75 results on '"van den Born M"'
Search Results
2. Confined placental mosaicism:Distribution of chromosomally abnormal cells over the term placenta
- Author
-
Eggenhuizen, G. M., van Veen, S., van Koetsveld, N., Go, A. T.J.I., Diderich, K. E.M., Joosten, M., van den Born, M., Srebniak, M. I., Van Opstal, D., Eggenhuizen, G. M., van Veen, S., van Koetsveld, N., Go, A. T.J.I., Diderich, K. E.M., Joosten, M., van den Born, M., Srebniak, M. I., and Van Opstal, D.
- Abstract
Objective: Non-invasive prenatal testing (NIPT) investigates placental DNA and may detect confined placental mosaicism (CPM). The aim of this study was to confirm CPM in the term placenta in cases with abnormal NIPT but normal follow-up cytogenetic studies of fetus and mother. Additionally we examined the distribution of abnormal cells over the placenta. Methods: Four chorionic villus (CV) biopsies from four placental quadrants were requested in cases where CPM was assumed. Both cell lineages of the CV, cytotrophoblast (CTB) and mesenchymal core (MC), were analyzed separately with SNP array. Results: The chromosome aberration was confirmed in 67 % of the placentas. Three quarters of the CTB and MC biopsies from these mosaic placentas were uniformly normal (57 %) or abnormal (20 %), and a minority showed mosaicism. Among 16 cases of CPM where first trimester CV were examined as well, 11 had chromosomally normal results during pregnancy. Discussion: Cytogenetic investigations of term placental biopsies suspected to be affected with CPM did not reveal the chromosome aberration in one third of the placentas. This is caused by the patchy pattern in which chromosomally abnormal cells are distributed over the placenta with the majority of the biopsies being uniformly normal. Further CPM research, including its clinical impact, requires the analysis of more than four biopsies to get insight into the extent of the affected part. Moreover, a subset of CPM type 1 and 3 seems to be only detectable with NIPT and not with first trimester CVS.
- Published
- 2024
3. Dysregulated cytokine expression in lesional and nonlesional skin in hidradenitis suppurativa
- Author
-
Kelly, G., Hughes, R., McGarry, T., van den Born, M., Adamzik, K., Fitzgerald, R., Lawlor, C., Tobin, A. M., Sweeney, C. M., and Kirby, B.
- Published
- 2015
- Full Text
- View/download PDF
4. Idiopathic noncirrhotic portal hypertension is associated with poor survival: results of a long-term cohort study
- Author
-
Schouten, J. N. L., Nevens, F., Hansen, B., Laleman, W., van den Born, M., Komuta, M., Roskams, T., Verheij, J., and Janssen, H. L. A.
- Published
- 2012
- Full Text
- View/download PDF
5. Correction to: The phenotypic spectrum of WWOX-related disorders: 20 additional cases of WOREE syndrome and review of the literature (Genetics in Medicine, (2018), 10.1038/s41436-018-0339-3)
- Author
-
Piard, J., Hawkes, L., Milh, M., Villard, L., Borgatti, Renato, Romaniello, R., Fradin, M., Capri, Y., Héron, D., Nougues, M. -C., Nava, Consuelo Rubina, Arsene, O. T., Shears, D., Taylor, J., Pagnamenta, A., Taylor, J. C., Sogawa, Y., Johnson, D., Firth, H., Vasudevan, P., Jones, G., Nguyen-Morel, M. -A., Busa, T., Roubertie, A., van den Born, M., Brischoux-Boucher, E., Koenig, M., Mignot, C., Kini, U., and Philippe, C.
- Subjects
WWOX-related disorders ,Settore M-PSI/08 - PSICOLOGIA CLINICA - Published
- 2019
6. Characterization of glycosylphosphatidylinositol biosynthesis defects by clinical features, flow cytometry, and automated image analysis
- Author
-
Knaus, A, Pantel, JT, Pendziwiat, M, Hajjir, N, Zhao, M, Hsieh, T-C, Schubach, M, Gurovich, Y, Fleischer, N, Jaeger, M, Koehler, S, Muhle, H, Korff, C, Moller, RS, Bayat, A, Calvas, P, Chassaing, N, Warren, H, Skinner, S, Louie, R, Evers, C, Bohn, M, Christen, H-J, van den Born, M, Obersztyn, E, Charzewska, A, Endziniene, M, Kortuem, F, Brown, N, Robinson, PN, Schelhaas, HJ, Weber, Y, Helbig, I, Mundlos, S, Horn, D, Krawitz, PM, Knaus, A, Pantel, JT, Pendziwiat, M, Hajjir, N, Zhao, M, Hsieh, T-C, Schubach, M, Gurovich, Y, Fleischer, N, Jaeger, M, Koehler, S, Muhle, H, Korff, C, Moller, RS, Bayat, A, Calvas, P, Chassaing, N, Warren, H, Skinner, S, Louie, R, Evers, C, Bohn, M, Christen, H-J, van den Born, M, Obersztyn, E, Charzewska, A, Endziniene, M, Kortuem, F, Brown, N, Robinson, PN, Schelhaas, HJ, Weber, Y, Helbig, I, Mundlos, S, Horn, D, and Krawitz, PM
- Abstract
BACKGROUND: Glycosylphosphatidylinositol biosynthesis defects (GPIBDs) cause a group of phenotypically overlapping recessive syndromes with intellectual disability, for which pathogenic mutations have been described in 16 genes of the corresponding molecular pathway. An elevated serum activity of alkaline phosphatase (AP), a GPI-linked enzyme, has been used to assign GPIBDs to the phenotypic series of hyperphosphatasia with mental retardation syndrome (HPMRS) and to distinguish them from another subset of GPIBDs, termed multiple congenital anomalies hypotonia seizures syndrome (MCAHS). However, the increasing number of individuals with a GPIBD shows that hyperphosphatasia is a variable feature that is not ideal for a clinical classification. METHODS: We studied the discriminatory power of multiple GPI-linked substrates that were assessed by flow cytometry in blood cells and fibroblasts of 39 and 14 individuals with a GPIBD, respectively. On the phenotypic level, we evaluated the frequency of occurrence of clinical symptoms and analyzed the performance of computer-assisted image analysis of the facial gestalt in 91 individuals. RESULTS: We found that certain malformations such as Morbus Hirschsprung and diaphragmatic defects are more likely to be associated with particular gene defects (PIGV, PGAP3, PIGN). However, especially at the severe end of the clinical spectrum of HPMRS, there is a high phenotypic overlap with MCAHS. Elevation of AP has also been documented in some of the individuals with MCAHS, namely those with PIGA mutations. Although the impairment of GPI-linked substrates is supposed to play the key role in the pathophysiology of GPIBDs, we could not observe gene-specific profiles for flow cytometric markers or a correlation between their cell surface levels and the severity of the phenotype. In contrast, it was facial recognition software that achieved the highest accuracy in predicting the disease-causing gene in a GPIBD. CONCLUSIONS: Due to the overlap
- Published
- 2018
7. Diabetes Risk Gene and Wnt Effector Tcf7l2/TCF4 Controls Hepatic Response to Perinatal and Adult Metabolic Demand
- Author
-
Boj, S.F., van Es, J.H., Huch, M., Li, V.S., Jose, A., Hatzis, P., Mokry, M., Haegebarth, A., van den Born, M., Chambon, P., Voshol, P., Dor, Y., Cuppen, E., Fillat, C., Clevers, H., Boj, S.F., van Es, J.H., Huch, M., Li, V.S., Jose, A., Hatzis, P., Mokry, M., Haegebarth, A., van den Born, M., Chambon, P., Voshol, P., Dor, Y., Cuppen, E., Fillat, C., and Clevers, H.
- Abstract
Most studies on TCF7L2 SNP variants in the pathogenesis of type 2 diabetes (T2D) focus on a role of the encoded transcription factor TCF4 in beta cells. Here, a mouse genetics approach shows that removal of TCF4 from beta cells does not affect their function, whereas manipulating TCF4 levels in the liver has major effects on metabolism. In Tcf7l2(-/-) mice, the immediate postnatal surge in liver metabolism does not occur. Consequently, pups die due to hypoglycemia. By combining chromatin immunoprecipitation with gene expression profiling, we identify a TCF4-controlled metabolic gene program that is acutely activated in the postnatal liver. In concordance, adult liver-specific Tcf7l2 knockout mice show reduced hepatic glucose production during fasting and display improved glucose homeostasis when maintained on high-fat diet. Furthermore, liver-specific TCF4 overexpression increases hepatic glucose production. These observations imply that TCF4 directly activates metabolic genes and that inhibition of Wnt signaling may be beneficial in metabolic disease., Most studies on TCF7L2 SNP variants in the pathogenesis of type 2 diabetes (T2D) focus on a role of the encoded transcription factor TCF4 in beta cells. Here, a mouse genetics approach shows that removal of TCF4 from beta cells does not affect their function, whereas manipulating TCF4 levels in the liver has major effects on metabolism. In Tcf7l2(-/-) mice, the immediate postnatal surge in liver metabolism does not occur. Consequently, pups die due to hypoglycemia. By combining chromatin immunoprecipitation with gene expression profiling, we identify a TCF4-controlled metabolic gene program that is acutely activated in the postnatal liver. In concordance, adult liver-specific Tcf7l2 knockout mice show reduced hepatic glucose production during fasting and display improved glucose homeostasis when maintained on high-fat diet. Furthermore, liver-specific TCF4 overexpression increases hepatic glucose production. These observations imply that TCF4 directly activates metabolic genes and that inhibition of Wnt signaling may be beneficial in metabolic disease.
- Published
- 2012
8. A critical role for the Wnt effector Tcf4 in adult intestinal homeostatic self-renewal
- Author
-
van Es, J.H., Haegebarth, A., Kujala, P., Itzkovitz, S., Koo, B.K., Boj, S.F., Korving, J., van den Born, M., van Oudenaarden, A., Robine, S., Clevers, H., van Es, J.H., Haegebarth, A., Kujala, P., Itzkovitz, S., Koo, B.K., Boj, S.F., Korving, J., van den Born, M., van Oudenaarden, A., Robine, S., and Clevers, H.
- Abstract
Throughout life, intestinal Lgr5+ stem cells give rise to proliferating transient amplifying cells in crypts, which subsequently differentiate into one of the five main cell types and migrate along the crypt-villus axis. These dynamic processes are coordinated by a relatively small number of evolutionarily conserved signaling pathways, which includes the Wnt signaling pathway. The DNA-binding proteins of the T-cell factor family, Tcf1/Tcf7, Lef, Tcf3/Tcf7l1, and Tcf4/Tcf7l2, constitute the downstream effectors of the Wnt signaling pathway. While Tcf4 is the major member active during embryogenesis, the role of these Wnt effectors in the homeostasis of the adult mouse intestinal epithelium is unresolved. Using Tcf1-/-, Tcf3(flox), and novel Tcf4(flox) mice, we demonstrate an essential role for Tcf4 during homeostasis of the adult mouse intestine., Throughout life, intestinal Lgr5+ stem cells give rise to proliferating transient amplifying cells in crypts, which subsequently differentiate into one of the five main cell types and migrate along the crypt-villus axis. These dynamic processes are coordinated by a relatively small number of evolutionarily conserved signaling pathways, which includes the Wnt signaling pathway. The DNA-binding proteins of the T-cell factor family, Tcf1/Tcf7, Lef, Tcf3/Tcf7l1, and Tcf4/Tcf7l2, constitute the downstream effectors of the Wnt signaling pathway. While Tcf4 is the major member active during embryogenesis, the role of these Wnt effectors in the homeostasis of the adult mouse intestinal epithelium is unresolved. Using Tcf1-/-, Tcf3(flox), and novel Tcf4(flox) mice, we demonstrate an essential role for Tcf4 during homeostasis of the adult mouse intestine.
- Published
- 2012
9. Lineage tracing reveals Lgr5+ stem cell activity in mouse intestinal adenomas
- Author
-
Schepers, A.G., Snippert, H.J.G., Stange, D.E., van den Born, M., van Es, J.H., van de Wetering, M., Clevers, H., Schepers, A.G., Snippert, H.J.G., Stange, D.E., van den Born, M., van Es, J.H., van de Wetering, M., and Clevers, H.
- Abstract
The concept that tumors are maintained by dedicated stem cells, the so-called cancer stem cell hypothesis, has attracted great interest but remains controversial. Studying mouse models, we provide direct, functional evidence for the presence of stem cell activity within primary intestinal adenomas, a precursor to intestinal cancer. By "lineage retracing" using the multicolor Cre-reporter R26R-Confetti, we demonstrate that the crypt stem cell marker Lgr5 (leucine-rich repeat-containing heterotrimeric guanine nucleotide-binding protein-coupled receptor 5) also marks a subpopulation of adenoma cells that fuel the growth of established intestinal adenomas. These Lgr5(+) cells, which represent about 5 to 10% of the cells in the adenomas, generate additional Lgr5(+) cells as well as all other adenoma cell types. The Lgr5(+) cells are intermingled with Paneth cells near the adenoma base, a pattern reminiscent of the architecture of the normal crypt niche., The concept that tumors are maintained by dedicated stem cells, the so-called cancer stem cell hypothesis, has attracted great interest but remains controversial. Studying mouse models, we provide direct, functional evidence for the presence of stem cell activity within primary intestinal adenomas, a precursor to intestinal cancer. By "lineage retracing" using the multicolor Cre-reporter R26R-Confetti, we demonstrate that the crypt stem cell marker Lgr5 (leucine-rich repeat-containing heterotrimeric guanine nucleotide-binding protein-coupled receptor 5) also marks a subpopulation of adenoma cells that fuel the growth of established intestinal adenomas. These Lgr5(+) cells, which represent about 5 to 10% of the cells in the adenomas, generate additional Lgr5(+) cells as well as all other adenoma cell types. The Lgr5(+) cells are intermingled with Paneth cells near the adenoma base, a pattern reminiscent of the architecture of the normal crypt niche.
- Published
- 2012
10. Dll1+ secretory progenitor cells revert to stem cells upon crypt damage
- Author
-
van Es, J.H., Sato, T., van de Wetering, M., Lyubimova, A., Nee, A.N., Gregorieff, A., Sasaki, N., Zeinstra, L., van den Born, M., Korving, J., Martens, A.C., Barker, N., van Oudenaarden, A., Clevers, H., van Es, J.H., Sato, T., van de Wetering, M., Lyubimova, A., Nee, A.N., Gregorieff, A., Sasaki, N., Zeinstra, L., van den Born, M., Korving, J., Martens, A.C., Barker, N., van Oudenaarden, A., and Clevers, H.
- Abstract
Lgr5+ intestinal stem cells generate enterocytes and secretory cells. Secretory lineage commitment requires Notch silencing. The Notch ligand Dll1 is expressed by a subset of immediate stem cell daughters. Lineage tracing in Dll1(GFP-ires-CreERT2) knock-in mice reveals that single Dll1(high) cells generate small, short-lived clones containing all four secretory cell types. Lineage specification thus occurs in immediate stem cell daughters through Notch lateral inhibition. Cultured Dll1(high) cells form long-lived organoids (mini-guts) on brief Wnt3A exposure. When Dll1(high) cells are genetically marked before tissue damage, stem cell tracing events occur. Thus, secretory progenitors exhibit plasticity by regaining stemness on damage., Lgr5+ intestinal stem cells generate enterocytes and secretory cells. Secretory lineage commitment requires Notch silencing. The Notch ligand Dll1 is expressed by a subset of immediate stem cell daughters. Lineage tracing in Dll1(GFP-ires-CreERT2) knock-in mice reveals that single Dll1(high) cells generate small, short-lived clones containing all four secretory cell types. Lineage specification thus occurs in immediate stem cell daughters through Notch lateral inhibition. Cultured Dll1(high) cells form long-lived organoids (mini-guts) on brief Wnt3A exposure. When Dll1(high) cells are genetically marked before tissue damage, stem cell tracing events occur. Thus, secretory progenitors exhibit plasticity by regaining stemness on damage.
- Published
- 2012
11. The BMP antagonist Follistatin-Like 1 is required for skeletal and lung organogenesis
- Author
-
Sylva, M., Li, V.S.W., Buffing, A.A.A., van Es, J.H., van den Born, M., van der Velden, S., Gunst, Q., Koolstra, J.H., Moorman, A.F.M., Clevers, H., van den Hoff, M.J.B., Sylva, M., Li, V.S.W., Buffing, A.A.A., van Es, J.H., van den Born, M., van der Velden, S., Gunst, Q., Koolstra, J.H., Moorman, A.F.M., Clevers, H., and van den Hoff, M.J.B.
- Published
- 2011
- Full Text
- View/download PDF
12. The High Diagnostic Yield of Prenatal Exome Sequencing Followed by 3400 Gene Panel Analysis in 629 Ongoing Pregnancies With Ultrasound Anomalies.
- Author
-
Diderich KEM, Bruggenwirth HT, Joosten M, Thurik F, Mijalkovic J, Polak M, Kromosoeto J, Somers-Bolman GM, van den Born M, Drost M, Galjaard RJH, Galjaard S, Hoefsloot LH, Knapen MFCM, van Minkelen R, van der Schoot V, van Slegtenhorst MA, Sleutels F, Stuurman KE, Weerts MJA, Go ATJI, Wilke M, and Srebniak MI
- Subjects
- Humans, Female, Pregnancy, Retrospective Studies, Adult, Abnormalities, Multiple genetics, Abnormalities, Multiple diagnostic imaging, Abnormalities, Multiple diagnosis, Prenatal Diagnosis methods, Prenatal Diagnosis statistics & numerical data, Genetic Testing methods, Exome Sequencing statistics & numerical data, Exome Sequencing methods, Ultrasonography, Prenatal
- Abstract
Background: The aim of this study was to evaluate the diagnostic yield of routine exome sequencing (ES) in fetuses with ultrasound anomalies., Methods: We performed a retrospective analysis of the ES results of 629 fetuses with isolated or multiple anomalies referred in 2019-2022. Variants in a gene panel consisting of approximately 3400 genes associated with multiple congenital anomalies and/or intellectual disability were analyzed. We used trio analysis and filtering for de novo variants, compound heterozygous variants, homozygous variants, X-linked variants, variants in imprinted genes, and known pathogenic variants., Results: Pathogenic and likely pathogenic variants (class five and four, respectively) were identified in 14.0% (88/629, 95% CI 11.5%-16.9%) of cases. In the current cohort, the probability of detecting a monogenetic disorder was ∼1:7 (88/629, 95% CI 1:8.7-1:5.9), ranging from 1:9 (49/424) in cases with one major anomaly to 1:5 (32/147) in cases with multiple system anomalies., Conclusions: Our results indicate that a notable number of fetuses (1:7) with ultrasound anomalies and a normal chromosomal microarray have a (likely) pathogenic variant that can be detected through prenatal ES. These results warrant implementation of exome sequencing in selected cases, including those with an isolated anomaly on prenatal ultrasound., (© 2024 The Author(s). Prenatal Diagnosis published by John Wiley & Sons Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
13. MSL2 variants lead to a neurodevelopmental syndrome with lack of coordination, epilepsy, specific dysmorphisms, and a distinct episignature.
- Author
-
Karayol R, Borroto MC, Haghshenas S, Namasivayam A, Reilly J, Levy MA, Relator R, Kerkhof J, McConkey H, Shvedunova M, Petersen AK, Magnussen K, Zweier C, Vasileiou G, Reis A, Savatt JM, Mulligan MR, Bicknell LS, Poke G, Abu-El-Haija A, Duis J, Hannig V, Srivastava S, Barkoudah E, Hauser NS, van den Born M, Hamiel U, Henig N, Baris Feldman H, McKee S, Krapels IPC, Lei Y, Todorova A, Yordanova R, Atemin S, Rogac M, McConnell V, Chassevent A, Barañano KW, Shashi V, Sullivan JA, Peron A, Iascone M, Canevini MP, Friedman J, Reyes IA, Kierstein J, Shen JJ, Ahmed FN, Mao X, Almoguera B, Blanco-Kelly F, Platzer K, Treu AB, Quilichini J, Bourgois A, Chatron N, Januel L, Rougeot C, Carere DA, Monaghan KG, Rousseau J, Myers KA, Sadikovic B, Akhtar A, and Campeau PM
- Subjects
- Adolescent, Child, Child, Preschool, Female, Humans, Male, Developmental Disabilities genetics, DNA Methylation genetics, Epigenesis, Genetic, Histones metabolism, Histones genetics, Induced Pluripotent Stem Cells metabolism, Intellectual Disability genetics, Phenotype, Epilepsy genetics, Neurodevelopmental Disorders genetics, Ubiquitin-Protein Ligases metabolism
- Abstract
Epigenetic dysregulation has emerged as an important etiological mechanism of neurodevelopmental disorders (NDDs). Pathogenic variation in epigenetic regulators can impair deposition of histone post-translational modifications leading to aberrant spatiotemporal gene expression during neurodevelopment. The male-specific lethal (MSL) complex is a prominent multi-subunit epigenetic regulator of gene expression and is responsible for histone 4 lysine 16 acetylation (H4K16ac). Using exome sequencing, here we identify a cohort of 25 individuals with heterozygous de novo variants in MSL complex member MSL2. MSL2 variants were associated with NDD phenotypes including global developmental delay, intellectual disability, hypotonia, and motor issues such as coordination problems, feeding difficulties, and gait disturbance. Dysmorphisms and behavioral and/or psychiatric conditions, including autism spectrum disorder, and to a lesser extent, seizures, connective tissue disease signs, sleep disturbance, vision problems, and other organ anomalies, were observed in affected individuals. As a molecular biomarker, a sensitive and specific DNA methylation episignature has been established. Induced pluripotent stem cells (iPSCs) derived from three members of our cohort exhibited reduced MSL2 levels. Remarkably, while NDD-associated variants in two other members of the MSL complex (MOF and MSL3) result in reduced H4K16ac, global H4K16ac levels are unchanged in iPSCs with MSL2 variants. Regardless, MSL2 variants altered the expression of MSL2 targets in iPSCs and upon their differentiation to early germ layers. Our study defines an MSL2-related disorder as an NDD with distinguishable clinical features, a specific blood DNA episignature, and a distinct, MSL2-specific molecular etiology compared to other MSL complex-related disorders., Competing Interests: Declaration of interests B.S. is a shareholder in EpiSign Inc, involved in commercial uses of EpiSign(TM) technology D.A.C. and K.G.M. are employees of GeneDx, LLC., (Copyright © 2024. Published by Elsevier Inc.)
- Published
- 2024
- Full Text
- View/download PDF
14. Limited additional value of karyotyping cultured amniotic fluid cell colonies in addition to microarray on uncultured cells for confirmation of abnormal non-invasive prenatal testing results.
- Author
-
Donze SH, Srebniak MI, Diderich KEM, van den Born M, Galjaard RJ, Govaerts LCP, van der Schoot V, Knapen MFCM, Joosten M, and Van Opstal D
- Subjects
- Pregnancy, Female, Humans, In Situ Hybridization, Fluorescence, Trisomy 13 Syndrome, Retrospective Studies, Placenta, Amniocentesis methods, Trisomy, Karyotyping, Mosaicism, Cells, Cultured, Prenatal Diagnosis methods, Amniotic Fluid
- Abstract
Objectives: Non-invasive prenatal testing (NIPT) allows the detection of placental chromosome aberrations. To verify whether the fetus also has the chromosome aberration, diagnostic follow-up testing is required. The aim of this retrospective study was to assess the added value of analyzing amniotic fluid (AF) cell cultures in addition to uncultured AF cells for the detection of fetal mosaicism., Method: NIPT was performed as part of the Dutch TRIDENT study. Cytogenetic studies in uncultured AF were performed using single nucleotide polymorphism (SNP)-array. Cultured AF cell colonies (in situ method) were investigated with fluorescent in situ hybridization and/or karyotyping. Clinical outcome data were collected in cases with discordant results., Results: Between April 2014 and December 2021, 368 amniocenteses were performed after a chromosomal aberration was detected with NIPT. Excluding 134 cases of common aneuploidies (confirmed by quantitative fluorescence polymerase chain reaction), 29 cases with investigation of uncultured cells only and 1 case without informed consent, 204 cases were eligible for this study. In 196 (96%) cases, the results in uncultured and cultured cells were concordant normal, abnormal or mosaic. Five cases (2%) showed mosaicism in cultured AF cells, whereas uncultured AF cells were normal. Two (1%) of these, one mosaic trisomy 13 and one mosaic trisomy 16, were considered true fetal mosaics., Conclusion: The added value of investigating AF cell cultures in addition to uncultured cells is limited to two of 204 (1%) cases in which true fetal mosaicsm would otherwise be missed. The clinical relevance of one (trisomy 13) remained unknown and the other case also showed ultrasound anomalies, which determined pregnancy management. This seems to justify limiting prenatal cytogenetic confirmatory testing to SNP arrays on uncultured AF cells, considerably shortening the reporting time., (© 2023 The Authors. Prenatal Diagnosis published by John Wiley & Sons Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
15. The role of confined placental mosaicism in fetal growth restriction: A retrospective cohort study.
- Author
-
Eggenhuizen GM, Go ATJI, Sauter Z, Hoffer MJV, Haak MC, Geeven G, Diderich KEM, Joosten M, van den Born M, Srebniak MI, and Van Opstal D
- Subjects
- Pregnancy, Female, Humans, Mosaicism, Fetal Growth Retardation diagnosis, Fetal Growth Retardation genetics, Cohort Studies, Birth Weight, Retrospective Studies, Chromosomes, Human, Pair 16, Placenta metabolism, Trisomy diagnosis, Trisomy genetics
- Abstract
Objective: To evaluate which cytogenetic characteristics of confined placental mosaicism (CPM) detected in the first trimester chorionic villi and/or placentas in terms of chromosome aberration, cell lineage involved and trisomy origin will lead to fetal growth restriction and low birthweight., Methods: Cohort study using routinely collected perinatal data and cytogenetic data of non-invasive prenatal testing, the first trimester chorionic villi sampling and postnatal placentas., Results: 215 CPM cases were found. Fetal growth restriction (FGR) and low birthweight below the 10
th percentile (BW < p10) were seen in 34.0% and 23.1%, respectively. Excluding cases of trisomy 16, 29.1% showed FGR and 17.9% had a BW < p10. The highest rate of FGR and BW < p10 was found in CPM type 3, but differences with type 1 and 2 were not significant. FGR and BW < p10 were significantly more often observed in cases with meiotic trisomies., Conclusion: There is an association between CPM and FGR and BW < p10. This association is not restricted to trisomy 16, neither to CPM type 3, nor to CPM involving a meiotic trisomy. Pregnancies with all CPM types and origins should be considered to be at increased risk of FGR and low BW < p10. A close prenatal fetal monitoring is indicated in all cases of CPM., (© 2024 The Authors. Prenatal Diagnosis published by John Wiley & Sons Ltd.)- Published
- 2024
- Full Text
- View/download PDF
16. Response to the comment on Diderich et al. "The role of a multidisciplinary team in managing variants of uncertain clinical significance in prenatal genetic diagnosis" (EJMG 66(10),104844).
- Author
-
Diderich KEM, Klapwijk JE, van der Schoot V, van den Born M, Wilke M, Joosten M, Stuurman KE, Hoefsloot LH, Van Opstal D, Brüggenwirth HT, and Srebniak MI
- Subjects
- Pregnancy, Female, Humans, Genetic Counseling, Patient Care Team, Clinical Relevance, Prenatal Diagnosis
- Abstract
Competing Interests: Declaration of competing interest All authors declare no conflict of interest.
- Published
- 2024
- Full Text
- View/download PDF
17. The role of a multidisciplinary team in managing variants of uncertain clinical significance in prenatal genetic diagnosis.
- Author
-
Diderich KEM, Klapwijk JE, van der Schoot V, van den Born M, Wilke M, Joosten M, Stuurman KE, Hoefsloot LH, Van Opstal D, Brüggenwirth HT, and Srebniak MI
- Subjects
- Female, Humans, Pregnancy, Patient Care Team, Prenatal Diagnosis, Prospective Studies, Retrospective Studies, Clinical Relevance, Genetic Testing
- Abstract
Background: Although in general prenatal exome sequencing only reports (likely) pathogenic variants, in some cases a variant of uncertain significance (VUS) is disclosed. The aims of this retrospective study were to evaluate the types of VUS that have been reported to prospective parents, possible reclassification and to design a standard flow chart to determine which types of VUS could be considered for reporting in prenatal settings. Furthermore, we investigated what the crucial elements are to facilitate rapid management of uncertain results in a prenatal setting., Material and Methods: We reviewed exome results from 451 pregnancies performed in 2019-2021. We analyzed which factors that were taken into account by the multidisciplinary team (MDT) contributed towards decision making on reporting VUS after prenatal exome sequencing., Results: In 9/451 (2%) pregnancies tested with exome sequencing using a broad panel analysis a VUS was reported. After birth 3/9 VUS could be reclassified to likely pathogenic variants based on new clinical follow up data. We considered reporting VUS in genes: 1) matching the fetal phenotype, 2) associated with a severe disorder when a functional test is available or 3) possibly associated with a disorder where early post-partum diagnosis and treatment are crucial for a better prognosis. Two flowcharts were designed to guide first the laboratory specialist and then the MDT in decisions on reporting VUS. The crucial elements that enabled timely decisions on VUS disclosure were regular meetings, appropriate expertise, professional connections with other experts and psychological safety within the MDT., Conclusion: In this study three out of nine VUS could be re-classified as likely pathogenic after clinical follow-up. In order to protect pregnant couples from the burden of uncertain results, the genetic professionals have to take the responsibility to limit the reporting of VUS. This can be done not only by automated filtering of data, by following professional guidelines and by building standardized decision flows, but also by discussing individual cases considering personal situations and the involved disease and by sharing professional experience and responsibility in a multidisciplinary prenatal team setting., Competing Interests: Declaration of competing interest All authors declare no conflict of interests., (Copyright © 2023. Published by Elsevier Masson SAS.)
- Published
- 2023
- Full Text
- View/download PDF
18. Discovering a new part of the phenotypic spectrum of Coffin-Siris syndrome in a fetal cohort.
- Author
-
van der Sluijs PJ, Joosten M, Alby C, Attié-Bitach T, Gilmore K, Dubourg C, Fradin M, Wang T, Kurtz-Nelson EC, Ahlers KP, Arts P, Barnett CP, Ashfaq M, Baban A, van den Born M, Borrie S, Busa T, Byrne A, Carriero M, Cesario C, Chong K, Cueto-González AM, Dempsey JC, Diderich KEM, Doherty D, Farholt S, Gerkes EH, Gorokhova S, Govaerts LCP, Gregersen PA, Hickey SE, Lefebvre M, Mari F, Martinovic J, Northrup H, O'Leary M, Parbhoo K, Patrier S, Popp B, Santos-Simarro F, Stoltenburg C, Thauvin-Robinet C, Thompson E, Vulto-van Silfhout AT, Zahir FR, Scott HS, Earl RK, Eichler EE, Vora NL, Wilnai Y, Giordano JL, Wapner RJ, Rosenfeld JA, Haak MC, and Santen GWE
- Published
- 2023
- Full Text
- View/download PDF
19. Chromosomal mosaicism in human blastocysts: a cytogenetic comparison of trophectoderm and inner cell mass after next-generation sequencing.
- Author
-
Chavli E, van den Born M, Eleveld C, Boter M, van Marion R, Hoefsloot L, Laven J, Baart E, and Van Opstal D
- Subjects
- Pregnancy, Female, Humans, Mosaicism, Aneuploidy, Blastocyst, High-Throughput Nucleotide Sequencing, Cytogenetic Analysis, Genetic Testing, Preimplantation Diagnosis
- Abstract
Research Question: What is the incidence of chromosomal mosaicism in human blastocysts and can a single trophectoderm (TE) biopsy accurately predict the chromosomal constitution of the inner cell mass (ICM)?, Design: Observational study in 46 surplus cryopreserved preimplantation embryos of unknown chromosomal constitution. For each embryo, a TE biopsy was performed and the ICM was collected separately. Both samples underwent next-generation sequencing (NGS) for cytogenetic analysis and were classified as chromosomally normal, abnormal or mosaic. Mosaic samples were classified as low or high mosaic, based on the majority dominance of either normal or abnormal cells in the biopsied sample. Findings within each embryo were compared., Results: Chromosomal mosaicism was detected in 59% (n = 27/46) of the embryos, with a cytogenetic concordance rate between TE and corresponding ICM of 48% (n = 22/46). Concordance was higher from a clinical perspective: in 86% of embryos with a high-mosaic or abnormal TE, the ICM was also high-mosaic or abnormal. In 88% of the blastocysts with a normal or low-mosaic TE biopsy, a normal or low-mosaic ICM was observed., Conclusion: Despite the low cytogenetic concordance rate due to chromosomal mosaicism present in blastocysts, it was found that a single TE biopsy could correctly predict whether the ICM consists of mostly normal or abnormal cells in the majority of cases., (Copyright © 2022 The Author(s). Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
20. Biallelic KIF24 Variants Are Responsible for a Spectrum of Skeletal Disorders Ranging From Lethal Skeletal Ciliopathy to Severe Acromesomelic Dysplasia.
- Author
-
Reilly ML, Ain NU, Muurinen M, Tata A, Huber C, Simon M, Ishaq T, Shaw N, Rusanen S, Pekkinen M, Högler W, Knapen MFCM, van den Born M, Saunier S, Naz S, Cormier-Daire V, Benmerah A, and Makitie O
- Subjects
- Animals, Humans, Mutation genetics, Pedigree, Phenotype, Ciliopathies diagnostic imaging, Ciliopathies genetics, Dwarfism diagnostic imaging, Dwarfism genetics, Osteochondrodysplasias diagnostic imaging, Osteochondrodysplasias genetics
- Abstract
Skeletal dysplasias comprise a large spectrum of mostly monogenic disorders affecting bone growth, patterning, and homeostasis, and ranging in severity from lethal to mild phenotypes. This study aimed to underpin the genetic cause of skeletal dysplasia in three unrelated families with variable skeletal manifestations. The six affected individuals from three families had severe short stature with extreme shortening of forelimbs, short long-bones, and metatarsals, and brachydactyly (family 1); mild short stature, platyspondyly, and metaphyseal irregularities (family 2); or a prenatally lethal skeletal dysplasia with kidney features suggestive of a ciliopathy (family 3). Genetic studies by whole genome, whole exome, and ciliome panel sequencing identified in all affected individuals biallelic missense variants in KIF24, which encodes a kinesin family member controlling ciliogenesis. In families 1 and 3, with the more severe phenotype, the affected subjects harbored homozygous variants (c.1457A>G; p.(Ile486Val) and c.1565A>G; p.(Asn522Ser), respectively) in the motor domain which plays a crucial role in KIF24 function. In family 2, compound heterozygous variants (c.1697C>T; p.(Ser566Phe)/c.1811C>T; p.(Thr604Met)) were found C-terminal to the motor domain, in agreement with a genotype-phenotype correlation. In vitro experiments performed on amnioblasts of one affected fetus from family 3 showed that primary cilia assembly was severely impaired, and that cytokinesis was also affected. In conclusion, our study describes novel forms of skeletal dysplasia associated with biallelic variants in KIF24. To our knowledge this is the first report implicating KIF24 variants as the cause of a skeletal dysplasia, thereby extending the genetic heterogeneity and the phenotypic spectrum of rare bone disorders and underscoring the wide range of monogenetic skeletal ciliopathies. © 2022 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR)., (© 2022 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).)
- Published
- 2022
- Full Text
- View/download PDF
21. Discovering a new part of the phenotypic spectrum of Coffin-Siris syndrome in a fetal cohort.
- Author
-
van der Sluijs PJ, Joosten M, Alby C, Attié-Bitach T, Gilmore K, Dubourg C, Fradin M, Wang T, Kurtz-Nelson EC, Ahlers KP, Arts P, Barnett CP, Ashfaq M, Baban A, van den Born M, Borrie S, Busa T, Byrne A, Carriero M, Cesario C, Chong K, Cueto-González AM, Dempsey JC, Diderich KEM, Doherty D, Farholt S, Gerkes EH, Gorokhova S, Govaerts LCP, Gregersen PA, Hickey SE, Lefebvre M, Mari F, Martinovic J, Northrup H, O'Leary M, Parbhoo K, Patrier S, Popp B, Santos-Simarro F, Stoltenburg C, Thauvin-Robinet C, Thompson E, Vulto-van Silfhout AT, Zahir FR, Scott HS, Earl RK, Eichler EE, Vora NL, Wilnai Y, Giordano JL, Wapner RJ, Rosenfeld JA, Haak MC, and Santen GWE
- Subjects
- Abnormalities, Multiple, Chromosomal Proteins, Non-Histone genetics, Face abnormalities, Genetic Association Studies, Humans, Neck abnormalities, Phenotype, Hand Deformities, Congenital genetics, Intellectual Disability genetics, Intellectual Disability pathology, Micrognathism genetics
- Abstract
Purpose: Genome-wide sequencing is increasingly being performed during pregnancy to identify the genetic cause of congenital anomalies. The interpretation of prenatally identified variants can be challenging and is hampered by our often limited knowledge of prenatal phenotypes. To better delineate the prenatal phenotype of Coffin-Siris syndrome (CSS), we collected clinical data from patients with a prenatal phenotype and a pathogenic variant in one of the CSS-associated genes., Methods: Clinical data was collected through an extensive web-based survey., Results: We included 44 patients with a variant in a CSS-associated gene and a prenatal phenotype; 9 of these patients have been reported before. Prenatal anomalies that were frequently observed in our cohort include hydrocephalus, agenesis of the corpus callosum, hypoplastic left heart syndrome, persistent left vena cava, diaphragmatic hernia, renal agenesis, and intrauterine growth restriction. Anal anomalies were frequently identified after birth in patients with ARID1A variants (6/14, 43%). Interestingly, pathogenic ARID1A variants were much more frequently identified in the current prenatal cohort (16/44, 36%) than in postnatal CSS cohorts (5%-9%)., Conclusion: Our data shed new light on the prenatal phenotype of patients with pathogenic variants in CSS genes., Competing Interests: Conflict of Interest All authors declare no conflicts of interest., (Copyright © 2022 American College of Medical Genetics and Genomics. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
22. Multi-Omics Profiling in Marfan Syndrome: Further Insights into the Molecular Mechanisms Involved in Aortic Disease.
- Author
-
Verhagen JMA, Burger J, Bekkers JA, den Dekker AT, von der Thüsen JH, Zajec M, Brüggenwirth HT, van der Sterre MLT, van den Born M, Luider TM, van IJcken WFJ, Wessels MW, Essers J, Roos-Hesselink JW, van der Pluijm I, van de Laar IMBH, and Brosens E
- Subjects
- Adult, Animals, Aorta metabolism, Aorta pathology, Aortic Diseases pathology, Cell Respiration, Female, Fibrillin-1 metabolism, Gene Expression Profiling, Gene Expression Regulation, Humans, Male, Marfan Syndrome pathology, Muscle, Smooth, Vascular pathology, Myocytes, Smooth Muscle metabolism, Myocytes, Smooth Muscle pathology, Signal Transduction, Transforming Growth Factor beta metabolism, Aortic Diseases genetics, Genomics, Marfan Syndrome genetics
- Abstract
Thoracic aortic aneurysm is a potentially life-threatening disease with a strong genetic contribution. Despite identification of multiple genes involved in aneurysm formation, little is known about the specific underlying mechanisms that drive the pathological changes in the aortic wall. The aim of our study was to unravel the molecular mechanisms underlying aneurysm formation in Marfan syndrome (MFS). We collected aortic wall samples from FBN1 variant-positive MFS patients (n = 6) and healthy donor hearts (n = 5). Messenger RNA (mRNA) expression levels were measured by RNA sequencing and compared between MFS patients and controls, and between haploinsufficient (HI) and dominant negative (DN) FBN1 variants. Immunohistochemical staining, proteomics and cellular respiration experiments were used to confirm our findings. FBN1 mRNA expression levels were highly variable in MFS patients and did not significantly differ from controls. Moreover, we did not identify a distinctive TGF-β gene expression signature in MFS patients. On the contrary, differential gene and protein expression analysis, as well as vascular smooth muscle cell respiration measurements, pointed toward inflammation and mitochondrial dysfunction. Our findings confirm that inflammatory and mitochondrial pathways play important roles in the pathophysiological processes underlying MFS-related aortic disease, providing new therapeutic options.
- Published
- 2021
- Full Text
- View/download PDF
23. Treatment of ARS deficiencies with specific amino acids.
- Author
-
Kok G, Tseng L, Schene IF, Dijsselhof ME, Salomons G, Mendes MI, Smith DEC, Wiedemann A, Canton M, Feillet F, de Koning TJ, Boothe M, Dean J, Kassel R, Ferreira EA, van den Born M, Nieuwenhuis EES, Rehmann H, Terheggen-Lagro SWJ, van Karnebeek CDM, and Fuchs SA
- Subjects
- Amino Acids, Aminoacylation, Humans, RNA, Transfer metabolism, Amino Acyl-tRNA Synthetases genetics
- Abstract
Purpose: Recessive cytosolic aminoacyl-tRNA synthetase (ARS) deficiencies are severe multiorgan diseases, with limited treatment options. By loading transfer RNAs (tRNAs) with their cognate amino acids, ARS are essential for protein translation. However, it remains unknown why ARS deficiencies lead to specific symptoms, especially early life and during infections. We set out to increase pathophysiological insight and improve therapeutic possibilities., Methods: In fibroblasts from patients with isoleucyl-RS (IARS), leucyl-RS (LARS), phenylalanyl-RS-beta-subunit (FARSB), and seryl-RS (SARS) deficiencies, we investigated aminoacylation activity, thermostability, and sensitivity to ARS-specific amino acid concentrations, and developed personalized treatments., Results: Aminoacylation activity was reduced in all patients, and further diminished at 38.5/40 °C (P
LARS and PFARSB ), consistent with infectious deteriorations. With lower cognate amino acid concentrations, patient fibroblast growth was severely affected. To prevent local and/or temporal deficiencies, we treated patients with corresponding amino acids (follow-up: 1/2-2 2/3rd years), and intensified treatment during infections. All patients showed beneficial treatment effects, most strikingly in growth (without tube feeding), head circumference, development, coping with infections, and oxygen dependency., Conclusion: For these four ARS deficiencies, we observed a common disease mechanism of episodic insufficient aminoacylation to meet translational demands and illustrate the power of amino acid supplementation for the expanding ARS patient group. Moreover, we provide a strategy for personalized preclinical functional evaluation., (© 2021. The Author(s), under exclusive licence to the American College of Medical Genetics and Genomics.)- Published
- 2021
- Full Text
- View/download PDF
24. De Novo Variants in CNOT1, a Central Component of the CCR4-NOT Complex Involved in Gene Expression and RNA and Protein Stability, Cause Neurodevelopmental Delay.
- Author
-
Vissers LELM, Kalvakuri S, de Boer E, Geuer S, Oud M, van Outersterp I, Kwint M, Witmond M, Kersten S, Polla DL, Weijers D, Begtrup A, McWalter K, Ruiz A, Gabau E, Morton JEV, Griffith C, Weiss K, Gamble C, Bartley J, Vernon HJ, Brunet K, Ruivenkamp C, Kant SG, Kruszka P, Larson A, Afenjar A, Billette de Villemeur T, Nugent K, Raymond FL, Venselaar H, Demurger F, Soler-Alfonso C, Li D, Bhoj E, Hayes I, Hamilton NP, Ahmad A, Fisher R, van den Born M, Willems M, Sorlin A, Delanne J, Moutton S, Christophe P, Mau-Them FT, Vitobello A, Goel H, Massingham L, Phornphutkul C, Schwab J, Keren B, Charles P, Vreeburg M, De Simone L, Hoganson G, Iascone M, Milani D, Evenepoel L, Revencu N, Ward DI, Burns K, Krantz I, Raible SE, Murrell JR, Wood K, Cho MT, van Bokhoven H, Muenke M, Kleefstra T, Bodmer R, and de Brouwer APM
- Subjects
- Alleles, Female, Genetic Variation genetics, Haploinsufficiency genetics, Heterozygote, Humans, Male, Nervous System Malformations genetics, Phenotype, Protein Stability, Developmental Disabilities genetics, Gene Expression genetics, Neurodevelopmental Disorders genetics, Nuclear Receptor Subfamily 4, Group A, Member 2 genetics, RNA genetics, Receptors, CCR4 genetics, Transcription Factors genetics
- Abstract
CNOT1 is a member of the CCR4-NOT complex, which is a master regulator, orchestrating gene expression, RNA deadenylation, and protein ubiquitination. We report on 39 individuals with heterozygous de novo CNOT1 variants, including missense, splice site, and nonsense variants, who present with a clinical spectrum of intellectual disability, motor delay, speech delay, seizures, hypotonia, and behavioral problems. To link CNOT1 dysfunction to the neurodevelopmental phenotype observed, we generated variant-specific Drosophila models, which showed learning and memory defects upon CNOT1 knockdown. Introduction of human wild-type CNOT1 was able to rescue this phenotype, whereas mutants could not or only partially, supporting our hypothesis that CNOT1 impairment results in neurodevelopmental delay. Furthermore, the genetic interaction with autism-spectrum genes, such as ASH1L, DYRK1A, MED13, and SHANK3, was impaired in our Drosophila models. Molecular characterization of CNOT1 variants revealed normal CNOT1 expression levels, with both mutant and wild-type alleles expressed at similar levels. Analysis of protein-protein interactions with other members indicated that the CCR4-NOT complex remained intact. An integrated omics approach of patient-derived genomics and transcriptomics data suggested only minimal effects on endonucleolytic nonsense-mediated mRNA decay components, suggesting that de novo CNOT1 variants are likely haploinsufficient hypomorph or neomorph, rather than dominant negative. In summary, we provide strong evidence that de novo CNOT1 variants cause neurodevelopmental delay with a wide range of additional co-morbidities. Whereas the underlying pathophysiological mechanism warrants further analysis, our data demonstrate an essential and central role of the CCR4-NOT complex in human brain development., (Copyright © 2020 American Society of Human Genetics. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
25. Enteroendocrine and tuft cells support Lgr5 stem cells on Paneth cell depletion.
- Author
-
van Es JH, Wiebrands K, López-Iglesias C, van de Wetering M, Zeinstra L, van den Born M, Korving J, Sasaki N, Peters PJ, van Oudenaarden A, and Clevers H
- Abstract
Cycling intestinal Lgr5
+ stem cells are intermingled with their terminally differentiated Paneth cell daughters at crypt bottoms. Paneth cells provide multiple secreted (e.g., Wnt, EGF) as well as surface-bound (Notch ligand) niche signals. Here we show that ablation of Paneth cells in mice, using a diphtheria toxin receptor gene inserted into the P-lysozyme locus, does not affect the maintenance of Lgr5+ stem cells. Flow cytometry, single-cell sequencing, and histological analysis showed that the ablated Paneth cells are replaced by enteroendocrine and tuft cells. As these cells physically occupy Paneth cell positions between Lgr5 stem cells, they serve as an alternative source of Notch signals, which are essential for Lgr5+ stem cell maintenance. Our combined in vivo results underscore the adaptive flexibility of the intestine in maintaining normal tissue homeostasis.- Published
- 2019
- Full Text
- View/download PDF
26. Biallelic Variants in ASNA1 , Encoding a Cytosolic Targeting Factor of Tail-Anchored Proteins, Cause Rapidly Progressive Pediatric Cardiomyopathy.
- Author
-
Verhagen JMA, van den Born M, van der Linde HC, G J Nikkels P, Verdijk RM, Kivlen MH, van Unen LMA, Baas AF, Ter Heide H, van Osch-Gevers L, Hoogeveen-Westerveld M, Herkert JC, Bertoli-Avella AM, van Slegtenhorst MA, Wessels MW, Verheijen FW, Hassel D, Hofstra RMW, Hegde RS, van Hasselt PM, van Ham TJ, and van de Laar IMBH
- Subjects
- Alleles, Amino Acid Sequence, Animals, Arsenite Transporting ATPases chemistry, Arsenite Transporting ATPases metabolism, Cardiomyopathies enzymology, Child, Preschool, Disease Models, Animal, Exome, Female, Genetic Variation, Humans, Protein Transport, Sequence Alignment, Zebrafish genetics, Zebrafish metabolism, Zebrafish Proteins chemistry, Zebrafish Proteins metabolism, Arsenite Transporting ATPases genetics, Cardiomyopathies genetics, Cytosol enzymology, Point Mutation, Zebrafish Proteins genetics
- Abstract
Background: Pediatric cardiomyopathies are a clinically and genetically heterogeneous group of heart muscle disorders associated with high morbidity and mortality. Although knowledge of the genetic basis of pediatric cardiomyopathy has improved considerably, the underlying cause remains elusive in a substantial proportion of cases., Methods: Exome sequencing was used to screen for the causative genetic defect in a pair of siblings with rapidly progressive dilated cardiomyopathy and death in early infancy. Protein expression was assessed in patient samples, followed by an in vitro tail-anchored protein insertion assay and functional analyses in zebrafish., Results: We identified compound heterozygous variants in the highly conserved ASNA1 gene (arsA arsenite transporter, ATP-binding, homolog), which encodes an ATPase required for post-translational membrane insertion of tail-anchored proteins. The c.913C>T variant on the paternal allele is predicted to result in a premature stop codon p.(Gln305*), and likely explains the decreased protein expression observed in myocardial tissue and skin fibroblasts. The c.488T>C variant on the maternal allele results in a valine to alanine substitution at residue 163 (p.Val163Ala). Functional studies showed that this variant leads to protein misfolding as well as less effective tail-anchored protein insertion. Loss of asna1 in zebrafish resulted in reduced cardiac contractility and early lethality. In contrast to wild-type mRNA, injection of either mutant mRNA failed to rescue this phenotype., Conclusions: Biallelic variants in ASNA1 cause severe pediatric cardiomyopathy and early death. Our findings point toward a critical role of the tail-anchored membrane protein insertion pathway in vertebrate cardiac function and disease.
- Published
- 2019
- Full Text
- View/download PDF
27. Correction: The phenotypic spectrum of WWOX-related disorders: 20 additional cases of WOREE syndrome and review of the literature.
- Author
-
Piard J, Hawkes L, Milh M, Villard L, Borgatti R, Romaniello R, Fradin M, Capri Y, Héron D, Nougues MC, Nava C, Arsene OT, Shears D, Taylor J, Pagnamenta A, Taylor JC, Sogawa Y, Johnson D, Firth H, Vasudevan P, Jones G, Nguyen-Morel MA, Busa T, Roubertie A, van den Born M, Brischoux-Boucher E, Koenig M, Mignot C, Kini U, and Philippe C
- Abstract
The article has been corrected to account for one patient being investigated through genome sequencing rather than exome sequencing as originally published; thus amendments to the Abstract and Methods have been made as well as addition of the relevant authors and acknowledgment.
- Published
- 2019
- Full Text
- View/download PDF
28. The phenotypic spectrum of WWOX-related disorders: 20 additional cases of WOREE syndrome and review of the literature.
- Author
-
Piard J, Hawkes L, Milh M, Villard L, Borgatti R, Romaniello R, Fradin M, Capri Y, Héron D, Nougues MC, Nava C, Arsene OT, Shears D, Taylor J, Pagnamenta A, Taylor JC, Sogawa Y, Johnson D, Firth H, Vasudevan P, Jones G, Nguyen-Morel MA, Busa T, Roubertie A, van den Born M, Brischoux-Boucher E, Koenig M, Mignot C, Kini U, and Philippe C
- Subjects
- Adolescent, Child, Child, Preschool, DNA Copy Number Variations genetics, Epilepsy genetics, Female, Genetic Association Studies methods, Humans, Infant, Male, Mutation genetics, Mutation, Missense genetics, Syndrome, Tumor Suppressor Proteins metabolism, WW Domain-Containing Oxidoreductase metabolism, Epileptic Syndromes genetics, Tumor Suppressor Proteins genetics, Tumor Suppressor Proteins physiology, WW Domain-Containing Oxidoreductase genetics, WW Domain-Containing Oxidoreductase physiology
- Abstract
Purpose: Germline WWOX pathogenic variants have been associated with disorder of sex differentiation (DSD), spinocerebellar ataxia (SCA), and WWOX-related epileptic encephalopathy (WOREE syndrome). We review clinical and molecular data on WWOX-related disorders, further describing WOREE syndrome and phenotype/genotype correlations., Methods: We report clinical and molecular findings in 20 additional patients from 18 unrelated families with WOREE syndrome and biallelic pathogenic variants in the WWOX gene. Different molecular screening approaches were used (quantitative polymerase chain reaction/multiplex ligation-dependent probe amplification [qPCR/MLPA], array comparative genomic hybridization [array-CGH], Sanger sequencing, epilepsy gene panel, exome sequencing), genome sequencing., Results: Two copy-number variations (CNVs) or two single-nucleotide variations (SNVs) were found respectively in four and nine families, with compound heterozygosity for one SNV and one CNV in five families. Eight novel missense pathogenic variants have been described. By aggregating our patients with all cases reported in the literature, 37 patients from 27 families with WOREE syndrome are known. This review suggests WOREE syndrome is a very severe epileptic encephalopathy characterized by absence of language development and acquisition of walking, early-onset drug-resistant seizures, ophthalmological involvement, and a high likelihood of premature death. The most severe clinical presentation seems to be associated with null genotypes., Conclusion: Germline pathogenic variants in WWOX are clearly associated with a severe early-onset epileptic encephalopathy. We report here the largest cohort of individuals with WOREE syndrome.
- Published
- 2019
- Full Text
- View/download PDF
29. Insight into the sulfur metabolism of Desulfurella amilsii by differential proteomics.
- Author
-
Florentino AP, Pereira IAC, Boeren S, van den Born M, Stams AJM, and Sánchez-Andrea I
- Subjects
- Oxidation-Reduction, Proteome metabolism, Sulfites metabolism, Thiosulfates metabolism, Deltaproteobacteria metabolism, Proteomics, Sulfur metabolism
- Abstract
Many questions regarding proteins involved in microbial sulfur metabolism remain unsolved. For sulfur respiration at low pH, the terminal electron acceptor is still unclear. Desulfurella amilsii is a sulfur-reducing bacterium that respires elemental sulfur (S
0 ) or thiosulfate, and grows by S0 disproportionation. Due to its versatility, comparative studies on D. amilsii may shed light on microbial sulfur metabolism. Requirement of physical contact between cells and S0 was analyzed. Sulfide production decreased by around 50% when S0 was trapped in dialysis membranes, suggesting that contact between cells and S0 is beneficial, but not strictly needed. Proteome analysis was performed under the aforementioned conditions. A Mo-oxidoreductase suggested from genome analysis to act as sulfur reductase was not detected in any growth condition. Thiosulfate and sulfite reductases showed increased abundance in thiosulfate-reducing cultures, while rhodanese-like sulfurtransferases were highly abundant in all conditions. DsrE and DsrL were abundantly detected during thiosulfate reduction, suggesting a modified mechanism of sulfite reduction. Proteogenomics suggest a different disproportionation pathway from what has been reported. This work points to an important role of rhodaneses in sulfur processes and these proteins should be considered in searches for sulfur metabolism in broader fields like meta-omics., (© 2018 The Authors. Environmental Microbiology published by Society for Applied Microbiology and John Wiley & Sons Ltd.)- Published
- 2019
- Full Text
- View/download PDF
30. Profiling proliferative cells and their progeny in damaged murine hearts.
- Author
-
Kretzschmar K, Post Y, Bannier-Hélaouët M, Mattiotti A, Drost J, Basak O, Li VSW, van den Born M, Gunst QD, Versteeg D, Kooijman L, van der Elst S, van Es JH, van Rooij E, van den Hoff MJB, and Clevers H
- Subjects
- Animals, Cells, Cultured, Disease Models, Animal, Female, Fibroblasts cytology, Fibroblasts metabolism, Follistatin-Related Proteins genetics, Follistatin-Related Proteins metabolism, Heart Injuries genetics, Heart Injuries metabolism, Humans, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Myocardium metabolism, Myocytes, Cardiac cytology, Myocytes, Cardiac metabolism, Pregnancy, Stem Cells cytology, Stem Cells metabolism, Cell Proliferation, Heart Injuries physiopathology
- Abstract
The significance of cardiac stem cell (CSC) populations for cardiac regeneration remains disputed. Here, we apply the most direct definition of stem cell function (the ability to replace lost tissue through cell division) to interrogate the existence of CSCs. By single-cell mRNA sequencing and genetic lineage tracing using two Ki67 knockin mouse models, we map all proliferating cells and their progeny in homoeostatic and regenerating murine hearts. Cycling cardiomyocytes were only robustly observed in the early postnatal growth phase, while cycling cells in homoeostatic and damaged adult myocardium represented various noncardiomyocyte cell types. Proliferative postdamage fibroblasts expressing follistatin-like protein 1 (FSTL1) closely resemble neonatal cardiac fibroblasts and form the fibrotic scar. Genetic deletion of Fstl1 in cardiac fibroblasts results in postdamage cardiac rupture. We find no evidence for the existence of a quiescent CSC population, for transdifferentiation of other cell types toward cardiomyocytes, or for proliferation of significant numbers of cardiomyocytes in response to cardiac injury., Competing Interests: Conflict of interest statement: H.C. is co-principal investigator (co-PI) on a Dutch grant on organs-on-a-chip with C.L.M. as co-PI, however, on an unrelated topic., (Copyright © 2018 the Author(s). Published by PNAS.)
- Published
- 2018
- Full Text
- View/download PDF
31. Homozygous Truncating Variant in PKP2 Causes Hypoplastic Left Heart Syndrome.
- Author
-
Verhagen JMA, van den Born M, Kurul S, Asimaki A, van de Laar IMBH, Frohn-Mulder IME, Kammeraad JAE, Yap SC, Bartelings MM, van Slegtenhorst MA, von der Thüsen JH, and Wessels MW
- Subjects
- Adult, Female, Homozygote, Humans, Infant, Newborn, Male, Mutation, Siblings, Hypoplastic Left Heart Syndrome genetics, Plakophilins genetics
- Published
- 2018
- Full Text
- View/download PDF
32. Long-Term Expansion of Functional Mouse and Human Hepatocytes as 3D Organoids.
- Author
-
Hu H, Gehart H, Artegiani B, LÖpez-Iglesias C, Dekkers F, Basak O, van Es J, Chuva de Sousa Lopes SM, Begthel H, Korving J, van den Born M, Zou C, Quirk C, Chiriboga L, Rice CM, Ma S, Rios A, Peters PJ, de Jong YP, and Clevers H
- Subjects
- Animals, Cell Culture Techniques, Cells, Cultured, Hepatocytes cytology, Humans, Mice, Mice, Inbred BALB C, Mice, Knockout, Organoids cytology, Stem Cells cytology, Stem Cells metabolism, Time Factors, Cell Proliferation, Hepatocytes metabolism, Organoids metabolism
- Abstract
The mammalian liver possesses a remarkable regenerative ability. Two modes of damage response have been described: (1) The "oval cell" response emanates from the biliary tree when all hepatocytes are affected by chronic liver disease. (2) A massive, proliferative response of mature hepatocytes occurs upon acute liver damage such as partial hepatectomy (PHx). While the oval cell response has been captured in vitro by growing organoids from cholangiocytes, the hepatocyte proliferative response has not been recapitulated in culture. Here, we describe the establishment of a long-term 3D organoid culture system for mouse and human primary hepatocytes. Organoids can be established from single hepatocytes and grown for multiple months, while retaining key morphological, functional and gene expression features. Transcriptional profiles of the organoids resemble those of proliferating hepatocytes after PHx. Human hepatocyte organoids proliferate extensively after engraftment into mice and thus recapitulate the proliferative damage-response of hepatocytes., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
33. N-Acetylglutamate Synthase Deficiency Due to a Recurrent Sequence Variant in the N-acetylglutamate Synthase Enhancer Region.
- Author
-
Williams M, Burlina A, Rubert L, Polo G, Ruijter GJG, van den Born M, Rüfenacht V, Haskins N, van Zutven LJCM, Tuchman M, Saris JJ, Häberle J, and Caldovic L
- Subjects
- Amino-Acid N-Acetyltransferase metabolism, Base Sequence, Child, Child, Preschool, Female, Humans, Hyperammonemia, Prognosis, Urea Cycle Disorders, Inborn metabolism, Urea Cycle Disorders, Inborn pathology, Amino-Acid N-Acetyltransferase genetics, Enhancer Elements, Genetic, Genetic Variation, Urea Cycle Disorders, Inborn etiology
- Abstract
N-acetylglutamate synthase deficiency (NAGSD, MIM #237310) is an autosomal recessive disorder of the urea cycle that results from absent or decreased production of N-acetylglutamate (NAG) due to either decreased NAGS gene expression or defective NAGS enzyme. NAG is essential for the activity of carbamylphosphate synthetase 1 (CPS1), the first and rate-limiting enzyme of the urea cycle. NAGSD is the only urea cycle disorder that can be treated with a single drug, N-carbamylglutamate (NCG), which can activate CPS1 and completely restore ureagenesis in patients with NAGSD. We describe a novel sequence variant NM_153006.2:c.-3026C > T in the NAGS enhancer that was found in three patients from two families with NAGSD; two patients had hyperammonemia that resolved upon treatment with NCG, while the third patient increased dietary protein intake after initiation of NCG therapy. Two patients were homozygous for the variant while the third patient had the c.-3026C > T variant and a partial uniparental disomy that encompassed the NAGS gene on chromosome 17. The c.-3026C > T sequence variant affects a base pair that is highly conserved in vertebrates; the variant is predicted to be deleterious by several bioinformatics tools. Functional assays in cultured HepG2 cells demonstrated that the c.-3026C > T substitution could result in reduced expression of the NAGS gene. These findings underscore the importance of analyzing NAGS gene regulatory regions when looking for molecular causes of NAGSD.
- Published
- 2018
- Full Text
- View/download PDF
34. Enteroendocrine cells switch hormone expression along the crypt-to-villus BMP signalling gradient.
- Author
-
Beumer J, Artegiani B, Post Y, Reimann F, Gribble F, Nguyen TN, Zeng H, Van den Born M, Van Es JH, and Clevers H
- Subjects
- Animals, Humans, Intestine, Small cytology, Mice, Inbred C57BL, Mice, Transgenic, Phenotype, Tissue Culture Techniques, Bone Morphogenetic Protein 4 pharmacology, Cell Differentiation drug effects, Cell Lineage, Cell Movement drug effects, Enteroendocrine Cells metabolism, Gastrointestinal Hormones metabolism, Intestine, Small metabolism, Signal Transduction drug effects
- Abstract
Enteroendocrine cells (EECs) control a wide range of physiological processes linked to metabolism
1 . We show that EEC hormones are differentially expressed between crypts (for example, Glp1) and villi (for example, secretin). As demonstrated by single-cell mRNA sequencing using murine Lgr5+ cell-derived organoids, BMP4 signals alter the hormone expression profiles of individual EECs to resemble those found in the villus. Accordingly, BMP4 induces hormone switching of EECs migrating up the crypt-villus axis in vivo. Our findings imply that EEC lineages in the small intestine exhibit a more flexible hormone repertoire than previously proposed. We also describe a protocol to generate human EECs in organoids and demonstrate a similar regulation of hormone expression by BMP signalling. These findings establish alternative strategies to target EECs with therapeutically relevant hormone production through BMP modulation.- Published
- 2018
- Full Text
- View/download PDF
35. Characterization of glycosylphosphatidylinositol biosynthesis defects by clinical features, flow cytometry, and automated image analysis.
- Author
-
Knaus A, Pantel JT, Pendziwiat M, Hajjir N, Zhao M, Hsieh TC, Schubach M, Gurovich Y, Fleischer N, Jäger M, Köhler S, Muhle H, Korff C, Møller RS, Bayat A, Calvas P, Chassaing N, Warren H, Skinner S, Louie R, Evers C, Bohn M, Christen HJ, van den Born M, Obersztyn E, Charzewska A, Endziniene M, Kortüm F, Brown N, Robinson PN, Schelhaas HJ, Weber Y, Helbig I, Mundlos S, Horn D, and Krawitz PM
- Subjects
- Abnormalities, Multiple metabolism, Automation, Biomarkers metabolism, Humans, Intellectual Disability metabolism, Phenotype, Phosphorus Metabolism Disorders metabolism, Syndrome, Flow Cytometry methods, Glycosylphosphatidylinositols biosynthesis, Image Processing, Computer-Assisted
- Abstract
Background: Glycosylphosphatidylinositol biosynthesis defects (GPIBDs) cause a group of phenotypically overlapping recessive syndromes with intellectual disability, for which pathogenic mutations have been described in 16 genes of the corresponding molecular pathway. An elevated serum activity of alkaline phosphatase (AP), a GPI-linked enzyme, has been used to assign GPIBDs to the phenotypic series of hyperphosphatasia with mental retardation syndrome (HPMRS) and to distinguish them from another subset of GPIBDs, termed multiple congenital anomalies hypotonia seizures syndrome (MCAHS). However, the increasing number of individuals with a GPIBD shows that hyperphosphatasia is a variable feature that is not ideal for a clinical classification., Methods: We studied the discriminatory power of multiple GPI-linked substrates that were assessed by flow cytometry in blood cells and fibroblasts of 39 and 14 individuals with a GPIBD, respectively. On the phenotypic level, we evaluated the frequency of occurrence of clinical symptoms and analyzed the performance of computer-assisted image analysis of the facial gestalt in 91 individuals., Results: We found that certain malformations such as Morbus Hirschsprung and diaphragmatic defects are more likely to be associated with particular gene defects (PIGV, PGAP3, PIGN). However, especially at the severe end of the clinical spectrum of HPMRS, there is a high phenotypic overlap with MCAHS. Elevation of AP has also been documented in some of the individuals with MCAHS, namely those with PIGA mutations. Although the impairment of GPI-linked substrates is supposed to play the key role in the pathophysiology of GPIBDs, we could not observe gene-specific profiles for flow cytometric markers or a correlation between their cell surface levels and the severity of the phenotype. In contrast, it was facial recognition software that achieved the highest accuracy in predicting the disease-causing gene in a GPIBD., Conclusions: Due to the overlapping clinical spectrum of both HPMRS and MCAHS in the majority of affected individuals, the elevation of AP and the reduced surface levels of GPI-linked markers in both groups, a common classification as GPIBDs is recommended. The effectiveness of computer-assisted gestalt analysis for the correct gene inference in a GPIBD and probably beyond is remarkable and illustrates how the information contained in human faces is pivotal in the delineation of genetic entities.
- Published
- 2018
- Full Text
- View/download PDF
36. Generation of an inducible colon-specific Cre enzyme mouse line for colon cancer research.
- Author
-
Tetteh PW, Kretzschmar K, Begthel H, van den Born M, Korving J, Morsink F, Farin H, van Es JH, Offerhaus GJ, and Clevers H
- Subjects
- Adenoma etiology, Adenoma metabolism, Adenoma pathology, Animals, Biomarkers, Tumor, Carbonic Anhydrase I genetics, Carbonic Anhydrase I metabolism, Cell Transformation, Neoplastic genetics, Cell Transformation, Neoplastic metabolism, Colonic Neoplasms metabolism, Colonic Neoplasms pathology, Disease Models, Animal, Disease Progression, Enzyme Activation, Gene Knock-In Techniques, Gene Targeting, Genes, APC, Genes, ras, Genetic Loci, Humans, Immunohistochemistry, Integrases metabolism, Intestinal Mucosa metabolism, Intestinal Mucosa pathology, Mice, Mice, Knockout, Mutation, Organ Specificity genetics, Research, Colonic Neoplasms etiology, Gene Expression Regulation, Integrases genetics, Mice, Transgenic
- Abstract
Current mouse models for colorectal cancer often differ significantly from human colon cancer, being largely restricted to the small intestine. Here, we aim to develop a colon-specific inducible mouse model that can faithfully recapitulate human colon cancer initiation and progression. Carbonic anhydrase I (Car1) is a gene expressed uniquely in colonic epithelial cells. We generated a colon-specific inducible Car1
CreER knock-in (KI) mouse with broad Cre activity in epithelial cells of the proximal colon and cecum. Deletion of the tumor suppressor gene Apc using the Car1CreER KI caused tumor formation in the cecum but did not yield adenomas in the proximal colon. Mutation of both Apc and Kras yielded microadenomas in both the cecum and the proximal colon, which progressed to macroadenomas with significant morbidity. Aggressive carcinomas with some invasion into lymph nodes developed upon combined induction of oncogenic mutations of Apc, Kras, p53, and Smad4 Importantly, no adenomas were observed in the small intestine. Additionally, we observed tumors from differentiated Car1-expressing cells with Apc/Kras mutations, suggesting that a top-down model of intestinal tumorigenesis can occur with multiple mutations. Our results establish the Car1CreER KI as a valuable mouse model to study colon-specific tumorigenesis and metastasis as well as cancer-cell-of-origin questions., Competing Interests: The authors declare no conflict of interest.- Published
- 2016
- Full Text
- View/download PDF
37. Reg4+ deep crypt secretory cells function as epithelial niche for Lgr5+ stem cells in colon.
- Author
-
Sasaki N, Sachs N, Wiebrands K, Ellenbroek SI, Fumagalli A, Lyubimova A, Begthel H, van den Born M, van Es JH, Karthaus WR, Li VS, López-Iglesias C, Peters PJ, van Rheenen J, van Oudenaarden A, and Clevers H
- Subjects
- Animals, Colon cytology, Colon growth & development, Colon metabolism, Colonic Neoplasms pathology, Epithelial Cells metabolism, Epithelial Cells pathology, Intestine, Small cytology, Intestine, Small metabolism, Mice, Neoplasm Proteins metabolism, Organoids growth & development, Organoids metabolism, Pancreatitis-Associated Proteins, Paneth Cells cytology, Paneth Cells metabolism, Receptors, G-Protein-Coupled metabolism, Receptors, Notch genetics, Stem Cell Niche genetics, Stem Cells cytology, Wnt Signaling Pathway genetics, Colonic Neoplasms metabolism, Neoplasm Proteins genetics, Receptors, G-Protein-Coupled genetics, Stem Cells metabolism
- Abstract
Leucine-rich repeat-containing G-protein coupled receptor 5-positive (Lgr5(+)) stem cells reside at crypt bottoms of the small and large intestine. Small intestinal Paneth cells supply Wnt3, EGF, and Notch signals to neighboring Lgr5(+) stem cells. Whereas the colon lacks Paneth cells, deep crypt secretory (DCS) cells are intermingled with Lgr5(+) stem cells at crypt bottoms. Here, we report regenerating islet-derived family member 4 (Reg4) as a marker of DCS cells. To investigate a niche function, we eliminated DCS cells by using the diphtheria-toxin receptor gene knocked into the murine Reg4 locus. Ablation of DCS cells results in loss of stem cells from colonic crypts and disrupts gut homeostasis and colon organoid growth. In agreement, sorted Reg4(+) DCS cells promote organoid formation of single Lgr5(+) colon stem cells. DCS cells can be massively produced from Lgr5(+) colon stem cells in vitro by combined Notch inhibition and Wnt activation. We conclude that Reg4(+) DCS cells serve as Paneth cell equivalents in the colon crypt niche., Competing Interests: The authors declare no conflict of interest.
- Published
- 2016
- Full Text
- View/download PDF
38. De Novo Prediction of Stem Cell Identity using Single-Cell Transcriptome Data.
- Author
-
Grün D, Muraro MJ, Boisset JC, Wiebrands K, Lyubimova A, Dharmadhikari G, van den Born M, van Es J, Jansen E, Clevers H, de Koning EJP, and van Oudenaarden A
- Subjects
- Adult, Algorithms, Animals, Bone Marrow Cells cytology, Bone Marrow Cells metabolism, Cell Lineage, Entropy, Hematopoietic Stem Cells cytology, Hematopoietic Stem Cells metabolism, Humans, Intestines cytology, Mice, Inbred C57BL, Multipotent Stem Cells cytology, Multipotent Stem Cells metabolism, Pancreatic Ducts cytology, Pluripotent Stem Cells cytology, Pluripotent Stem Cells metabolism, Reproducibility of Results, Single-Cell Analysis methods, Stem Cells cytology, Transcriptome genetics
- Abstract
Adult mitotic tissues like the intestine, skin, and blood undergo constant turnover throughout the life of an organism. Knowing the identity of the stem cell is crucial to understanding tissue homeostasis and its aberrations upon disease. Here we present a computational method for the derivation of a lineage tree from single-cell transcriptome data. By exploiting the tree topology and the transcriptome composition, we establish StemID, an algorithm for identifying stem cells among all detectable cell types within a population. We demonstrate that StemID recovers two known adult stem cell populations, Lgr5+ cells in the small intestine and hematopoietic stem cells in the bone marrow. We apply StemID to predict candidate multipotent cell populations in the human pancreas, a tissue with largely uncharacterized turnover dynamics. We hope that StemID will accelerate the search for novel stem cells by providing concrete markers for biological follow-up and validation., (Copyright © 2016 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
39. Replacement of Lost Lgr5-Positive Stem Cells through Plasticity of Their Enterocyte-Lineage Daughters.
- Author
-
Tetteh PW, Basak O, Farin HF, Wiebrands K, Kretzschmar K, Begthel H, van den Born M, Korving J, de Sauvage F, van Es JH, van Oudenaarden A, and Clevers H
- Subjects
- Alkaline Phosphatase metabolism, Animals, Biomarkers metabolism, Cell Dedifferentiation, Cell Line, Cell Proliferation, Enterocytes pathology, Integrases metabolism, Intestinal Neoplasms pathology, Mice, Mutation genetics, Organoids, Paneth Cells metabolism, Paneth Cells pathology, Regeneration genetics, Single-Cell Analysis, beta-Galactosidase metabolism, Cell Lineage, Enterocytes metabolism, Receptors, G-Protein-Coupled metabolism, Stem Cells metabolism
- Abstract
Intestinal crypts display robust regeneration upon injury. The relatively rare secretory precursors can replace lost stem cells, but it is unknown if the abundant enterocyte progenitors that express the Alkaline phosphate intestinal (Alpi) gene also have this capacity. We created an Alpi-IRES-CreERT2 (Alpi(CreER)) knockin allele for lineage tracing. Marked clones consist entirely of enterocytes and are all lost from villus tips within days. Genetic fate-mapping of Alpi(+) cells before or during targeted ablation of Lgr5-expressing stem cells generated numerous long-lived crypt-villus "ribbons," indicative of dedifferentiation of enterocyte precursors into Lgr5(+) stems. By single-cell analysis of dedifferentiating enterocytes, we observed the generation of Paneth-like cells and proliferative stem cells. We conclude that the highly proliferative, short-lived enterocyte precursors serve as a large reservoir of potential stem cells during crypt regeneration., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
40. Porcupine inhibitor suppresses paracrine Wnt-driven growth of Rnf43;Znrf3-mutant neoplasia.
- Author
-
Koo BK, van Es JH, van den Born M, and Clevers H
- Subjects
- Acyltransferases, Animals, Basic Helix-Loop-Helix Transcription Factors deficiency, Basic Helix-Loop-Helix Transcription Factors genetics, Humans, Intestinal Neoplasms genetics, Intestinal Neoplasms pathology, Mice, Mice, Knockout, Mice, Transgenic, Mutation, Paracrine Communication drug effects, Ubiquitin-Protein Ligases deficiency, Wnt3 Protein deficiency, Wnt3 Protein genetics, Zinc Fingers genetics, Benzeneacetamides pharmacology, Intestinal Neoplasms drug therapy, Membrane Proteins antagonists & inhibitors, Pyridines pharmacology, Ubiquitin-Protein Ligases genetics, Wnt Signaling Pathway drug effects
- Abstract
Rnf43 (RING finger protein 43) and Znrf3 (zinc/RING finger protein 3) (RZ) are two closely related transmembrane E3 ligases, encoded by Wnt target genes, that remove surface Wnt (wingless-int) receptors. The two genes are mutated in various human cancers. Such tumors are predicted to be hypersensitive to, yet still depend on, secreted Wnts. We previously showed that mutation of RZ in the intestine yields rapidly growing adenomas containing LGR5(+) (leucine-rich repeat-containing G-protein coupled receptor 5) stem cells and Wnt3-producing Paneth cells. We now show that removal of Paneth cells by Math1 mutation inhibits RZ(-/-) tumor formation. Similarly, deletion of Wnt3 inhibits tumorigenesis. Treatment of mice carrying RZ(-/-) intestinal neoplasia with a small molecule Wnt secretion inhibitor (porcupine inhibitor C59) strongly inhibited growth, whereas adjacent normal crypts remained intact. These results establish that paracrine Wnt secretion is an essential driver of RZ(-/-) tumor growth and imply that a therapeutic window exists for the use of porcupine inhibitors for RZ-mutant cancers.
- Published
- 2015
- Full Text
- View/download PDF
41. A20 controls intestinal homeostasis through cell-specific activities.
- Author
-
Vereecke L, Vieira-Silva S, Billiet T, van Es JH, Mc Guire C, Slowicka K, Sze M, van den Born M, De Hertogh G, Clevers H, Raes J, Rutgeerts P, Vermeire S, Beyaert R, and van Loo G
- Subjects
- Animals, Apoptosis, Colitis enzymology, Colitis genetics, Colitis pathology, Colitis physiopathology, Cysteine Endopeptidases genetics, Epithelial Cells cytology, Epithelial Cells pathology, Female, Goblet Cells cytology, Goblet Cells enzymology, Goblet Cells pathology, Homeostasis, Humans, Intestines pathology, Intracellular Signaling Peptides and Proteins genetics, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Paneth Cells cytology, Paneth Cells enzymology, Paneth Cells pathology, Species Specificity, Tumor Necrosis Factor alpha-Induced Protein 3, Cysteine Endopeptidases metabolism, Epithelial Cells enzymology, Intestines enzymology, Intracellular Signaling Peptides and Proteins metabolism
- Abstract
The transcription factor NF-κB is indispensable for intestinal immune homeostasis, but contributes to chronic inflammation and inflammatory bowel disease (IBD). A20, an inhibitor of both NF-κB and apoptotic signalling, was identified as a susceptibility gene for multiple inflammatory diseases, including IBD. Despite absence of spontaneous intestinal inflammation in intestinal epithelial cell (IEC) specific A20 knockout mice, we found additional myeloid-specific A20 deletion to synergistically drive intestinal pathology through cell-specific mechanisms. A20 ensures intestinal barrier stability by preventing cytokine-induced IEC apoptosis, while A20 prevents excessive cytokine production in myeloid cells. Combining IEC and myeloid A20 deletion induces ileitis and severe colitis, characterized by IEC apoptosis, Paneth and goblet cell loss, epithelial hyperproliferation and intestinal microbiota dysbiosis. Continuous epithelial cell death and regeneration in an inflammatory environment sensitizes cells for neoplastic transformation and the development of colorectal tumours in aged mice.
- Published
- 2014
- Full Text
- View/download PDF
42. Diabetes risk gene and Wnt effector Tcf7l2/TCF4 controls hepatic response to perinatal and adult metabolic demand.
- Author
-
Boj SF, van Es JH, Huch M, Li VS, José A, Hatzis P, Mokry M, Haegebarth A, van den Born M, Chambon P, Voshol P, Dor Y, Cuppen E, Fillat C, and Clevers H
- Subjects
- Animals, Animals, Newborn, Diet, High-Fat, Fasting metabolism, Islets of Langerhans metabolism, Mice, Mice, Knockout, Transcription Factor 7-Like 2 Protein genetics, Transcriptional Activation, Diabetes Mellitus genetics, Diabetes Mellitus metabolism, Glucose metabolism, Liver metabolism, Metabolic Networks and Pathways, Transcription Factor 7-Like 2 Protein metabolism
- Abstract
Most studies on TCF7L2 SNP variants in the pathogenesis of type 2 diabetes (T2D) focus on a role of the encoded transcription factor TCF4 in β cells. Here, a mouse genetics approach shows that removal of TCF4 from β cells does not affect their function, whereas manipulating TCF4 levels in the liver has major effects on metabolism. In Tcf7l2(-/-) mice, the immediate postnatal surge in liver metabolism does not occur. Consequently, pups die due to hypoglycemia. By combining chromatin immunoprecipitation with gene expression profiling, we identify a TCF4-controlled metabolic gene program that is acutely activated in the postnatal liver. In concordance, adult liver-specific Tcf7l2 knockout mice show reduced hepatic glucose production during fasting and display improved glucose homeostasis when maintained on high-fat diet. Furthermore, liver-specific TCF4 overexpression increases hepatic glucose production. These observations imply that TCF4 directly activates metabolic genes and that inhibition of Wnt signaling may be beneficial in metabolic disease., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
43. Dll1+ secretory progenitor cells revert to stem cells upon crypt damage.
- Author
-
van Es JH, Sato T, van de Wetering M, Lyubimova A, Yee Nee AN, Gregorieff A, Sasaki N, Zeinstra L, van den Born M, Korving J, Martens ACM, Barker N, van Oudenaarden A, and Clevers H
- Subjects
- Animals, Calcium-Binding Proteins, Cell Lineage, Cells, Cultured, Gene Knock-In Techniques, Intercellular Signaling Peptides and Proteins analysis, Intercellular Signaling Peptides and Proteins genetics, Mice, Organoids metabolism, Receptors, Notch antagonists & inhibitors, Receptors, Notch metabolism, Wnt3A Protein pharmacology, Intercellular Signaling Peptides and Proteins metabolism, Intestinal Mucosa metabolism, Stem Cells metabolism
- Abstract
Lgr5+ intestinal stem cells generate enterocytes and secretory cells. Secretory lineage commitment requires Notch silencing. The Notch ligand Dll1 is expressed by a subset of immediate stem cell daughters. Lineage tracing in Dll1(GFP-ires-CreERT2) knock-in mice reveals that single Dll1(high) cells generate small, short-lived clones containing all four secretory cell types. Lineage specification thus occurs in immediate stem cell daughters through Notch lateral inhibition. Cultured Dll1(high) cells form long-lived organoids (mini-guts) on brief Wnt3A exposure. When Dll1(high) cells are genetically marked before tissue damage, stem cell tracing events occur. Thus, secretory progenitors exhibit plasticity by regaining stemness on damage.
- Published
- 2012
- Full Text
- View/download PDF
44. Lineage tracing reveals Lgr5+ stem cell activity in mouse intestinal adenomas.
- Author
-
Schepers AG, Snippert HJ, Stange DE, van den Born M, van Es JH, van de Wetering M, and Clevers H
- Subjects
- Adenoma genetics, Adenoma metabolism, Animals, Biomarkers analysis, Cell Lineage, Cell Transformation, Neoplastic, Gene Expression Profiling, Gene Knock-In Techniques, Genes, Reporter, Intestinal Mucosa metabolism, Intestinal Mucosa pathology, Intestinal Neoplasms genetics, Mice, Multipotent Stem Cells pathology, Multipotent Stem Cells physiology, Paneth Cells pathology, Stem Cell Niche, Tamoxifen pharmacology, Tumor Stem Cell Assay, Adenoma pathology, Intestinal Neoplasms pathology, Neoplastic Stem Cells pathology, Neoplastic Stem Cells physiology, Receptors, G-Protein-Coupled analysis
- Abstract
The concept that tumors are maintained by dedicated stem cells, the so-called cancer stem cell hypothesis, has attracted great interest but remains controversial. Studying mouse models, we provide direct, functional evidence for the presence of stem cell activity within primary intestinal adenomas, a precursor to intestinal cancer. By "lineage retracing" using the multicolor Cre-reporter R26R-Confetti, we demonstrate that the crypt stem cell marker Lgr5 (leucine-rich repeat-containing heterotrimeric guanine nucleotide-binding protein-coupled receptor 5) also marks a subpopulation of adenoma cells that fuel the growth of established intestinal adenomas. These Lgr5(+) cells, which represent about 5 to 10% of the cells in the adenomas, generate additional Lgr5(+) cells as well as all other adenoma cell types. The Lgr5(+) cells are intermingled with Paneth cells near the adenoma base, a pattern reminiscent of the architecture of the normal crypt niche.
- Published
- 2012
- Full Text
- View/download PDF
45. A critical role for the Wnt effector Tcf4 in adult intestinal homeostatic self-renewal.
- Author
-
van Es JH, Haegebarth A, Kujala P, Itzkovitz S, Koo BK, Boj SF, Korving J, van den Born M, van Oudenaarden A, Robine S, and Clevers H
- Subjects
- Animals, Basic Helix-Loop-Helix Leucine Zipper Transcription Factors genetics, Cell Differentiation, Cell Proliferation, Gene Expression Regulation, Developmental, Intestinal Mucosa cytology, Intestinal Mucosa metabolism, Mice, Stem Cells cytology, Transcription Factor 4, Basic Helix-Loop-Helix Leucine Zipper Transcription Factors metabolism, Intestines cytology, Stem Cells metabolism, Wnt Signaling Pathway
- Abstract
Throughout life, intestinal Lgr5+ stem cells give rise to proliferating transient amplifying cells in crypts, which subsequently differentiate into one of the five main cell types and migrate along the crypt-villus axis. These dynamic processes are coordinated by a relatively small number of evolutionarily conserved signaling pathways, which includes the Wnt signaling pathway. The DNA-binding proteins of the T-cell factor family, Tcf1/Tcf7, Lef, Tcf3/Tcf7l1, and Tcf4/Tcf7l2, constitute the downstream effectors of the Wnt signaling pathway. While Tcf4 is the major member active during embryogenesis, the role of these Wnt effectors in the homeostasis of the adult mouse intestinal epithelium is unresolved. Using Tcf1-/-, Tcf3(flox), and novel Tcf4(flox) mice, we demonstrate an essential role for Tcf4 during homeostasis of the adult mouse intestine.
- Published
- 2012
- Full Text
- View/download PDF
46. Lgr5 intestinal stem cells have high telomerase activity and randomly segregate their chromosomes.
- Author
-
Schepers AG, Vries R, van den Born M, van de Wetering M, and Clevers H
- Subjects
- Animals, Cell Cycle, Cell Differentiation, Epithelial Cells enzymology, Epithelial Cells physiology, Mice, Time Factors, Chromosome Segregation, Intestinal Mucosa cytology, Intestinal Mucosa enzymology, Stem Cell Niche, Stem Cells enzymology, Stem Cells physiology, Telomerase metabolism
- Abstract
Somatic cells have been proposed to be limited in the number of cell divisions they can undergo. This is thought to be a mechanism by which stem cells retain their integrity preventing disease. However, we have recently discovered intestinal crypt stem cells that persist for the lifetime of a mouse, yet divide every day. We now demonstrate biochemically that primary isolated Lgr5+ve stem cells contain significant telomerase activity. Telomerase activity rapidly decreases in the undifferentiated progeny of these stem cells and is entirely lost in differentiated villus cells. Conversely, asymmetric segregation of chromosomes has been proposed as a mechanism for stem cells to protect their genomes against damage. We determined the average cell cycle length of Lgr5+ve stem cells at 21.5 h and find that Lgr5+ve intestinal stem cells randomly segregate newly synthesized DNA strands, opposing the 'immortal strand' hypothesis.
- Published
- 2011
- Full Text
- View/download PDF
47. Paneth cells constitute the niche for Lgr5 stem cells in intestinal crypts.
- Author
-
Sato T, van Es JH, Snippert HJ, Stange DE, Vries RG, van den Born M, Barker N, Shroyer NF, van de Wetering M, and Clevers H
- Subjects
- Animals, CD24 Antigen metabolism, Cell Count, Cell Proliferation, Coculture Techniques, Humans, Mice, Paneth Cells metabolism, Stem Cell Niche metabolism, Wnt Proteins metabolism, Wnt3 Protein, Intestines cytology, Multipotent Stem Cells cytology, Multipotent Stem Cells metabolism, Paneth Cells cytology, Receptors, G-Protein-Coupled metabolism, Stem Cell Niche cytology
- Abstract
Homeostasis of self-renewing small intestinal crypts results from neutral competition between Lgr5 stem cells, which are small cycling cells located at crypt bottoms. Lgr5 stem cells are interspersed between terminally differentiated Paneth cells that are known to produce bactericidal products such as lysozyme and cryptdins/defensins. Single Lgr5-expressing stem cells can be cultured to form long-lived, self-organizing crypt-villus organoids in the absence of non-epithelial niche cells. Here we find a close physical association of Lgr5 stem cells with Paneth cells in mice, both in vivo and in vitro. CD24(+) Paneth cells express EGF, TGF-α, Wnt3 and the Notch ligand Dll4, all essential signals for stem-cell maintenance in culture. Co-culturing of sorted stem cells with Paneth cells markedly improves organoid formation. This Paneth cell requirement can be substituted by a pulse of exogenous Wnt. Genetic removal of Paneth cells in vivo results in the concomitant loss of Lgr5 stem cells. In colon crypts, CD24(+) cells residing between Lgr5 stem cells may represent the Paneth cell equivalents. We conclude that Lgr5 stem cells compete for essential niche signals provided by a specialized daughter cell, the Paneth cell.
- Published
- 2011
- Full Text
- View/download PDF
48. The BMP antagonist follistatin-like 1 is required for skeletal and lung organogenesis.
- Author
-
Sylva M, Li VS, Buffing AA, van Es JH, van den Born M, van der Velden S, Gunst Q, Koolstra JH, Moorman AF, Clevers H, and van den Hoff MJ
- Subjects
- Animals, Female, Follistatin-Related Proteins genetics, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Organogenesis genetics, Bone Morphogenetic Proteins antagonists & inhibitors, Follistatin-Related Proteins metabolism, Lung embryology, Lung metabolism, Muscle, Skeletal embryology, Muscle, Skeletal metabolism, Organogenesis physiology
- Abstract
Follistatin-like 1 (Fstl1) is a secreted protein of the BMP inhibitor class. During development, expression of Fstl1 is already found in cleavage stage embryos and becomes gradually restricted to mesenchymal elements of most organs during subsequent development. Knock down experiments in chicken and zebrafish demonstrated a role as a BMP antagonist in early development. To investigate the role of Fstl1 during mouse development, a conditional Fstl1 KO allele as well as a Fstl1-GFP reporter mouse were created. KO mice die at birth from respiratory distress and show multiple defects in lung development. Also, skeletal development is affected. Endochondral bone development, limb patterning as well as patterning of the axial skeleton are perturbed in the absence of Fstl1. Taken together, these observations show that Fstl1 is a crucial regulator in BMP signalling during mouse development.
- Published
- 2011
- Full Text
- View/download PDF
49. Intestinal crypt homeostasis results from neutral competition between symmetrically dividing Lgr5 stem cells.
- Author
-
Snippert HJ, van der Flier LG, Sato T, van Es JH, van den Born M, Kroon-Veenboer C, Barker N, Klein AM, van Rheenen J, Simons BD, and Clevers H
- Subjects
- Animals, Clone Cells, Mice, Models, Biological, Receptors, G-Protein-Coupled metabolism, Cell Lineage, Intestine, Small cytology, Stem Cells cytology
- Abstract
Intestinal stem cells, characterized by high Lgr5 expression, reside between Paneth cells at the small intestinal crypt base and divide every day. We have carried out fate mapping of individual stem cells by generating a multicolor Cre-reporter. As a population, Lgr5(hi) stem cells persist life-long, yet crypts drift toward clonality within a period of 1-6 months. We have collected short- and long-term clonal tracing data of individual Lgr5(hi) cells. These reveal that most Lgr5(hi) cell divisions occur symmetrically and do not support a model in which two daughter cells resulting from an Lgr5(hi) cell division adopt divergent fates (i.e., one Lgr5(hi) cell and one transit-amplifying [TA] cell per division). The cellular dynamics are consistent with a model in which the resident stem cells double their numbers each day and stochastically adopt stem or TA fates. Quantitative analysis shows that stem cell turnover follows a pattern of neutral drift dynamics., (Copyright © 2010 Elsevier Inc. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
50. Lgr6 marks stem cells in the hair follicle that generate all cell lineages of the skin.
- Author
-
Snippert HJ, Haegebarth A, Kasper M, Jaks V, van Es JH, Barker N, van de Wetering M, van den Born M, Begthel H, Vries RG, Stange DE, Toftgård R, and Clevers H
- Subjects
- Animals, Epidermal Cells, Gene Expression Profiling, Gene Knock-In Techniques, Hair cytology, Hair embryology, Hair growth & development, Hair Follicle embryology, Hair Follicle growth & development, Mice, Mice, Nude, Sebaceous Glands cytology, Signal Transduction, Stem Cell Transplantation, Stem Cells metabolism, Wound Healing, Cell Lineage, Hair Follicle cytology, Receptors, G-Protein-Coupled genetics, Receptors, G-Protein-Coupled metabolism, Skin cytology, Stem Cells cytology
- Abstract
Mammalian epidermis consists of three self-renewing compartments: the hair follicle, the sebaceous gland, and the interfollicular epidermis. We generated knock-in alleles of murine Lgr6, a close relative of the Lgr5 stem cell gene. Lgr6 was expressed in the earliest embryonic hair placodes. In adult hair follicles, Lgr6+ cells resided in a previously uncharacterized region directly above the follicle bulge. They expressed none of the known bulge stem cell markers. Prenatal Lgr6+ cells established the hair follicle, sebaceous gland, and interfollicular epidermis. Postnatally, Lgr6+ cells generated sebaceous gland and interfollicular epidermis, whereas contribution to hair lineages gradually diminished with age. Adult Lgr6+ cells executed long-term wound repair, including the formation of new hair follicles. We conclude that Lgr6 marks the most primitive epidermal stem cell.
- 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.