observed in individuals with protein truncating or missense mutations clustered in the highly conserved paired-type homeodomain. Results. We have identified two novel point mutations in the R379 residue of the ARX homeodomain; c.1135C>A, p.R379S in a patient with infantile spasms and intellectual disability and c.1136G>T, p.R379L in a patient with XLAG. We investigated these and other missense mutations (R332P, R332H, R332C, T333N: associated with XLAG and Proud syndrome) predicted to affect the nuclear localisation sequences (NLS) flanking either end of the ARX homeodomain. The NLS regions are required for correct nuclear import facilitated by Importin 13 (IPO13). We demonstrate that missense mutations in either the N- or C-terminal NLS regions of the homeodomain cause significant disruption to nuclear
- Published
- 2010
- Full Text
- View/download PDF
14. Mutations in the nuclear localization sequence of the Aristaless related homeobox; sequestration of mutant ARX with IPO13 disrupts normal subcellular distribution of the transcription factor and retards cell division
- Author
-
Shoubridge, C, Tan, MH, Fullston, T, Cloosterman, D, Coman, D, McGillivray, G, Verheijen - Mancini, Grazia, Kleefstra, T, Gecz, J, Shoubridge, C, Tan, MH, Fullston, T, Cloosterman, D, Coman, D, McGillivray, G, Verheijen - Mancini, Grazia, Kleefstra, T, and Gecz, J
- Published
- 2010
15. A systematic, large-scale resequencing screen of X-chromosome coding exons in mental retardation.
- Author
-
Tarpey, P.S., Smith, R., Pleasance, E., Whibley, A., Edkins, S., Hardy, C., O'Meara, S., Latimer, C., Dicks, E., Menzies, A., Stephens, P., Blow, M., Greenman, C., Xue, Y., Tyler-Smith, C., Thompson, D., Gray, K., Andrews, J., Barthorpe, S., Buck, G., Cole, J., Dunmore, R., Jones, D., Maddison, M., Mironenko, T., Turner, R., Turrell, K., Varian, J., West, S., Widaa, S., Wray, P., Teague, J., Butler, A., Jenkinson, A., Jia, M., Richardson, D., Shepherd, R., Wooster, R., Tejada, M.I., Martinez, F., Carvill, G., Goliath, R., Brouwer, A.P.M. de, Bokhoven, H. van, Esch, H. van, Chelly, J., Raynaud, M., Ropers, H.H., Abidi, F.E., Srivastava, A.K., Cox, J., Luo, Y., Mallya, U., Moon, J., Parnau, J., Mohammed, S., Tolmie, J.L., Shoubridge, C., Corbett, M., Gardner, A., Haan, E., Rujirabanjerd, S., Shaw, M.A., Vandeleur, L., Fullston, T., Easton, D.F., Boyle, J., Partington, M., Hackett, A., Field, M., Skinner, C., Stevenson, R.E., Bobrow, M., Turner, G., Schwartz, C.E., Gecz, J., Raymond, F.L., Futreal, P.A., Stratton, M.R., Tarpey, P.S., Smith, R., Pleasance, E., Whibley, A., Edkins, S., Hardy, C., O'Meara, S., Latimer, C., Dicks, E., Menzies, A., Stephens, P., Blow, M., Greenman, C., Xue, Y., Tyler-Smith, C., Thompson, D., Gray, K., Andrews, J., Barthorpe, S., Buck, G., Cole, J., Dunmore, R., Jones, D., Maddison, M., Mironenko, T., Turner, R., Turrell, K., Varian, J., West, S., Widaa, S., Wray, P., Teague, J., Butler, A., Jenkinson, A., Jia, M., Richardson, D., Shepherd, R., Wooster, R., Tejada, M.I., Martinez, F., Carvill, G., Goliath, R., Brouwer, A.P.M. de, Bokhoven, H. van, Esch, H. van, Chelly, J., Raynaud, M., Ropers, H.H., Abidi, F.E., Srivastava, A.K., Cox, J., Luo, Y., Mallya, U., Moon, J., Parnau, J., Mohammed, S., Tolmie, J.L., Shoubridge, C., Corbett, M., Gardner, A., Haan, E., Rujirabanjerd, S., Shaw, M.A., Vandeleur, L., Fullston, T., Easton, D.F., Boyle, J., Partington, M., Hackett, A., Field, M., Skinner, C., Stevenson, R.E., Bobrow, M., Turner, G., Schwartz, C.E., Gecz, J., Raymond, F.L., Futreal, P.A., and Stratton, M.R.
- Abstract
Contains fulltext : 79687.pdf (publisher's version ) (Closed access), Large-scale systematic resequencing has been proposed as the key future strategy for the discovery of rare, disease-causing sequence variants across the spectrum of human complex disease. We have sequenced the coding exons of the X chromosome in 208 families with X-linked mental retardation (XLMR), the largest direct screen for constitutional disease-causing mutations thus far reported. The screen has discovered nine genes implicated in XLMR, including SYP, ZNF711 and CASK reported here, confirming the power of this strategy. The study has, however, also highlighted issues confronting whole-genome sequencing screens, including the observation that loss of function of 1% or more of X-chromosome genes is compatible with apparently normal existence.
- Published
- 2009
16. Mutation frequencies of X-linked mental retardation genes in families from the EuroMRX consortium.
- Author
-
Brouwer, A.P.M. de, Yntema, H.G., Kleefstra, T., Lugtenberg, D., Oudakker, A.R., Vries, B. de, Bokhoven, J.H.L.M. van, Esch, H. van, Frints, S.G., Froyen, G., Fryns, J.P., Raynaud, M., Moizard, M.P., Ronce, N., Bensalem, A., Moraine, C., Poirier, K., Castelnau, L., Saillour, Y., Bienvenu, T., Beldjord, C., Portes, V. des, Chelly, J., Turner, G., Fullston, T., Gecz, J., Kuss, A.W., Tzschach, A., Jensen, L.R., Lenzner, S., Kalscheuer, V.M.M., Ropers, H.H., Hamel, B.C.J., Brouwer, A.P.M. de, Yntema, H.G., Kleefstra, T., Lugtenberg, D., Oudakker, A.R., Vries, B. de, Bokhoven, J.H.L.M. van, Esch, H. van, Frints, S.G., Froyen, G., Fryns, J.P., Raynaud, M., Moizard, M.P., Ronce, N., Bensalem, A., Moraine, C., Poirier, K., Castelnau, L., Saillour, Y., Bienvenu, T., Beldjord, C., Portes, V. des, Chelly, J., Turner, G., Fullston, T., Gecz, J., Kuss, A.W., Tzschach, A., Jensen, L.R., Lenzner, S., Kalscheuer, V.M.M., Ropers, H.H., and Hamel, B.C.J.
- Abstract
Contains fulltext : 53461.pdf (publisher's version ) (Closed access), The EuroMRX family cohort consists of about 400 families with non-syndromic and 200 families with syndromic X-linked mental retardation (XLMR). After exclusion of Fragile X (Fra X) syndrome, probands from these families were tested for mutations in the coding sequence of 90 known and candidate XLMR genes. In total, 73 causative mutations were identified in 21 genes. For 42% of the families with obligate female carriers, the mental retardation phenotype could be explained by a mutation. There was no difference between families with (lod score >2) or without (lod score <2) significant linkage to the X chromosome. For families with two to five affected brothers (brother pair=BP families) only 17% of the MR could be explained. This is significantly lower (P=0.0067) than in families with obligate carrier females and indicates that the MR in about 40% (17/42) of the BP families is due to a single genetic defect on the X chromosome. The mutation frequency of XLMR genes in BP families is lower than can be expected on basis of the male to female ratio of patients with MR or observed recurrence risks. This might be explained by genetic risk factors on the X chromosome, resulting in a more complex etiology in a substantial portion of XLMR patients. The EuroMRX effort is the first attempt to unravel the molecular basis of cognitive dysfunction by large-scale approaches in a large patient cohort. Our results show that it is now possible to identify 42% of the genetic defects in non-syndromic and syndromic XLMR families with obligate female carriers.
- Published
- 2007
17. 103. THE EFFECT OF PATERNAL OBESITY IN MICE ON REPRODUCTIVE AND METABOLIC FITNESS OF F1 MALE OFFSPRING
- Author
-
Mitchell, M., primary, Fullston, T., additional, Palmer, N. O., additional, Bakos, H. W., additional, Owens, J. A., additional, and Lane, M., additional
- Published
- 2010
- Full Text
- View/download PDF
18. 181. SIRT6 PROTEIN IS REDUCED IN TESTES AND SPERM FROM OBESE MALE MICE
- Author
-
Palmer, N. O., primary, Fullston, T., additional, Mitchell, M., additional, and Lane, M., additional
- Published
- 2010
- Full Text
- View/download PDF
19. 131. SIRT3 IN OVARIAN CELLS IS ALTERED BY MATERNAL AGE AND OVARIAN RESERVE
- Author
-
Pacella, L., primary, Zander-Fox, D., additional, Hussein, T., additional, Fullston, T., additional, and Lane, M., additional
- Published
- 2010
- Full Text
- View/download PDF
20. 130. MICROARRAY ANALYSIS OF FOETAL MOUSE BRAIN FOLLOWING INDUCTION OF MITOCHONDRIAL DYSFUNCTION DURING PRE-IMPLANTATION EMBRYO DEVELOPMENT
- Author
-
Fullston, T., primary, Mitchell, M., additional, Wakefield, S., additional, Filby, A., additional, and Lane, M., additional
- Published
- 2009
- Full Text
- View/download PDF
21. A review of fundamental principles for animal models of DOHaD research: an Australian perspective
- Author
-
Margaret J. Morris, Tod Fullston, Timothy J. M. Moss, Lisa K. Akison, Brendan J. Waddell, Christopher A. Maloney, John E. Schjenken, Kent L. Thornburg, Deanne H. Hryciw, Sarah A. Robertson, Amy L. Wooldridge, Caitlin S. Wyrwoll, Peter J. Mark, Beverly S. Muhlhausler, Kathryn L. Gatford, Stacey J. Ellery, Karen M. Moritz, Janna L. Morrison, Hayley Dickinson, Dickinson, H, Moss, TJ, Gatford, KL, Moritz, KM, Akison, L, Fullston, T, Hryciw, DH, Maloney, CA, Morris, MJ, Wooldridge, AL, Schjenken, JE, Robertson, SA, Waddell, BJ, Mark, PJ, Wyrwoll, CS, Ellery, SJ, Thornburg, KL, Muhlhausler, BS, and Morrison, JL
- Subjects
0301 basic medicine ,Developmental stage ,Perspective (graphical) ,Medicine (miscellaneous) ,Physiology ,Biology ,programming ,developmental stage ,03 medical and health sciences ,030104 developmental biology ,outcome/system ,Relevance (law) ,Engineering ethics ,developmental origins of health and disease ,Animal testing ,Developmental programming - Abstract
Epidemiology formed the basis of ‘the Barker hypothesis’, the concept of ‘developmental programming’ and today’s discipline of the Developmental Origins of Health and Disease (DOHaD). Animal experimentation provided proof of the underlying concepts, and continues to generate knowledge of underlying mechanisms. Interventions in humans, based on DOHaD principles, will be informed by experiments in animals. As knowledge in this discipline has accumulated, from studies of humans and other animals, the complexity of interactions between genome, environment and epigenetics, has been revealed. The vast nature of programming stimuli and breadth of effects is becoming known. As a result of our accumulating knowledge we now appreciate the impact of many variables that contribute to programmed outcomes. To guide further animal research in this field, the Australia and New Zealand DOHaD society (ANZ DOHaD) Animals Models of DOHaD Research Working Group convened at the 2nd Annual ANZ DOHaD Congress in Melbourne, Australia in April 2015. This review summarizes the contributions of animal research to the understanding of DOHaD, and makes recommendations for the design and conduct of animal experiments to maximize relevance, reproducibility and translation of knowledge into improving health and well-being. Refereed/Peer-reviewed
- Published
- 2016
22. Maternal high-fat diet changes DNA methylation in the early embryo by disrupting the TCA cycle intermediary alpha ketoglutarate.
- Author
-
Penn A, McPherson N, Fullston T, Arman B, and Zander-Fox D
- Subjects
- Animals, Female, Humans, Mice, Pregnancy, 5-Methylcytosine metabolism, Cytosine metabolism, Diet, High-Fat, Ketoglutaric Acids pharmacology, Zygote metabolism, DNA Methylation, Obesity, Maternal metabolism
- Abstract
In Brief: Maternal obesity can impair metabolism in the embryo and the resulting offspring. This study shows that metabolic disruptions through α-ketoglutarate may link altered metabolism with epigenetic changes in embryos., Abstract: Maternal obesity can impair offspring metabolic health; however, the precise mechanism underpinning programming is unknown. Ten-Eleven translocase (TET) enzymes demethylate DNA using the TCA cycle intermediary α-ketoglutarate and may be involved in programming offspring health. Whether TETs are disrupted by maternal obesity is unknown. Five to six week-old C57Bl/6 female mice were fed a control diet (CD; 6% fat, n = 175) or a high-fat diet (HFD; 21% fat, n = 158) for 6 weeks. After superovulation, oocytes were collected for metabolic assessment, or females were mated and zygotes were cultured for embryo development, fetal growth, and assessment of global DNA methylation (5-methylcytosine (5mC), 5-hydroxymethylcytosine (5hmC), 5-formylcytosine (5fC), and 5-carboxycytosine (5caC)) in the two-cell embryo. Zygotes collected from superovulated CBAF1 females were cultured in media containing α-ketoglutarate (0, 1.4, 3.5, or 14.0 mM) or with 2-hydroxyglutarate (2HG) (0 or 20 mM), a competitive inhibitor of α-ketoglutarate, with methylation and blastocyst differentiation assessed. After HFD, oocytes showed increased pyruvate oxidation and intracellular ROS, with no changes in Tet3 expression, while two-cell embryo global 5hmC DNA methylation was reduced and 5fC increased. Embryos cultured with 1.4 mM α-ketoglutarate had decreased two-cell 5mC, while 14.0 mM α-ketoglutarate increased the 5hmC:5mC ratio. In contrast, supplementation with 20 mM 2HG increased 5mC and decreased 5fC:5mC and 5caC:5mC ratios. α-ketoglutarate up to 3.5 mM did not alter embryo development, while culturing in 14.0 mM α-ketoglutarate blocked development at the two-cell. Culture with 2HG delayed embryo development past the four-cell and decreased blastocyst total cell number. In conclusion, disruptions in metabolic intermediates in the preimplantation embryo may provide a link between maternal obesity and programming offspring for ill health.
- Published
- 2023
- Full Text
- View/download PDF
23. High-fat Diet Alters Male Seminal Plasma Composition to Impair Female Immune Adaptation for Pregnancy in Mice.
- Author
-
Schjenken JE, Moldenhauer LM, Sharkey DJ, Chan HY, Chin PY, Fullston T, McPherson NO, and Robertson SA
- Subjects
- Adiposity, Animals, Body Composition, Cytokines metabolism, Diet, High-Fat, Female, Lymphocyte Subsets, Male, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Nude, Phenotype, Pregnancy, Pregnancy, Animal, Protein Isoforms, Reproduction, Semen physiology, Spermatozoa physiology, T-Lymphocytes cytology, T-Lymphocytes, Regulatory immunology, Transforming Growth Factor beta1 metabolism, Uterus pathology, Plasma metabolism, Semen metabolism
- Abstract
Paternal experiences and exposures before conception can influence fetal development and offspring phenotype. The composition of seminal plasma contributes to paternal programming effects through modulating the female reproductive tract immune response after mating. To investigate whether paternal obesity affects seminal plasma immune-regulatory activity, C57Bl/6 male mice were fed an obesogenic high-fat diet (HFD) or control diet (CD) for 14 weeks. Although HFD consumption caused only minor changes to parameters of sperm quality, the volume of seminal vesicle fluid secretions was increased by 65%, and the concentrations and total content of immune-regulatory TGF-β isoforms were decreased by 75% to 80% and 43% to 55%, respectively. Mating with BALB/c females revealed differences in the strength and properties of the postmating immune response elicited. Transcriptional analysis showed >300 inflammatory genes were similarly regulated in the uterine endometrium by mating independently of paternal diet, and 13 were dysregulated by HFD-fed compared with CD-fed males. Seminal vesicle fluid factors reduced in HFD-fed males, including TGF-β1, IL-10, and TNF, were among the predicted upstream regulators of differentially regulated genes. Additionally, the T-cell response induced by mating with CD-fed males was blunted after mating with HFD-fed males, with 27% fewer CD4+ T cells, 26% fewer FOXP3+CD4+ regulatory T cells (Treg) cells, and 19% fewer CTLA4+ Treg cells, particularly within the NRP1+ thymic Treg cell population. These findings demonstrate that an obesogenic HFD alters the composition of seminal vesicle fluid and impairs seminal plasma capacity to elicit a favorable pro-tolerogenic immune response in females at conception., (© The Author(s) 2021. Published by Oxford University Press on behalf of the Endocrine Society. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2021
- Full Text
- View/download PDF
24. It takes a community to conceive: an analysis of the scope, nature and accuracy of online sources of health information for couples trying to conceive.
- Author
-
Kedzior SGE, Bianco-Miotto T, Breen J, Diener KR, Donnelley M, Dunning KR, Penno MAS, Schjenken JE, Sharkey DJ, Hodyl NA, Fullston T, Gardiner M, Brown HM, and Rumbold AR
- Abstract
This study examined the nature and accuracy of information available across online platforms for couples trying to conceive. A consumer simulation-based investigation of English websites and social media (Facebook, Twitter, Instagram) was undertaken using common search terms identified in a pilot study. Claims about fertility and pregnancy health were then extracted from the results and analysed thematically. The accuracy of each claim was assessed independently by six fertility and conception experts, rated on a scale of 1 (not factual) to 4 (highly factual), with scores collated to produce a median rating. Claims with a median score < 3 were classified as inaccurate. The use of the terms 'trying to conceive' and '#TTC' were common identifiers on online platforms. Claims were extracted predominantly from websites ( n = 89) rather than social media, with Twitter and Instagram comprising commercial elements and Facebook focused on community-based support. Thematic analysis revealed three major themes among the claims across all platforms: conception behaviour and monitoring, lifestyle and exposures, and medical. Fact-checking by the experts revealed that 40% of the information assessed was inaccurate, and that inaccuracies were more likely to be present in the conception behaviour and monitoring advice, the topics most amenable to modification. Since online information is a readily accessible and commonly utilized resource, there is opportunity for improved dissemination of evidence-based material to reach interested couples. Further cross-disciplinary and consumer-based research, such as a user survey, is required to understand how best to provide the 'trying to conceive' community with accurate information., (Crown Copyright © 2019 Published by Elsevier Ltd.)
- Published
- 2019
- Full Text
- View/download PDF
25. Dietary Micronutrient Supplementation for 12 Days in Obese Male Mice Restores Sperm Oxidative Stress.
- Author
-
McPherson NO, Shehadeh H, Fullston T, Zander-Fox DL, and Lane M
- Subjects
- Animals, Diet, High-Fat, Disease Models, Animal, Embryonic Development drug effects, Infertility, Male etiology, Infertility, Male therapy, Male, Mice, Mice, Inbred C57BL, Mice, Obese, Obesity complications, Obesity therapy, Sperm Motility drug effects, Spermatozoa drug effects, Dietary Supplements, Infertility, Male metabolism, Micronutrients pharmacology, Obesity metabolism, Oxidative Stress drug effects
- Abstract
Male obesity, which often co-presents with micronutrient deficiencies, is associated with sub-fertility. Here we investigate whether short-term dietary supplementation of micronutrients (zinc, selenium, lycopene, vitamins E and C, folic acid, and green tea extract) to obese mice for 12 days (designed to span the epididymal transit) could improve sperm quality and fetal outcomes. Five-week-old C57BL6 males were fed a control diet (CD, n = 24) or high fat diet (HFD, n = 24) for 10 weeks before allocation to the 12-day intervention of maintaining their original diets (CD, n = 12, HFD n = 12) or with micronutrient supplementation (CD + S, n = 12, HFD + S, n = 12). Measures of sperm quality (motility, morphology, capacitation, binding), sperm oxidative stress (DCFDA, MSR, and 8OHdG), early embryo development (2-cell cleavage, 8OHdG), and fetal outcomes were assessed. HFD + S males had reduced sperm intracellular reactive oxygen species (ROS) concentrations and 8OHdG lesions, which resulted in reduced 8OHdG lesions in the male pronucleus, increased 2-cell cleavage rates, and partial restoration of fetal weight similar to controls. Sub-fertility associated with male obesity may be restored with very short-term micronutrient supplementation that targets the timing of the transit of sperm through the epididymis, which is the developmental window where sperm are the most susceptible to oxidative damage.
- Published
- 2019
- Full Text
- View/download PDF
26. X-Linked Lissencephaly With Absent Corpus Callosum and Abnormal Genitalia: An Evolving Multisystem Syndrome With Severe Congenital Intestinal Diarrhea Disease.
- Author
-
Coman D, Fullston T, Shoubridge C, Leventer R, Wong F, Nazaretian S, Simpson I, Gecz J, and McGillivray G
- Abstract
X-linked lissencephaly with abnormal genitalia is a rare and devastating syndrome. The authors present an infant with a multisystem phenotype where the intestinal manifestations were as life limiting as the central nervous system features. Severe chronic diarrhea resulted in failure to thrive, dehydration, electrolyte derangements, long-term hospitalization, and prompted transition to palliative care. Other multisystem manifestations included megacolon, colitis, pancreatic insufficiency hypothalamic dysfunction, hypothyroidism, and hypophosphatasia. A novel aristaless-related homeobox gene mutation, c.1136G>T/p.R379L, was identified. This case contributes to the clinical, histological, and molecular understanding of the multisystem nature of this disorder, especially the role of ARX in the development of the enteroendocrine system., Competing Interests: Declaration of Conflicting Interests: The authors declared no potential conflict of interest with respect to the research, authorship, and/or publication of this article.
- Published
- 2017
- Full Text
- View/download PDF
27. MicroRNA regulation of immune events at conception.
- Author
-
Robertson SA, Zhang B, Chan H, Sharkey DJ, Barry SC, Fullston T, and Schjenken JE
- Subjects
- Animals, Female, Humans, Mice, Pregnancy, Fertility immunology, Immune Tolerance, MicroRNAs immunology, Placentation immunology, T-Lymphocytes, Regulatory immunology
- Abstract
The reproductive tract environment at conception programs the developmental trajectory of the embryo, sets the course of pregnancy, and impacts offspring phenotype and health. Despite the fundamental importance of this stage of reproduction, the rate-limiting regulatory mechanisms operating locally to control fertility and fecundity are incompletely understood. Emerging studies highlight roles for microRNAs (miRNAs) in regulating reproductive and developmental processes and in modulating the quality and strength of the female immune response. Since endometrial receptivity and robust placentation require specific adaptation of the immune response, we hypothesize that miRNAs participate in establishing pregnancy through effects on key gene networks in immune cells. Our recent studies investigated miRNAs that are induced in the peri-conception environment, focusing on miRNAs that have immune-regulatory roles-particularly miR-223, miR-155, and miR-146a. Genetic mouse models deficient in individual miRNAs are proving informative in defining roles for these miRNAs in the generation and stabilization of regulatory T cells (Treg cells) that confer adaptive immune tolerance. Overlapping and redundant functions between miRNAs that target multiple genes, combined with multiple miRNAs targeting individual genes, indicate complex and sensitive regulatory networks. Although to date most data on miRNA regulation of reproductive events are from mice, conserved functions of miRNAs across species imply similar biological pathways operate in all mammals. Understanding the regulation and roles of miRNAs in the peri-conception immune response will advance our knowledge of how environmental determinants act at conception, and could have practical applications for animal breeding as well as human fertility., (© 2017 Wiley Periodicals, Inc.)
- Published
- 2017
- Full Text
- View/download PDF
28. The most common vices of men can damage fertility and the health of the next generation.
- Author
-
Fullston T, McPherson NO, Zander-Fox D, and Lane M
- Subjects
- Animals, Epigenesis, Genetic, Humans, Male, Alcohol Drinking adverse effects, Infertility, Male etiology, Overweight complications, Paternal Behavior, Smoking adverse effects
- Abstract
Animal and human studies demonstrate that acquired paternal traits can impair both a male's fertility and the health of his offspring, including advanced age, smoking, stress, trauma, under-nutrition, infection, toxin exposure, and obesity. Many of these factors lead to similar changes to neurological, behavioural, and/or metabolic functioning in offspring. The molecular mechanisms that both respond to the paternal environment and act to transmit traits to offspring are beginning to emerge. This review focuses on three vices of men (alcohol consumption, overweight/obesity, and tobacco smoking) that damage fertility and pose risks to offspring health. These vices are not only the three most prevalent but are also leading risk factors for death and disability adjusted life years (DALYs) worldwide. Moreover, given that these vices are predominantly self-inflicted, interventions aimed at mitigating their consequences are readily identified., (© 2017 Society for Endocrinology.)
- Published
- 2017
- Full Text
- View/download PDF
29. An Exercise-Only Intervention in Obese Fathers Restores Glucose and Insulin Regulation in Conjunction with the Rescue of Pancreatic Islet Cell Morphology and MicroRNA Expression in Male Offspring.
- Author
-
McPherson NO, Lane M, Sandeman L, Owens JA, and Fullston T
- Subjects
- Animals, Blood Glucose physiology, Cellular Reprogramming, Cellular Reprogramming Techniques methods, Diet, High-Fat adverse effects, Fathers, Fatty Acids, Nonesterified blood, Female, Insulin physiology, Islets of Langerhans physiopathology, Male, Mice, Mice, Inbred C57BL, Obesity genetics, Paternal Inheritance, Islets of Langerhans cytology, MicroRNAs metabolism, Obesity physiopathology, Obesity therapy, Physical Conditioning, Animal methods
- Abstract
Paternal obesity programs metabolic syndrome in offspring. Low-impact exercise in obese males improves the metabolic health of female offspring, however whether this occurred in male offspring remained unknown. C57BL/6NHsd (Harlan) mice were fed a control diet (CD; 6% fat, n = 7) or a high-fat diet (HFD; 21% fat, n = 16) for 18 weeks. After 9 weeks, HFD-fed mice either remained sedentary (HH, n = 8) or undertook low-moderate exercise (HE, n = 8) for another 9 weeks. Male offspring were assessed for glucose/insulin tolerance, body composition, plasma lipids, pancreatic islet cell morphology and microRNA expression. Founder HH induced glucose intolerance, insulin insensitivity, and hyperlipidaemia in male offspring (p < 0.05). Metabolic health was fully restored in male offspring by founder exercise to control levels. Founder HH reduced pancreatic β-cell area and islet cell size in male offspring, and altered the expression of 13 pancreatic microRNAs (p < 0.05). Founder HE led to partial restoration of pancreatic islet cell morphology and the expression of two pancreatic microRNAs (let7d-5p, 194-5p) in male offspring. Founder HE reduced male offspring adiposity, increased muscle mass, reduced plasma free fatty acids (FFAs), and further altered pancreatic microRNAs (35 vs. HH; 32 vs. CD) (p < 0.05). Low-impact exercise in obese fathers prior to conception, without dietary change, may be a viable intervention strategy to reduce the illeffects of obesity-induced paternal programming in male offspring.
- Published
- 2017
- Full Text
- View/download PDF
30. Sperm microRNA Content Is Altered in a Mouse Model of Male Obesity, but the Same Suite of microRNAs Are Not Altered in Offspring's Sperm.
- Author
-
Fullston T, Ohlsson-Teague EM, Print CG, Sandeman LY, and Lane M
- Subjects
- Adiposity genetics, Animals, Diet, High-Fat adverse effects, Disease Models, Animal, Epigenesis, Genetic genetics, Fathers, Fertilization genetics, Male, Mice, Mice, Inbred C57BL, Obesity metabolism, Reproduction genetics, Transcriptome genetics, MicroRNAs genetics, Obesity genetics, Spermatozoa metabolism
- Abstract
The prevalence of obesity is increasing worldwide and has tripled in men of reproductive age since the 1970s. Concerningly, obesity is not only comorbid with other chronic diseases, but there is mounting evidence that it increases the non-communicable disease load in their children (eg mortality, obesity, autism). Animal studies have demonstrated that paternal obesity increases the risk of metabolic (eg glucose metabolism defects, obesity) and reproductive disorders in offspring. Epigenetic changes within sperm are clear mechanistic candidates that are associated with both changes to the father's environment and offspring phenotype. Specifically there is emerging evidence that a father's sperm microRNA content both responds to paternal environmental cues and alters the gene expression profile and subsequent development of the early embryo. We used a mouse model of high fat diet (HFD) induced obesity to investigate whether male obesity could modulate sperm microRNA content. We also investigated whether this alteration to a father's sperm microRNA content lead to a similar change in the sperm of male offspring. Our investigations were initially guided by a Taqman PCR array, which indicated the differential abundance of 28 sperm borne microRNAs in HFD mice. qPCR confirmation in a much larger cohort of founder males demonstrated that 13 of these microRNAs were differentially abundant (11 up-regulated; 2 down-regulated) due to HFD feeding. Despite metabolic and reproductive phenotypes also being observed in grand-offspring fathered via the male offspring lineage, there was no evidence that any of the 13 microRNAs were also dysregulated in male offspring sperm. This was presumably due to the variation seen within both groups of offspring and suggests other mechanisms might act between offspring and grand-offspring. Thus 13 sperm borne microRNAs are modulated by a father's HFD and the presumed transfer of this altered microRNA payload to the embryo at fertilisation potentially acts to alter the embryonic molecular makeup post-fertilisation, altering its growth trajectory, ultimately affecting adult offspring phenotype and may contribute to paternal programming., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2016
- Full Text
- View/download PDF
31. Paternal under-nutrition programs metabolic syndrome in offspring which can be reversed by antioxidant/vitamin food fortification in fathers.
- Author
-
McPherson NO, Fullston T, Kang WX, Sandeman LY, Corbett MA, Owens JA, and Lane M
- Subjects
- Adiposity, Animals, Antioxidants administration & dosage, Body Composition, Embryonic Development, Female, Food, Fortified, Founder Effect, Infertility, Male etiology, Infertility, Male genetics, Insulin blood, Leptin blood, Lipids blood, Male, Malnutrition complications, Metabolic Syndrome blood, Metabolic Syndrome etiology, Mice, Inbred C57BL, Paternal Inheritance, Reactive Oxygen Species metabolism, Sperm Count, Sperm Motility, Malnutrition genetics, Metabolic Syndrome genetics
- Abstract
There is an ever increasing body of evidence that demonstrates that paternal over-nutrition prior to conception programs impaired metabolic health in offspring. Here we examined whether paternal under-nutrition can also program impaired health in offspring and if any detrimental health outcomes in offspring could be prevented by micronutrient supplementation (vitamins and antioxidants). We discovered that restricting the food intake of male rodents reduced their body weight, fertility, increased sperm oxidative DNA lesions and reduced global sperm methylation. Under-nourished males then sired offspring with reduced postnatal weight and growth but somewhat paradoxically increased adiposity and dyslipidaemia, despite being fed standard chow. Paternal vitamin/antioxidant food fortification during under-nutrition not only normalised founder oxidative sperm DNA lesions but also prevented early growth restriction, fat accumulation and dyslipidaemia in offspring. This demonstrates that paternal under-nutrition reduces postnatal growth but increases the risk of obesity and metabolic disease in the next generation and that micronutrient supplementation during this period of under-nutrition is capable of restoring offspring metabolic health.
- Published
- 2016
- Full Text
- View/download PDF
32. miRNA Regulation of Immune Tolerance in Early Pregnancy.
- Author
-
Schjenken JE, Zhang B, Chan HY, Sharkey DJ, Fullston T, and Robertson SA
- Subjects
- Animals, Coitus physiology, Female, Humans, Male, Semen immunology, Endometrium immunology, Gene Expression Regulation immunology, Immune Tolerance physiology, MicroRNAs immunology, Pregnancy immunology, T-Lymphocytes, Regulatory immunology
- Abstract
To support embryo implantation, the female reproductive tract must provide a tolerogenic immune environment. Seminal fluid contact at conception contributes to activating the endometrial gene expression and immune cell changes required for robust implantation, influencing not only the quality of the ensuing pregnancy but also the health of offspring. miRNAs are small non-coding RNAs that play important regulatory roles in biological processes, including regulation of the immune environment. miRNAs are known to contribute to gene regulation in pregnancy and are altered in pregnancy pathologies. Recent studies indicate that miRNAs participate in establishing immune tolerance at conception, and may contribute to the regulatory effects of seminal fluid in generating tolerogenic dendritic cells and T regulatory cells. This review highlights those miRNAs implicated in programming immune cells that are critical during the peri-conception period and explores how seminal fluid may regulate female tract miRNA expression following coitus., (© 2016 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)
- Published
- 2016
- Full Text
- View/download PDF
33. When two obese parents are worse than one! Impacts on embryo and fetal development.
- Author
-
McPherson NO, Bell VG, Zander-Fox DL, Fullston T, Wu LL, Robker RL, and Lane M
- Subjects
- Animals, Blastocyst, Cell Count, Diet, High-Fat, Female, Fetal Weight, Male, Mice, Mitochondria, Organ Size, Placenta, Pregnancy, Embryonic Development, Fetal Development, Gene Expression Regulation, Developmental, Maternal Exposure, Obesity, Paternal Exposure
- Abstract
The prevalence of overweight and obesity in reproductive-age adults is increasing worldwide. While the effects of either paternal or maternal obesity on gamete health and subsequent fertility and pregnancy have been reported independently, the combination of having both parents overweight/obese on fecundity and offspring health has received minimal attention. Using a 2 × 2 study design in rodents we established the relative contributions of paternal and maternal obesity on fetal and embryo development and whether combined paternal and maternal obesity had an additive effect. Here, we show that parental obesity reduces fetal and placental weights without altering pregnancy establishment and is not dependent on an in utero exposure to a high-fat diet. Interestingly combined parental obesity seemed to accumulate both the negative influences of paternal and maternal obesity had alone on embryo and fetal health rather than an amplification, manifested as reduced embryo developmental competency, reduced blastocyst cell numbers, impaired mitochondrial function, and alterations to active and repressive embryonic chromatin marks, resulting in aberrant placental gene expression and reduced fetal liver mtDNA copy numbers. Further understanding both the maternal cytoplasmic and paternal genetic interactions during this early developmental time frame will be vital for understanding how developmental programming is regulated and for the proposition of interventions to mitigate their effects., (Copyright © 2015 the American Physiological Society.)
- Published
- 2015
- Full Text
- View/download PDF
34. Female offspring sired by diet induced obese male mice display impaired blastocyst development with molecular alterations to their ovaries, oocytes and cumulus cells.
- Author
-
Fullston T, Shehadeh H, Sandeman LY, Kang WX, Wu LL, Robker RL, McPherson NO, and Lane M
- Subjects
- Animals, Blastocyst metabolism, Cumulus Cells pathology, Embryo, Mammalian cytology, Embryo, Mammalian metabolism, Female, Male, Mice, Mice, Inbred C57BL, Mice, Obese, Obesity etiology, Oocytes pathology, Ovary pathology, Pregnancy, Real-Time Polymerase Chain Reaction, Semen chemistry, Blastocyst pathology, Cumulus Cells metabolism, Diet, High-Fat adverse effects, Obesity physiopathology, Oocytes metabolism, Ovary metabolism
- Abstract
Purpose: To investigate the impacts that a paternal high fat diet (HFD) has on embryology, ovarian/cumulus cell gene expression and COC metabolism from female offspring, using a mouse model., Methods: Founder male mice were either fed a control diet (CD) or a HFD for 12 weeks. The HFD induced obesity but not diabetes, and founder males were then mated to normal weight CD fed female mice. Female offspring were maintained on a CD, super-ovulated, mated and the resultant zygotes were cultured to the blastocyst stage for embryo morphology, blastocyst cell number and apoptosis assessment. Ovaries and cumulus cells from offspring were collected for gene expression analysis of selected genes that maintain chromatin remodeling and endoplasmic reticulum (ER), metabolic and inflammatory homeostasis. Cumulus/oocyte complexes were also investigated for glucose uptake and lipid accumulation., Results: Female offspring sired by obese fathers produced embryos with delayed development and impaired quality, displayed increases in ovarian expression of Glut1, Glut3 and Glut4, and an increase in cumulus cell expression of Glut4. Interestingly their COCs did take up more glucose, but did accumulate more lipid., Conclusions: A paternal HFD is associated with subfertility in female offspring despite the offspring being fed a CD and this subfertility is concomitant with ovarian/cumulus cell molecular alterations and increased lipid accumulation.
- Published
- 2015
- Full Text
- View/download PDF
35. Preconception diet or exercise intervention in obese fathers normalizes sperm microRNA profile and metabolic syndrome in female offspring.
- Author
-
McPherson NO, Owens JA, Fullston T, and Lane M
- Subjects
- Animals, Diet, Female, Fertilization physiology, Infertility, Male genetics, Infertility, Male prevention & control, Male, Metabolic Syndrome metabolism, Mice, Mice, Inbred C57BL, Mice, Obese, Pregnancy, Prenatal Exposure Delayed Effects genetics, Prenatal Exposure Delayed Effects metabolism, Transcriptome, Animal Nutritional Physiological Phenomena, Fathers, Metabolic Syndrome prevention & control, MicroRNAs genetics, Obesity genetics, Physical Conditioning, Animal physiology, Spermatozoa metabolism
- Abstract
Obesity and type 2 diabetes are increasingly prevalent across all demographics. Paternal obesity in humans and rodents can program obesity and impair insulin sensitivity in female offspring. It remains to be determined whether these perturbed offspring phenotypes can be improved through targeted lifestyle interventions in the obese father. Using a mouse model, we demonstrate that diet or exercise interventions for 8 wk (2 rounds of spermatogenesis) in obese founder males restores insulin sensitivity and normalized adiposity in female offspring. Founder diet and/or exercise also normalizes abundance of X-linked sperm microRNAs that target genes regulating cell cycle and apoptosis, pathways central to oocyte and early embryogenesis. Additionally, obesity-associated comorbidities, including inflammation, glucose intolerance, stress, and hypercholesterolemia, were good predictors for sperm microRNA abundance and offspring phenotypes. Interventions aimed at improving paternal metabolic health during specific windows prior to conception can partially normalize aberrant epigenetic signals in sperm and improve the metabolic health of female offspring., (Copyright © 2015 the American Physiological Society.)
- Published
- 2015
- Full Text
- View/download PDF
36. Reduction of Mitochondrial Function by FCCP During Mouse Cleavage Stage Embryo Culture Reduces Birth Weight and Impairs the Metabolic Health of Offspring.
- Author
-
Zander-Fox DL, Fullston T, McPherson NO, Sandeman L, Kang WX, Good SB, Spillane M, and Lane M
- Subjects
- Adenosine Triphosphate genetics, Adenosine Triphosphate metabolism, Adiposity drug effects, Animals, Birth Weight, Embryo Culture Techniques, Embryo Transfer, Embryonic Development drug effects, Female, Gene Expression Regulation, Developmental drug effects, Litter Size, Male, Metalloproteases genetics, Metalloproteases metabolism, Mice, Mitochondria metabolism, Pregnancy, Prenatal Exposure Delayed Effects, Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone toxicity, Cleavage Stage, Ovum drug effects, Mitochondria drug effects
- Abstract
The periconceptual environment represents a critical window for programming fetal growth trajectories and susceptibility to disease; however, the underlying mechanism responsible for programming remains elusive. This study demonstrates a causal link between reduction of precompaction embryonic mitochondrial function and perturbed offspring growth trajectories and subsequent metabolic dysfunction. Incubation of embryos with carbonyl cyanide 4-(trifluoromethoxy) phenylhydrazone (FCCP), which uncouples mitochondrial oxidative phosphorylation, significantly reduced mitochondrial membrane potential and ATP production in 8-cell embryos and the number of inner cell mass cells within blastocysts; however, blastocyst development was unchanged. This perturbed embryonic mitochondrial function was concomitant with reduced birth weight in female offspring following embryo transfer, which persisted until weaning. FCCP-treated females also exhibited increased adiposity at 4 wk, increased adiposity gain between 4 and 14 wk, glucose intolerance at 8 wk, and insulin resistance at 14 wk. Although FCCP-treated males also exhibited reduced glucose tolerance, but their insulin sensitivity and adiposity gain between 4 and 14 wk was unchanged. To our knowledge, this is one of the first studies to demonstrate that reducing mitochondrial function and, thus, decreasing ATP output in the precompacting embryo can influence offspring phenotype. This is of great significance as a large proportion of patients requiring assisted reproductive technologies are of advanced maternal age or have a high body mass index, both of which have been independently linked with perturbed early embryonic mitochondrial function., (© 2015 by the Society for the Study of Reproduction, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
37. Paternal obesity induces metabolic and sperm disturbances in male offspring that are exacerbated by their exposure to an "obesogenic" diet.
- Author
-
Fullston T, McPherson NO, Owens JA, Kang WX, Sandeman LY, and Lane M
- Abstract
Obesity and related comorbidities are becoming increasingly prevalent globally. In mice preconception paternal exposure to a high fat diet (HFD) impairs the metabolic and reproductive health of male offspring, despite their control diet (CD) consumption. However, offspring share lifestyle, including diet, with parents. We assessed if male offspring from HFD fathers have a heightened susceptibility to HFD-induced metabolic and reproductive derangements. This 2 × 2 design saw founder males (F0) and their offspring (F1) fed either a HFD or a nutritionally matched CD. Regardless of paternal diet, HFD fed male offspring had greater total body weight and adiposity. Offspring sired by a HFD male and fed a HFD were the heaviest, had the greatest adiposity and had the greatest concentration of serum cholesterol, triglyceride, HDL, and NEFA compared with CD sired/fed littermates. A synergistic increase in serum insulin was unmasked by both father/son HFD consumption, concomitant with increased sera glucose. Either a paternal or offspring HFD was associated with similar reductions to offspring sperm motility. Whereas sperm ROS concentrations and sperm-oocyte binding saw detrimental effects of both F0 HFD and F1 HFD with an interaction evident between both, culminating in the most impaired sperm parameters in this group. This indicates that metabolic and fertility disturbances in male offspring sired by HFD fathers are exacerbated by a "second-hit" of exposure to the same obesogenic environment postnatally. If translatable to human health, this suggests that adverse reproductive and metabolic outcomes may be amplified across generations through a shared calorie dense diet, relevant to the current worldwide obesity epidemic., (© 2015 The Authors. Physiological Reports published by Wiley Periodicals, Inc. on behalf of the American Physiological Society and The Physiological Society.)
- Published
- 2015
- Full Text
- View/download PDF
38. Oxidative stress in mouse sperm impairs embryo development, fetal growth and alters adiposity and glucose regulation in female offspring.
- Author
-
Lane M, McPherson NO, Fullston T, Spillane M, Sandeman L, Kang WX, and Zander-Fox DL
- Subjects
- Animals, Blastocyst cytology, Female, Fertilization, Fetal Development, Glucose Intolerance, Hydrogen Peroxide pharmacology, Male, Mice, Inbred C57BL, Mice, Inbred CBA, Pregnancy, Adiposity, Embryonic Development, Glucose metabolism, Oxidative Stress, Spermatozoa metabolism
- Abstract
Paternal health cues are able to program the health of the next generation however the mechanism for this transmission is unknown. Reactive oxygen species (ROS) are increased in many paternal pathologies, some of which program offspring health, and are known to induce DNA damage and alter the methylation pattern of chromatin. We therefore investigated whether a chemically induced increase of ROS in sperm impairs embryo, pregnancy and offspring health. Mouse sperm was exposed to 1500 µM of hydrogen peroxide (H2O2), which induced oxidative damage, however did not affect sperm motility or the ability to bind and fertilize an oocyte. Sperm treated with H2O2 delayed on-time development of subsequent embryos, decreased the ratio of inner cell mass cells (ICM) in the resulting blastocyst and reduced implantation rates. Crown-rump length at day 18 of gestation was also reduced in offspring produced by H2O2 treated sperm. Female offspring from H2O2 treated sperm were smaller, became glucose intolerant and accumulated increased levels of adipose tissue compared to control female offspring. Interestingly male offspring phenotype was less severe with increases in fat depots only seen at 4 weeks of age, which was restored to that of control offspring later in life, demonstrating sex-specific impacts on offspring. This study implicates elevated sperm ROS concentrations, which are common to many paternal health pathologies, as a mediator of programming offspring for metabolic syndrome and obesity.
- Published
- 2014
- Full Text
- View/download PDF
39. Obese father's metabolic state, adiposity, and reproductive capacity indicate son's reproductive health.
- Author
-
McPherson NO, Fullston T, Bakos HW, Setchell BP, and Lane M
- Subjects
- Animals, Blood Glucose metabolism, Diet, High-Fat methods, Disease Models, Animal, Fathers, Female, Insulin Resistance physiology, Male, Mice, Obesity pathology, Pregnancy, Prenatal Exposure Delayed Effects pathology, Random Allocation, Adiposity physiology, Diet, High-Fat adverse effects, Obesity metabolism, Prenatal Exposure Delayed Effects metabolism, Reproduction physiology, Sperm Motility physiology
- Abstract
Objective: To determine whether dietary and exercise regimes in obese males can provide a novel intervention window for improving the reproductive health of the next generation., Design: Experimental animal study., Setting: University research facilities., Animal(s): C57BL6 male and female mice., Intervention(s): Mice were fed a control diet (6% fat) or high-fat diet (21% fat) for 9 weeks. After the initial feeding, high-fat-diet males were allocated to diet and/or exercise interventions for a further 9 weeks. After intervention males were mated with females fed standard chow (4% fat) before and during pregnancy., Main Outcome Measure(s): F1 sperm motility, count, morphology, capacitation, mitochondrial function, and sperm binding and weight of reproductive organs., Result(s): Our primary finding was that diet intervention alone in founders improved offspring sperm motility and mitochondrial markers of sperm health (decreased reactive oxygen species and mitochondrial membrane potential), ultimately improving sperm binding. Sperm binding and capacitation was also improved in F1 males born to a combined diet and exercise intervention in founders. Founder sperm parameters and metabolic measures as a response to diet and/or exercise (i.e., lipid/glucose homeostasis, sperm count and morphology) correlated with offspring's sperm function, independent of founder treatment. This implicates paternal metabolic and reproductive status in predicting male offspring's reproductive function., Conclusion(s): This is the first study to show that improvements to both metabolic (lipids, glucose and insulin sensitivity) and reproductive function (sperm motility and morphology) in obese fathers via diet and exercise interventions can improve subsequent reproductive health in offspring., (Copyright © 2014 American Society for Reproductive Medicine. Published by Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
40. Paternal obesity, interventions, and mechanistic pathways to impaired health in offspring.
- Author
-
McPherson NO, Fullston T, Aitken RJ, and Lane M
- Subjects
- Animals, Child, Child Nutritional Physiological Phenomena, DNA Damage, Epigenesis, Genetic, Fathers, Female, Fetal Development, Humans, Life Style, Male, Nutrition Policy, Obesity epidemiology, Obesity pathology, Paternal Behavior, Pregnancy, Spermatozoa pathology, Diet adverse effects, Family Health, Health Promotion, Motor Activity, Obesity etiology, Obesity prevention & control, Patient Compliance
- Abstract
Background: The global rates of male overweight/obesity are rising, approaching 70% of the total adult population in Western nations. Overweight/obesity increases the risk of chronic diseases; however, there is increasing awareness that male obesity negatively impacts fertility, subsequent pregnancy, and the offspring health burden. Developmental programming is well defined in mothers; however, it is becoming increasingly evident that developmental programming can be paternally initiated and mediated through paternal obesity., Key Messages: Both human and rodent models have established that paternal obesity impairs sex hormones, basic sperm function, and molecular composition. This results in perturbed embryo development and health and an increased subsequent offspring disease burden in both sexes. The reversibility of obesity-induced parental programming has only recently received attention. Promising results in animal models utilizing diet and exercise interventions have shown improvements in sperm function and molecular composition, resulting in restorations of both embryo and fetal health and subsequent male offspring fertility. The direct mode for paternal inheritance is likely mediated via spermatozoa. We propose two main theories for the origin of male obesity-induced paternal programming: (1) accumulation of sperm DNA damage resulting in de novo mutations in the embryo and (2) changes in sperm epigenetic marks (microRNA, methylation, or acetylation) altering the access, transcription, and translation of paternally derived genes during early embryogenesis., Conclusions: Paternal overweight/obesity induces paternal programming of offspring phenotypes likely mediated through genetic and epigenetic changes in spermatozoa. These programmed changes to offspring health appear to be partially restored via diet/exercise interventions in obese fathers preconception, which have been shown to improve aspects of sperm DNA integrity. However, the majority of data surrounding paternal obesity and offspring phenotypes have come from rodent models; therefore, we contend that it will be increasingly important to study population-based data to determine the likely mode of inheritance in humans., (© 2014 S. Karger AG, Basel.)
- Published
- 2014
- Full Text
- View/download PDF
41. Paternal obesity initiates metabolic disturbances in two generations of mice with incomplete penetrance to the F2 generation and alters the transcriptional profile of testis and sperm microRNA content.
- Author
-
Fullston T, Ohlsson Teague EM, Palmer NO, DeBlasio MJ, Mitchell M, Corbett M, Print CG, Owens JA, and Lane M
- Subjects
- Animals, Energy Metabolism, Female, Gene Expression Regulation physiology, Glucose Tolerance Test, Insulin Resistance, Male, Mice, Mice, Inbred C57BL, MicroRNAs genetics, Obesity genetics, Reactive Oxygen Species, Sex Factors, MicroRNAs metabolism, Obesity metabolism, Spermatozoa metabolism, Testis metabolism, Transcriptome physiology
- Abstract
Obesity is highly prevalent, and its incidence is increasing. The previous study showing a major effect of paternal obesity on metabolic health of offspring is confounded by comorbidity with diabetes. Therefore, we investigated the effect of diet-induced paternal obesity, in the absence of diabetes, on the metabolic health of two resultant generations and the molecular profiles of the testes and sperm. Founder (F0) male C57BL6 mice were fed either a high-fat diet (HFD) or a control diet (CD); n = 10/diet for a period of 10 wk. Testis expression of mRNA/microRNAs was analyzed by microarray and qPCR and sperm microRNA abundance by qPCR. Two subsequent generations were generated by mating F0 and then F1 mice to CD mice, and their metabolic health was investigated. All mice, other than F0 males, were maintained on a CD. HFD feeding induced paternal obesity with a 21% increase in adiposity, but not overt diabetes, and initiated intergenerational transmission of obesity and insulin resistance in two generations of offspring. This distinct phenotypic constellation is either partially or fully transmitted to both female and male F1 offspring and further transmitted through both parental lineages to the F2 generation, with a heightened effect on female F1 offspring (+67% in adiposity) and their F2 sons (+24% in adiposity). Founder male obesity altered the testes expression of 414 mRNAs by microarray and 11 microRNAs by qPCR, concomitant with alterations in sperm microRNA content and a 25% reduction in global methylation of germ cell DNA. Diet-induced paternal obesity modulates sperm microRNA content and germ cell methylation status, which are potential signals that program offspring health and initiate the transmission of obesity and impaired metabolic health to future generations. This study implicates paternal obesity in the transgenerational amplification of obesity and type 2 diabetes in humans.
- Published
- 2013
- Full Text
- View/download PDF
42. Impact of obesity on male fertility, sperm function and molecular composition.
- Author
-
Palmer NO, Bakos HW, Fullston T, and Lane M
- Abstract
Male obesity in reproductive-age men has nearly tripled in the past 30 y and coincides with an increase in male infertility worldwide. There is now emerging evidence that male obesity impacts negatively on male reproductive potential not only reducing sperm quality, but in particular altering the physical and molecular structure of germ cells in the testes and ultimately mature sperm. Recent data has shown that male obesity also impairs offspring metabolic and reproductive health suggesting that paternal health cues are transmitted to the next generation with the mediator mostly likely occurring via the sperm. Interestingly the molecular profile of germ cells in the testes and sperm from obese males is altered with changes to epigenetic modifiers. The increasing prevalence of male obesity calls for better public health awareness at the time of conception, with a better understanding of the molecular mechanism involved during spermatogenesis required along with the potential of interventions in reversing these deleterious effects. This review will focus on how male obesity affects fertility and sperm quality with a focus on proposed mechanisms and the potential reversibility of these adverse effects.
- Published
- 2012
- Full Text
- View/download PDF
43. Inherited balanced translocation t(9;17)(q33.2;q25.3) concomitant with a 16p13.1 duplication in a patient with schizophrenia.
- Author
-
Fullston T, Gabb B, Callen D, Ullmann R, Woollatt E, Bain S, Ropers HH, Cooper M, Chandler D, Carter K, Jablensky A, Kalaydjieva L, and Gecz J
- Subjects
- Adult, Alleles, Family, Humans, Male, Mutation genetics, Nerve Tissue Proteins genetics, Pedigree, Protein Processing, Post-Translational, Schizophrenia metabolism, Schizophrenia pathology, Chromosomes, Human, Pair 16, Chromosomes, Human, Pair 17 genetics, Chromosomes, Human, Pair 9 genetics, Schizophrenia genetics, Segmental Duplications, Genomic, Translocation, Genetic
- Abstract
We report two rare genetic aberrations in a schizophrenia patient that may act together to confer disease susceptibility. A previously unreported balanced t(9;17)(q33.2;q25.3) translocation was observed in two schizophrenia-affected members of a small family with diverse psychiatric disorders. The proband also carried a 1.5 Mbp microduplication at 16p13.1 that could not be investigated in other family members. The duplication has been reported to predispose to schizophrenia, autism and mental retardation, with incomplete penetrance and variable expressivity. The t(9;17) (q33.2;q25.3) translocation breakpoint occurs within the open reading frames of KIAA1618 on 17q25.3, and TTLL11 (tyrosine tubulin ligase like 11) on 9q33.2, causing no change in the expression level of KIAA1618 but leading to loss of expression of one TTLL11 allele. TTLL11 belongs to a family of enzymes catalyzing polyglutamylation, an unusual neuron-specific post-translational modification of microtubule proteins, which modulates microtubule development and dynamics. The 16p13.1 duplication resulted in increased expression of NDE1, encoding a DISC1 protein partner mediating DISC1 functions in microtubule dynamics. We hypothesize that concomitant TTLL11-NDE1 deregulation may increase mutation load, among others, also on the DISC1 pathway, which could contribute to disease pathogenesis through multiple effects on neuronal development, synaptic plasticity, and neurotransmission. Our data illustrate the difficulties in interpreting the contribution of multiple potentially pathogenic changes likely to emerge in future next-generation sequencing studies, where access to extended families will be increasingly important., (Copyright © 2011 Wiley-Liss, Inc.)
- Published
- 2011
- Full Text
- View/download PDF
44. SIRT6 in mouse spermatogenesis is modulated by diet-induced obesity.
- Author
-
Palmer NO, Fullston T, Mitchell M, Setchell BP, and Lane M
- Subjects
- Acetylation, Acrosome metabolism, Acrosome ultrastructure, Animals, Cell Nucleus metabolism, Cell Nucleus ultrastructure, DNA Damage, Dietary Fats adverse effects, Down-Regulation, Epididymis metabolism, Epididymis ultrastructure, Infertility, Male etiology, Male, Mice, Mice, Inbred C57BL, Obesity pathology, Obesity physiopathology, Protein Transport, RNA, Messenger metabolism, Random Allocation, Sirtuins genetics, Sperm Capacitation, Spermatids metabolism, Spermatids ultrastructure, Spermatozoa ultrastructure, Testis ultrastructure, Obesity metabolism, Sirtuins metabolism, Spermatogenesis, Spermatozoa metabolism, Testis metabolism
- Abstract
Male obesity is associated with reduced sperm function and increased incidence of sperm DNA damage; however, the underlying molecular mechanisms have not yet been identified. Mammalian SIRT6 protein is involved in caloric-dependant DNA damage repair in other tissue types, yet a possible role for SIRT6 in male obesity and subfertility has not been investigated previously. To assess SIRT6 levels and activity in the testes, male mice (n=12 per diet) were fed either a control diet (CD; 6% fat) or a high-fat diet (HFD; 21% fat) for 16 weeks before the collection of testes and spermatozoa. SIRT6 protein was localised to the nucleus of transitional spermatids and the acrosome of mature spermatozoa, with levels significantly decreased in HFD-fed male mice (P<0.05). This decrease in SIRT6 protein was associated with transitional spermatids having increased levels of acetylated H3K9 in the nucleus (P<0.01) and increased DNA damage (P<0.001). We propose a role for SIRT6 in spermiogenesis and potentially protamination processes, which are known to be compromised by male obesity.
- Published
- 2011
- Full Text
- View/download PDF
45. Mitochondrial inhibition during preimplantation embryogenesis shifts the transcriptional profile of fetal mouse brain.
- Author
-
Fullston T, Mitchell M, Wakefield S, and Lane M
- Subjects
- Animals, Blastocyst metabolism, Brain embryology, Brain metabolism, Embryo Culture Techniques, Embryo Transfer, Epigenesis, Genetic drug effects, Gene Expression Profiling methods, Gene Regulatory Networks drug effects, Gestational Age, Mice, Mice, Inbred C57BL, Mice, Inbred CBA, Mitochondria metabolism, Oligonucleotide Array Sequence Analysis, Polymerase Chain Reaction, Aminooxyacetic Acid pharmacology, Blastocyst drug effects, Brain drug effects, Gene Expression Regulation, Developmental drug effects, Mitochondria drug effects, RNA, Messenger metabolism, Transcription, Genetic drug effects
- Abstract
Environmental stress results in perturbations to mitochondrial function in the preimplantation embryo and hinders subsequent embryo and possibly offspring development. Global gene expression in fetal mouse brain was investigated following targeted mitochondrial inhibition by amino-oxyacetate (AOA) from the 2-cell to the blastocyst stage. Blastocysts were transferred to pseudopregnant recipients and RNA extracted from Day 18 fetal brains for microarray interrogation. Exposure to 5 μM AOA during preimplantation embryo development induced differential expression of 166 genes (>1.25 fold) in the fetal brain, relative to control medium-cultured embryos. Altered expression pathways included carbohydrate metabolism, neurological development, cellular proliferation and death, DNA replication, recombination and repair. Of 28 genes exhibiting the greatest change in expression, qPCR confirmed that 16 were significantly altered. Targeted qPCR assessment of a further 20 genes associated with methylation, acetylation and mitochondrial dysfunction revealed that three were significantly altered (Immp1l, Nars2, Sat2) and Dmap1 exhibited a sex-specific response to AOA exposure. Only 2/48 genes had significantly altered expression by qPCR (Nola3, Timm8b) in fetal brains exposed to 50 μM AOA embryo culture, excluding an AOA dose-dependent response. It was concluded that perturbation of mitochondrial function induced by 5 μM AOA during preimplantation embryo development alters gene expression in the neonatal brain in a manner that suggests that proper brain development may be compromised.
- Published
- 2011
- Full Text
- View/download PDF
46. Fine-scale survey of X chromosome copy number variants and indels underlying intellectual disability.
- Author
-
Whibley AC, Plagnol V, Tarpey PS, Abidi F, Fullston T, Choma MK, Boucher CA, Shepherd L, Willatt L, Parkin G, Smith R, Futreal PA, Shaw M, Boyle J, Licata A, Skinner C, Stevenson RE, Turner G, Field M, Hackett A, Schwartz CE, Gecz J, Stratton MR, and Raymond FL
- Subjects
- Chromosome Breakage, Chromosome Segregation genetics, Cohort Studies, Disease genetics, Female, Gene Rearrangement genetics, Genes, X-Linked genetics, Humans, Male, Oligonucleotide Array Sequence Analysis, Pedigree, Reproducibility of Results, Retroelements genetics, Sequence Deletion genetics, Chromosomes, Human, X genetics, DNA Copy Number Variations genetics, INDEL Mutation genetics, Intellectual Disability genetics
- Abstract
Copy number variants and indels in 251 families with evidence of X-linked intellectual disability (XLID) were investigated by array comparative genomic hybridization on a high-density oligonucleotide X chromosome array platform. We identified pathogenic copy number variants in 10% of families, with mutations ranging from 2 kb to 11 Mb in size. The challenge of assessing causality was facilitated by prior knowledge of XLID-associated genes and the ability to test for cosegregation of variants with disease through extended pedigrees. Fine-scale analysis of rare variants in XLID families leads us to propose four additional genes, PTCHD1, WDR13, FAAH2, and GSPT2, as candidates for XLID causation and the identification of further deletions and duplications affecting X chromosome genes but without apparent disease consequences. Breakpoints of pathogenic variants were characterized to provide insight into the underlying mutational mechanisms and indicated a predominance of mitotic rather than meiotic events. By effectively bridging the gap between karyotype-level investigations and X chromosome exon resequencing, this study informs discussion of alternative mutational mechanisms, such as noncoding variants and non-X-linked disease, which might explain the shortfall of mutation yield in the well-characterized International Genetics of Learning Disability (IGOLD) cohort, where currently disease remains unexplained in two-thirds of families.
- Published
- 2010
- Full Text
- View/download PDF
47. ARX spectrum disorders: making inroads into the molecular pathology.
- Author
-
Shoubridge C, Fullston T, and Gécz J
- Subjects
- Doublecortin Protein, Genes, X-Linked, Genetic Association Studies, Humans, Islets of Langerhans metabolism, Mutation genetics, Homeodomain Proteins genetics, Intellectual Disability genetics, Intellectual Disability pathology, Transcription Factors genetics
- Abstract
The Aristaless-related homeobox gene (ARX) is one of the most frequently mutated genes in a spectrum of X-chromosome phenotypes with intellectual disability (ID) as their cardinal feature. To date, close to 100 families and isolated cases have been reported to carry 44 different mutations, the majority of these (59%) being a result of polyalanine tract expansions. At least 10 well-defined clinical entities, including Ohtahara, Partington, and Proud syndromes, X-linked infantile spasms, X-linked lissencephaly with ambiguous genitalia, X-linked myoclonic epilepsy and nonsyndromic intellectual disability have been ascertained from among the patients with ARX mutations. The striking intra- and interfamilial pleiotropy together with genetic heterogeneity (same clinical entities associated with different ARX mutations) are becoming a hallmark of ARX mutations. Although males are predominantly affected, some mutations associated with malformation phenotypes in males also show a phenotype in carrier females. Recent progress in the study of the effect of ARX mutations through sophisticated animal (mice) and cellular models begins to provide crucial insights into the molecular function of ARX and associated molecular pathology, thus guiding future inquiries into therapeutic interventions.
- Published
- 2010
- Full Text
- View/download PDF
48. Cyclin-dependent kinase-like 5 (CDKL5) mutation screening in Rett syndrome and related disorders.
- Author
-
White R, Ho G, Schmidt S, Scheffer IE, Fischer A, Yendle SC, Bienvenu T, Nectoux J, Ellaway CJ, Darmanian A, Tong X, Cloosterman D, Bennetts B, Kalra V, Fullston T, Gecz J, Cox TC, and Christodoulou J
- Subjects
- Amino Acid Sequence, Cognition Disorders diagnosis, Cognition Disorders enzymology, Cohort Studies, Cyclin-Dependent Kinase 5 metabolism, Female, Genetic Testing, Humans, Male, Molecular Sequence Data, Rett Syndrome enzymology, Seizures diagnosis, Seizures enzymology, Cognition Disorders genetics, Cyclin-Dependent Kinase 5 genetics, Mutation, Rett Syndrome diagnosis, Rett Syndrome genetics, Seizures genetics
- Abstract
Rett syndrome (RTT) is a severe neurodevelopmental disorder affecting females almost exclusively and is characterized by a wide spectrum of clinical manifestations. Mutations in the X-linked methyl-CpG-binding protein 2 (MECP2) gene have been found in up to 95% of classical RTT cases and a lesser proportion of atypical cases. Recently, mutations in another X-linked gene, CDKL5 (cyclin-dependent kinase-like 5) have been found to cause atypical RTT, in particular the early onset seizure (Hanefeld variant) and one female with autism. In this study we screened several cohorts of children for CDKL5 mutations, totaling 316 patients, including individuals with a clinical diagnosis of RTT but who were negative for MECP2 mutations (n=102), males with X-linked mental retardation (n=9), patients with West syndrome (n=52), patients with autism (n=59), patients with epileptic encephalopathy (n=33), patients with Aicardi syndrome (n=7) and other patients with intellectual disability with or without seizures (n=54). In all, seven polymorphic variations and four de novo mutations (c.586C>T [p.S196L]; c.58G>C [p.G20R]; c.2504delC [p.P835fs]; deletion of exons 1-3) were identified, and in all instances of the latter the clinical phenotype was that of an epileptic encephalopathy. These results suggest that pathogenic CDKL5 mutations are unlikely to be identified in the absence of severe early-onset seizures and highlight the importance of screening for large intragenic and whole gene deletions.
- Published
- 2010
- Full Text
- View/download PDF
49. Ohtahara syndrome in a family with an ARX protein truncation mutation (c.81C>G/p.Y27X).
- Author
-
Fullston T, Brueton L, Willis T, Philip S, MacPherson L, Finnis M, Gecz J, and Morton J
- Subjects
- Adult, Arrhythmias, Cardiac genetics, Base Sequence, Cesarean Section, Child, DNA Mutational Analysis, Epilepsy genetics, Female, Genetic Carrier Screening, Humans, Infant, Newborn, Intellectual Disability genetics, Male, Pedigree, Pregnancy, Sequence Deletion, Homeodomain Proteins genetics, Mental Retardation, X-Linked genetics, Mutation, Spasms, Infantile genetics, Transcription Factors genetics
- Abstract
Aristaless-related homeobox (ARX) gene mutations cause a diverse spectrum of disorders of the human brain, including lissencephaly, various forms of epilepsy and non-syndromic mental retardation. We have identified a novel mutation, c.81C>G (p.Y27X), within the ARX gene in a family with two affected male cousins. One of the boys was diagnosed with an early infantile epileptic encephalopathy also known as Ohtahara syndrome, whereas his cousin had been diagnosed with West syndrome (WS). Both patients have normal genitalia and neither have lissencephaly. The ARX mutation identified is predicted to yield a severely truncated protein of only 26 amino acids and can be considered as a null mutation. Somewhat surprisingly, however, it does not yield the X-linked lissencephaly with ambiguous genitalia (XLAG) syndrome. We proposed that the ARX mRNA translation re-initiated at the next AUG codon at position c.121-123 (aa 41) and, thus, partly rescued these patients from XLAG. Our in vitro studies show that this N-terminally truncated ARX protein (p.M41_C562) is detected by western immunoblot in lysates from cells transiently transfected with an ARX over-expression construct containing the c.81C>G mutation. Although these findings widen the spectrum of clinical phenotypes because of mutations in the ARX gene, they also emphasize the molecular pathogenetic effect of individual mutations as well as the effect of genetic background resulting in intrafamilial clinical heterogeneity for these mutations.
- Published
- 2010
- Full Text
- View/download PDF
50. Mutations in the nuclear localization sequence of the Aristaless related homeobox; sequestration of mutant ARX with IPO13 disrupts normal subcellular distribution of the transcription factor and retards cell division.
- Author
-
Shoubridge C, Tan MH, Fullston T, Cloosterman D, Coman D, McGillivray G, Mancini GM, Kleefstra T, and Gécz J
- Abstract
Background: Aristaless related homeobox (ARX) is a paired-type homeobox gene. ARX function is frequently affected by naturally occurring mutations. Nonsense mutations, polyalanine tract expansions and missense mutations in ARX cause a range of intellectual disability and epilepsy phenotypes with or without additional features including hand dystonia, lissencephaly, autism or dysarthria. Severe malformation phenotypes, such as X-linked lissencephaly with ambiguous genitalia (XLAG), are frequently observed in individuals with protein truncating or missense mutations clustered in the highly conserved paired-type homeodomain., Results: We have identified two novel point mutations in the R379 residue of the ARX homeodomain; c.1135C>A, p.R379S in a patient with infantile spasms and intellectual disability and c.1136G>T, p.R379L in a patient with XLAG. We investigated these and other missense mutations (R332P, R332H, R332C, T333N: associated with XLAG and Proud syndrome) predicted to affect the nuclear localisation sequences (NLS) flanking either end of the ARX homeodomain. The NLS regions are required for correct nuclear import facilitated by Importin 13 (IPO13). We demonstrate that missense mutations in either the N- or C-terminal NLS regions of the homeodomain cause significant disruption to nuclear localisation of the ARX protein in vitro. Surprisingly, none of these mutations abolished the binding of ARX to IPO13. This was confirmed by co-immunoprecipitation and immmuno fluorescence studies. Instead, tagged and endogenous IPO13 remained bound to the mutant ARX proteins, even in the RanGTP rich nuclear environment. We also identify the microtubule protein TUBA1A as a novel interacting protein for ARX and show cells expressing mutant ARX protein accumulate in mitosis, indicating normal cell division may be disrupted., Conclusions: We show that the most likely, common pathogenic mechanism of the missense mutations in NLS regions of the ARX homeodomain is inadequate accumulation and distribution of the ARX transcription factor within the nucleus due to sequestration of ARX with IPO13.
- Published
- 2010
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.