101 results on '"Stals, K."'
Search Results
2. De novo missense variants in FBXO11 alter its protein expression and subcellular localization
- Author
-
Gregor, A, Meerbrei, T, Gerstner, T, Toutain, A, Lynch, SA, Stals, K, Maxton, C, Lemke, JR, Bernat, JA, Bombei, HM, Foulds, N, Hunt, D, Kuechler, A, Beygo, J, Stobe, P, Bouman, A, Palomares-Bralo, M, Santos-Simarro, F, Garcia-Minaur, S, Pacio-Miguez, M, Popp, B, Vasileiou, G, Hebebrand, M, Reis, A, Schuhmann, S, Krumbiegel, M, Brown, NJ, Sparber, P, Melikyan, L, Bessonova, L, Cherevatova, T, Sharkov, A, Shcherbakova, N, Dabir, T, Kini, U, Schwaibold, EMC, Haack, TB, Bertoli, M, Hoffjan, S, Falb, R, Shinawi, M, Sticht, H, Zweier, C, Gregor, A, Meerbrei, T, Gerstner, T, Toutain, A, Lynch, SA, Stals, K, Maxton, C, Lemke, JR, Bernat, JA, Bombei, HM, Foulds, N, Hunt, D, Kuechler, A, Beygo, J, Stobe, P, Bouman, A, Palomares-Bralo, M, Santos-Simarro, F, Garcia-Minaur, S, Pacio-Miguez, M, Popp, B, Vasileiou, G, Hebebrand, M, Reis, A, Schuhmann, S, Krumbiegel, M, Brown, NJ, Sparber, P, Melikyan, L, Bessonova, L, Cherevatova, T, Sharkov, A, Shcherbakova, N, Dabir, T, Kini, U, Schwaibold, EMC, Haack, TB, Bertoli, M, Hoffjan, S, Falb, R, Shinawi, M, Sticht, H, and Zweier, C
- Abstract
Recently, others and we identified de novo FBXO11 (F-Box only protein 11) variants as causative for a variable neurodevelopmental disorder (NDD). We now assembled clinical and mutational information on 23 additional individuals. The phenotypic spectrum remains highly variable, with developmental delay and/or intellectual disability as the core feature and behavioral anomalies, hypotonia and various facial dysmorphism as frequent aspects. The mutational spectrum includes intragenic deletions, likely gene disrupting and missense variants distributed across the protein. To further characterize the functional consequences of FBXO11 missense variants, we analyzed their effects on protein expression and localization by overexpression of 17 different mutant constructs in HEK293 and HeLa cells. We found that the majority of missense variants resulted in subcellular mislocalization and/or reduced FBXO11 protein expression levels. For instance, variants located in the nuclear localization signal and the N-terminal F-Box domain lead to altered subcellular localization with exclusion from the nucleus or the formation of cytoplasmic aggregates and to reduced protein levels in western blot. In contrast, variants localized in the C-terminal Zn-finger UBR domain lead to an accumulation in the cytoplasm without alteration of protein levels. Together with the mutational data, our functional results suggest that most missense variants likely lead to a loss of the original FBXO11 function and thereby highlight haploinsufficiency as the most likely disease mechanism for FBXO11-associated NDDs.
- Published
- 2022
3. Dominant and recessive SLC12A2-syndrome
- Author
-
McNeill, A., Aurora, P., Rajput, K., Nash, R., Stals, K., Robinson, H., and Wakeling, E.
- Published
- 2022
4. Correction: A restricted spectrum of missense KMT2D variants cause a multiple malformations disorder distinct from Kabuki syndrome (Genetics in Medicine, (2020), 22, 5, (867-877), 10.1038/s41436-019-0743-3)
- Author
-
Cuvertino S., Hartill V., Colyer A., Garner T., Nair N., Al-Gazali L., Canham N., Faundes V., Flinter F., Hertecant J., Holder-Espinasse M., Jackson B., Lynch S. A., Nadat F., Narasimhan V. M., Peckham M., Sellers R., Seri M., Montanari F., Southgate L., Squeo G. M., Trembath R., van Heel D., Venuto S., Weisberg D., Stals K., Ellard S., Barton A., Kimber S. J., Sheridan E., Merla G., Stevens A., Johnson C. A., Banka S., Cuvertino, S., Hartill, V., Colyer, A., Garner, T., Nair, N., Al-Gazali, L., Canham, N., Faundes, V., Flinter, F., Hertecant, J., Holder-Espinasse, M., Jackson, B., Lynch, S. A., Nadat, F., Narasimhan, V. M., Peckham, M., Sellers, R., Seri, M., Montanari, F., Southgate, L., Squeo, G. M., Trembath, R., van Heel, D., Venuto, S., Weisberg, D., Stals, K., Ellard, S., Barton, A., Kimber, S. J., Sheridan, E., Merla, G., Stevens, A., Johnson, C. A., and Banka, S.
- Abstract
Correction to: Genetics in Medicine 22:2020 https://doi.org/10.1038/s41436-019-0743-3 published online 17 January 2020. An incorrect reference was cited in the fourth sentence of the Results section. The correct citation should have been Al-Gazali LI, Hamid Z, Hertecant J et al. An autosomal recessive syndrome of choanal atresia, hypothelia/athelia and thyroid gland anomalies overlapping Bamforth syndrome, ANOTHER syndrome and methimazole embryopathy. Clin Dysmorphol 2002;2:79–85. This has now been corrected in both the PDF and HTML versions of the Article.
- Published
- 2020
5. SPEN haploinsufficiency causes a neurodevelopmental disorder overlapping proximal 1p36 deletion syndrome with an episignature of X chromosomes in females
- Author
-
Radio, F.C., Pang, K., Ciolfi, A., Levy, M.A., Hernández-García, A., Pedace, L., Pantaleoni, F., Liu, Z, Boer, E. de, Jackson, A., Bruselles, A., McConkey, H., Stellacci, E., Cicero, S. Lo, Motta, M., Carrozzo, R., Dentici, M.L., McWalter, K., Desai, M., Monaghan, K.G., Telegrafi, A., Philippe, C., Vitobello, A., Au, M., Grand, K., Sanchez-Lara, P.A., Baez, J., Lindstrom, K., Kulch, P., Sebastian, J., Madan-Khetarpal, S., Roadhouse, C., MacKenzie, J.J., Monteleone, B., Saunders, C.J., Cuevas, J.K. Jean, Cross, L., Zhou, D., Hartley, T., Sawyer, S.L., Monteiro, F.P., Secches, T.V., Kok, F., Schultz-Rogers, L.E., Macke, E.L., Morava, E., Klee, E.W., Kemppainen, J., Iascone, M., Selicorni, A., Tenconi, R., Amor, D.J., Pais, L., Gallacher, L., Turnpenny, P.D., Stals, K., Ellard, S., Cabet, S., Lesca, G., Pascal, J., Steindl, K., Ravid, S., Weiss, K., Castle, A.M.R., Carter, M.T., Kalsner, L., Vries, B.B.A. de, Bon, B.W. van, Wevers, M.R., Pfundt, R.P., Stegmann, A.P.A., Kerr, B., Kingston, H.M., Chandler, K.E., Sheehan, W., Elias, A.F., Shinde, D.N., Towne, M.C., Robin, N.H., Goodloe, D., Vanderver, A., Sherbini, O., Bluske, K., Hagelstrom, R.T., Zanus, C., Faletra, F., Musante, L., Kurtz-Nelson, E.C., Earl, R.K., Anderlid, B.M., Morin, G., Slegtenhorst, M. van, Diderich, K.E.M., Brooks, A.S., Gribnau, J., Boers, R.G., Finestra, T.R., Carter, L.B., Rauch, A., Gasparini, P., Vissers, L.E.L.M., Lloyd Holder, J., Tartaglia, M., Radio, F.C., Pang, K., Ciolfi, A., Levy, M.A., Hernández-García, A., Pedace, L., Pantaleoni, F., Liu, Z, Boer, E. de, Jackson, A., Bruselles, A., McConkey, H., Stellacci, E., Cicero, S. Lo, Motta, M., Carrozzo, R., Dentici, M.L., McWalter, K., Desai, M., Monaghan, K.G., Telegrafi, A., Philippe, C., Vitobello, A., Au, M., Grand, K., Sanchez-Lara, P.A., Baez, J., Lindstrom, K., Kulch, P., Sebastian, J., Madan-Khetarpal, S., Roadhouse, C., MacKenzie, J.J., Monteleone, B., Saunders, C.J., Cuevas, J.K. Jean, Cross, L., Zhou, D., Hartley, T., Sawyer, S.L., Monteiro, F.P., Secches, T.V., Kok, F., Schultz-Rogers, L.E., Macke, E.L., Morava, E., Klee, E.W., Kemppainen, J., Iascone, M., Selicorni, A., Tenconi, R., Amor, D.J., Pais, L., Gallacher, L., Turnpenny, P.D., Stals, K., Ellard, S., Cabet, S., Lesca, G., Pascal, J., Steindl, K., Ravid, S., Weiss, K., Castle, A.M.R., Carter, M.T., Kalsner, L., Vries, B.B.A. de, Bon, B.W. van, Wevers, M.R., Pfundt, R.P., Stegmann, A.P.A., Kerr, B., Kingston, H.M., Chandler, K.E., Sheehan, W., Elias, A.F., Shinde, D.N., Towne, M.C., Robin, N.H., Goodloe, D., Vanderver, A., Sherbini, O., Bluske, K., Hagelstrom, R.T., Zanus, C., Faletra, F., Musante, L., Kurtz-Nelson, E.C., Earl, R.K., Anderlid, B.M., Morin, G., Slegtenhorst, M. van, Diderich, K.E.M., Brooks, A.S., Gribnau, J., Boers, R.G., Finestra, T.R., Carter, L.B., Rauch, A., Gasparini, P., Vissers, L.E.L.M., Lloyd Holder, J., and Tartaglia, M.
- Abstract
Contains fulltext : 231702.pdf (Publisher’s version ) (Closed access), Deletion 1p36 (del1p36) syndrome is the most common human disorder resulting from a terminal autosomal deletion. This condition is molecularly and clinically heterogeneous. Deletions involving two non-overlapping regions, known as the distal (telomeric) and proximal (centromeric) critical regions, are sufficient to cause the majority of the recurrent clinical features, although with different facial features and dysmorphisms. SPEN encodes a transcriptional repressor commonly deleted in proximal del1p36 syndrome and is located centromeric to the proximal 1p36 critical region. Here, we used clinical data from 34 individuals with truncating variants in SPEN to define a neurodevelopmental disorder presenting with features that overlap considerably with those of proximal del1p36 syndrome. The clinical profile of this disease includes developmental delay/intellectual disability, autism spectrum disorder, anxiety, aggressive behavior, attention deficit disorder, hypotonia, brain and spine anomalies, congenital heart defects, high/narrow palate, facial dysmorphisms, and obesity/increased BMI, especially in females. SPEN also emerges as a relevant gene for del1p36 syndrome by co-expression analyses. Finally, we show that haploinsufficiency of SPEN is associated with a distinctive DNA methylation episignature of the X chromosome in affected females, providing further evidence of a specific contribution of the protein to the epigenetic control of this chromosome, and a paradigm of an X chromosome-specific episignature that classifies syndromic traits. We conclude that SPEN is required for multiple developmental processes and SPEN haploinsufficiency is a major contributor to a disorder associated with deletions centromeric to the previously established 1p36 critical regions.
- Published
- 2021
6. Significant benefits of AIP testing and clinical screening in familial isolated and young-onset pituitary tumors
- Author
-
Marques, P, Caimari, F, Hernández-Ramírez, LC, Collier, D, Iacovazzo, D, Ronaldson, A, Magid, K, Lim, CT, Stals, K, Ellard, S, Grossman, AB, Korbonits, M, Abraham, P, Aflorei, E, Agha, A, Ahlquist, J, Akker, SA, Alexandraki, K, Alföldi, S, Anselmo, J, Arlt, W, Atkinson, B, Aulinas-Masó, A, Aylwin, SJ, Baborie, A, Backeljauw, PF, Badiu, C, Baldeweg, S, Ball, S, Bano, G, Barkan, A, Barton, J, Barwell, J, Bates, P, Bernal-González, C, Besser, M, Bevan, JS, Bickerton, A, Blair, J, Bolanowski, M, Bouloux, P, Bradley, L, Bradley, K, Brain, C, Brooke, A, Brown, R, Buchfelder, M, Burren, C, Cakir, M, Canham, N, Capraro, J, Carroll, P, Carter, P, Carty, D, Cavlan, D, Chahal, HS, Cheetham, T, Chentli, F, Choong, C, Christ-Crain, M, Chung, T-T, Clayton, P, Clayton, RN, Cohen, M, Courtney, H, Cove, D, Crowne, E, Cuthbertson, D, Dal, J, Dalantaeva, N, Damjanovic, S, Daousi, C, Darzy, K, Dattani, M, Davies, M, Davies, J, Davis, J, de Castro, M, de Marinis, L, Deal, C, Dénes, J, Dimitri, P, Dorward, N, Dow, G, Drake, W, Druce, M, Drummond, J, Dutta, P, Dzeranova, L, Edén-Engström, B, Eeles, R, Elfving, M, Ellis, K, Elston, M, Emmerson, L, Ezzat, S, Fersht, N, Fica, S, Fischli, S, Fleseriu, M, Forsythe, E, Foulkes, W, Freda, P, Friedman, T, Gadelha, M, Gainsborough, M, Gallacher, S, Gallego, P, Gan, H-W, Georgescu, C, Gevers, E, Gilkes, C, Glynn, N, Goldman, JE, Goldstone, AP, Góth, M, Green, A, Greenhalgh, L, Grieve, J, Griz, L, Guitelman, M, Gürlek, A, Gurnell, M, Hamblin, PS, Hana, V, Harding, P, Hay, E, Hilton, DA, Ho, W, Hong, G, Horváth, K, Howell, S, Howlett, TA, Höybye, C, Hunter, S, Idampitiya, C, Igaz, P, Imran, A, Inder, WJ, Iwata, T, Izatt, L, Jagadeesh, S, Johnston, C, Jose, B, Kaltsas, G, Kaplan, F, Karavitaki, N, Kastelan, D, Katz, M, Kearney, T, Kershaw, M, Khoo, B, Kiraly-Borri, C, Knispelis, R, Kovács, GL, Kumar, A, Kumar, AV, Kun, IZ, Kyriaku, A, Lambrescu, I, Lampe, AK, Laws, ER, Lebek-Szatanska, A, Lechan, RM, Leese, G, Levy, A, Levy, MJ, Lewandowski, K, Lin, E, Lo, J, Lyons, C, Maartens, N, Maghnie, M, Makaya, T, Marcus, H, Niedziela, M, Martin, N, Matsuno, A, McGowan, B, McQuaid, SE, Medic-Stojanoska, M, Mendoza, N, Mercado-Atri, M, Mettananda, S, Mezősi, E, Miljic, D, Miller, KK, Modenesi, S, Molitch, ME, Monson, J, Morris, DG, Morrison, PJ, Mosterman, B, Munir, A, Murray, RD, Musat, M, Musolino, N, Nachtigall, L, Nagi, D, Nair, R, Nelson, R, Newell-Price, J, Nikookam, K, Ogilivie, A, Orme, SM, O´Weickert, M, Pal, A, Pascanu, I, Patócs, A, Patterson, C, Pearce, SH, Giraldi, FP, Penney, L, Perez-Rivas, LG, Pfeifer, M, Pirie, F, Poplawski, N, Popovic, V, Powell, M, Pullan, P, Quinton, R, Radian, S, Randeva, H, Reddy, N, Rees, A, Renals, V, de Oliveira, AR, Richardson, T, Rodd, C, Ross, RJM, Roncaroli, F, Ryan, F, Salvatori, R, Schöfl, C, Shears, D, Shotliff, K, Skelly, R, Snape, K, Soares, BS, Somasundaram, N, Spada, A, Sperber, J, Spoudeas, H, Stelmachowska-Banas, M, Stewart, S, Storr, HL, Strasburger, C, Street, ME, Suter-Widmer, I, Suthers, G, Swords, F, Syro, LV, Swantje, B, Sze, C, Taylor, J, Thakker, RV, Tham, E, Thompson, C, Thorner, MO, Tóth, M, Trainer, PJ, Tsagarakis, S, Twine, G, Tzanela, M, Vadasz, J, Vaidya, B, Vaks, V, Vance, ML, Verkauskiene, R, Von Esch, H, Wass, JA, Waterhouse, M, Webb, S, Weber, A, Wernig, F, Widell, H, Yamada, S, Yap, P, Yarman, S, Yeoh, P, Yoshimoto, K, Yuen, K, and Zammitt, NN
- Abstract
Context\ud \ud Germline mutations in the aryl hydrocarbon receptor-interacting protein (AIP) gene are responsible for a subset of familial isolated pituitary adenoma (FIPA) cases and sporadic pituitary neuroendocrine tumors (PitNETs).\ud \ud \ud \ud Objective\ud \ud To compare prospectively diagnosed AIP mutation-positive (AIPmut) PitNET patients with clinically presenting patients and to compare the clinical characteristics of AIPmut and AIPneg PitNET patients.\ud \ud \ud \ud Design\ud \ud 12-year prospective, observational study.\ud \ud \ud \ud Participants & Setting\ud \ud We studied probands and family members of FIPA kindreds and sporadic patients with disease onset ≤18 years or macroadenomas with onset ≤30 years (n = 1477). This was a collaborative study conducted at referral centers for pituitary diseases.\ud \ud \ud \ud Interventions & Outcome\ud \ud AIP testing and clinical screening for pituitary disease. Comparison of characteristics of prospectively diagnosed (n = 22) vs clinically presenting AIPmut PitNET patients (n = 145), and AIPmut (n = 167) vs AIPneg PitNET patients (n = 1310).\ud \ud \ud \ud Results\ud \ud Prospectively diagnosed AIPmut PitNET patients had smaller lesions with less suprasellar extension or cavernous sinus invasion and required fewer treatments with fewer operations and no radiotherapy compared with clinically presenting cases; there were fewer cases with active disease and hypopituitarism at last follow-up. When comparing AIPmut and AIPneg cases, AIPmut patients were more often males, younger, more often had GH excess, pituitary apoplexy, suprasellar extension, and more patients required multimodal therapy, including radiotherapy. AIPmut patients (n = 136) with GH excess were taller than AIPneg counterparts (n = 650).\ud \ud \ud \ud Conclusions\ud \ud Prospectively diagnosed AIPmut patients show better outcomes than clinically presenting cases, demonstrating the benefits of genetic and clinical screening. AIP-related pituitary disease has a wide spectrum ranging from aggressively growing lesions to stable or indolent disease course.
- Published
- 2020
7. HNF1B deletions in patients with young-onset diabetes but no known renal disease
- Author
-
Edghill, E. L., Stals, K., Oram, R. A., Shepherd, M. H., Hattersley, A. T., and Ellard, S.
- Published
- 2013
- Full Text
- View/download PDF
8. JAG1 mutations are found in approximately one third of patients presenting with only one or two clinical features of Alagille syndrome
- Author
-
Guegan, K, Stals, K, Day, M, Turnpenny, P, and Ellard, S
- Published
- 2012
- Full Text
- View/download PDF
9. Is HNF1B really a MODY gene?: A64 (P430)
- Author
-
Shepherd, M, Edghill, E, Stals, K, Oram, R A, Larsson, H, Ellard, S, and Hattersley, A T
- Published
- 2011
10. Supplement to: AIP mutation in pituitary adenomas in 18th century and today.
- Author
-
Chahal, H S, Stals, K, and Unterländer, M
- Published
- 2011
11. A novel cause of hyperinsulinaemic hypoglycaemia with increased birthweight due to heterozygous mutations in the HNF4A gene: A8
- Author
-
Steele, A M, Barrett, T G, Stals, K, Shield, J PH, Tysoe, C, Ellard, S, Hattersley, A T, and Pearson, E R
- Published
- 2006
12. Kir6.2 gene mutations are a common cause of diabetes before 6 months and the presence of neurological features varies with the site of the mutation: A37
- Author
-
FLANAGAN, S E, EDGHILL, E L, GLOYN, A L, STALS, K L, ELLARD, S, and HATTERSLEY, A T
- Published
- 2005
13. A hypomorphic allele of SLC35D1 results in Schneckenbecken-like dysplasia
- Author
-
Rautengarten, C, Quarrell, OW, Stals, K, Caswell, RC, De Franco, E, Baple, E, Burgess, N, Jokhi, R, Heazlewood, JL, Offiah, AC, Ebert, B, Ellard, S, Rautengarten, C, Quarrell, OW, Stals, K, Caswell, RC, De Franco, E, Baple, E, Burgess, N, Jokhi, R, Heazlewood, JL, Offiah, AC, Ebert, B, and Ellard, S
- Abstract
We report the case of a consanguineous couple who lost four pregnancies associated with skeletal dysplasia. Radiological examination of one fetus was inconclusive. Parental exome sequencing showed that both parents were heterozygous for a novel missense variant, p.(Pro133Leu), in the SLC35D1 gene encoding a nucleotide sugar transporter. The affected fetus was homozygous for the variant. The radiological features were reviewed, and being similar, but atypical, the phenotype was classified as a 'Schneckenbecken-like dysplasia.' The effect of the missense change was assessed using protein modelling techniques and indicated alterations in the mouth of the solute channel. A detailed biochemical investigation of SLC35D1 transport function and that of the missense variant p.(Pro133Leu) revealed that SLC35D1 acts as a general UDP-sugar transporter and that the p.(Pro133Leu) mutation resulted in a significant decrease in transport activity. The reduced transport activity observed for p.(Pro133Leu) was contrasted with in vitro activity for SLC35D1 p.(Thr65Pro), the loss-of-function mutation was associated with Schneckenbecken dysplasia. The functional classification of SLC35D1 as a general nucleotide sugar transporter of the endoplasmic reticulum suggests an expanded role for this transporter beyond chondroitin sulfate biosynthesis to a variety of important glycosylation reactions occurring in the endoplasmic reticulum.
- Published
- 2019
14. Phenotype of CNTNAP1:a study of patients demonstrating a specific severe congenital hypomyelinating neuropathy with survival beyond infancy
- Author
-
Low, K. J., Stals, K., Caswell, R., Wakeling, M., Clayton-Smith, J., Donaldson, A., Foulds, N., Norman, A., Splitt, M., Urankar, K., Vijayakumar, K., Majumdar, A., Study, DDD, Ellard, S., and Smithson, S. F.
- Abstract
CHN is genetically heterogeneous and its genetic basis is difficult to determine on features alone. CNTNAP1 encodes CASPR, integral in the paranodal junction high molecular mass complex. Nineteen individuals with biallelic variants have been described in association with severe congenital hypomyelinating neuropathy, respiratory compromise, profound intellectual disability and death within the first year. We report 7 additional patients ascertained through exome sequencing. We identified 9 novel CNTNAP1 variants in 6 families: three missense variants, four nonsense variants, one frameshift variant and one splice site variant. Significant polyhydramnios occurred in 6/7 pregnancies. Severe respiratory compromise was seen in 6/7 (tracheostomy in 5). A complex neurological phenotype was seen in all patients who had marked brain hypomyelination/demyelination and profound developmental delay. Additional neurological findings included cranial nerve compromise: orobulbar dysfunction in 5/7, facial nerve weakness in 4/7 and vocal cord paresis in 5/7. Dystonia occurred in 2/7 patients and limb contractures in 5/7. All had severe gastroesophageal reflux, and a gastrostomy was required in 5/7. In contrast to most previous reports, only one patient died in the first year of life. Protein modelling was performed for all detected CNTNAP1 variants. We propose a genotype–phenotype correlation, whereby hypomorphic missense variants partially ameliorate the phenotype, prolonging survival. This study suggests that biallelic variants in CNTNAP1 cause a distinct recognisable syndrome, which is not caused by other genes associated with CHN. Neonates presenting with this phenotype will benefit from early genetic definition to inform clinical management and enable essential genetic counselling for their families.
- Published
- 2018
15. Phenotype of CNTNAP1: a study of patients demonstrating a specific severe congenital hypomyelinating neuropathy with survival beyond infancy
- Author
-
Low, KJ, primary, Stals, K, additional, Caswell, R, additional, Wakeling, M, additional, Clayton-Smith, J, additional, Donaldson, A, additional, Foulds, N, additional, Norman, A, additional, Splitt, M, additional, Urankar, K, additional, Vijayakumar, K, additional, Majumdar, A, additional, Study, DDD, additional, Ellard, S, additional, and Smithson, SF, additional
- Published
- 2018
- Full Text
- View/download PDF
16. In-frame seven amino-acid duplication in AIP arose over the last 3000 years, disrupts protein interaction and stability and is associated with gigantism
- Author
-
Salvatori, R, Radian, S, Diekmann, Y, Iacovazzo, D, David, A, Gabrovska, P, Grassi, G, Bussell, A-M, Stals, K, Weber, A, Quinton, R, Crowne, EC, Corazzini, V, Metherell, L, Kearney, T, Du Plessis, D, Sinha, AK, Baborie, A, Lecoq, A-L, Chanson, P, Ansorge, O, Ellard, S, Trainer, PJ, Balding, D, Thomas, MG, Korbonits, M, Salvatori, R, Radian, S, Diekmann, Y, Iacovazzo, D, David, A, Gabrovska, P, Grassi, G, Bussell, A-M, Stals, K, Weber, A, Quinton, R, Crowne, EC, Corazzini, V, Metherell, L, Kearney, T, Du Plessis, D, Sinha, AK, Baborie, A, Lecoq, A-L, Chanson, P, Ansorge, O, Ellard, S, Trainer, PJ, Balding, D, Thomas, MG, and Korbonits, M
- Abstract
OBJECTIVE: Mutations in the aryl hydrocarbon receptor-interacting protein (AIP) gene are associated with pituitary adenoma, acromegaly and gigantism. Identical alleles in unrelated pedigrees could be inherited from a common ancestor or result from recurrent mutation events. DESIGN AND METHODS: Observational, inferential and experimental study, including: AIP mutation testing; reconstruction of 14 AIP-region (8.3 Mbp) haplotypes; coalescent-based approximate Bayesian estimation of the time to most recent common ancestor (tMRCA) of the derived allele; forward population simulations to estimate current number of allele carriers; proposal of mutation mechanism; protein structure predictions; co-immunoprecipitation and cycloheximide chase experiments. RESULTS: Nine European-origin, unrelated c.805_825dup-positive pedigrees (four familial, five sporadic from the UK, USA and France) included 16 affected (nine gigantism/four acromegaly/two non-functioning pituitary adenoma patients and one prospectively diagnosed acromegaly patient) and nine unaffected carriers. All pedigrees shared a 2.79 Mbp haploblock around AIP with additional haploblocks privately shared between subsets of the pedigrees, indicating the existence of an evolutionarily recent common ancestor, the 'English founder', with an estimated median tMRCA of 47 generations (corresponding to 1175 years) with a confidence interval (9-113 generations, equivalent to 225-2825 years). The mutation occurred in a small tandem repeat region predisposed to slipped strand mispairing. The resulting seven amino-acid duplication disrupts interaction with HSP90 and leads to a marked reduction in protein stability. CONCLUSIONS: The c.805_825dup allele, originating from a common ancestor, associates with a severe clinical phenotype and a high frequency of gigantism. The mutation is likely to be the result of slipped strand mispairing and affects protein-protein interactions and AIP protein stability.
- Published
- 2017
17. Increased Population Risk of AIP-Related Acromegaly and Gigantism in Ireland
- Author
-
Radian, S, Diekmann, Y, Gabrovska, P, Holland, B, Bradley, L, Wallace, H, Stals, K, Bussell, A-M, McGurren, K, Cuesta, M, Ryan, AW, Herincs, M, Hernandez-Ramirez, LC, Holland, A, Samuels, J, Aflorei, ED, Barry, S, Denes, J, Pernicova, I, Stiles, CE, Trivellin, G, McCloskey, R, Ajzensztejn, M, Abid, N, Akker, SA, Mercado, M, Cohen, M, Thakker, RV, Baldeweg, S, Barkan, A, Musat, M, Levy, M, Orme, SM, Unterlaender, M, Burger, J, Kumar, AV, Ellard, S, McPartlin, J, McManus, R, Linden, GJ, Atkinson, B, Balding, DJ, Agha, A, Thompson, CJ, Hunter, SJ, Thomas, MG, Morrison, PJ, Korbonits, M, Radian, S, Diekmann, Y, Gabrovska, P, Holland, B, Bradley, L, Wallace, H, Stals, K, Bussell, A-M, McGurren, K, Cuesta, M, Ryan, AW, Herincs, M, Hernandez-Ramirez, LC, Holland, A, Samuels, J, Aflorei, ED, Barry, S, Denes, J, Pernicova, I, Stiles, CE, Trivellin, G, McCloskey, R, Ajzensztejn, M, Abid, N, Akker, SA, Mercado, M, Cohen, M, Thakker, RV, Baldeweg, S, Barkan, A, Musat, M, Levy, M, Orme, SM, Unterlaender, M, Burger, J, Kumar, AV, Ellard, S, McPartlin, J, McManus, R, Linden, GJ, Atkinson, B, Balding, DJ, Agha, A, Thompson, CJ, Hunter, SJ, Thomas, MG, Morrison, PJ, and Korbonits, M
- Abstract
The aryl hydrocarbon receptor interacting protein (AIP) founder mutation R304* (or p.R304* ; NM_003977.3:c.910C>T, p.Arg304Ter) identified in Northern Ireland (NI) predisposes to acromegaly/gigantism; its population health impact remains unexplored. We measured R304* carrier frequency in 936 Mid Ulster, 1,000 Greater Belfast (both in NI) and 2,094 Republic of Ireland (ROI) volunteers and in 116 NI or ROI acromegaly/gigantism patients. Carrier frequencies were 0.0064 in Mid Ulster (95%CI = 0.0027-0.013; P = 0.0005 vs. ROI), 0.001 in Greater Belfast (0.00011-0.0047) and zero in ROI (0-0.0014). R304* prevalence was elevated in acromegaly/gigantism patients in NI (11/87, 12.6%, P < 0.05), but not in ROI (2/29, 6.8%) versus non-Irish patients (0-2.41%). Haploblock conservation supported a common ancestor for all the 18 identified Irish pedigrees (81 carriers, 30 affected). Time to most recent common ancestor (tMRCA) was 2550 (1,275-5,000) years. tMRCA-based simulations predicted 432 (90-5,175) current carriers, including 86 affected (18-1,035) for 20% penetrance. In conclusion, R304* is frequent in Mid Ulster, resulting in numerous acromegaly/gigantism cases. tMRCA is consistent with historical/folklore accounts of Irish giants. Forward simulations predict many undetected carriers; geographically targeted population screening improves asymptomatic carrier identification, complementing clinical testing of patients/relatives. We generated disease awareness locally, necessary for early diagnosis and improved outcomes of AIP-related disease.
- Published
- 2017
18. Inzet van netwerken
- Author
-
Wilschut, M., Goense, P., Margot Fleuren, Stals, K., Goossens, F., Boendermaker, L., and Clinical Psychology
- Subjects
Implementern ,Organisaties ,Life ,Richtlijnen ,Health ,CH - Child Health ,ELSS - Earth, Life and Social Sciences ,Healthy for Life ,Healthy Living - Abstract
Hoe zorgen we dat een beleidsmaatregel effect heeft in de praktijk? Hoe zorgen we dat organisaties werken volgens aanbevolen richtlijnen? Het programma was toch bewezen effectief, waarom zie ik in de praktijk dan geen effect?
- Published
- 2016
19. CNTNAP1: Extending the phenotype of congenital hypomyelinating neuropathy in 6 further patients
- Author
-
Low, K., primary, Stals, K., additional, Caswell, R., additional, Clayton-Smith, J., additional, Donaldson, A., additional, Foulds, N., additional, Splitt, M., additional, Norman, A., additional, Urankar, K., additional, Vijayakumar, K., additional, Study, D., additional, Ellard, S., additional, Majumdar, A., additional, and Smithson, S., additional
- Published
- 2017
- Full Text
- View/download PDF
20. Landscape of familial isolated and young-onset pituitary adenomas: Prospective diagnosis in AIP mutation carriers
- Author
-
Hernandez-Ramirez, L.C., Gabrovska, P., Denes, J., Stals, K., Trivellin, G., Tilley, D., Ferrau, F., Evanson, J., Ellard, S., Grossman, A.B., Roncaroli, F., Gadelha, M.R., Korbonits, M., Agha, A., Akker, S.A., Aflorei, E.D., Alföldi, S., Arlt, W., Atkinson, B., Aulinas-Masó, A., Aylwin, S.J., Backeljauw, P.F., Badiu, C., Baldeweg, S., Bano, G., Barkan, A., Barwell, J., Bernal-González, C., Besser, G., Bevan, J.S., Blair, J., Bouloux, P., Bradley, L., Buchfelder, M., Cakir, M., Canham N, ., Carroll, P., Chahal, H.S., Cheetham, T., Chentli, F., Clayton, R.N., Cohen, M., Cole, T., Courtney, H., Crowne, E., Cuthbertson, D., Dal J, ., Dalantaeva, N., Daousi, C., Darzy, K., Dattani, M., Davies, J.H., Davis, J., De Castro, M., De Marinis, L., Drake, W., Dutta, P., Dzeranova, L., Edén-Engström, B., Eeles, R., Elfving, M., Elston, M., Emmerson, L., Fersht, N., Fica, S., Fischli, S., Flanagan, D., Fleseriu, M., Freda, P.U., Friedman, T., Frohman, L.A., Gallego, P., Gevers, E., Gláz, E., Goldman, J.A., Goldstone, A.P., Goth, M., Greenhalgh, L., Grieve, J., Guitelman, M., Gürlek, A., Gurnell, M., Horvath, K., Howlett, T.A., Höybye, C., Hunter S, ., Iacovazzo D, ., Igaz, P., Inder, W.J., Iwata, T., Izatt, L., Jagadeesh, S., Kaltsas, G., Kaplan F, ., Karavitaki, N., Kastelan, D., Katz, M., Kearney, T., Khoo, B., Kiraly-Borri, C., Knispelis, R., Kovács, G.L., Kumar, A.V., Laws, E.R., Lechan, R.M., Levy, J., Lewandowski, K., Lo, J., Maartens, N., Matsuno, A., Mcgowan, B., Mcquaid, S.E., Medic-Stojanoska, M., Mercado-Atri, M., Mezősi, E., Miljic, D., Miller, K.K., Modenesi, S., Molitch, M.E., Monson, J., Morris, D.G., Morrison, P.J., Munir, A., Murray, R.D., Musat, M., Musolino, N., Nachtigall, L., Newell-Price, J., Ogilvie, A., Orme, S.M., Paşcanu, I., Patócs, A., Patterson, C., Pearce, S.H., Pecori Giraldi, F., Pfeifer, M., Popovic, V., Poplawski, N., Powell, M., Pullan, P., Quinton, R., Radian, S., Randeva, H., Ribeiro-Oliveira, A., Rodd, C., Ryan, F., Salvatori, R., Schöfl, C., Shears, D., Shotliff, K., Soares, B.S., Spada, A., Sperber, J., Spoudeas, H.A., Stewart, S., Storr, H., Strasburger, C., Street, M.E., Swords, F., Thakker, R.V., Tham, E., Thompson, C., Thorner, M.O., Tóth, M., Trainer, P.J., Tsagarakis, S., Tzanela, M., Vadász, J., Vaks, V., Verkauskiene, R., Wass, J.A., Webb, S.M., Weber, A., Yamada, S., Yarman, S., Yeoh, P., Yoshimoto, K., Zammitt, N.N., and İç hastalıkları
- Subjects
Adenoma ,Adult ,Male ,Adolescent ,Aged ,Aged, 80 and over ,Child ,Child, Preschool ,Female ,Genetic Testing ,Germ-Line Mutation ,Growth Hormone-Secreting Pituitary Adenoma ,Humans ,Intracellular Signaling Peptides and Proteins ,Longitudinal Studies ,Middle Aged ,Mutation ,Pituitary Neoplasms ,Prospective Studies ,Young Adult ,Endocrinology, Diabetes and Metabolism ,Biochemistry ,Endocrinology ,Clinical Biochemistry ,Biochemistry (medical) ,Observational Study ,Settore MED/13 - Endocrinologia ,Journal Article ,80 and over ,Preschool ,JCEM Online: Advances in Genetics ,Research Support, Non-U.S. Gov't - Abstract
Context:Familial isolated pituitary adenoma (FIPA) due to aryl hydrocarbon receptor interacting protein (AIP) gene mutations is an autosomal dominant disease with incomplete penetrance. Clinical screening of apparently unaffected AIP mutation (AIPmut) carriers could identify previously unrecognized disease.Objective:To determine the AIP mutational status of FIPA and young pituitary adenoma patients, analyzing their clinical characteristics, and to perform clinical screening of apparently unaffected AIPmut carrier family members.Design:This was an observational, longitudinal study conducted over 7 years.Setting:International collaborative study conducted at referral centers for pituitary diseases.Participants:FIPA families (n = 216) and sporadic young-onset (≤30 y) pituitary adenoma patients (n = 404) participated in the study.Interventions:We performed genetic screening of patients for AIPmuts, clinical assessment of their family members, and genetic screening for somatic GNAS1 mutations and the germline FGFR4 p.G388R variant.Main Outcome Measure(s):We assessed clinical disease in mutation carriers, comparison of characteristics of AIPmut positive and negative patients, results of GNAS1, and FGFR4 analysis.Results:Thirty-seven FIPA families and 34 sporadic patients had AIPmuts. Patients with truncating AIPmuts had a younger age at disease onset and diagnosis, compared with patients with nontruncating AIPmuts. Somatic GNAS1 mutations were absent in tumors from AIPmut-positive patients, and the studied FGFR4 variant did not modify the disease behavior or penetrance in AIPmut-positive individuals. A total of 164 AIPmut-positive unaffected family members were identified; pituitary disease was detected in 18 of those who underwent clinical screening.Conclusions:A quarter of the AIPmut carriers screened were diagnosed with pituitary disease, justifying this screening and suggesting a variable clinical course for AIPmut-positive pituitary adenomas.
- Published
- 2015
21. Richtlijnen in de jeugdgezondheidszorg : onderbouwing voor landelijke invoering
- Author
-
Fleuren, M.A.H., Stals, K., Ooms, H., and Weeda, C.
- Subjects
Behavioural Changes ,Health ,ELSS - Earth, Life and Social Sciences ,Jeugdgezondheidszorg ,Healthy for Life ,Standaarden ,LS - Life Style ,Healthy Living - Published
- 2014
22. De cirkel is rond. Onderzoek naar succesvolle implementatie van interventies in de jeugdzorg
- Author
-
Stals, K., Adolescent development: Characteristics and determinants, Afd Jeugd en Gezin, van Yperen, TA, Stams, G.J.J.M., Reith, W.J.M., Wortel, E., and University Utrecht
- Abstract
Providers of youth care encounter problems with the implementation of interventions. This dissertation aimed to gain insight into factors that lead to successful and sustainable implementation of interventions in youth care. The first part of the dissertation provides a definition and a theoretical model that incorporates the main concepts related to implementation. Implementation is considered to be a systematic approach with the aim of a successful and sustainable implementation of an intervention. Regarding the success of implementation, specific attention is paid to the distinction between (1) implementation in the narrow sense – to carry out the intervention as intended, designated as program integrity - and (2) implementation in a broad sense - to achieve the intended outcomes. Through the following four steps a planned and phased approach is guaranteed: (1) define the desired outcome in a narrow and broad sense, (2) analyze the determinants (3) select strategies and (4) monitor, evaluate and adjust the process. Interviews with experts show that although various implementation activities were used in practice, these activities do not seem to be selected very systematically. The second part of the dissertation shows the ‘implementation monitor’, a schematic representation of an implementation process of a particular intervention in a specific organization. The monitor can be used to design, conduct, analyze and present research on implementation. This part also focuses on psychometric quality of two research instruments, the Decision Determinants Questionnaire (DDQ) and the questionnaire ‘Vragenlijst Kerncomponenten’. The DDQ measures readiness to change of employees in an organization. The ‘Vragenlijst Kerncomponenten’ is designed to gain insight into skills of youth care professionals and as a tool to discuss these skills between the professionals and their supervisors. In the third part of the dissertation contains two empirical studies on implementation. Firstly, the implementation of the intervention ‘Methodiek Ambulante Hulp’ within Jeugdformaat. Data were collected using questionnaires and structured interviews with 253 clients, 59 professionals and their supervisors. This study gave insight into the process of implementing an intervention. Furthermore, it shows relevant correlations between determinants of that implementation process and implementation in the narrow sense (program integrity) and in the broad sense (child outcomes). Secondly, a study on the implementation in the narrow sense of the intervention ‘Deltamethode Gezinsvoogdij’, a method for family guardianship at Bureau Jeugdzorg. The dissertation ends with a discussion, containing conclusions regarding the research questions, a summary of main findings for research and practice, limitations of the study and implications for further research and for the practice of youth care in the Netherlands. Amongst other findings, it is argued that the evidence for determinants of successful implementation is scarce, that this dissertation shows that implementation is a complex process in which various characteristics influence how the process proceeds and that an intervention can only help children and parents if it fits the needs of the client and if providers of the intervention are properly supervised. To conclude: implementation of an intervention should be well-thought
- Published
- 2012
23. Brief Report: AIP Mutation in Pituitary Adenomas in the 18th Century and Today
- Author
-
Chahal, H, Stals, K, Unterlander, M, Balding, D, Thomas, MG, Kumar, A, Besser, G, Atkinson, AB, Morrison, P, Howlett, T, Levy, M, Orme, S, Akker, SA, Abel, R, Grossman, AB, Burger, J, Ellard, S, and Korbonits, M
- Published
- 2011
24. The burden of AIP mutations in pituitary adenoma patients from the UK
- Author
-
Caimari, F, primary, Dang, M N, additional, Gabrovska, P, additional, Hernandez-Ramirez, L C, additional, Stals, K, additional, Bussell, A M, additional, Cranston, T, additional, Karavitaki, N, additional, Kumar, A V, additional, Hunter, S, additional, Kearney, T, additional, Trainer, P J, additional, Izatt, I, additional, Bevan, J, additional, Quinton, R, additional, Grieve, J, additional, Baldeweg, S E, additional, Grossman, A B, additional, Morrison, P, additional, and Korbonits, M, additional
- Published
- 2015
- Full Text
- View/download PDF
25. Planmatig implementeren leidt tot effectievere interventies in de praktijk
- Author
-
Stals, K., van Yperen, T.A., Reith, W.J.M., Stams, G.J.J.M., and Forensic Child and Youth Care (RICDE, FMG)
- Published
- 2010
26. Kennissynthese van condities voor effectieve invoering van jeugdinterventies
- Author
-
Mikolajczak, J., Stals, K., Fleuren, M.A.H., Wilde, E.J. de, Paulussen, T.G.W.M., and TNO Kwaliteit van Leven
- Subjects
Jeugd en Gezondheid ,Health - Published
- 2009
27. Jeugdzorg kan nog veel leren over implementeren: gebruik van kennis over implementatie vergroot de kans op succes
- Author
-
Stals, K., van Yperen, T.A., Reith, W.J.M., Stams, G.J.J.M., Forensic Child and Youth Care (RICDE, FMG), Adolescent development: Characteristics and determinants, Afd ASW, and Dep Educatie & Pedagogiek
- Abstract
Een mooi omschreven interventie die uiteindelijk niet meer van de boekenplank komt. Een praktisch protocol dat alleen de binnenkant van een bureaulade te zien krijgt. Herkenbaar? Waarschijnlijk wel. Want ondanks de groeiende aandacht voor effectieve interventies in de jeugdzorg verloopt de invoering in de praktijk vaak moeizaam. Een goed gepland implementatieproces vergroot de kans op succes.
- Published
- 2009
28. Effectieve en duurzame implementatie in de jeugdzorg: Een literatuurrapportage over belemmerende en bevorderende factoren op implementatie van interventies in de jeugdzorg
- Author
-
Stals, K., van Yperen, T., Reith, W., Stams, G.J., and Kohnstamm instituut
- Abstract
Effectieve en duurzame implementatie van jeugdzorginterventies is een belangrijke schakel om te komen tot effectieve jeugdzorg. Aangezien er geen kennis beschikbaar is over effectieve implementatie van interventies in de jeugdzorg, is er een literatuurstudie verricht naar effectieve implementatie van interventies in de gezondheidszorg, het onderwijs en het bedrijfsleven. Er is gebruik gemaakt van reviews, metastudies en primaire studies die zijn gepubliceerd in (inter)nationale wetenschappelijke tijdschriften, maar ook van rapporten en verslagen van organisatie in Nederland die zich met implementatievraagstukken bezighouden. In dit rapport wordt beschikbare kennis uit onderzoek en theorie toepasbaar gemaakt op de situatie in de jeugdzorg, zodat deze kennis benut kan worden voor een succesvol verloop van implementatietrajecten.
- Published
- 2008
29. P.171 - CNTNAP1: Extending the phenotype of congenital hypomyelinating neuropathy in 6 further patients
- Author
-
Low, K., Stals, K., Caswell, R., Clayton-Smith, J., Donaldson, A., Foulds, N., Splitt, M., Norman, A., Urankar, K., Vijayakumar, K., Study, D., Ellard, S., Majumdar, A., and Smithson, S.
- Published
- 2017
- Full Text
- View/download PDF
30. De cirkel is rond. Onderzoek naar succesvolle implementatie van interventies in de jeugdzorg
- Author
-
Adolescent development: Characteristics and determinants, Afd Jeugd en Gezin, van Yperen, TA, Stams, G.J.J.M., Reith, W.J.M., Wortel, E., Stals, K., Adolescent development: Characteristics and determinants, Afd Jeugd en Gezin, van Yperen, TA, Stams, G.J.J.M., Reith, W.J.M., Wortel, E., and Stals, K.
- Published
- 2012
31. Jeugdzorg kan nog veel leren over implementeren: Gebruik van kennis over implementatie vergroot de kans op succes
- Author
-
Adolescent development: Characteristics and determinants, Afd ASW, Dep Educatie & Pedagogiek, Stals, K., van Yperen, T.A., Reith, W.J.M., Stams, G.J.J.M., Adolescent development: Characteristics and determinants, Afd ASW, Dep Educatie & Pedagogiek, Stals, K., van Yperen, T.A., Reith, W.J.M., and Stams, G.J.J.M.
- Published
- 2009
32. HNF1B deletions in patients with young‐onset diabetes but no known renal disease
- Author
-
Edghill, E. L., primary, Stals, K., additional, Oram, R. A., additional, Shepherd, M. H., additional, Hattersley, A. T., additional, and Ellard, S., additional
- Published
- 2012
- Full Text
- View/download PDF
33. JAG1 mutations are found in approximately one third of patients presenting with only one or two clinical features of Alagille syndrome
- Author
-
Guegan, K, primary, Stals, K, additional, Day, M, additional, Turnpenny, P, additional, and Ellard, S, additional
- Published
- 2011
- Full Text
- View/download PDF
34. Hepatocyte nuclear factor-1 gene deletions--a common cause of renal disease
- Author
-
Edghill, E. L., primary, Oram, R. A., additional, Owens, M., additional, Stals, K. L., additional, Harries, L. W., additional, Hattersley, A. T., additional, Ellard, S., additional, and Bingham, C., additional
- Published
- 2007
- Full Text
- View/download PDF
35. The Tax Treatment of Corporate Losses: A Comparative Study.
- Author
-
Post, D. R. and Stals, K. P. E.
- Subjects
COMPARATIVE studies ,CORPORATE taxes ,BUSINESS losses ,EARNED income tax credit ,GROSS domestic product - Abstract
The article outlines the findings of a comparative study on tax treatment on corporate losses among 50 countries. It demonstrates that in many countries, the total corporate income tax losses of many businesses have increased significantly to as high as 25% of gross domestic product (GDP). It highlights certain important aspects relating to the vaporization of tax losses and tax loss refresher strategies.
- Published
- 2012
36. Een schuld teveel, een onderzoek naar huishoudens met problematische schulden in Rotterdam
- Author
-
Purperhart, J. and Stals, K.
- Subjects
Household and Consumer Studies ,Life Science ,Huishoudstudies - Published
- 1988
37. HNF1B deletions in patients with young-onset diabetes but no known renal disease
- Author
-
Oram, R. A., Ellard, S., Hattersley, A. T., Stals, K., Shepherd, M. H., and Edghill, E. L.
38. The authors and a colleague reply
- Author
-
Stals, K., Trivellin, G., and Marta Korbonits
39. A restricted spectrum of missense KMT2D variants cause a multiple malformations disorder distinct fromKabuki syndrome
- Author
-
Karen Stals, Sara Cuvertino, Víctor Faundes, Frances Flinter, Lihadh Al-Gazali, Santina Venuto, Vagheesh M. Narasimhan, Laura Southgate, Colin A. Johnson, Eamonn Sheridan, Nisha Nair, Anne Barton, Alice Colyer, Susan J. Kimber, Brian R. Jackson, Adam Stevens, Daniel Weisberg, Natalie Canham, Giuseppe Merla, Gabriella Maria Squeo, Richard C. Trembath, Sally Ann Lynch, Fatima Nadat, Terence Garner, Robert Sellers, Sian Ellard, Muriel Holder-Espinasse, David A. van Heel, Michelle Peckham, Francesca Montanari, Siddharth Banka, Verity L. Hartill, Marco Seri, Jozef Hertecant, Cuvertino, S., Hartill, V., Colyer, A., Garner, T., Nair, N., Al-Gazali, L., Canham, N., Faundes, V. Flinter F., Hertecant, J., Holder-Espinasse, M., Jackson, B., Lynch, Sa, Nadat, F., Narasimhan, V., Peckham, M., Sellers, R., Seri, M., Montanari, F., Southgate, L., Squeo, Gm, Trembath, R., van Heel, D., Venuto, S., Weisberg, D., Stals, K., Ellard, S., The, 100, Barton, A., Kimber, S., Sheridan, E., Merla, G, Stevens, A., Johnson, Ca, Banka, S., Cuvertino S., Hartill V., Colyer A., Garner T., Nair N., Al-Gazali L., Canham N., Faundes V., Flinter F., Hertecant J., Holder-Espinasse M., Jackson B., Lynch S.A., Nadat F., Narasimhan V.M., Peckham M., Sellers R., Seri M., Montanari F., Southgate L., Squeo G.M., Trembath R., van Heel D., Venuto S., Weisberg D., Stals K., Ellard S., Barton A., Kimber S.J., Sheridan E., Merla G., Stevens A., Johnson C.A., and Banka S.
- Subjects
Genetics ,0303 health sciences ,Kabuki syndrome ,Hearing loss ,KMT2D ,Choanal atresia ,Biology ,intrinsically disordered region ,medicine.disease ,Article ,Human genetics ,multiple congenital anomaly ,03 medical and health sciences ,Exon ,0302 clinical medicine ,030220 oncology & carcinogenesis ,DNA methylation ,medicine ,Missense mutation ,medicine.symptom ,histone 3 lysine 4 methyltransferase ,Haploinsufficiency ,Genetics (clinical) ,030304 developmental biology - Abstract
Purpose: To investigate if specific exon 38 or 39 KMT2D missense variants (MVs) cause a condition distinct from Kabuki syndrome type 1 (KS1). Methods: Multiple individuals, with MVs in exons 38 or 39 of KMT2D that encode a highly conserved region of 54 amino acids flanked by Val3527 and Lys3583, were identified and phenotyped. Functional tests were performed to study their pathogenicity and understand the disease mechanism. Results: The consistent clinical features of the affected individuals, from seven unrelated families, included choanal atresia, athelia or hypoplastic nipples, branchial sinus abnormalities, neck pits, lacrimal duct anomalies, hearing loss, external ear malformations and thyroid abnormalities. None of the individuals had intellectual disability. The frequency of clinical features, objective software-based facial analysis metrics and genome-wide peripheral blood DNA methylation patterns in these patients were significantly different from that of KS1. Circular dichroism spectroscopy indicated that these MVs perturb KMT2D secondary structure through an increased disordered to alpha helical transition. Conclusion: KMT2D MVs located in a specific region spanning exons 38 and 39 and affecting highly conserved residues cause a novel multiple malformations syndrome distinct from KS1. Unlike KMT2D haploinsufficiency in KS1, these MVs likely result in disease through a dominant negative mechanism.
- Published
- 2020
40. SPEN haploinsufficiency causes a neurodevelopmental disorder overlapping proximal 1p36 deletion syndrome with an episignature of X chromosomes in females
- Author
-
Gilles Morin, Krista Bluske, Nathaniel H. Robin, Laurence Faivre, Manuela Priolo, Dihong Zhou, Evangeline Kurtz-Nelson, Tianyun Wang, Omar Sherbini, Daryl A. Scott, Karen Stals, Fabíola Paoli Monteiro, Kaifang Pang, Sara Cabet, Francesca Clementina Radio, Bruno Dallapiccola, Marjon van Slegtenhorst, Rachel K. Earl, Katheryn Grand, Maria Iascone, Alice S. Brooks, Angelo Selicorni, July K. Jean Cuevas, Paolo Gasparini, Maria Lisa Dentici, Marialetizia Motta, Britt-Marie Anderlid, Kristin Lindstrom, Berrin Monteleone, Andrea Ciolfi, Karin Weiss, Katharina Steindl, Kirsty McWalter, Rosalba Carrozzo, Ruben Boers, Helen Kingston, Kym M. Boycott, Bekim Sadikovic, Laura Schultz-Rogers, Evan E. Eichler, Laura A Cross, Alison M R Castle, Louisa Kalsner, Lucia Pedace, Marijke R. Wevers, John M. Graham, Jessica Sebastian, Antonio Vitobello, Gaetan Lesca, Alexander P.A. Stegmann, Suneeta Madan-Khetarpal, Tahsin Stefan Barakat, Abdallah F. Elias, Teresa Robert Finestra, Adeline Vanderver, Peter D. Turnpenny, Bregje W.M. van Bon, Aida Telegrafi, David J. Amor, Deepali N. Shinde, Pedro A. Sanchez-Lara, Lisenka E.L.M. Vissers, Adam Jackson, Rolph Pfundt, Alessandro Bruselles, Andres Hernandez-Garcia, Karin E. M. Diderich, Flavio Faletra, Dana H. Goodloe, Joanne Baez, Sarit Ravid, Romano Tenconi, Sarah L. Sawyer, Lynn Pais, Bronwyn Kerr, Joost Gribnau, Lauren Carter, Melissa T. Carter, Zhandong Liu, Jennifer L. Kemppainen, Jennifer MacKenzie, Jimmy Holder, Elke de Boer, Margaret Au, Taila Hartley, Carol J Saunders, Luciana Musante, Bert B.A. de Vries, Tania Vertemati Secches, Haley McConkey, Willow Sheehan, Francesca Pantaleoni, Caterina Zanus, Christophe Philippe, Chelsea Roadhouse, Stefania Lo Cicero, Sian Ellard, R. Tanner Hagelstrom, Megha Desai, Fernando Kok, Joset Pascal, Marco Tartaglia, Eric W. Klee, Eva Morava, Michael A. Levy, Peggy Kulch, Lyndon Gallacher, Erica L. Macke, Emilia Stellacci, Siddharth Banka, Kristin G. Monaghan, Anita Rauch, Meghan C. Towne, Kate Chandler, Clinical Genetics, Developmental Biology, Radio, F. C., Pang, K., Ciolfi, A., Levy, M. A., Hernandez-Garcia, A., Pedace, L., Pantaleoni, F., Liu, Z., de Boer, E., Jackson, A., Bruselles, A., Mcconkey, H., Stellacci, E., Lo Cicero, S., Motta, M., Carrozzo, R., Dentici, M. L., Mcwalter, K., Desai, M., Monaghan, K. G., Telegrafi, A., Philippe, C., Vitobello, A., Au, M., Grand, K., Sanchez-Lara, P. A., Baez, J., Lindstrom, K., Kulch, P., Sebastian, J., Madan-Khetarpal, S., Roadhouse, C., Mackenzie, J. J., Monteleone, B., Saunders, C. J., Jean Cuevas, J. K., Cross, L., Zhou, D., Hartley, T., Sawyer, S. L., Monteiro, F. P., Secches, T. V., Kok, F., Schultz-Rogers, L. E., Macke, E. L., Morava, E., Klee, E. W., Kemppainen, J., Iascone, M., Selicorni, A., Tenconi, R., Amor, D. J., Pais, L., Gallacher, L., Turnpenny, P. D., Stals, K., Ellard, S., Cabet, S., Lesca, G., Pascal, J., Steindl, K., Ravid, S., Weiss, K., Castle, A. M. R., Carter, M. T., Kalsner, L., de Vries, B. B. A., van Bon, B. W., Wevers, M. R., Pfundt, R., Stegmann, A. P. A., Kerr, B., Kingston, H. M., Chandler, K. E., Sheehan, W., Elias, A. F., Shinde, D. N., Towne, M. C., Robin, N. H., Goodloe, D., Vanderver, A., Sherbini, O., Bluske, K., Hagelstrom, R. T., Zanus, C., Faletra, F., Musante, L., Kurtz-Nelson, E. C., Earl, R. K., Anderlid, B. -M., Morin, G., van Slegtenhorst, M., Diderich, K. E. M., Brooks, A. S., Gribnau, J., Boers, R. G., Finestra, T. R., Carter, L. B., Rauch, A., Gasparini, P., Boycott, K. M., Barakat, T. S., Graham, J. M., Faivre, L., Banka, S., Wang, T., Eichler, E. E., Priolo, M., Dallapiccola, B., Vissers, L. E. L. M., Sadikovic, B., Scott, D. A., Holder, J. L., Tartaglia, M., MUMC+: DA KG Lab Centraal Lab (9), and RS: FHML non-thematic output
- Subjects
0301 basic medicine ,SHARP ,Male ,obesity ,genotype-phenotype correlations ,Autism Spectrum Disorder ,PROTEIN ,Chromosome Disorders ,Haploinsufficiency ,RNA-Binding Protein ,PHENOTYPE CORRELATIONS ,1p36 ,distal 1p36 deletion syndrome ,DNA methylome analysis ,episignature ,neurodevelopmental disorder ,proximal 1p36 deletion syndrome ,SPEN ,X chromosome ,Adolescent ,Child ,Child, Preschool ,Chromosome Deletion ,Chromosomes, Human, Pair 1 ,Chromosomes, Human, X ,DNA Methylation ,DNA-Binding Proteins ,Epigenesis, Genetic ,Female ,Humans ,Intellectual Disability ,Neurodevelopmental Disorders ,Phenotype ,RNA-Binding Proteins ,Young Adult ,0302 clinical medicine ,Neurodevelopmental disorder ,Neurodevelopmental Disorder ,Intellectual disability ,MOLECULAR CHARACTERIZATION ,Genetics (clinical) ,Genetics ,DNA methylome analysi ,SPLIT-ENDS ,Hypotonia ,Autism spectrum disorder ,MONOSOMY 1P36 ,Pair 1 ,medicine.symptom ,Rare cancers Radboud Institute for Health Sciences [Radboudumc 9] ,Human ,DNA-Binding Protein ,Biology ,genotype-phenotype correlation ,Chromosomes ,03 medical and health sciences ,Genetic ,SDG 3 - Good Health and Well-being ,Report ,REVEALS ,medicine ,Epigenetics ,Preschool ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,1p36 deletion syndrome ,IDENTIFICATION ,MUTATIONS ,medicine.disease ,GENE ,030104 developmental biology ,Chromosome Disorder ,030217 neurology & neurosurgery ,Epigenesis - Abstract
Contains fulltext : 231702.pdf (Publisher’s version ) (Closed access) Deletion 1p36 (del1p36) syndrome is the most common human disorder resulting from a terminal autosomal deletion. This condition is molecularly and clinically heterogeneous. Deletions involving two non-overlapping regions, known as the distal (telomeric) and proximal (centromeric) critical regions, are sufficient to cause the majority of the recurrent clinical features, although with different facial features and dysmorphisms. SPEN encodes a transcriptional repressor commonly deleted in proximal del1p36 syndrome and is located centromeric to the proximal 1p36 critical region. Here, we used clinical data from 34 individuals with truncating variants in SPEN to define a neurodevelopmental disorder presenting with features that overlap considerably with those of proximal del1p36 syndrome. The clinical profile of this disease includes developmental delay/intellectual disability, autism spectrum disorder, anxiety, aggressive behavior, attention deficit disorder, hypotonia, brain and spine anomalies, congenital heart defects, high/narrow palate, facial dysmorphisms, and obesity/increased BMI, especially in females. SPEN also emerges as a relevant gene for del1p36 syndrome by co-expression analyses. Finally, we show that haploinsufficiency of SPEN is associated with a distinctive DNA methylation episignature of the X chromosome in affected females, providing further evidence of a specific contribution of the protein to the epigenetic control of this chromosome, and a paradigm of an X chromosome-specific episignature that classifies syndromic traits. We conclude that SPEN is required for multiple developmental processes and SPEN haploinsufficiency is a major contributor to a disorder associated with deletions centromeric to the previously established 1p36 critical regions.
- Published
- 2021
41. PSMF1 variants cause a phenotypic spectrum from early-onset Parkinson's disease to perinatal lethality by disrupting mitochondrial pathways.
- Author
-
Magrinelli F, Tesson C, Angelova PR, Salazar-Villacorta A, Rodriguez JA, Scardamaglia A, Chung BH, Jaconelli M, Vona B, Esteras N, Kwong AK, Courtin T, Maroofian R, Alavi S, Nirujogi R, Severino M, Lewis PA, Efthymiou S, O'Callaghan B, Buchert R, Sofan L, Lis P, Pinon C, Breedveld GJ, Chui MM, Murphy D, Pitz V, Makarious MB, Cassar M, Hassan BA, Iftikhar S, Rocca C, Bauer P, Tinazzi M, Svetel M, Samanci B, Hanağası HA, Bilgiç B, Obeso JA, Kurtis MM, Cogan G, Başak AN, Kiziltan G, Gül T, Yalçın G, Elibol B, Barišić N, Ng EW, Fan SS, Hershkovitz T, Weiss K, Raza Alvi J, Sultan T, Azmi Alkhawaja I, Froukh T, E Alrukban HA, Fauth C, Schatz UA, Zöggeler T, Zech M, Stals K, Varghese V, Gandhi S, Blauwendraat C, Hardy JA, Lesage S, Bonifati V, Haack TB, Bertoli-Avella AM, Steinfeld R, Alessi DR, Steller H, Brice A, Abramov AY, Bhatia KP, and Houlden H
- Abstract
Dissecting biological pathways highlighted by Mendelian gene discovery has provided critical insights into the pathogenesis of Parkinson's disease (PD) and neurodegeneration. This approach ultimately catalyzes the identification of potential biomarkers and therapeutic targets. Here, we identify PSMF1 as a new gene implicated in PD and childhood neurodegeneration. We find that biallelic PSMF1 missense and loss-of-function variants co-segregate with phenotypes from early-onset PD and parkinsonism to perinatal lethality with neurological manifestations across 15 unrelated pedigrees with 22 affected subjects, showing clear genotype-phenotype correlation. PSMF1 encodes the proteasome regulator PSMF1/PI31, a highly conserved, ubiquitously expressed partner of the 20S proteasome and neurodegeneration-associated F-box-O 7 and valosin-containing proteins. We demonstrate that PSMF1 variants impair mitochondrial membrane potential, dynamics and mitophagy in patient-derived fibroblasts. Additionally, we develop models of psmf1 knockdown Drosophila and Psmf1 conditional knockout mouse exhibiting age-dependent motor impairment, with diffuse gliosis in mice. These findings unequivocally link defective PSMF1 to early-onset PD and neurodegeneration and suggest mitochondrial dysfunction as a mechanistic contributor.
- Published
- 2024
- Full Text
- View/download PDF
42. Biallelic Variants in MNS1 Are Associated with Laterality Defects and Respiratory Involvement.
- Author
-
Hjeij R, Leslie J, Rizk H, Dworniczak B, Olbrich H, Raidt J, Bode SFN, Gardham A, Stals K, Al-Haggar M, Osman E, Crosby A, Eldesoky T, Baple E, and Omran H
- Subjects
- Child, Child, Preschool, Female, Humans, Male, Cilia pathology, Cilia genetics, Ciliopathies genetics, Ciliopathies pathology, Pedigree, Phenotype, Infant, Adolescent, Alleles
- Abstract
Defects in motile cilia, termed motile ciliopathies, result in clinical manifestations affecting the respiratory and reproductive system, as well as laterality defects and hydrocephalus. We previously defined biallelic MNS1 variants causing situs inversus and male infertility, mirroring the findings in Mns1
-/- mice. Here, we present clinical and genomic findings in five newly identified individuals from four unrelated families affected by MNS1 -related disorder. Ciliopathy panel testing and whole exome sequencing identified one previously reported and two novel MNS1 variants extending the genotypic spectrum of disease. A broad spectrum of laterality defects including situs inversus totalis and heterotaxia was confirmed. Interestingly, a single affected six-year-old girl homozygous for an MNS1 nonsense variant presented with a history of neonatal respiratory distress syndrome, recurrent respiratory tract infections, chronic rhinitis, and wet cough. Accordingly, immunofluorescence analysis showed the absence of MNS1 from the respiratory epithelial cells of this individual. Two other individuals with hypomorphic variants showed laterality defects and mild respiratory phenotype. This study represents the first observation of heterotaxia and respiratory disease in individuals with biallelic MNS1 variants, an important extension of the phenotype associated with MNS1-related motile ciliopathy disorder.- Published
- 2024
- Full Text
- View/download PDF
43. Biallelic PKP2 loss of function variants are associated with a lethal perinatal-onset biventricular dilated cardiomyopathy with excessive trabeculations and ventricular septal defects.
- Author
-
Gibb J, Wall E, Fields E, Seale A, Armstrong C, Bamber A, Daubeney P, Jacobs-Pearson M, Marton T, Stals K, Low K, Kaski JP, and Spentzou G
- Subjects
- Humans, Plakophilins genetics, Homozygote, Cardiomyopathy, Dilated genetics, Cardiomyopathies genetics, Heart Septal Defects, Ventricular
- Abstract
Homozygous plakophilin-2 ( PKP2 ) variants have been identified as a cause of a lethal form of dilated cardiomyopathy with excessive trabeculations (DCM-ET) in three cases. We report three more cases from two families with homozygous pathogenic PKP2 variants and perinatal-onset, lethal DCM-ET. Identification of the genetic abnormalities played a key role in decision-making and family counselling in these cases. This case series supports the published evidence that biallelic loss of function PKP2 variants cause a lethal, perinatal-onset cardiomyopathy., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2024. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2024
- Full Text
- View/download PDF
44. Rapid genome sequencing for infantile-onset epilepsy within a national health-care setting.
- Author
-
Robinson HK, Stals K, Hill S, Parrish A, and Baple EL
- Subjects
- Humans, Infant, Mutation, Epilepsy diagnosis, Epilepsy genetics, Spasms, Infantile
- Abstract
Competing Interests: We declare no competing interests.
- Published
- 2024
- Full Text
- View/download PDF
45. Biallelic CRELD1 variants cause a multisystem syndrome, including neurodevelopmental phenotypes, cardiac dysrhythmias, and frequent infections.
- Author
-
Jeffries L, Mis EK, McWalter K, Donkervoort S, Brodsky NN, Carpier JM, Ji W, Ionita C, Roy B, Morrow JS, Darbinyan A, Iyer K, Aul RB, Banka S, Chao KR, Cobbold L, Cohen S, Custodio HM, Drummond-Borg M, Elmslie F, Finanger E, Hainline BE, Helbig I, Hewson S, Hu Y, Jackson A, Josifova D, Konstantino M, Leach ME, Mak B, McCormick D, McGee E, Nelson S, Nguyen J, Nugent K, Ortega L, Goodkin HP, Roeder E, Roy S, Sapp K, Saade D, Sisodiya SM, Stals K, Towner S, Wilson W, Khokha MK, Bönnemann CG, Lucas CL, and Lakhani SA
- Subjects
- Humans, Leukocytes, Mononuclear, Syndrome, Phenotype, Arrhythmias, Cardiac genetics, Cell Adhesion Molecules genetics, Extracellular Matrix Proteins genetics, Reinfection, Neurodevelopmental Disorders genetics
- Abstract
Purpose: We sought to delineate a multisystem disorder caused by recessive cysteine-rich with epidermal growth factor-like domains 1 (CRELD1) gene variants., Methods: The impact of CRELD1 variants was characterized through an international collaboration utilizing next-generation DNA sequencing, gene knockdown, and protein overexpression in Xenopus tropicalis, and in vitro analysis of patient immune cells., Results: Biallelic variants in CRELD1 were found in 18 participants from 14 families. Affected individuals displayed an array of phenotypes involving developmental delay, early-onset epilepsy, and hypotonia, with about half demonstrating cardiac arrhythmias and some experiencing recurrent infections. Most harbored a frameshift in trans with a missense allele, with 1 recurrent variant, p.(Cys192Tyr), identified in 10 families. X tropicalis tadpoles with creld1 knockdown displayed developmental defects along with increased susceptibility to induced seizures compared with controls. Additionally, human CRELD1 harboring missense variants from affected individuals had reduced protein function, indicated by a diminished ability to induce craniofacial defects when overexpressed in X tropicalis. Finally, baseline analyses of peripheral blood mononuclear cells showed similar proportions of immune cell subtypes in patients compared with healthy donors., Conclusion: This patient cohort, combined with experimental data, provide evidence of a multisystem clinical syndrome mediated by recessive variants in CRELD1., Competing Interests: Conflict of Interest Two authors report part ownership of startup companies unrelated to this work: Qiyas Higher Health (Saquib A. Lakhani) and Victory Genomics (Saquib A. Lakhani and Mustafa K. Khokha). Kirsty McWalter is an employee of GeneDx. Kimberly Nugent is currently an employee of Cooper Surgical. Bryan Mak is currently an employee of Genome Medical. All other authors declare no conflicts of interest., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
46. Case Report: ISG15 deficiency caused by novel variants in two families and effective treatment with Janus kinase inhibition.
- Author
-
Burleigh A, Moraitis E, Al Masroori E, Al-Abadi E, Hong Y, Omoyinmi E, Titheradge H, Stals K, Jones WD, Gait A, Jayarajan V, Di WL, Sebire N, Solman L, Ogboli M, Welch SB, Sudarsanam A, Wacogne I, Price-Kuehne F, Jensen B, Brogan PA, and Eleftheriou D
- Subjects
- Humans, Interferons, Ubiquitin Thiolesterase genetics, Ubiquitin Thiolesterase metabolism, Ubiquitins metabolism, Cytokines metabolism
- Abstract
ISG15 deficiency is a rare disease caused by autosomal recessive variants in the ISG15 gene, which encodes the ISG15 protein. The ISG15 protein plays a dual role in both the type I and II interferon (IFN) immune pathways. Extracellularly, the ISG15 protein is essential for IFN-γ-dependent anti-mycobacterial immunity, while intracellularly, ISG15 is necessary for USP18-mediated downregulation of IFN-α/β signalling. Due to this dual role, ISG15 deficiency can present with various clinical phenotypes, ranging from susceptibility to mycobacterial infection to autoinflammation characterised by necrotising skin lesions, intracerebral calcification, and pulmonary involvement. In this report, we describe novel variants found in two different families that result in complete ISG15 deficiency and severe skin ulceration. Whole exome sequencing identified a heterozygous missense p.Q16X ISG15 variant and a heterozygous multigene 1p36.33 deletion in the proband from the first family. In the second family, a homozygous total ISG15 gene deletion was detected in two siblings. We also conducted further analysis, including characterisation of cytokine dysregulation, interferon-stimulated gene expression, and p-STAT1 activation in lymphocytes and lesional tissue. Finally, we demonstrate the complete and rapid resolution of clinical symptoms associated with ISG15 deficiency in one sibling from the second family following treatment with the Janus kinase (JAK) inhibitor baricitinib., Competing Interests: PB has received institutional grants from: Novartis, SOBI, Roche, Chemocentryx, and Novimmune; consultancy fees from Roche, Novartis and SOBI; and speaker fees from UCB. DE has received institutional grants from Lilly, Sobi, Roche and Pfizer. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Burleigh, Moraitis, Al Masroori, Al-Abadi, Hong, Omoyinmi, Titheradge, Stals, Jones, Gait, Jayarajan, Di, Sebire, Solman, Ogboli, Welch, Sudarsanam, Wacogne, Price-Kuehne, Jensen, Brogan and Eleftheriou.)
- Published
- 2023
- Full Text
- View/download PDF
47. Dominant negative variants in KIF5B cause osteogenesis imperfecta via down regulation of mTOR signaling.
- Author
-
Marom R, Zhang B, Washington ME, Song IW, Burrage LC, Rossi VC, Berrier AS, Lindsey A, Lesinski J, Nonet ML, Chen J, Baldridge D, Silverman GA, Sutton VR, Rosenfeld JA, Tran AA, Hicks MJ, Murdock DR, Dai H, Weis M, Jhangiani SN, Muzny DM, Gibbs RA, Caswell R, Pottinger C, Cilliers D, Stals K, Eyre D, Krakow D, Schedl T, Pak SC, and Lee BH
- Subjects
- Animals, Humans, Mice, Caenorhabditis elegans genetics, Caenorhabditis elegans metabolism, Carrier Proteins genetics, Down-Regulation, NIH 3T3 Cells, Proteomics, Signal Transduction genetics, TOR Serine-Threonine Kinases genetics, TOR Serine-Threonine Kinases metabolism, Kinesins genetics, Kinesins metabolism, Osteogenesis Imperfecta
- Abstract
Background: Kinesin motor proteins transport intracellular cargo, including mRNA, proteins, and organelles. Pathogenic variants in kinesin-related genes have been implicated in neurodevelopmental disorders and skeletal dysplasias. We identified de novo, heterozygous variants in KIF5B, encoding a kinesin-1 subunit, in four individuals with osteogenesis imperfecta. The variants cluster within the highly conserved kinesin motor domain and are predicted to interfere with nucleotide binding, although the mechanistic consequences on cell signaling and function are unknown., Methods: To understand the in vivo genetic mechanism of KIF5B variants, we modeled the p.Thr87Ile variant that was found in two patients in the C. elegans ortholog, unc-116, at the corresponding position (Thr90Ile) by CRISPR/Cas9 editing and performed functional analysis. Next, we studied the cellular and molecular consequences of the recurrent p.Thr87Ile variant by microscopy, RNA and protein analysis in NIH3T3 cells, primary human fibroblasts and bone biopsy., Results: C. elegans heterozygous for the unc-116 Thr90Ile variant displayed abnormal body length and motility phenotypes that were suppressed by additional copies of the wild type allele, consistent with a dominant negative mechanism. Time-lapse imaging of GFP-tagged mitochondria showed defective mitochondria transport in unc-116 Thr90Ile neurons providing strong evidence for disrupted kinesin motor function. Microscopy studies in human cells showed dilated endoplasmic reticulum, multiple intracellular vacuoles, and abnormal distribution of the Golgi complex, supporting an intracellular trafficking defect. RNA sequencing, proteomic analysis, and bone immunohistochemistry demonstrated down regulation of the mTOR signaling pathway that was partially rescued with leucine supplementation in patient cells., Conclusion: We report dominant negative variants in the KIF5B kinesin motor domain in individuals with osteogenesis imperfecta. This study expands the spectrum of kinesin-related disorders and identifies dysregulated signaling targets for KIF5B in skeletal development., Competing Interests: The authors have declared that no competing interests exist., (Copyright: © 2023 Marom et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.)
- Published
- 2023
- Full Text
- View/download PDF
48. Clinical and molecular characterization of novel FARS2 variants causing neonatal mitochondrial disease.
- Author
-
Chen W, Rehsi P, Thompson K, Yeo M, Stals K, He L, Schimmel P, Chrzanowska-Lightowlers ZMA, Wakeling E, Taylor RW, and Kuhle B
- Subjects
- Humans, Infant, Infant, Newborn, Mitochondria metabolism, Mitochondrial Proteins genetics, Mutation, RNA, Transfer genetics, RNA, Transfer, Phe metabolism, Epilepsy pathology, Mitochondrial Diseases metabolism, Phenylalanine-tRNA Ligase genetics, Phenylalanine-tRNA Ligase chemistry
- Abstract
FARS2 encodes the mitochondrial phenylalanyl-tRNA synthetase (mtPheRS), which is essential for charging mitochondrial (mt-) tRNA
Phe with phenylalanine for use in intramitochondrial translation. Many biallelic, pathogenic FARS2 variants have been described previously, which are mostly associated with two distinct clinical phenotypes; an early onset epileptic mitochondrial encephalomyopathy or a later onset spastic paraplegia. In this study, we report on a patient who presented at 3 weeks of age with tachypnoea and poor feeding, which progressed to severe metabolic decompensation with lactic acidosis and seizure activity followed by death at 9 weeks of age. Rapid trio whole exome sequencing identified compound heterozygous FARS2 variants including a pathogenic exon 2 deletion on one allele and a rare missense variant (c.593G > T, p.(Arg198Leu)) on the other allele, necessitating further work to aid variant classification. Assessment of patient fibroblasts demonstrated severely decreased steady-state levels of mtPheRS, but no obvious defect in any components of the oxidative phosphorylation system. To investigate the potential pathogenicity of the missense variant, we determined its high-resolution crystal structure, demonstrating a local structural destabilization in the catalytic domain. Moreover, the R198L mutation reduced the thermal stability and impaired the enzymatic activity of mtPheRS due to a lower binding affinity for tRNAPhe and a slower turnover rate. Together these data confirm the pathogenicity of this FARS2 variant in causing early-onset mitochondrial epilepsy., Competing Interests: Declaration of Competing Interest The authors have no conflicts of interest to disclose., (Copyright © 2023 Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
49. Heterozygous rare variants in NR2F2 cause a recognizable multiple congenital anomaly syndrome with developmental delays.
- Author
-
Ganapathi M, Matsuoka LS, March M, Li D, Brokamp E, Benito-Sanz S, White SM, Lachlan K, Ahimaz P, Sewda A, Bastarache L, Thomas-Wilson A, Stoler JM, Bramswig NC, Baptista J, Stals K, Demurger F, Cogne B, Isidor B, Bedeschi MF, Peron A, Amiel J, Zackai E, Schacht JP, Iglesias AD, Morton J, Schmetz A, Seidel V, Lucia S, Baskin SM, Thiffault I, Cogan JD, Gordon CT, Chung WK, Bowdin S, and Bhoj E
- Subjects
- Animals, Humans, COUP Transcription Factor II genetics, Muscle Hypotonia, Syndrome, Abnormalities, Multiple genetics, Abnormalities, Multiple diagnosis, Heart Defects, Congenital genetics, Hernias, Diaphragmatic, Congenital genetics, Intellectual Disability genetics
- Abstract
Nuclear receptor subfamily 2 group F member 2 (NR2F2 or COUP-TF2) encodes a transcription factor which is expressed at high levels during mammalian development. Rare heterozygous Mendelian variants in NR2F2 were initially identified in individuals with congenital heart disease (CHD), then subsequently in cohorts of congenital diaphragmatic hernia (CDH) and 46,XX ovotesticular disorders/differences of sexual development (DSD); however, the phenotypic spectrum associated with pathogenic variants in NR2F2 remains poorly characterized. Currently, less than 40 individuals with heterozygous pathogenic variants in NR2F2 have been reported. Here, we review the clinical and molecular details of 17 previously unreported individuals with rare heterozygous NR2F2 variants, the majority of which were de novo. Clinical features were variable, including intrauterine growth restriction (IUGR), CHD, CDH, genital anomalies, DSD, developmental delays, hypotonia, feeding difficulties, failure to thrive, congenital and acquired microcephaly, dysmorphic facial features, renal failure, hearing loss, strabismus, asplenia, and vascular malformations, thus expanding the phenotypic spectrum associated with NR2F2 variants. The variants seen were predicted loss of function, including a nonsense variant inherited from a mildly affected mosaic mother, missense and a large deletion including the NR2F2 gene. Our study presents evidence for rare, heterozygous NR2F2 variants causing a highly variable syndrome of congenital anomalies, commonly associated with heart defects, developmental delays/intellectual disability, dysmorphic features, feeding difficulties, hypotonia, and genital anomalies. Based on the new and previous cases, we provide clinical recommendations for evaluating individuals diagnosed with an NR2F2-associated disorder., (© 2023. The Author(s), under exclusive licence to European Society of Human Genetics.)
- Published
- 2023
- Full Text
- View/download PDF
50. Biallelic alterations in PLXND1 cause common arterial trunk and other cardiac malformations in humans.
- Author
-
Guimier A, de Pontual L, Braddock SR, Torti E, Pérez-Jurado LA, Muñoz-Cabello P, Arumí M, Monaghan KG, Lee H, Wang LK, Pluym ID, Lynch SA, Stals K, Ellard S, Muller C, Houyel L, Cohen L, Lyonnet S, Bajolle F, Amiel J, and Gordon CT
- Subjects
- Humans, Heart Defects, Congenital, Truncus Arteriosus, Persistent
- Published
- 2023
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.