1
|
Sim SY, Baek IC, Cho WK, Jung MH, Kim TG, Suh BK. Immune Gene Expression Profiling in Individuals with Turner Syndrome, Graves' Disease, and a Healthy Female by Single-Cell RNA Sequencing: A Comparative Study. Cells 2025; 14:93. [PMID: 39851522 PMCID: PMC11764232 DOI: 10.3390/cells14020093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 01/07/2025] [Accepted: 01/08/2025] [Indexed: 01/26/2025] Open
Abstract
Turner syndrome (TS) can be determined by karyotype analysis, marked by the loss of one X chromosome in females. However, the genes involved in autoimmunity in TS patients remain unclear. In this study, we aimed to analyze differences in immune gene expression between a patient with TS, a healthy female, and a female patient with Graves' disease using single-cell RNA sequencing (scRNA-seq) analysis of antigen-specific CD4(+) T cells. We identified 43 differentially expressed genes in the TS patient compared with the healthy female and the female patient with Graves' disease. Many of these genes have previously been suggested to play a role in immune system regulation. This study provides valuable insights into the differences in immune-related gene expression between TS patients, healthy individuals, and those with autoimmune diseases.
Collapse
Affiliation(s)
- Soo Yeun Sim
- Department of Pediatrics, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (S.Y.S.); (B.-K.S.)
| | - In-Cheol Baek
- Catholic Hematopoietic Stem Cell Bank, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Won Kyoung Cho
- Department of Pediatrics, St. Vincent’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 16247, Republic of Korea
| | - Min Ho Jung
- Department of Pediatrics, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 07345, Republic of Korea;
| | - Tai-Gyu Kim
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea;
| | - Byung-Kyu Suh
- Department of Pediatrics, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (S.Y.S.); (B.-K.S.)
| |
Collapse
|
2
|
Huang T, Fakurazi S, Cheah PS, Ling KH. Chromosomal and cellular therapeutic approaches for Down syndrome: A research update. Biochem Biophys Res Commun 2024; 735:150664. [PMID: 39260337 DOI: 10.1016/j.bbrc.2024.150664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 08/20/2024] [Accepted: 09/03/2024] [Indexed: 09/13/2024]
Abstract
In individuals with Down syndrome (DS), an additional HSA21 chromosome copy leads to the overexpression of a myriad of HSA21 genes, disrupting the transcription of the entire genome. This dysregulation in transcription and post-transcriptional modifications contributes to abnormal phenotypes across nearly all tissues and organs in DS individuals. The array of severe clinical symptoms associated with trisomy 21 poses a considerable challenge in the quest for a cure for DS. Fortunately, a wealth of research suggests that chromosome therapy, hinging on cutting-edge genome editing technologies, can potentially eliminate the extra copy of the human chromosome 21. Genome editing tools have demonstrated their efficacy in restoring trisomy to a normal diploid state in vitro DS cell models. Furthermore, we delve into the noteworthy findings in cellular therapy for DS, with recent studies showcasing the increasing feasibility of strategies involving stem cells and CAR T-cells to address corresponding clinical phenotypes.
Collapse
Affiliation(s)
- Tan Huang
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Sharida Fakurazi
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Pike-See Cheah
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia; Malaysian Research Institute on Ageing (MyAgeing(TM)), Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - King-Hwa Ling
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia; Malaysian Research Institute on Ageing (MyAgeing(TM)), Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia.
| |
Collapse
|
3
|
Murphy AJ, Wilton SD, Aung-Htut MT, McIntosh CS. Down syndrome and DYRK1A overexpression: relationships and future therapeutic directions. Front Mol Neurosci 2024; 17:1391564. [PMID: 39114642 PMCID: PMC11303307 DOI: 10.3389/fnmol.2024.1391564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 07/15/2024] [Indexed: 08/10/2024] Open
Abstract
Down syndrome is a genetic-based disorder that results from the triplication of chromosome 21, leading to an overexpression of many triplicated genes, including the gene encoding Dual-Specificity Tyrosine Phosphorylation-Regulated Kinase 1A (DYRK1A). This protein has been observed to regulate numerous cellular processes, including cell proliferation, cell functioning, differentiation, and apoptosis. Consequently, an overexpression of DYRK1A has been reported to result in cognitive impairment, a key phenotype of individuals with Down syndrome. Therefore, downregulating DYRK1A has been explored as a potential therapeutic strategy for Down syndrome, with promising results observed from in vivo mouse models and human clinical trials that administered epigallocatechin gallate. Current DYRK1A inhibitors target the protein function directly, which tends to exhibit low specificity and selectivity, making them unfeasible for clinical or research purposes. On the other hand, antisense oligonucleotides (ASOs) offer a more selective therapeutic strategy to downregulate DYRK1A expression at the gene transcript level. Advances in ASO research have led to the discovery of numerous chemical modifications that increase ASO potency, specificity, and stability. Recently, several ASOs have been approved by the U.S. Food and Drug Administration to address neuromuscular and neurological conditions, laying the foundation for future ASO therapeutics. The limitations of ASOs, including their high production cost and difficulty delivering to target tissues can be overcome by further advances in ASO design. DYRK1A targeted ASOs could be a viable therapeutic approach to improve the quality of life for individuals with Down syndrome and their families.
Collapse
Affiliation(s)
- Aidan J. Murphy
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA, Australia
- Perron Institute for Neurological and Translational Science, Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Perth, WA, Australia
| | - Steve D. Wilton
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA, Australia
- Perron Institute for Neurological and Translational Science, Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Perth, WA, Australia
| | - May T. Aung-Htut
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA, Australia
- Perron Institute for Neurological and Translational Science, Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Perth, WA, Australia
| | - Craig S. McIntosh
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA, Australia
- Perron Institute for Neurological and Translational Science, Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Perth, WA, Australia
| |
Collapse
|
4
|
Lamantia J, Sloan K, Wallace JM, Roper RJ. Compromised femoral and lumbovertebral bone in the Dp(16)1Yey Down syndrome mouse model. Bone 2024; 181:117046. [PMID: 38336158 PMCID: PMC11000152 DOI: 10.1016/j.bone.2024.117046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/30/2024] [Accepted: 02/06/2024] [Indexed: 02/12/2024]
Abstract
Down syndrome (DS), affecting ∼1 in 800 live births, is caused by the triplication of human chromosome 21 (Hsa21). Individuals with DS have skeletal features including craniofacial abnormalities and decreased bone mineral density (BMD). Lowered BMD can lead to increased fracture risk, with common fracture points at the femoral neck and lumbar spine. While the femur has been studied in DS mouse models, there is little research done on the vertebrae despite evidence that humans with DS have affected vertebrae. Additionally, it is important to establish when skeletal deficits occur to find times of potential intervention. The Dp(16)1Yey DS mouse model has all genes triplicated on mouse chromosome 16 orthologous to Hsa21 and displayed deficits in long bone, including trabecular and cortical deficits in male but not female mice, at 12 weeks. We hypothesized that the long bone and lumbovertebral microarchitecture would exhibit sexually dimorphic deficits in Dp(16)1Yey mice compared to control mice and long bone strength would be diminished in Dp(16)1Yey mice at 6 weeks. The trabecular region of the 4th lumbar (L4) vertebra and the trabecular and cortical regions of the femur were analyzed via micro-computed tomography and 3-point bending in 6-week-old male and female Dp(16)1Yey and control mice. Trabecular and cortical deficits were observed in femurs from male Dp(16)1Yey mice, and cortical deficits were seen in femurs of male and female Dp(16)1Yey mice. Male Dp(16)1Yey femurs had more deficits in bone strength at whole bone and tissue-estimate level properties, but female Dp(16)1Yey mice were also affected. Additionally, the L4 of male and female Dp(16)1Yey mice show trabecular deficits, which have not been previously reported in a DS mouse model. Our results indicate that skeletal deficits associated with DS occur early in skeletal development, are dependent on skeletal compartment and site, are sex dependent, and potential interventions should likely begin early in skeletal development of DS mouse models.
Collapse
Affiliation(s)
- Joshua Lamantia
- Department of Biology, Indiana University-Purdue University Indianapolis (IUPUI), United States of America
| | - Kourtney Sloan
- Department of Biology, Indiana University-Purdue University Indianapolis (IUPUI), United States of America
| | - Joseph M Wallace
- Department of Biomedical Engineering, Indiana University-Purdue University Indianapolis (IUPUI), United States of America
| | - Randall J Roper
- Department of Biology, Indiana University-Purdue University Indianapolis (IUPUI), United States of America.
| |
Collapse
|
5
|
Llambrich S, Tielemans B, Saliën E, Atzori M, Wouters K, Van Bulck V, Platt M, Vanherp L, Gallego Fernandez N, Grau de la Fuente L, Poptani H, Verlinden L, Himmelreich U, Croitor A, Attanasio C, Callaerts-Vegh Z, Gsell W, Martínez-Abadías N, Vande Velde G. Pleiotropic effects of trisomy and pharmacologic modulation on structural, functional, molecular, and genetic systems in a Down syndrome mouse model. eLife 2024; 12:RP89763. [PMID: 38497812 PMCID: PMC10948151 DOI: 10.7554/elife.89763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2024] Open
Abstract
Down syndrome (DS) is characterized by skeletal and brain structural malformations, cognitive impairment, altered hippocampal metabolite concentration and gene expression imbalance. These alterations were usually investigated separately, and the potential rescuing effects of green tea extracts enriched in epigallocatechin-3-gallate (GTE-EGCG) provided disparate results due to different experimental conditions. We overcame these limitations by conducting the first longitudinal controlled experiment evaluating genotype and GTE-EGCG prenatal chronic treatment effects before and after treatment discontinuation. Our findings revealed that the Ts65Dn mouse model reflected the pleiotropic nature of DS, exhibiting brachycephalic skull, ventriculomegaly, neurodevelopmental delay, hyperactivity, and impaired memory robustness with altered hippocampal metabolite concentration and gene expression. GTE-EGCG treatment modulated most systems simultaneously but did not rescue DS phenotypes. On the contrary, the treatment exacerbated trisomic phenotypes including body weight, tibia microarchitecture, neurodevelopment, adult cognition, and metabolite concentration, not supporting the therapeutic use of GTE-EGCG as a prenatal chronic treatment. Our results highlight the importance of longitudinal experiments assessing the co-modulation of multiple systems throughout development when characterizing preclinical models in complex disorders and evaluating the pleiotropic effects and general safety of pharmacological treatments.
Collapse
Affiliation(s)
- Sergi Llambrich
- Biomedical MRI, Department of Imaging and Pathology, KU LeuvenLeuvenBelgium
| | - Birger Tielemans
- Biomedical MRI, Department of Imaging and Pathology, KU LeuvenLeuvenBelgium
| | - Ellen Saliën
- Biomedical MRI, Department of Imaging and Pathology, KU LeuvenLeuvenBelgium
| | - Marta Atzori
- Department of Human Genetics, KU LeuvenLeuvenBelgium
| | - Kaat Wouters
- Laboratory of Biological Psychology, KU LeuvenLeuvenBelgium
| | | | - Mark Platt
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer Medicine, University of LiverpoolLiverpoolUnited Kingdom
| | - Laure Vanherp
- Biomedical MRI, Department of Imaging and Pathology, KU LeuvenLeuvenBelgium
| | - Nuria Gallego Fernandez
- Departament de Biologia Evolutiva, Ecologia i Ciències Ambientals (BEECA), Facultat de Biologia, Universitat de BarcelonaBarcelonaSpain
| | - Laura Grau de la Fuente
- Departament de Biologia Evolutiva, Ecologia i Ciències Ambientals (BEECA), Facultat de Biologia, Universitat de BarcelonaBarcelonaSpain
| | - Harish Poptani
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer Medicine, University of LiverpoolLiverpoolUnited Kingdom
| | - Lieve Verlinden
- Clinical and Experimental Endocrinology, KU LeuvenLeuvenBelgium
| | - Uwe Himmelreich
- Biomedical MRI, Department of Imaging and Pathology, KU LeuvenLeuvenBelgium
| | - Anca Croitor
- Biomedical MRI, Department of Imaging and Pathology, KU LeuvenLeuvenBelgium
| | | | | | - Willy Gsell
- Biomedical MRI, Department of Imaging and Pathology, KU LeuvenLeuvenBelgium
| | - Neus Martínez-Abadías
- Departament de Biologia Evolutiva, Ecologia i Ciències Ambientals (BEECA), Facultat de Biologia, Universitat de BarcelonaBarcelonaSpain
| | | |
Collapse
|
6
|
Chapman LR, Ramnarine IVP, Zemke D, Majid A, Bell SM. Gene Expression Studies in Down Syndrome: What Do They Tell Us about Disease Phenotypes? Int J Mol Sci 2024; 25:2968. [PMID: 38474215 DOI: 10.3390/ijms25052968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 02/27/2024] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
Down syndrome is a well-studied aneuploidy condition in humans, which is associated with various disease phenotypes including cardiovascular, neurological, haematological and immunological disease processes. This review paper aims to discuss the research conducted on gene expression studies during fetal development. A descriptive review was conducted, encompassing all papers published on the PubMed database between September 1960 and September 2022. We found that in amniotic fluid, certain genes such as COL6A1 and DSCR1 were found to be affected, resulting in phenotypical craniofacial changes. Additionally, other genes such as GSTT1, CLIC6, ITGB2, C21orf67, C21orf86 and RUNX1 were also identified to be affected in the amniotic fluid. In the placenta, dysregulation of genes like MEST, SNF1LK and LOX was observed, which in turn affected nervous system development. In the brain, dysregulation of genes DYRK1A, DNMT3L, DNMT3B, TBX1, olig2 and AQP4 has been shown to contribute to intellectual disability. In the cardiac tissues, dysregulated expression of genes GART, ETS2 and ERG was found to cause abnormalities. Furthermore, dysregulation of XIST, RUNX1, SON, ERG and STAT1 was observed, contributing to myeloproliferative disorders. Understanding the differential expression of genes provides insights into the genetic consequences of DS. A better understanding of these processes could potentially pave the way for the development of genetic and pharmacological therapies.
Collapse
Affiliation(s)
- Laura R Chapman
- Sheffield Children's NHS Foundation Trust, Clarkson St, Sheffield S10 2TH, UK
- Sheffield Institute of Translational Neuroscience, University of Sheffield, Glossop Road, Sheffield S10 2GF, UK
| | - Isabela V P Ramnarine
- Sheffield Institute of Translational Neuroscience, University of Sheffield, Glossop Road, Sheffield S10 2GF, UK
| | - Dan Zemke
- Sheffield Institute of Translational Neuroscience, University of Sheffield, Glossop Road, Sheffield S10 2GF, UK
| | - Arshad Majid
- Sheffield Institute of Translational Neuroscience, University of Sheffield, Glossop Road, Sheffield S10 2GF, UK
- Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Glossop Road, Sheffield S10 2GJ, UK
| | - Simon M Bell
- Sheffield Institute of Translational Neuroscience, University of Sheffield, Glossop Road, Sheffield S10 2GF, UK
- Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Glossop Road, Sheffield S10 2GJ, UK
| |
Collapse
|
7
|
Barwe SP, Kolb EA, Gopalakrishnapillai A. Down syndrome and leukemia: An insight into the disease biology and current treatment options. Blood Rev 2024; 64:101154. [PMID: 38016838 DOI: 10.1016/j.blre.2023.101154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/31/2023] [Accepted: 11/19/2023] [Indexed: 11/30/2023]
Abstract
Children with Down syndrome (DS) have a 10- to 20-fold greater predisposition to develop acute leukemia compared to the general population, with a skew towards myeloid leukemia (ML-DS). While ML-DS is known to be a subtype with good outcome, patients who relapse face a dismal prognosis. Acute lymphocytic leukemia in DS (DS-ALL) is considered to have poor prognosis. The relapse rate is high in DS-ALL compared to their non-DS counterparts. We have a better understanding about the mutational spectrum of DS leukemia. Studies using animal, embryonic stem cell- and induced pluripotent stem cell-based models have shed light on the mechanism by which these mutations contribute to disease initiation and progression. In this review, we list the currently available treatment strategies for DS-leukemias along with their outcome with emphasis on challenges with chemotherapy-related toxicities in children with DS. We focus on the mechanisms of initiation and progression of leukemia in children with DS and highlight the novel molecular targets with greater success in preclinical trials that have the potential to progress to the clinic.
Collapse
Affiliation(s)
- Sonali P Barwe
- Lisa Dean Moseley Institute for Cancer and Blood Disorders, Nemours Children's Health, Wilmington, Delaware, 19803, USA
| | - E Anders Kolb
- Lisa Dean Moseley Institute for Cancer and Blood Disorders, Nemours Children's Health, Wilmington, Delaware, 19803, USA
| | - Anilkumar Gopalakrishnapillai
- Lisa Dean Moseley Institute for Cancer and Blood Disorders, Nemours Children's Health, Wilmington, Delaware, 19803, USA.
| |
Collapse
|
8
|
Lana-Elola E, Aoidi R, Llorian M, Gibbins D, Buechsenschuetz C, Bussi C, Flynn H, Gilmore T, Watson-Scales S, Haugsten Hansen M, Hayward D, Song OR, Brault V, Herault Y, Deau E, Meijer L, Snijders AP, Gutierrez MG, Fisher EMC, Tybulewicz VLJ. Increased dosage of DYRK1A leads to congenital heart defects in a mouse model of Down syndrome. Sci Transl Med 2024; 16:eadd6883. [PMID: 38266108 PMCID: PMC7615651 DOI: 10.1126/scitranslmed.add6883] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 12/22/2023] [Indexed: 01/26/2024]
Abstract
Down syndrome (DS) is caused by trisomy of human chromosome 21 (Hsa21). DS is a gene dosage disorder that results in multiple phenotypes including congenital heart defects. This clinically important cardiac pathology is the result of a third copy of one or more of the approximately 230 genes on Hsa21, but the identity of the causative dosage-sensitive genes and hence mechanisms underlying this cardiac pathology remain unclear. Here, we show that hearts from human fetuses with DS and embryonic hearts from the Dp1Tyb mouse model of DS show reduced expression of mitochondrial respiration genes and cell proliferation genes. Using systematic genetic mapping, we determined that three copies of the dual-specificity tyrosine phosphorylation-regulated kinase 1A (Dyrk1a) gene, encoding a serine/threonine protein kinase, are associated with congenital heart disease pathology. In embryos from Dp1Tyb mice, reducing Dyrk1a gene copy number from three to two reversed defects in cellular proliferation and mitochondrial respiration in cardiomyocytes and rescued heart septation defects. Increased dosage of DYRK1A protein resulted in impairment of mitochondrial function and congenital heart disease pathology in mice with DS, suggesting that DYRK1A may be a useful therapeutic target for treating this common human condition.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Véronique Brault
- Université de Strasbourg, CNRS UMR7104, INSERM U1258, Institut de Génétique et de Biologie Moléculaire et Cellulaire, IGBMC, BP 10142, 1 rue Laurent Fries, 67404 Illkirch CEDEX, France
| | - Yann Herault
- Université de Strasbourg, CNRS UMR7104, INSERM U1258, Institut de Génétique et de Biologie Moléculaire et Cellulaire, IGBMC, BP 10142, 1 rue Laurent Fries, 67404 Illkirch CEDEX, France
| | - Emmanuel Deau
- Perha Pharmaceuticals, Presqu'île de Perharidy, 29680 Roscoff, France
| | - Laurent Meijer
- Perha Pharmaceuticals, Presqu'île de Perharidy, 29680 Roscoff, France
| | | | | | - Elizabeth M C Fisher
- Department of Neuromuscular Diseases, UCL Institute of Neurology, London WC1N 3BG, UK
| | | |
Collapse
|
9
|
Zhegalova IV, Vasiluev PA, Flyamer IM, Shtompel AS, Glazyrina E, Shilova N, Minzhenkova M, Markova Z, Petrova NV, Dashinimaev EB, Razin SV, Ulianov SV. Trisomies Reorganize Human 3D Genome. Int J Mol Sci 2023; 24:16044. [PMID: 38003233 PMCID: PMC10671006 DOI: 10.3390/ijms242216044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 11/03/2023] [Accepted: 11/05/2023] [Indexed: 11/26/2023] Open
Abstract
Trisomy is the presence of one extra copy of an entire chromosome or its part in a cell nucleus. In humans, autosomal trisomies are associated with severe developmental abnormalities leading to embryonic lethality, miscarriage or pronounced deviations of various organs and systems at birth. Trisomies are characterized by alterations in gene expression level, not exclusively on the trisomic chromosome, but throughout the genome. Here, we applied the high-throughput chromosome conformation capture technique (Hi-C) to study chromatin 3D structure in human chorion cells carrying either additional chromosome 13 (Patau syndrome) or chromosome 16 and in cultured fibroblasts with extra chromosome 18 (Edwards syndrome). The presence of extra chromosomes results in systematic changes of contact frequencies between small and large chromosomes. Analyzing the behavior of individual chromosomes, we found that a limited number of chromosomes change their contact patterns stochastically in trisomic cells and that it could be associated with lamina-associated domains (LAD) and gene content. For trisomy 13 and 18, but not for trisomy 16, the proportion of compacted loci on a chromosome is correlated with LAD content. We also found that regions of the genome that become more compact in trisomic cells are enriched in housekeeping genes, indicating a possible decrease in chromatin accessibility and transcription level of these genes. These results provide a framework for understanding the mechanisms of pan-genome transcription dysregulation in trisomies in the context of chromatin spatial organization.
Collapse
Affiliation(s)
- Irina V. Zhegalova
- Center for Molecular and Cellular Biology, Skolkovo Institute of Science and Technology, 143026 Moscow, Russia
- A.A. Kharkevich Institute for Information Transmission Problems, Russian Academy of Sciences, 127051 Moscow, Russia
| | | | - Ilya M. Flyamer
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, 4058 Basel, Switzerland
| | - Anastasia S. Shtompel
- Department of Molecular Biology, Faculty of Biology, M.V. Lomonosov Moscow State University, 119234 Moscow, Russia
- Laboratory of Structural-Functional Organization of Chromosomes, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia;
| | | | | | | | - Zhanna Markova
- Research Centre for Medical Genetics, 115522 Moscow, Russia
| | - Natalia V. Petrova
- Laboratory of Structural-Functional Organization of Chromosomes, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia;
| | - Erdem B. Dashinimaev
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Sergey V. Razin
- Department of Molecular Biology, Faculty of Biology, M.V. Lomonosov Moscow State University, 119234 Moscow, Russia
- Laboratory of Structural-Functional Organization of Chromosomes, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia;
| | - Sergey V. Ulianov
- Department of Molecular Biology, Faculty of Biology, M.V. Lomonosov Moscow State University, 119234 Moscow, Russia
- Laboratory of Structural-Functional Organization of Chromosomes, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia;
| |
Collapse
|
10
|
Arthun J, Aldaz S, Hayes P, Roberts JS, Olson LE. Volume of the thalamus and hypothalamus in the Ts1Rhr and Ms1Rhr mouse models relevant to Down syndrome. MICROPUBLICATION BIOLOGY 2023; 2023:10.17912/micropub.biology.000981. [PMID: 38021171 PMCID: PMC10644112 DOI: 10.17912/micropub.biology.000981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 09/26/2023] [Accepted: 10/29/2023] [Indexed: 12/01/2023]
Abstract
A variety of mouse models for Down syndrome (Trisomy 21) have been created to test hypotheses about the correlation of phenotypes to gene content and copy number. Ts1Rhr mice are trisomic for a region on mouse chromosome 16 that is homologous to 5.3 Mb of human chromosome 21. Ms1Rhr mice are monosomic for this region. Magnetic Resonance Imaging (MRI) has revealed characteristic volumetric changes in the brains of humans with Down syndrome such as reductions in the cerebellum, hippocampus, and brain stem, and increases in the ventricles and thalamus. We used MRI with region of interest analysis to measure the volume of the thalamus and hypothalamus in Ts1Rhr, Ms1Rhr, and euploid control mice (n = 10-11 per group). Ts1Rhr mice had a 6.6% reduction and Ms1Rhr mice had an 8.2% reduction in the volume of the thalamus. Ts1Rhr and Ms1Rhr hypothalamic volumes were equivalent to controls. Conflicting data in mouse models show a lack of clarity on causative roles of regions homologous to human chromosome 21 in phenotypes related to the thalamus and hypothalamus in Down syndrome.
Collapse
Affiliation(s)
| | | | - Pete Hayes
- Biology, University of Redlands, Redlands, CA USA
| | | | | |
Collapse
|
11
|
Redhead Y, Gibbins D, Lana-Elola E, Watson-Scales S, Dobson L, Krause M, Liu KJ, Fisher EMC, Green JBA, Tybulewicz VLJ. Craniofacial dysmorphology in Down syndrome is caused by increased dosage of Dyrk1a and at least three other genes. Development 2023; 150:dev201077. [PMID: 37102702 PMCID: PMC10163349 DOI: 10.1242/dev.201077] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 03/21/2023] [Indexed: 04/28/2023]
Abstract
Down syndrome (DS), trisomy of human chromosome 21 (Hsa21), occurs in 1 in 800 live births and is the most common human aneuploidy. DS results in multiple phenotypes, including craniofacial dysmorphology, which is characterised by midfacial hypoplasia, brachycephaly and micrognathia. The genetic and developmental causes of this are poorly understood. Using morphometric analysis of the Dp1Tyb mouse model of DS and an associated mouse genetic mapping panel, we demonstrate that four Hsa21-orthologous regions of mouse chromosome 16 contain dosage-sensitive genes that cause the DS craniofacial phenotype, and identify one of these causative genes as Dyrk1a. We show that the earliest and most severe defects in Dp1Tyb skulls are in bones of neural crest (NC) origin, and that mineralisation of the Dp1Tyb skull base synchondroses is aberrant. Furthermore, we show that increased dosage of Dyrk1a results in decreased NC cell proliferation and a decrease in size and cellularity of the NC-derived frontal bone primordia. Thus, DS craniofacial dysmorphology is caused by an increased dosage of Dyrk1a and at least three other genes.
Collapse
Affiliation(s)
- Yushi Redhead
- Centre for Craniofacial Biology and Regenerative Biology, King's College London, London SE1 9RT, UK
- The Francis Crick Institute, London NW1 1AT, UK
| | | | | | | | - Lisa Dobson
- Centre for Craniofacial Biology and Regenerative Biology, King's College London, London SE1 9RT, UK
- Randall Centre for Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK
| | - Matthias Krause
- Randall Centre for Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK
| | - Karen J. Liu
- Centre for Craniofacial Biology and Regenerative Biology, King's College London, London SE1 9RT, UK
| | | | - Jeremy B. A. Green
- Centre for Craniofacial Biology and Regenerative Biology, King's College London, London SE1 9RT, UK
| | | |
Collapse
|
12
|
Sloan K, Thomas J, Blackwell M, Voisard D, Lana-Elola E, Watson-Scales S, Roper DL, Wallace JM, Fisher EMC, Tybulewicz VLJ, Roper RJ. Genetic dissection of triplicated chromosome 21 orthologs yields varying skeletal traits in Down syndrome model mice. Dis Model Mech 2023; 16:dmm049927. [PMID: 36939025 PMCID: PMC10163323 DOI: 10.1242/dmm.049927] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 03/02/2023] [Indexed: 03/21/2023] Open
Abstract
Down syndrome (DS) phenotypes result from triplicated genes, but the effects of three copy genes are not well known. A mouse mapping panel genetically dissecting human chromosome 21 (Hsa21) syntenic regions was used to investigate the contributions and interactions of triplicated Hsa21 orthologous genes on mouse chromosome 16 (Mmu16) on skeletal phenotypes. Skeletal structure and mechanical properties were assessed in femurs of male and female Dp9Tyb, Dp2Tyb, Dp3Tyb, Dp4Tyb, Dp5Tyb, Dp6Tyb, Ts1Rhr and Dp1Tyb;Dyrk1a+/+/- mice. Dp1Tyb mice, with the entire Hsa21 homologous region of Mmu16 triplicated, display bone deficits similar to those of humans with DS and served as a baseline for other strains in the panel. Bone phenotypes varied based on triplicated gene content, sex and bone compartment. Three copies of Dyrk1a played a sex-specific, essential role in trabecular deficits and may interact with other genes to influence cortical deficits related to DS. Triplicated genes in Dp9Tyb and Dp2Tyb mice improved some skeletal parameters. As triplicated genes can both improve and worsen bone deficits, it is important to understand the interaction between and molecular mechanisms of skeletal alterations affected by these genes.
Collapse
Affiliation(s)
- Kourtney Sloan
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Jared Thomas
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Matthew Blackwell
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Deanna Voisard
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | | | | | | | - Joseph M. Wallace
- Department of Biomedical Engineering, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | | | | | - Randall J. Roper
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| |
Collapse
|
13
|
Sarver DC, Xu C, Velez LM, Aja S, Jaffe AE, Seldin MM, Reeves RH, Wong GW. Dysregulated systemic metabolism in a Down syndrome mouse model. Mol Metab 2023; 68:101666. [PMID: 36587842 PMCID: PMC9841171 DOI: 10.1016/j.molmet.2022.101666] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/14/2022] [Accepted: 12/26/2022] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVE Trisomy 21 is one of the most complex genetic perturbations compatible with postnatal survival. Dosage imbalance arising from the triplication of genes on human chromosome 21 (Hsa21) affects multiple organ systems. Much of Down syndrome (DS) research, however, has focused on addressing how aneuploidy dysregulates CNS function leading to cognitive deficit. Although obesity, diabetes, and associated sequelae such as fatty liver and dyslipidemia are well documented in the DS population, only limited studies have been conducted to determine how gene dosage imbalance affects whole-body metabolism. Here, we conduct a comprehensive and systematic analysis of key metabolic parameters across different physiological states in the Ts65Dn trisomic mouse model of DS. METHODS Ts65Dn mice and euploid littermates were subjected to comprehensive metabolic phenotyping under basal (chow-fed) state and the pathophysiological state of obesity induced by a high-fat diet (HFD). RNA sequencing of liver, skeletal muscle, and two major fat depots were conducted to determine the impact of aneuploidy on tissue transcriptome. Pathway enrichments, gene-centrality, and key driver estimates were performed to provide insights into tissue autonomous and non-autonomous mechanisms contributing to the dysregulation of systemic metabolism. RESULTS Under the basal state, chow-fed Ts65Dn mice of both sexes had elevated locomotor activity and energy expenditure, reduced fasting serum cholesterol levels, and mild glucose intolerance. Sexually dimorphic deterioration in metabolic homeostasis became apparent when mice were challenged with a high-fat diet. While obese Ts65Dn mice of both sexes exhibited dyslipidemia, male mice also showed impaired systemic insulin sensitivity, reduced mitochondrial activity, and elevated fibrotic and inflammatory gene signatures in the liver and adipose tissue. Systems-level analysis highlighted conserved pathways and potential endocrine drivers of adipose-liver crosstalk that contribute to dysregulated glucose and lipid metabolism. CONCLUSIONS A combined alteration in the expression of trisomic and disomic genes in peripheral tissues contribute to metabolic dysregulations in Ts65Dn mice. These data lay the groundwork for understanding the impact of aneuploidy on in vivo metabolism.
Collapse
Affiliation(s)
- Dylan C Sarver
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Cheng Xu
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Leandro M Velez
- Department of Biological Chemistry, University of California, Irvine, Irvine, USA; Center for Epigenetics and Metabolism, University of California Irvine, Irvine, USA
| | - Susan Aja
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Andrew E Jaffe
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA; The Lieber Institute for Brain Development, Baltimore, MD, USA; Center for Computational Biology, Johns Hopkins University, Baltimore, MD, USA; Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Marcus M Seldin
- Department of Biological Chemistry, University of California, Irvine, Irvine, USA; Center for Epigenetics and Metabolism, University of California Irvine, Irvine, USA
| | - Roger H Reeves
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - G William Wong
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
14
|
Hormann FM, Hoogkamer AQ, Boeree A, Sonneveld E, Escherich G, den Boer ML, Boer JM. Integrating copy number data of 64 iAMP21 BCP-ALL patients narrows the common region of amplification to 1.57 Mb. Front Oncol 2023; 13:1128560. [PMID: 36910655 PMCID: PMC9996016 DOI: 10.3389/fonc.2023.1128560] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 02/10/2023] [Indexed: 02/25/2023] Open
Abstract
Background and purpose Intrachromosomal amplification of chromosome 21 (iAMP21) is a rare subtype of B-cell precursor acute lymphoblastic leukaemia (BCP-ALL). It is unknown how iAMP21 contributes to leukaemia. The currently known commonly amplified region is 5.1 Mb. Methods We aimed to narrow down the common region of amplification by using high resolution techniques. Array comparative genomic hybridization (aCGH) was used to determine copy number aberrations, Affymetrix U133 Plus2 expression arrays were used to determine gene expression. Genome-wide expression correlations were evaluated using Globaltest. Results We narrowed down the common region of amplification by combining copy number data from 12 iAMP21 cases with 52 cases from literature. The combined common region of amplification was 1.57 Mb, located from 36.07 to 37.64 Mb (GRCh38). This region is located telomeric from, but not including, RUNX1, which is the locus commonly used to diagnose iAMP21. This narrow region, which falls inside the Down Syndrome critical region, includes 13 genes of which the expression of eight genes was significantly upregulated compared with 143 non-iAMP21 B-other cases. Among these, transcriptional repressor RIPPLY3 (also known as DSCR6) was the highest overexpressed gene (fold change = 4.2, FDR < 0.001) and most strongly correlated (R = 0.58) with iAMP21-related genome-wide expression changes. Discussion The more precise definition of the common region of amplification could be beneficial in the diagnosis of iAMP21 based on copy number analysis from DNA sequencing or arrays as well as stimulate functional research into the role of the included genes in iAMP21 biology.
Collapse
Affiliation(s)
- Femke M Hormann
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands.,Oncode Institute, Utrecht, Netherlands.,Erasmus Medical Center (MC) - Sophia Children's Hospital, Department of Pediatric Oncology and Hematology, Rotterdam, Netherlands
| | - Alex Q Hoogkamer
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands.,Oncode Institute, Utrecht, Netherlands
| | - Aurélie Boeree
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands.,Oncode Institute, Utrecht, Netherlands
| | - Edwin Sonneveld
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands.,Dutch Childhood Oncology Group, Utrecht, Netherlands
| | - Gabriele Escherich
- Cooperative study group for childhood acute lymphoblastic leukaemia (COALL) - German Cooperative Study Group for Childhood Acute Lymphoblastic Leukemia, Hamburg, Germany.,Clinic of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Monique L den Boer
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands.,Oncode Institute, Utrecht, Netherlands.,Erasmus Medical Center (MC) - Sophia Children's Hospital, Department of Pediatric Oncology and Hematology, Rotterdam, Netherlands.,Dutch Childhood Oncology Group, Utrecht, Netherlands
| | - Judith M Boer
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands.,Oncode Institute, Utrecht, Netherlands
| |
Collapse
|
15
|
Krivega M, Stiefel CM, Storchova Z. Consequences of chromosome gain: A new view on trisomy syndromes. Am J Hum Genet 2022; 109:2126-2140. [PMID: 36459979 PMCID: PMC9808507 DOI: 10.1016/j.ajhg.2022.10.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Chromosome gains are detrimental for the development of the human embryo. As such, autosomal trisomies almost always result in spontaneous abortion, and the rare embryos surviving until live birth suffer from a plethora of pathological defects. There is no treatment currently available to ameliorate the consequences of trisomies, such as Down syndrome (trisomy of chromosome 21). Identifying the source of the phenotypes observed in cells with extra chromosomes is crucial for understanding the underlying molecular causes of trisomy syndromes. Although increased expression of the genes localized on the extra chromosome triggers several pathological phenotypes, an alternative model suggests that global, aneuploidy-associated changes in cellular physiology also contribute to the pathology. Here, we compare the molecular consequences of trisomy syndromes in vivo against engineered cell lines carrying various chromosome gains in vitro. We point out several phenotypes that are shared by variable trisomies and, therefore, might be caused by the presence of an extra chromosome per se, independent of its identity. This alternative view may provide useful insights for understanding Down syndrome pathology and open additional opportunities for diagnostics and treatments.
Collapse
Affiliation(s)
- Maria Krivega
- Reproduction Genetics, Department of Endocrinology and Infertility Disorders, Women Hospital, Heidelberg University, Im Neuenheimer Feld 440, 69120 Heidelberg, Germany.
| | - Clara M Stiefel
- Department of Radiation Oncology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Zuzana Storchova
- Department of Molecular Genetics, Faculty of Biology, TU Kaiserslautern, Paul-Ehrlich-Str. 24, 67663 Kaiserslautern, Germany
| |
Collapse
|
16
|
Sherman KM, Williams DK, Welsh CA, Cooper AM, Falck A, Huggins S, Bokhari RS, Gaddy D, McKelvey KD, Dawson LA, Suva LJ. Low bone mass and impaired fracture healing in mouse models of Trisomy21 (Down syndrome). Bone 2022; 162:116471. [PMID: 35716916 PMCID: PMC9356441 DOI: 10.1016/j.bone.2022.116471] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 06/01/2022] [Accepted: 06/09/2022] [Indexed: 11/16/2022]
Abstract
Individuals with Down syndrome (DS), the result of trisomy of human chromosome Hsa21 (Ts21), present with an array of skeletal abnormalities typified by altered craniofacial features, short stature and low bone mineral density (BMD). While bone deficits progress with age in both sexes, low bone mass is more pronounced in DS men than women and osteopenia appears earlier. In the current study, the reproductive hormone status (FSH, LH, testosterone) of 17 DS patients (males, ages range 19-52 years) was measured. Although testosterone was consistently low, the hypothalamic-pituitary-gonadal axis was intact with corresponding rises in FSH and LH. To provide further insight into the heterogeneity of the bone mass in DS, the skeletal phenotypes of three of the most used murine DS models, Ts65Dn (Ts65), TC1, and Dp16(Yey1) (Dp16) were characterized and contrasted. Evaluation of the bone phenotype of both male and female 3-month-old Dp16 mice demonstrated sexual dimorphism, with low bone mass apparent in males, as it is in Ts65, but not in female Dp16. In contrast, male TC1 mice had no apparent bone phenotype. To determine whether low bone mass in DS impacted fracture healing, fractures of the middle phalanx (P2) digits were generated in both male and female Dp16 mice at 15 weeks of age, an age where the sexually dimorphic low BMD persisted. Fracture healing was assessed via in vivo microCT over (13 weeks) 93 days post fracture (DPF). At 93 DPF, 0 % of DS male (n = 12) or female (n = 8) fractures healed, compared to 50 % of the male (n = 28) or female (n = 8) WT littermate fractures. MicroCT revealed periosteal unbridged mineralized callus formation across the fracture gap in Dp16 mice, which was confirmed by subsequent histology. These studies provide the first direct evidence of significantly impaired fracture healing in the setting of DS.
Collapse
Affiliation(s)
- Kirby M Sherman
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, United States of America
| | - Diarra K Williams
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, United States of America
| | - Casey A Welsh
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, United States of America
| | - Alexis M Cooper
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, United States of America
| | - Alyssa Falck
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, United States of America
| | - Shannon Huggins
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, United States of America
| | - Rihana S Bokhari
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, United States of America
| | - Dana Gaddy
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, United States of America
| | - Kent D McKelvey
- Department of Family Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States of America; Department of Medical Genetics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States of America
| | - Lindsay A Dawson
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, United States of America
| | - Larry J Suva
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, United States of America.
| |
Collapse
|
17
|
Wang J, Chen Z, He F, Lee T, Cai W, Chen W, Miao N, Zeng Z, Hussain G, Yang Q, Guo Q, Sun T. Single-Cell Transcriptomics of Cultured Amniotic Fluid Cells Reveals Complex Gene Expression Alterations in Human Fetuses With Trisomy 18. Front Cell Dev Biol 2022; 10:825345. [PMID: 35392164 PMCID: PMC8980718 DOI: 10.3389/fcell.2022.825345] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 02/24/2022] [Indexed: 12/12/2022] Open
Abstract
Trisomy 18, commonly known as Edwards syndrome, is the second most common autosomal trisomy among live born neonates. Multiple tissues including cardiac, abdominal, and nervous systems are affected by an extra chromosome 18. To delineate the complexity of anomalies of trisomy 18, we analyzed cultured amniotic fluid cells from two euploid and three trisomy 18 samples using single-cell transcriptomics. We identified 6 cell groups, which function in development of major tissues such as kidney, vasculature and smooth muscle, and display significant alterations in gene expression as detected by single-cell RNA-sequencing. Moreover, we demonstrated significant gene expression changes in previously proposed trisomy 18 critical regions, and identified three new regions such as 18p11.32, 18q11 and 18q21.32, which are likely associated with trisomy 18 phenotypes. Our results indicate complexity of trisomy 18 at the gene expression level and reveal genetic reasoning of diverse phenotypes in trisomy 18 patients.
Collapse
Affiliation(s)
- Jing Wang
- Center for Precision Medicine, School of Medicine and School of Biomedical Sciences, Huaqiao University, Xiamen, China
- College of Materials Science and Engineering, Huaqiao University, Xiamen, China
| | - Zixi Chen
- Shenzhen Key Laboratory of Marine Bioresource and Eco- Environmental Science, Shenzhen Engineering Laboratory for Marine Algal Biotechnology, Guangdong Provincial Key Laboratory for Plant Epigenetics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Fei He
- Genergy Bio-Technology (Shanghai) Co., Ltd, Shanghai, China
| | - Trevor Lee
- Department of Cell and Developmental Biology, Cornell University Weill Medical College, New York, NY, United States
| | - Wenjie Cai
- Department of Radiation Oncology, First Hospital of Quanzhou, Fujian Medical University, Quanzhou, China
| | - Wanhua Chen
- Department of Clinical Laboratory, First Hospital of Quanzhou, Fujian Medical University, Quanzhou, China
| | - Nan Miao
- Center for Precision Medicine, School of Medicine and School of Biomedical Sciences, Huaqiao University, Xiamen, China
| | - Zhiwei Zeng
- Center for Precision Medicine, School of Medicine and School of Biomedical Sciences, Huaqiao University, Xiamen, China
| | - Ghulam Hussain
- Neurochemical Biology and Genetics Laboratory, Department of Physiology, Faculty of Life Sciences, Government College University, Faisalabad, Pakistan
| | - Qingwei Yang
- Department of Neurology, School of Medicine, Zhongshan Hospital, Xiamen University, Xiamen, China
| | - Qiwei Guo
- United Diagnostic and Research Center for Clinical Genetics, School of Medicine and School of Public Health, Women and Children’s Hospital, Xiamen University, Xiamen, China
- *Correspondence: Qiwei Guo, ; Tao Sun,
| | - Tao Sun
- Center for Precision Medicine, School of Medicine and School of Biomedical Sciences, Huaqiao University, Xiamen, China
- *Correspondence: Qiwei Guo, ; Tao Sun,
| |
Collapse
|
18
|
Venegas-Zamora L, Bravo-Acuña F, Sigcho F, Gomez W, Bustamante-Salazar J, Pedrozo Z, Parra V. New Molecular and Organelle Alterations Linked to Down Syndrome Heart Disease. Front Genet 2022; 12:792231. [PMID: 35126461 PMCID: PMC8808411 DOI: 10.3389/fgene.2021.792231] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 12/13/2021] [Indexed: 12/13/2022] Open
Abstract
Down syndrome (DS) is a genetic disorder caused by a trisomy of the human chromosome 21 (Hsa21). Overexpression of Hsa21 genes that encode proteins and non-coding RNAs (ncRNAs) can disrupt several cellular functions and biological processes, especially in the heart. Congenital heart defects (CHDs) are present in 45–50% of individuals with DS. Here, we describe the genetic background of this condition (Hsa21 and non-Hsa21 genes), including the role of ncRNAs, and the relevance of these new players in the study of the pathophysiology of DS heart diseases. Additionally, we discuss several distinct pathways in cardiomyocytes which help maintain a functional heart, but that might trigger hypertrophy and oxidative stress when altered. Moreover, we highlight the importance of investigating how mitochondrial and lysosomal dysfunction could eventually contribute to understanding impaired heart function and development in subjects with the Hsa21 trisomy. Altogether, this review focuses on the newest insights about the gene expression, molecular pathways, and organelle alterations involved in the cardiac phenotype of DS.
Collapse
Affiliation(s)
- Leslye Venegas-Zamora
- Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas y Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Francisco Bravo-Acuña
- Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas y Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Francisco Sigcho
- Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas y Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Wileidy Gomez
- Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas y Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Laboratory of Neuroprotection and Autophagy, Center for Integrative Biology, Faculty of Science, Universidad Mayor, Santiago, Chile
| | - José Bustamante-Salazar
- Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas y Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Zully Pedrozo
- Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas y Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Programa de Fisiología y Biofísica, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Red para El Estudio de Enfermedades Cardiopulmonares de Alta Letalidad (REECPAL), Universidad de Chile, Santiago, Chile
- *Correspondence: Zully Pedrozo, ; Valentina Parra,
| | - Valentina Parra
- Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas y Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
- Red para El Estudio de Enfermedades Cardiopulmonares de Alta Letalidad (REECPAL), Universidad de Chile, Santiago, Chile
- *Correspondence: Zully Pedrozo, ; Valentina Parra,
| |
Collapse
|
19
|
Altered cell and RNA isoform diversity in aging Down syndrome brains. Proc Natl Acad Sci U S A 2021; 118:2114326118. [PMID: 34795060 PMCID: PMC8617492 DOI: 10.1073/pnas.2114326118] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/08/2021] [Indexed: 12/11/2022] Open
Abstract
Down syndrome (DS) neurocognitive disabilities associated with trisomy 21 are known; however, gene changes within individual brain cells occurring with age are unknown. Here, we interrogated >170,000 cells from 29 aging DS and control brains using single-nucleus RNA sequencing. We observed increases in inhibitory-over-excitatory neurons, microglial activation in the youngest DS brains coinciding with overexpression of genes associated with microglial-mediated synaptic pruning, and overexpression of the chromosome 21 gene RUNX1 that may be a potential driving factor in microglial activation. Single-nucleus long-read sequencing revealed hundreds of thousands of unannotated RNA transcripts. These included diverse species for the Alzheimer’s disease gene—amyloid precursor protein—that contained intra-exonic junctions previously associated with somatic gene recombination, which was also identified in ∼8,000 other genes. Down syndrome (DS), trisomy of human chromosome 21 (HSA21), is characterized by lifelong cognitive impairments and the development of the neuropathological hallmarks of Alzheimer’s disease (AD). The cellular and molecular modifications responsible for these effects are not understood. Here we performed single-nucleus RNA sequencing (snRNA-seq) employing both short- (Illumina) and long-read (Pacific Biosciences) sequencing technologies on a total of 29 DS and non-DS control prefrontal cortex samples. In DS, the ratio of inhibitory-to-excitatory neurons was significantly increased, which was not observed in previous reports examining sporadic AD. DS microglial transcriptomes displayed AD-related aging and activation signatures in advance of AD neuropathology, with increased microglial expression of C1q complement genes (associated with dendritic pruning) and the HSA21 transcription factor gene RUNX1. Long-read sequencing detected vast RNA isoform diversity within and among specific cell types, including numerous sequences that differed between DS and control brains. Notably, over 8,000 genes produced RNAs containing intra-exonic junctions, including amyloid precursor protein (APP) that had previously been associated with somatic gene recombination. These and related results illuminate large-scale cellular and transcriptomic alterations as features of the aging DS brain.
Collapse
|
20
|
Lana-Elola E, Cater H, Watson-Scales S, Greenaway S, Müller-Winkler J, Gibbins D, Nemes M, Slender A, Hough T, Keskivali-Bond P, Scudamore CL, Herbert E, Banks GT, Mobbs H, Canonica T, Tosh J, Noy S, Llorian M, Nolan PM, Griffin JL, Good M, Simon M, Mallon AM, Wells S, Fisher EMC, Tybulewicz VLJ. Comprehensive phenotypic analysis of the Dp1Tyb mouse strain reveals a broad range of Down syndrome-related phenotypes. Dis Model Mech 2021; 14:dmm049157. [PMID: 34477842 PMCID: PMC8543064 DOI: 10.1242/dmm.049157] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 08/26/2021] [Indexed: 12/24/2022] Open
Abstract
Down syndrome (DS), trisomy 21, results in many complex phenotypes including cognitive deficits, heart defects and craniofacial alterations. Phenotypes arise from an extra copy of human chromosome 21 (Hsa21) genes. However, these dosage-sensitive causative genes remain unknown. Animal models enable identification of genes and pathological mechanisms. The Dp1Tyb mouse model of DS has an extra copy of 63% of Hsa21-orthologous mouse genes. In order to establish whether this model recapitulates DS phenotypes, we comprehensively phenotyped Dp1Tyb mice using 28 tests of different physiological systems and found that 468 out of 1800 parameters were significantly altered. We show that Dp1Tyb mice have wide-ranging DS-like phenotypes, including aberrant erythropoiesis and megakaryopoiesis, reduced bone density, craniofacial changes, altered cardiac function, a pre-diabetic state, and deficits in memory, locomotion, hearing and sleep. Thus, Dp1Tyb mice are an excellent model for investigating complex DS phenotype-genotype relationships for this common disorder.
Collapse
Affiliation(s)
| | - Heather Cater
- MRC Harwell Institute, Harwell Campus, Didcot, OX11 0RD, UK
| | | | | | | | | | | | - Amy Slender
- The Francis Crick Institute, London NW1 1AT, UK
| | - Tertius Hough
- MRC Harwell Institute, Harwell Campus, Didcot, OX11 0RD, UK
| | | | | | | | | | - Helene Mobbs
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge CB2 1QW, UK
| | - Tara Canonica
- School of Psychology, Cardiff University, Cardiff CF10 3AT, UK
| | - Justin Tosh
- The Francis Crick Institute, London NW1 1AT, UK
- UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Suzanna Noy
- UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | | | | | - Julian L. Griffin
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge CB2 1QW, UK
- Imperial College Dementia Research Institute, Imperial College London, London W12 7TA, UK
| | - Mark Good
- School of Psychology, Cardiff University, Cardiff CF10 3AT, UK
| | - Michelle Simon
- MRC Harwell Institute, Harwell Campus, Didcot, OX11 0RD, UK
| | | | - Sara Wells
- MRC Harwell Institute, Harwell Campus, Didcot, OX11 0RD, UK
| | | | - Victor L. J. Tybulewicz
- The Francis Crick Institute, London NW1 1AT, UK
- Department of Immunology and Inflammation, Imperial College London, London W12 0NN, UK
| |
Collapse
|
21
|
Novel Approaches to an Integrated Route for Trisomy 21 Evaluation. Biomolecules 2021; 11:biom11091328. [PMID: 34572541 PMCID: PMC8465311 DOI: 10.3390/biom11091328] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/16/2021] [Accepted: 09/06/2021] [Indexed: 12/31/2022] Open
Abstract
Trisomy 21 (T21) is one of the most commonly occurring genetic disorders, caused by the partial or complete triplication of chromosome 21. Despite the significant progress in the diagnostic tools applied for prenatal screening, commonly used methods are still imprecise and involve invasive diagnostic procedures that are related to a maternal risk of miscarriage. In this case, novel prenatal biomarkers are still being evaluated using highly specialized techniques, which could increase the diagnostic usefulness of biochemical prenatal screening for T21. From the other hand, the T21′s pathogenesis, caused by the improper division of genetic material, disrupting many metabolic pathways, could be further evaluated with the use of omics methods, which could result in bringing relevant insights for the evaluation of potential medical targets. Accordingly, a literature search was undertaken to collect novel information about prenatal screening for Down syndrome with the use of advanced technology, with a particular emphasis on the evaluation of novel screening biomarkers and the discovery of potential medical targets. These meta-analyses are focused on novel approaches designed with the use of omics techniques, representing the most rapidly developing and promising field in research today. Considering the limitations and progress of these methods, the use of omics techniques in evaluating T21 pathogenesis could bring beneficial results in prenatal screening, simultaneously uncovering novel potential medical targets.
Collapse
|
22
|
Li Y, Xing Z, Yu T, Pao A, Daadi M, Yu YE. Coat Color-Facilitated Efficient Generation and Analysis of a Mouse Model of Down Syndrome Triplicated for All Human Chromosome 21 Orthologous Regions. Genes (Basel) 2021; 12:genes12081215. [PMID: 34440389 PMCID: PMC8393392 DOI: 10.3390/genes12081215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 07/31/2021] [Accepted: 08/05/2021] [Indexed: 11/16/2022] Open
Abstract
Down syndrome (DS) is one of the most complex genetic disorders in humans and a leading genetic cause of developmental delays and intellectual disabilities. The mouse remains an essential model organism in DS research because human chromosome 21 (Hsa21) is orthologously conserved with three regions in the mouse genome. Recent studies have revealed complex interactions among different triplicated genomic regions and Hsa21 gene orthologs that underlie major DS phenotypes. Because we do not know conclusively which triplicated genes are indispensable in such interactions for a specific phenotype, it is desirable that all evolutionarily conserved Hsa21 gene orthologs are triplicated in a complete model. For this reason, the Dp(10)1Yey/+;Dp(16)1Yey/+;Dp(17)1Yey/+ mouse is the most complete model of DS to reflect gene dosage effects because it is the only mutant triplicated for all Hsa21 orthologous regions. Recently, several groups have expressed concerns that efforts needed to generate the triple compound model would be so overwhelming that it may be impractical to take advantage of its unique strength. To alleviate these concerns, we developed a strategy to drastically improve the efficiency of generating the triple compound model with the aid of a targeted coat color, and the results confirmed that the mutant mice generated via this approach exhibited cognitive deficits.
Collapse
Affiliation(s)
- Yichen Li
- The Children’s Guild Foundation Down Syndrome Research Program, Genetics and Genomics Program and Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; (Y.L.); (Z.X.); (T.Y.); (A.P.)
| | - Zhuo Xing
- The Children’s Guild Foundation Down Syndrome Research Program, Genetics and Genomics Program and Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; (Y.L.); (Z.X.); (T.Y.); (A.P.)
| | - Tao Yu
- The Children’s Guild Foundation Down Syndrome Research Program, Genetics and Genomics Program and Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; (Y.L.); (Z.X.); (T.Y.); (A.P.)
| | - Annie Pao
- The Children’s Guild Foundation Down Syndrome Research Program, Genetics and Genomics Program and Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; (Y.L.); (Z.X.); (T.Y.); (A.P.)
| | - Marcel Daadi
- Regenerative Medicine and Aging Unit, Texas Biomedical Research Institute, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78245, USA;
| | - Y. Eugene Yu
- The Children’s Guild Foundation Down Syndrome Research Program, Genetics and Genomics Program and Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; (Y.L.); (Z.X.); (T.Y.); (A.P.)
- Genetics, Genomics and Bioinformatics Program, State University of New York at Buffalo, Buffalo, NY 14203, USA
- Correspondence:
| |
Collapse
|
23
|
Abstract
PURPOSE Down syndrome (DS) is caused by trisomy 21 (Ts21) and results in skeletal deficits including shortened stature, low bone mineral density, and a predisposition to early onset osteoporosis. Ts21 causes significant alterations in skeletal development, morphology of the appendicular skeleton, bone homeostasis, age-related bone loss, and bone strength. However, the genetic or cellular origins of DS skeletal phenotypes remain unclear. RECENT FINDINGS New studies reveal a sexual dimorphism in characteristics and onset of skeletal deficits that differ between DS and typically developing individuals. Age-related bone loss occurs earlier in the DS as compared to general population. Perturbations of DS skeletal quality arise from alterations in cellular and molecular pathways affected by the overexpression of trisomic genes. Sex-specific alterations occur in critical developmental pathways that disrupt bone accrual, remodeling, and homeostasis and are compounded by aging, resulting in increased risks for osteopenia, osteoporosis, and fracture in individuals with DS.
Collapse
Affiliation(s)
- Jared R Thomas
- Department of Biology, Indiana University-Purdue University Indianapolis, 723 West Michigan Street, SL 306, Indianapolis, IN, 46202-3275, USA
| | - Randall J Roper
- Department of Biology, Indiana University-Purdue University Indianapolis, 723 West Michigan Street, SL 306, Indianapolis, IN, 46202-3275, USA.
| |
Collapse
|
24
|
Duchon A, Del Mar Muniz Moreno M, Martin Lorenzo S, Silva de Souza MP, Chevalier C, Nalesso V, Meziane H, Loureiro de Sousa P, Noblet V, Armspach JP, Brault V, Herault Y. Multi-influential genetic interactions alter behaviour and cognition through six main biological cascades in Down syndrome mouse models. Hum Mol Genet 2021; 30:771-788. [PMID: 33693642 PMCID: PMC8161522 DOI: 10.1093/hmg/ddab012] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 12/13/2022] Open
Abstract
Down syndrome (DS) is the most common genetic form of intellectual disability caused by the presence of an additional copy of human chromosome 21 (Hsa21). To provide novel insights into genotype–phenotype correlations, we used standardized behavioural tests, magnetic resonance imaging and hippocampal gene expression to screen several DS mouse models for the mouse chromosome 16 region homologous to Hsa21. First, we unravelled several genetic interactions between different regions of chromosome 16 and how they contribute significantly to altering the outcome of the phenotypes in brain cognition, function and structure. Then, in-depth analysis of misregulated expressed genes involved in synaptic dysfunction highlighted six biological cascades centred around DYRK1A, GSK3β, NPY, SNARE, RHOA and NPAS4. Finally, we provide a novel vision of the existing altered gene–gene crosstalk and molecular mechanisms targeting specific hubs in DS models that should become central to better understanding of DS and improving the development of therapies.
Collapse
Affiliation(s)
- Arnaud Duchon
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), department of translational medicine and neurogenetics 1 rue Laurent Fries, 67404 Illkirch Graffenstaden, France
| | - Maria Del Mar Muniz Moreno
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), department of translational medicine and neurogenetics 1 rue Laurent Fries, 67404 Illkirch Graffenstaden, France
| | - Sandra Martin Lorenzo
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), department of translational medicine and neurogenetics 1 rue Laurent Fries, 67404 Illkirch Graffenstaden, France
| | - Marcia Priscilla Silva de Souza
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), department of translational medicine and neurogenetics 1 rue Laurent Fries, 67404 Illkirch Graffenstaden, France
| | - Claire Chevalier
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), department of translational medicine and neurogenetics 1 rue Laurent Fries, 67404 Illkirch Graffenstaden, France
| | - Valérie Nalesso
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), department of translational medicine and neurogenetics 1 rue Laurent Fries, 67404 Illkirch Graffenstaden, France
| | - Hamid Meziane
- Université de Strasbourg, CNRS, INSERM, Institut Clinique de la Souris (ICS), CELPHEDIA, PHENOMIN, 1 rue Laurent Fries, 67404 Illkirch Graffenstaden, France
| | | | - Vincent Noblet
- Université de Strasbourg, CNRS UMR 7357, ICube, FMTS, 67000 Strasbourg, France
| | - Jean-Paul Armspach
- Université de Strasbourg, CNRS UMR 7357, ICube, FMTS, 67000 Strasbourg, France
| | - Veronique Brault
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), department of translational medicine and neurogenetics 1 rue Laurent Fries, 67404 Illkirch Graffenstaden, France
| | - Yann Herault
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), department of translational medicine and neurogenetics 1 rue Laurent Fries, 67404 Illkirch Graffenstaden, France.,Université de Strasbourg, CNRS, INSERM, Institut Clinique de la Souris (ICS), CELPHEDIA, PHENOMIN, 1 rue Laurent Fries, 67404 Illkirch Graffenstaden, France
| |
Collapse
|
25
|
Moreau M, Benhaddou S, Dard R, Tolu S, Hamzé R, Vialard F, Movassat J, Janel N. Metabolic Diseases and Down Syndrome: How Are They Linked Together? Biomedicines 2021; 9:biomedicines9020221. [PMID: 33671490 PMCID: PMC7926648 DOI: 10.3390/biomedicines9020221] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 02/15/2021] [Accepted: 02/17/2021] [Indexed: 12/13/2022] Open
Abstract
Down syndrome is a genetic disorder caused by the presence of a third copy of chromosome 21, associated with intellectual disabilities. Down syndrome is associated with anomalies of both the nervous and endocrine systems. Over the past decades, dramatic advances in Down syndrome research and treatment have helped to extend the life expectancy of these patients. Improved life expectancy is obviously a positive outcome, but it is accompanied with the need to address previously overlooked complications and comorbidities of Down syndrome, including obesity and diabetes, in order to improve the quality of life of Down syndrome patients. In this focused review, we describe the associations between Down syndrome and comorbidities, obesity and diabetes, and we discuss the understanding of proposed mechanisms for the association of Down syndrome with metabolic disorders. Drawing molecular mechanisms through which Type 1 diabetes and Type 2 diabetes could be linked to Down syndrome could allow identification of novel drug targets and provide therapeutic solutions to limit the development of metabolic and cognitive disorders.
Collapse
Affiliation(s)
- Manon Moreau
- Laboratoire Processus Dégénératifs, Université de Paris, BFA, UMR 8251, CNRS, Stress et Vieillissemen, F-75013 Paris, France; (M.M.); (S.B.); (R.D.)
| | - Soukaina Benhaddou
- Laboratoire Processus Dégénératifs, Université de Paris, BFA, UMR 8251, CNRS, Stress et Vieillissemen, F-75013 Paris, France; (M.M.); (S.B.); (R.D.)
| | - Rodolphe Dard
- Laboratoire Processus Dégénératifs, Université de Paris, BFA, UMR 8251, CNRS, Stress et Vieillissemen, F-75013 Paris, France; (M.M.); (S.B.); (R.D.)
- Genetics Deptartment, CHI Poissy St Germain-en-Laye, F-78300 Poissy, France;
- Université Paris-Saclay, UVSQ, INRAE, ENVA, BREED, F-78350 Jouy-en-Josas, France
| | - Stefania Tolu
- Laboratoire de Biologie et Pathologie du Pancréas Endocrine, Université de Paris, BFA, UMR 8251, CNRS, F-75013 Paris, France; (S.T.); (R.H.); (J.M.)
| | - Rim Hamzé
- Laboratoire de Biologie et Pathologie du Pancréas Endocrine, Université de Paris, BFA, UMR 8251, CNRS, F-75013 Paris, France; (S.T.); (R.H.); (J.M.)
| | - François Vialard
- Genetics Deptartment, CHI Poissy St Germain-en-Laye, F-78300 Poissy, France;
- Université Paris-Saclay, UVSQ, INRAE, ENVA, BREED, F-78350 Jouy-en-Josas, France
| | - Jamileh Movassat
- Laboratoire de Biologie et Pathologie du Pancréas Endocrine, Université de Paris, BFA, UMR 8251, CNRS, F-75013 Paris, France; (S.T.); (R.H.); (J.M.)
| | - Nathalie Janel
- Laboratoire Processus Dégénératifs, Université de Paris, BFA, UMR 8251, CNRS, Stress et Vieillissemen, F-75013 Paris, France; (M.M.); (S.B.); (R.D.)
- Correspondence: ; Tel.: +33-1-57-27-83-60; Fax: +33-1-57-27-83-54
| |
Collapse
|
26
|
Proteomics Study of Peripheral Blood Mononuclear Cells in Down Syndrome Children. Antioxidants (Basel) 2020; 9:antiox9111112. [PMID: 33187268 PMCID: PMC7696178 DOI: 10.3390/antiox9111112] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/22/2020] [Accepted: 11/09/2020] [Indexed: 02/07/2023] Open
Abstract
Down syndrome (DS) is the most common chromosomal disorder and the leading genetic cause of intellectual disability in humans, which results from the triplication of chromosome 21. To search for biomarkers for the early detection and exploration of the disease mechanisms, here, we investigated the protein expression signature of peripheral blood mononuclear cells (PBMCs) in DS children compared with healthy donors (HD) by using an in-depth label-free shotgun proteomics approach. Identified proteins are found associated with metabolic pathways, cellular trafficking, DNA structure, stress response, cytoskeleton network, and signaling pathways. The results showed that a well-defined number of dysregulated pathways retain a prominent role in mediating DS pathological features. Further, proteomics results are consistent with published study in DS and provide evidences that increased oxidative stress and the increased induction of stress related response, is a participant in DS pathology. In addition, the expression levels of some key proteins have been validated by Western blot analysis while protein carbonylation, as marker of protein oxidation, was investigated. The results of this study propose that PBMCs from DS children might be in an activated state where endoplasmic reticulum stress and increased production of radical species are one of the primary events contributing to multiple DS pathological features.
Collapse
|
27
|
Arima-Yoshida F, Raveau M, Shimohata A, Amano K, Fukushima A, Watanave M, Kobayashi S, Hattori S, Usui M, Sago H, Mataga N, Miyakawa T, Yamakawa K, Manabe T. Impairment of spatial memory accuracy improved by Cbr1 copy number resumption and GABA B receptor-dependent enhancement of synaptic inhibition in Down syndrome model mice. Sci Rep 2020; 10:14187. [PMID: 32843708 PMCID: PMC7447763 DOI: 10.1038/s41598-020-71085-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 08/10/2020] [Indexed: 11/09/2022] Open
Abstract
Down syndrome is a complex genetic disorder caused by the presence of three copies of the chromosome 21 in humans. The most common models, carrying extra-copies of overlapping fragments of mouse chromosome 16 that is syntenic to human chromosome 21, are Ts2Cje, Ts1Cje and Ts1Rhr mice. In electrophysiological analyses using hippocampal slices, we found that the later phase of the depolarization during tetanic stimulation, which was regulated by GABAB receptors, was significantly smaller in Ts1Cje and Ts2Cje mice than that in WT controls but not in Ts1Rhr mice. Furthermore, isolated GABAB receptor-mediated inhibitory synaptic responses were larger in Ts1Cje mice. To our knowledge, this is the first report that directly shows the enhancement of GABAB receptor-mediated synaptic currents in Ts1Cje mice. These results suggest that GABAB receptor-mediated synaptic inhibition was enhanced in Ts1Cje and Ts2Cje mice but not in Ts1Rhr mice. The Cbr1 gene, which is present in three copies in Ts1Cje and Ts2Cje but not in Ts1Rhr, encodes carbonyl reductase that may facilitate GABAB-receptor activity through a reduction of prostaglandin E2 (PGE2). Interestingly, we found that a reduction of PGE2 and an memory impairment in Ts1Cje mice were alleviated when only Cbr1 was set back to two copies (Ts1Cje;Cbr1+/+/-). However, the GABAB receptor-dependent enhancement of synaptic inhibition in Ts1Cje was unaltered in Ts1Cje;Cbr1+/+/- mice. These results indicate that Cbr1 is one of the genes responsible for DS cognitive impairments and the gene(s) other than Cbr1, which is included in Ts1Cje but not in Ts1Rhr, is responsible for the GABAB receptor-dependent over-inhibition.
Collapse
Affiliation(s)
- Fumiko Arima-Yoshida
- Division of Neuronal Network, Institute of Medical Science, University of Tokyo, Tokyo, 108-8639, Japan
| | - Matthieu Raveau
- Laboratory for Neurogenetics, RIKEN Center for Brain Science, Wako, Saitama, 351-0198, Japan
| | - Atsushi Shimohata
- Laboratory for Neurogenetics, RIKEN Center for Brain Science, Wako, Saitama, 351-0198, Japan
| | - Kenji Amano
- Laboratory for Neurogenetics, RIKEN Center for Brain Science, Wako, Saitama, 351-0198, Japan
| | - Akihiro Fukushima
- Division of Neuronal Network, Institute of Medical Science, University of Tokyo, Tokyo, 108-8639, Japan
| | - Masashi Watanave
- Division of Neuronal Network, Institute of Medical Science, University of Tokyo, Tokyo, 108-8639, Japan
| | - Shizuka Kobayashi
- Division of Neuronal Network, Institute of Medical Science, University of Tokyo, Tokyo, 108-8639, Japan
| | - Satoko Hattori
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Aichi, 470-1192, Japan
| | - Masaya Usui
- Research Resources Division, RIKEN Center for Brain Science, Wako, Saitama, 351-0198, Japan
| | - Haruhiko Sago
- Center for Maternal-Fetal, Neonatal and Reproductive Medicine, National Center for Child Health and Development, Tokyo, 157-8535, Japan
| | - Nobuko Mataga
- Research Resources Division, RIKEN Center for Brain Science, Wako, Saitama, 351-0198, Japan
| | - Tsuyoshi Miyakawa
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Aichi, 470-1192, Japan
| | - Kazuhiro Yamakawa
- Laboratory for Neurogenetics, RIKEN Center for Brain Science, Wako, Saitama, 351-0198, Japan. .,Department of Neurodevelopmental Disorder Genetics, Institute of Brain Sciences, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, 467-8601, Japan.
| | - Toshiya Manabe
- Division of Neuronal Network, Institute of Medical Science, University of Tokyo, Tokyo, 108-8639, Japan.
| |
Collapse
|
28
|
Roper RJ, Goodlett CR, Martínez de Lagrán M, Dierssen M. Behavioral Phenotyping for Down Syndrome in Mice. ACTA ACUST UNITED AC 2020; 10:e79. [PMID: 32780566 DOI: 10.1002/cpmo.79] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Down syndrome (DS) is the most frequent genetic cause of intellectual disability, characterized by alterations in different behavioral symptom domains: neurodevelopment, motor behavior, and cognition. As mouse models have the potential to generate data regarding the neurological basis for the specific behavioral profile of DS, and may indicate pharmacological treatments with the potential to affect their behavioral phenotype, it is important to be able to assess disease-relevant behavioral traits in animal models in order to provide biological plausibility to the potential findings. The field is at a juncture that requires assessments that may effectively translate the findings acquired in mouse models to humans with DS. In this article, behavioral tests are described that are relevant to the domains affected in DS. A neurodevelopmental behavioral screen, the balance beam test, and the Multivariate Concentric Square Field test to assess multiple behavioral phenotypes and locomotion are described, discussing the ways to merge these findings to more fully understand cognitive strengths and weaknesses in this population. New directions for approaches to cognitive assessment in mice and humans are discussed. © 2020 Wiley Periodicals LLC. Basic Protocol 1: Preweaning neurodevelopmental battery Basic Protocol 2: Balance beam Basic Protocol 3: Multivariate concentric square field test (MCSF).
Collapse
Affiliation(s)
| | | | - María Martínez de Lagrán
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Mara Dierssen
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain.,Biomedical Research Networking Center on Rare Diseases (CIBERER), Institute of Health Carlos III, Madrid, Spain
| |
Collapse
|
29
|
Laurent AP, Kotecha RS, Malinge S. Gain of chromosome 21 in hematological malignancies: lessons from studying leukemia in children with Down syndrome. Leukemia 2020; 34:1984-1999. [PMID: 32433508 PMCID: PMC7387246 DOI: 10.1038/s41375-020-0854-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 04/22/2020] [Accepted: 04/28/2020] [Indexed: 12/31/2022]
Abstract
Structural and numerical alterations of chromosome 21 are extremely common in hematological malignancies. While the functional impact of chimeric transcripts from fused chromosome 21 genes such as TEL-AML1, AML1-ETO, or FUS-ERG have been extensively studied, the role of gain of chromosome 21 remains largely unknown. Gain of chromosome 21 is a frequently occurring aberration in several types of acute leukemia and can be found in up to 35% of cases. Children with Down syndrome (DS), who harbor constitutive trisomy 21, highlight the link between gain of chromosome 21 and leukemogenesis, with an increased risk of developing acute leukemia compared with other children. Clinical outcomes for DS-associated leukemia have improved over the years through the development of uniform treatment protocols facilitated by international cooperative groups. The genetic landscape has also recently been characterized, providing an insight into the molecular pathogenesis underlying DS-associated leukemia. These studies emphasize the key role of trisomy 21 in priming a developmental stage and cellular context susceptible to transformation, and have unveiled its cooperative function with additional genetic events that occur during leukemia progression. Here, using DS-leukemia as a paradigm, we aim to integrate our current understanding of the role of trisomy 21, of critical dosage-sensitive chromosome 21 genes, and of associated mechanisms underlying the development of hematological malignancies. This review will pave the way for future investigations on the broad impact of gain of chromosome 21 in hematological cancer, with a view to discovering new vulnerabilities and develop novel targeted therapies to improve long term outcomes for DS and non-DS patients.
Collapse
Affiliation(s)
- Anouchka P Laurent
- INSERM U1170, Gustave Roussy Institute, Université Paris Saclay, Villejuif, France
- Université Paris Diderot, Paris, France
| | - Rishi S Kotecha
- School of Pharmacy and Biomedical Sciences, Curtin University, Perth, Western Australia, Australia
- Department of Clinical Haematology, Oncology and Bone Marrow Transplantation, Perth Children's Hospital, Perth, Western Australia, Australia
- Telethon Kids Cancer Centre, Telethon Kids Institute, University of Western Australia, Perth, Western Australia, Australia
| | - Sébastien Malinge
- INSERM U1170, Gustave Roussy Institute, Université Paris Saclay, Villejuif, France.
- Telethon Kids Cancer Centre, Telethon Kids Institute, University of Western Australia, Perth, Western Australia, Australia.
| |
Collapse
|
30
|
Zedníková I, Chylíková B, Šeda O, Korabečná M, Pazourková E, Břešťák M, Krkavcová M, Calda P, Hořínek A. Genome-wide miRNA profiling in plasma of pregnant women with down syndrome fetuses. Mol Biol Rep 2020; 47:4531-4540. [PMID: 32472298 PMCID: PMC7295716 DOI: 10.1007/s11033-020-05545-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 05/23/2020] [Indexed: 12/14/2022]
Abstract
Down syndrome (DS) is one of the most common causes of intellectual disability and new approaches allowing its rapid and effective prenatal detection are being explored. In this study, we investigated the diagnostic potential of plasma microRNAs (miRNAs). This study builds upon our previous study in DS placentas, where seven miRNAs were found to be significantly up-regulated. A total of 70 first-trimester plasma samples from pregnant women were included in the present study (35 samples with DS fetuses; 35 with euploid fetuses). Genome-wide miRNA profiling was performed in the pilot study using Affymetrix GeneChip™ miRNA 4.1 Array Strips (18 samples). Selected miRNAs were then analysed in the validation study using quantitative reverse transcription PCR (RT-qPCR; 52 samples). Based on the current pilot study results (12 miRNAs), our previous research on chorionic villi samples (7 miRNAs) and the literature (4 miRNAs), a group of 23 miRNAs was selected for the validation study. Although the results of the pilot study were promising, the validation study using the more sensitive RT-qPCR technique and a larger group of samples revealed no significant differences in miRNA profiles between the compared groups. Our results suggest that testing of the first-trimester plasma miRNAs is probably not suitable for non-invasive prenatal testing (NIPT). Different results could be theoretically achieved at later gestational ages; however, such a result probably would have limited use in clinical practice.
Collapse
Affiliation(s)
- Iveta Zedníková
- Institute of Biology and Medical Genetics of the First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic.
| | - Blanka Chylíková
- Institute of Biology and Medical Genetics of the First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Ondřej Šeda
- Institute of Biology and Medical Genetics of the First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Marie Korabečná
- Institute of Biology and Medical Genetics of the First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Eva Pazourková
- Institute of Biology and Medical Genetics of the First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Miroslav Břešťák
- Department of Obstetrics and Gynecology of the First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
- Screening Center ProfiG2, Prague, Czech Republic
| | | | - Pavel Calda
- Department of Obstetrics and Gynecology of the First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Aleš Hořínek
- Institute of Biology and Medical Genetics of the First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
- 3rd Department of Medicine, Department of Endocrinology and Metabolism, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| |
Collapse
|
31
|
LaCombe JM, Roper RJ. Skeletal dynamics of Down syndrome: A developing perspective. Bone 2020; 133:115215. [PMID: 31887437 PMCID: PMC7044033 DOI: 10.1016/j.bone.2019.115215] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 12/05/2019] [Accepted: 12/24/2019] [Indexed: 01/14/2023]
Abstract
Individuals with Down syndrome (DS) display distinctive skeletal morphology compared to the general population, but disparate descriptions, methodologies, analyses, and populations sampled have led to diverging conclusions about this unique skeletal phenotype. As individuals with DS are living longer, they may be at a higher risk of aging disorders such as osteoporosis and increased fracture risk. Sexual dimorphism has been suggested between males and females with DS in which males, not females, experience an earlier decline in bone mineral density (BMD). Unfortunately, studies focusing on skeletal health related to Trisomy 21 (Ts21) are few in number and often too underpowered to answer questions about skeletal development, resultant osteoporosis, and sexual dimorphism, especially in stages of bone accrual. Further confounding the field are the varied methods of bone imaging, analysis, and data interpretation. This review takes a critical look at the current knowledge of DS skeletal phenotypes, both from human and mouse studies, and presents knowledge gaps that need to be addressed, differences in research methodologies and analyses that affect the interpretation of results, and proposes guidelines for overcoming obstacles to understand skeletal traits associated with DS. By examining our current knowledge of bone in individuals with Ts21, a trajectory for future studies may be established to provide meaningful solutions for understanding the development of and improving skeletal structures in individuals with and without DS.
Collapse
Affiliation(s)
- Jonathan M LaCombe
- Department of Biology, Indiana University-Purdue University Indianapolis, United States of America
| | - Randall J Roper
- Department of Biology, Indiana University-Purdue University Indianapolis, United States of America.
| |
Collapse
|
32
|
Laurent AP, Siret A, Ignacimouttou C, Panchal K, Diop M, Jenni S, Tsai YC, Roos-Weil D, Aid Z, Prade N, Lagarde S, Plassard D, Pierron G, Daudigeos E, Lecluse Y, Droin N, Bornhauser BC, Cheung LC, Crispino JD, Gaudry M, Bernard OA, Macintyre E, Barin Bonnigal C, Kotecha RS, Geoerger B, Ballerini P, Bourquin JP, Delabesse E, Mercher T, Malinge S. Constitutive Activation of RAS/MAPK Pathway Cooperates with Trisomy 21 and Is Therapeutically Exploitable in Down Syndrome B-cell Leukemia. Clin Cancer Res 2020; 26:3307-3318. [PMID: 32220889 DOI: 10.1158/1078-0432.ccr-19-3519] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 02/20/2020] [Accepted: 03/23/2020] [Indexed: 12/20/2022]
Abstract
PURPOSE Children with Down syndrome (constitutive trisomy 21) that develop acute lymphoblastic leukemia (DS-ALL) have a 3-fold increased likelihood of treatment-related mortality coupled with a higher cumulative incidence of relapse, compared with other children with B-cell acute lymphoblastic leukemia (B-ALL). This highlights the lack of suitable treatment for Down syndrome children with B-ALL. EXPERIMENTAL DESIGN To facilitate the translation of new therapeutic agents into clinical trials, we built the first preclinical cohort of patient-derived xenograft (PDX) models of DS-ALL, comprehensively characterized at the genetic and transcriptomic levels, and have proven its suitability for preclinical studies by assessing the efficacy of drug combination between the MEK inhibitor trametinib and conventional chemotherapy agents. RESULTS Whole-exome and RNA-sequencing experiments revealed a high incidence of somatic alterations leading to RAS/MAPK pathway activation in our cohort of DS-ALL, as well as in other pediatric B-ALL presenting somatic gain of the chromosome 21 (B-ALL+21). In murine and human B-cell precursors, activated KRASG12D functionally cooperates with trisomy 21 to deregulate transcriptional networks that promote increased proliferation and self renewal, as well as B-cell differentiation blockade. Moreover, we revealed that inhibition of RAS/MAPK pathway activation using the MEK1/2 inhibitor trametinib decreased leukemia burden in several PDX models of B-ALL+21, and enhanced survival of DS-ALL PDX in combination with conventional chemotherapy agents such as vincristine. CONCLUSIONS Altogether, using novel and suitable PDX models, this study indicates that RAS/MAPK pathway inhibition represents a promising strategy to improve the outcome of Down syndrome children with B-cell precursor leukemia.
Collapse
Affiliation(s)
- Anouchka P Laurent
- INSERM U1170, Gustave Roussy Institute, Université Paris Saclay, Villejuif, France.,Université Paris Diderot, Paris, France
| | - Aurélie Siret
- INSERM U1170, Gustave Roussy Institute, Université Paris Saclay, Villejuif, France
| | - Cathy Ignacimouttou
- INSERM U1170, Gustave Roussy Institute, Université Paris Saclay, Villejuif, France
| | - Kunjal Panchal
- Telethon Kids Cancer Centre, Telethon Kids Institute, University of Western Australia, Perth, Australia
| | - M'Boyba Diop
- Gustave Roussy Institute Cancer Campus, Department of Pediatric and Adolescent Oncology, INSERM U1015, Equipe Labellisée Ligue Nationale Contre le Cancer, Université Paris-Saclay, Villejuif, France
| | - Silvia Jenni
- Department of Pediatric Oncology, Children's Research Centre, University Children's Hospital Zurich, Zurich, Switzerland
| | - Yi-Chien Tsai
- Department of Pediatric Oncology, Children's Research Centre, University Children's Hospital Zurich, Zurich, Switzerland
| | - Damien Roos-Weil
- INSERM U1170, Gustave Roussy Institute, Université Paris Saclay, Villejuif, France
| | - Zakia Aid
- INSERM U1170, Gustave Roussy Institute, Université Paris Saclay, Villejuif, France
| | - Nais Prade
- Centre of Research on Cancer of Toulouse (CRCT), CHU Toulouse, Université Toulouse III, Toulouse, France
| | - Stephanie Lagarde
- Centre of Research on Cancer of Toulouse (CRCT), CHU Toulouse, Université Toulouse III, Toulouse, France
| | | | | | - Estelle Daudigeos
- Gustave Roussy Institute Cancer Campus, Department of Pediatric and Adolescent Oncology, INSERM U1015, Equipe Labellisée Ligue Nationale Contre le Cancer, Université Paris-Saclay, Villejuif, France
| | - Yann Lecluse
- Gustave Roussy Institute Cancer Campus, Department of Pediatric and Adolescent Oncology, INSERM U1015, Equipe Labellisée Ligue Nationale Contre le Cancer, Université Paris-Saclay, Villejuif, France
| | - Nathalie Droin
- INSERM U1170, Gustave Roussy Institute, Université Paris Saclay, Villejuif, France
| | - Beat C Bornhauser
- Department of Pediatric Oncology, Children's Research Centre, University Children's Hospital Zurich, Zurich, Switzerland
| | - Laurence C Cheung
- Telethon Kids Cancer Centre, Telethon Kids Institute, University of Western Australia, Perth, Australia.,School of Pharmacy and Biomedical Sciences, Curtin University, Bentley, Australia
| | - John D Crispino
- Division of Hematology/Oncology, Northwestern University, Chicago, Illinois
| | - Muriel Gaudry
- INSERM U1170, Gustave Roussy Institute, Université Paris Saclay, Villejuif, France
| | - Olivier A Bernard
- INSERM U1170, Gustave Roussy Institute, Université Paris Saclay, Villejuif, France
| | - Elizabeth Macintyre
- Hematology, Université de Paris, Institut Necker-Enfants Malades and Assistance Publique-Hopitaux de Paris, Paris, France
| | | | - Rishi S Kotecha
- Telethon Kids Cancer Centre, Telethon Kids Institute, University of Western Australia, Perth, Australia.,School of Pharmacy and Biomedical Sciences, Curtin University, Bentley, Australia.,Department of Clinical Haematology, Oncology and Bone Marrow Transplantation, Perth Children's Hospital, Perth, Australia
| | - Birgit Geoerger
- Gustave Roussy Institute Cancer Campus, Department of Pediatric and Adolescent Oncology, INSERM U1015, Equipe Labellisée Ligue Nationale Contre le Cancer, Université Paris-Saclay, Villejuif, France
| | - Paola Ballerini
- Laboratoire d'Hématologie, Hôpital Trousseau, APHP, Paris-Sorbonne, Paris, France
| | - Jean-Pierre Bourquin
- Department of Pediatric Oncology, Children's Research Centre, University Children's Hospital Zurich, Zurich, Switzerland
| | - Eric Delabesse
- Centre of Research on Cancer of Toulouse (CRCT), CHU Toulouse, Université Toulouse III, Toulouse, France
| | - Thomas Mercher
- INSERM U1170, Gustave Roussy Institute, Université Paris Saclay, Villejuif, France.,Equipe Labellisée Ligue Nationale Contre le Cancer, Paris, France
| | - Sebastien Malinge
- INSERM U1170, Gustave Roussy Institute, Université Paris Saclay, Villejuif, France. .,Telethon Kids Cancer Centre, Telethon Kids Institute, University of Western Australia, Perth, Australia
| |
Collapse
|
33
|
Chuluun B, Pittaras E, Hong H, Fisher N, Colas D, Ruby NF, Heller HC. Suprachiasmatic lesions restore object recognition in down syndrome model mice. Neurobiol Sleep Circadian Rhythms 2020; 8:100049. [PMID: 32195448 PMCID: PMC7075983 DOI: 10.1016/j.nbscr.2020.100049] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 02/04/2020] [Accepted: 02/12/2020] [Indexed: 01/03/2023] Open
Abstract
The Ts65Dn mouse is a well-studied model of trisomy 21, Down syndrome. This mouse strain has severe learning disability as measured by several rodent learning tests that depend on hippocampal spatial memory function. Hippocampal long-term potentiation (LTP) is deficient in these mice. Short-term daily treatment with low-dose GABA receptor antagonists rescue spatial learning and LTP in Ts65Dn mice leading to the hypothesis that the learning disability is due to GABAergic over-inhibition of hippocampal circuits. The fact that the GABA receptor antagonists were only effective if delivered during the daily light phase suggested that the source of the excess GABA was controlled directly or indirectly by the circadian system. The central circadian pacemaker of mammals is the suprachiasmatic nucleus (SCN), which is largely a GABAergic nucleus. In this study we investigated whether elimination of the SCN in Ts65Dn mice would restore their ability to form recognition memories as tested by the novel object recognition (NOR) task. Full, but not partial lesions of the SCN of Ts65Dn mice normalized their ability to perform on the NOR test. These results suggest that the circadian system modulates neuroplasticity over the time frame involved in the process of consolidation of recognition memories.
Collapse
|
34
|
Antonarakis SE, Skotko BG, Rafii MS, Strydom A, Pape SE, Bianchi DW, Sherman SL, Reeves RH. Down syndrome. Nat Rev Dis Primers 2020; 6:9. [PMID: 32029743 PMCID: PMC8428796 DOI: 10.1038/s41572-019-0143-7] [Citation(s) in RCA: 463] [Impact Index Per Article: 92.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/17/2019] [Indexed: 12/11/2022]
Abstract
Trisomy 21, the presence of a supernumerary chromosome 21, results in a collection of clinical features commonly known as Down syndrome (DS). DS is among the most genetically complex of the conditions that are compatible with human survival post-term, and the most frequent survivable autosomal aneuploidy. Mouse models of DS, involving trisomy of all or part of human chromosome 21 or orthologous mouse genomic regions, are providing valuable insights into the contribution of triplicated genes or groups of genes to the many clinical manifestations in DS. This endeavour is challenging, as there are >200 protein-coding genes on chromosome 21 and they can have direct and indirect effects on homeostasis in cells, tissues, organs and systems. Although this complexity poses formidable challenges to understanding the underlying molecular basis for each of the many clinical features of DS, it also provides opportunities for improving understanding of genetic mechanisms underlying the development and function of many cell types, tissues, organs and systems. Since the first description of trisomy 21, we have learned much about intellectual disability and genetic risk factors for congenital heart disease. The lower occurrence of solid tumours in individuals with DS supports the identification of chromosome 21 genes that protect against cancer when overexpressed. The universal occurrence of the histopathology of Alzheimer disease and the high prevalence of dementia in DS are providing insights into the pathology and treatment of Alzheimer disease. Clinical trials to ameliorate intellectual disability in DS signal a new era in which therapeutic interventions based on knowledge of the molecular pathophysiology of DS can now be explored; these efforts provide reasonable hope for the future.
Collapse
Affiliation(s)
- Stylianos E Antonarakis
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland.
| | - Brian G Skotko
- Down Syndrome Program, Division of Medical Genetics, Department of Pediatrics, Massachusetts General Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Michael S Rafii
- Keck School of Medicine of University of Southern California, California, CA, USA
| | - Andre Strydom
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Sarah E Pape
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Diana W Bianchi
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Stephanie L Sherman
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Roger H Reeves
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
35
|
Cannavo C, Tosh J, Fisher EMC, Wiseman FK. Using mouse models to understand Alzheimer's disease mechanisms in the context of trisomy of chromosome 21. PROGRESS IN BRAIN RESEARCH 2019; 251:181-208. [PMID: 32057307 DOI: 10.1016/bs.pbr.2019.10.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
People who have Down syndrome are at significantly elevated risk of developing early onset Alzheimer's disease that causes dementia (AD-DS). Here we review recent progress in modeling the development of AD-DS in mouse models. These studies provide insight into mechanisms underlying Alzheimer's disease and generate new clinical research questions. In addition, they suggest potential new targets for disease prevention therapies.
Collapse
Affiliation(s)
- Claudia Cannavo
- Department of Neuromuscular Diseases, UCL Institute of Neurology, London, United Kingdom; UK Dementia Research Institute at University College, London, United Kingdom
| | - Justin Tosh
- Department of Neuromuscular Diseases, UCL Institute of Neurology, London, United Kingdom
| | - Elizabeth M C Fisher
- Department of Neuromuscular Diseases, UCL Institute of Neurology, London, United Kingdom; The London Down Syndrome Consortium (LonDownS), London, United Kingdom
| | - Frances K Wiseman
- Department of Neuromuscular Diseases, UCL Institute of Neurology, London, United Kingdom; The London Down Syndrome Consortium (LonDownS), London, United Kingdom; UK Dementia Research Institute at University College, London, United Kingdom.
| |
Collapse
|
36
|
Roper RJ, Hawley L, Goodlett CR. Influence of allelic differences in Down syndrome. PROGRESS IN BRAIN RESEARCH 2019; 251:29-54. [PMID: 32057311 PMCID: PMC7500172 DOI: 10.1016/bs.pbr.2019.09.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Both trisomic and non-trisomic genes may affect the incidence and severity of phenotypes associated with Down syndrome (DS). The importance of extra (trisomic) genetic material is emphasized in DS, with less emphasis to the allelic composition of candidate trisomic genes in defining the trisomic gene-phenotype relationship in DS. Allelic differences in non-trisomic genes have been shown to be important moderators of cardiac, leukemia, and developmental phenotypes associated with DS. Trisomic mouse models provide an in vivo genetic platform for examining the gene-phenotype relationship, including the influence of allelic variants, on DS-like phenotypes. DS mouse models have differing trisomic genetic makeup, and optimal development, viability and translational value of these mouse models may require a non-inbred genetic background with heterogeneity at many loci. Additionally, understanding the contribution of specific genes or regions to DS phenotypes often requires the utilization of genetically manipulated mice that may be established on a different inbred background than the trisomic mice. The impact of allelic differences of trisomic and background genes in human and model systems may offer insight into the variability in occurrence and severity of trisomic phenotypes.
Collapse
Affiliation(s)
- Randall J Roper
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, United States.
| | - Laura Hawley
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, United States
| | - Charles R Goodlett
- Department of Psychology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, United States
| |
Collapse
|
37
|
Muñiz Moreno MDM, Brault V, Birling MC, Pavlovic G, Herault Y. Modeling Down syndrome in animals from the early stage to the 4.0 models and next. PROGRESS IN BRAIN RESEARCH 2019; 251:91-143. [PMID: 32057313 DOI: 10.1016/bs.pbr.2019.08.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The genotype-phenotype relationship and the physiopathology of Down Syndrome (DS) have been explored in the last 20 years with more and more relevant mouse models. From the early age of transgenesis to the new CRISPR/CAS9-derived chromosomal engineering and the transchromosomic technologies, mouse models have been key to identify homologous genes or entire regions homologous to the human chromosome 21 that are necessary or sufficient to induce DS features, to investigate the complexity of the genetic interactions that are involved in DS and to explore therapeutic strategies. In this review we report the new developments made, how genomic data and new genetic tools have deeply changed our way of making models, extended our panel of animal models, and increased our understanding of the neurobiology of the disease. But even if we have made an incredible progress which promises to make DS a curable condition, we are facing new research challenges to nurture our knowledge of DS pathophysiology as a neurodevelopmental disorder with many comorbidities during ageing.
Collapse
Affiliation(s)
- Maria Del Mar Muñiz Moreno
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
| | - Véronique Brault
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
| | - Marie-Christine Birling
- Université de Strasbourg, CNRS, INSERM, PHENOMIN Institut Clinique de la Souris, Illkirch, France
| | - Guillaume Pavlovic
- Université de Strasbourg, CNRS, INSERM, PHENOMIN Institut Clinique de la Souris, Illkirch, France
| | - Yann Herault
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France; Université de Strasbourg, CNRS, INSERM, PHENOMIN Institut Clinique de la Souris, Illkirch, France.
| |
Collapse
|
38
|
Baburamani AA, Patkee PA, Arichi T, Rutherford MA. New approaches to studying early brain development in Down syndrome. Dev Med Child Neurol 2019; 61:867-879. [PMID: 31102269 PMCID: PMC6618001 DOI: 10.1111/dmcn.14260] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/28/2019] [Indexed: 12/19/2022]
Abstract
Down syndrome is the most common genetic developmental disorder in humans and is caused by partial or complete triplication of human chromosome 21 (trisomy 21). It is a complex condition which results in multiple lifelong health problems, including varying degrees of intellectual disability and delays in speech, memory, and learning. As both length and quality of life are improving for individuals with Down syndrome, attention is now being directed to understanding and potentially treating the associated cognitive difficulties and their underlying biological substrates. These have included imaging and postmortem studies which have identified decreased regional brain volumes and histological anomalies that accompany early onset dementia. In addition, advances in genome-wide analysis and Down syndrome mouse models are providing valuable insight into potential targets for intervention that could improve neurogenesis and long-term cognition. As little is known about early brain development in human Down syndrome, we review recent advances in magnetic resonance imaging that allow non-invasive visualization of brain macro- and microstructure, even in utero. It is hoped that together these advances may enable Down syndrome to become one of the first genetic disorders to be targeted by antenatal treatments designed to 'normalize' brain development. WHAT THIS PAPER ADDS: Magnetic resonance imaging can provide non-invasive characterization of early brain development in Down syndrome. Down syndrome mouse models enable study of underlying pathology and potential intervention strategies. Potential therapies could modify brain structure and improve early cognitive levels. Down syndrome may be the first genetic disorder to have targeted therapies which alter antenatal brain development.
Collapse
Affiliation(s)
- Ana A Baburamani
- Centre for the Developing BrainDepartment of Perinatal Imaging and HealthSchool of Biomedical Engineering & Imaging SciencesKing's College LondonKing's Health PartnersSt Thomas’ HospitalLondonUK
| | - Prachi A Patkee
- Centre for the Developing BrainDepartment of Perinatal Imaging and HealthSchool of Biomedical Engineering & Imaging SciencesKing's College LondonKing's Health PartnersSt Thomas’ HospitalLondonUK
| | - Tomoki Arichi
- Centre for the Developing BrainDepartment of Perinatal Imaging and HealthSchool of Biomedical Engineering & Imaging SciencesKing's College LondonKing's Health PartnersSt Thomas’ HospitalLondonUK,Department of BioengineeringImperial College LondonLondonUK,Children's NeurosciencesEvelina London Children's HospitalLondonUK
| | - Mary A Rutherford
- Centre for the Developing BrainDepartment of Perinatal Imaging and HealthSchool of Biomedical Engineering & Imaging SciencesKing's College LondonKing's Health PartnersSt Thomas’ HospitalLondonUK
| |
Collapse
|
39
|
Ishihara K, Shimizu R, Takata K, Kawashita E, Amano K, Shimohata A, Low D, Nabe T, Sago H, Alexander WS, Ginhoux F, Yamakawa K, Akiba S. Perturbation of the immune cells and prenatal neurogenesis by the triplication of the Erg gene in mouse models of Down syndrome. Brain Pathol 2019; 30:75-91. [PMID: 31206867 DOI: 10.1111/bpa.12758] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 06/05/2019] [Indexed: 12/15/2022] Open
Abstract
Some mouse models of Down syndrome (DS), including Ts1Cje mice, exhibit impaired prenatal neurogenesis with yet unknown molecular mechanism. To gain insights into the impaired neurogenesis, a transcriptomic and flow cytometry analysis of E14.5 Ts1Cje embryo brain was performed. Our analysis revealed that the neutrophil and monocyte ratios in the CD45-positive hematopoietic cells were relatively increased, in agreement with the altered expression of inflammation/immune-related genes, in Ts1Cje embryonic brain, whereas the relative number of brain macrophages was decreased in comparison to wild-type mice. Similar upregulation of inflammation-associated mRNAs was observed in other DS mouse models, with variable trisomic region lengths. We used genetic manipulation to assess the contribution of Erg, a trisomic gene in these DS models, known to regulation hemato-immune cells. The perturbed proportions of immune cells in Ts1Cje mouse brain were restored in Ts1Cje-Erg+/+/Mld2 mice, which are disomic for functional Erg but otherwise trisomic on a Ts1Cje background. Moreover, the embryonic neurogenesis defects observed in Ts1Cje cortex were reduced in Ts1Cje-Erg+/+/Mld2 embryos. Our findings suggest that Erg gene triplication contributes to the dysregulation of the homeostatic proportion of the populations of immune cells in the embryonic brain and decreased prenatal cortical neurogenesis in the prenatal brain with DS.
Collapse
Affiliation(s)
- Keiichi Ishihara
- Department of Pathological Biochemistry, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Ryohei Shimizu
- Department of Pathological Biochemistry, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Kazuyuki Takata
- Department of Clinical and Translational Physiology, Division of Biological Sciences, Kyoto Pharmaceutical University, Kyoto, Japan.,Division of Integrated Pharmaceutical Sciences, Kyoto Pharmaceutical University, Kyoto, Japan.,Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Eri Kawashita
- Department of Pathological Biochemistry, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Kenji Amano
- Laboratory for Neurogenetics, RIKEN Center for Brain Science, Saitama, Japan
| | - Atsushi Shimohata
- Laboratory for Neurogenetics, RIKEN Center for Brain Science, Saitama, Japan
| | - Donovan Low
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Takeshi Nabe
- Laboratory of Immunopharmacology, Faculty of Pharmaceutical Sciences, Setsunan University, Osaka, Japan
| | - Haruhiko Sago
- Center for Maternal-Fetal, Neonatal and Reproductive Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Warren S Alexander
- Cancer and Haematology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.,Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kazuhiro Yamakawa
- Laboratory for Neurogenetics, RIKEN Center for Brain Science, Saitama, Japan
| | - Satoshi Akiba
- Department of Pathological Biochemistry, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto, Japan
| |
Collapse
|
40
|
Ahlfors H, Anyanwu N, Pakanavicius E, Dinischiotu N, Lana-Elola E, Watson-Scales S, Tosh J, Wiseman F, Briscoe J, Page K, Fisher EMC, Tybulewicz VLJ. Gene expression dysregulation domains are not a specific feature of Down syndrome. Nat Commun 2019; 10:2489. [PMID: 31171815 PMCID: PMC6554309 DOI: 10.1038/s41467-019-10129-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 04/18/2019] [Indexed: 11/19/2022] Open
Abstract
Down syndrome (DS), trisomy of human chromosome 21 (Hsa21), results in a broad range of phenotypes. A recent study reported that DS cells show genome-wide transcriptional changes in which up- or down-regulated genes are clustered in gene expression dysregulation domains (GEDDs). GEDDs were also reported in fibroblasts derived from a DS mouse model duplicated for some Hsa21-orthologous genes, indicating cross-species conservation of this phenomenon. Here we investigate GEDDs using the Dp1Tyb mouse model of DS, which is duplicated for the entire Hsa21-orthologous region of mouse chromosome 16. Our statistical analysis shows that GEDDs are present both in DS cells and in Dp1Tyb mouse fibroblasts and hippocampus. However, we find that GEDDs do not depend on the DS genotype but occur whenever gene expression changes. We conclude that GEDDs are not a specific feature of DS but instead result from the clustering of co-regulated genes, a function of mammalian genome organisation. Gene expression dysregulation domains (GEDDs) have been reported in Down syndrome (DS) cells, where changes in gene expression are clustered. Here the authors find that, while GEDDs are present in DS cells and in the Dp1Tyb mouse model of DS, GEDDs do not depend on the DS genotype and occur whenever gene expression changes, suggesting they result from the clustering of co-regulated genes as a function of mammalian genome organisation.
Collapse
Affiliation(s)
- Helena Ahlfors
- NE Thames Regional Genetics Laboratory, GOSH NHS Foundation Trust, London, WC1N 3JH, UK
| | | | | | | | | | | | - Justin Tosh
- UCL Institute of Neurology, London, WC1N 3BG, UK
| | | | | | - Karen Page
- Department of Mathematics, University College London, London, WC1E 6BT, UK
| | | | - Victor L J Tybulewicz
- The Francis Crick Institute, London, NW1 1AT, UK. .,Imperial College, London, W12 0NN, UK.
| |
Collapse
|
41
|
Partial trisomy 21 contributes to T-cell malignancies induced by JAK3-activating mutations in murine models. Blood Adv 2019; 2:1616-1627. [PMID: 29986854 DOI: 10.1182/bloodadvances.2018016089] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 05/17/2018] [Indexed: 02/05/2023] Open
Abstract
JAK3-activating mutations are commonly seen in chronic or acute hematologic malignancies affecting the myeloid, megakaryocytic, lymphoid, and natural killer (NK) cell compartment. Overexpression models of mutant JAK3 or pharmacologic inhibition of its kinase activity have highlighted the role that these constitutively activated mutants play in the T-cell, NK cell, and megakaryocytic lineages, but to date, the functional impact of JAK3 mutations at an endogenous level remains unknown. Here, we report a JAK3A572V knockin mouse model and demonstrate that activated JAK3 leads to a progressive and dose-dependent expansion of CD8+ T cells in the periphery before colonization of the bone marrow. This phenotype is dependent on the γc chain of cytokine receptors and presents several features of the human leukemic form of cutaneous T-cell lymphoma (L-CTCL), including skin involvements. We also showed that the JAK3A572V-positive malignant cells are transplantable and phenotypically heterogeneous in bone marrow transplantation assays. Interestingly, we revealed that activated JAK3 functionally cooperates with partial trisomy 21 in vivo to enhance the L-CTCL phenotype, ultimately leading to a lethal and fully penetrant disorder. Finally, we assessed the efficacy of JAK3 inhibition and showed that CTCL JAK3A572V-positive T cells are sensitive to tofacitinib, which provides additional preclinical insights into the use of JAK3 inhibitors in these disorders. Altogether, this JAK3A572V knockin model is a relevant new tool for testing the efficacy of JAK inhibitors in JAK3-related hematopoietic malignancies.
Collapse
|
42
|
Xie X, Pang L, Yao A, Ye S, Wang D. Nanocement Produced from Borosilicate Bioactive Glass Nanoparticles Composited with Alginate. Aust J Chem 2019. [DOI: 10.1071/ch18410] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
A novel injectable bone cement was prepared using sol–gel derived borosilicate bioactive glass nanoparticles as a solid phase and sodium alginate solution as a liquid phase. The gelation reaction of the alginate was modulated by Ca2+ ions released from the borosilicate glass phase, which in turn greatly depended on the boron content of the borosilicate glass phase. Such a gelation reaction not only significantly enhanced the anti-washout property of the bone cements, but also allowed control of the setting, handling properties, and compressive strength of the composite bone cements. Consequently, bone cements with controllable performances can be developed by simply adjusting the B2O3/SiO2 ratio of the borosilicate glass phase. Borosilicate bioactive glass with 20–30 mol-% borate contents exhibit a short setting time, good compressive strength, injectability, and anti-washout properties. With controllable performances and excellent bioactivity, the borosilicate bioactive glass/sodium alginate (BSBG/SA) composite bone cements are highly attractive for bone filling and regeneration applications.
Collapse
|
43
|
Herault Y, Delabar JM, Fisher EMC, Tybulewicz VLJ, Yu E, Brault V. Rodent models in Down syndrome research: impact and future opportunities. Dis Model Mech 2018; 10:1165-1186. [PMID: 28993310 PMCID: PMC5665454 DOI: 10.1242/dmm.029728] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Down syndrome is caused by trisomy of chromosome 21. To date, a multiplicity of mouse models with Down-syndrome-related features has been developed to understand this complex human chromosomal disorder. These mouse models have been important for determining genotype-phenotype relationships and identification of dosage-sensitive genes involved in the pathophysiology of the condition, and in exploring the impact of the additional chromosome on the whole genome. Mouse models of Down syndrome have also been used to test therapeutic strategies. Here, we provide an overview of research in the last 15 years dedicated to the development and application of rodent models for Down syndrome. We also speculate on possible and probable future directions of research in this fast-moving field. As our understanding of the syndrome improves and genome engineering technologies evolve, it is necessary to coordinate efforts to make all Down syndrome models available to the community, to test therapeutics in models that replicate the whole trisomy and design new animal models to promote further discovery of potential therapeutic targets. Summary: Mouse models have boosted therapeutic options for Down syndrome, and improved models are being developed to better understand the pathophysiology of this genetic condition.
Collapse
Affiliation(s)
- Yann Herault
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, 1 rue Laurent Fries, 67404 Illkirch, France .,Centre National de la Recherche Scientifique, UMR7104, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France.,Université de Strasbourg, 67404 Illkirch, France.,T21 Research Society, Brain and Spine Institute (ICM), 75013 Paris
| | - Jean M Delabar
- T21 Research Society, Brain and Spine Institute (ICM), 75013 Paris.,Université Paris Diderot, Sorbonne Paris Cité, Unité de Biologie Fonctionnelle et Adaptative, UMR8251, CNRS, 75205 Paris, France.,INSERM U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et la Moelle épinière, ICM, 75013 Paris, France.,Brain and Spine Institute (ICM) CNRS UMR7225, INSERM UMRS 975, 75013 Paris, France
| | - Elizabeth M C Fisher
- T21 Research Society, Brain and Spine Institute (ICM), 75013 Paris.,Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, WC1N 3BG, UK.,LonDownS Consortium, London, W1T 7NF UK
| | - Victor L J Tybulewicz
- T21 Research Society, Brain and Spine Institute (ICM), 75013 Paris.,LonDownS Consortium, London, W1T 7NF UK.,The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK.,Department of Medicine, Imperial College, London, SW7 2AZ, UK
| | - Eugene Yu
- T21 Research Society, Brain and Spine Institute (ICM), 75013 Paris.,The Children's Guild Foundation Down Syndrome Research Program, Department of Cancer Genetics and Genetics Program, Roswell Park Cancer Institute, Buffalo, NY 14263, USA.,Department of Cellular and Molecular Biology, Roswell Park Division of Graduate School, Genetics, Genomics and Bioinformatics Program, State University of New York at Buffalo, Buffalo, NY 14263, USA
| | - Veronique Brault
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, 1 rue Laurent Fries, 67404 Illkirch, France.,Centre National de la Recherche Scientifique, UMR7104, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France.,Université de Strasbourg, 67404 Illkirch, France
| |
Collapse
|
44
|
Cipriani G, Danti S, Carlesi C, Di Fiorino M. Aging With Down Syndrome: The Dual Diagnosis: Alzheimer's Disease and Down Syndrome. Am J Alzheimers Dis Other Demen 2018; 33:253-262. [PMID: 29504408 PMCID: PMC10852513 DOI: 10.1177/1533317518761093] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND People with Down syndrome (DS) enjoy a longer life expectancy now than they ever have before and are therefore at greater risk of developing conditions associated with aging, including dementia. OBJECTIVES To explore the phenomenon of dementia in DS. METHODS Medline and Google Scholar searches were conducted for relevant articles, chapters, and books published until 2017. Search terms included Alzheimer's disease, cognitive impairment, dementia, DS, and trisomy 21. Publications found through this indexed search were reviewed for further references. RESULTS AND CONCLUSIONS Virtually, all subject aged 35 to 40 show key neuropathologic changes characteristic of Alzheimer's disease, but only a part of them show clinical signs of dementia, usually around the age of 50 years. Early signs of dementia in people with DS may be different from those experienced by the general population. Failure to recognize this can delay diagnosis and subsequent interventions.
Collapse
Affiliation(s)
- Gabriele Cipriani
- Neurology Unit, Hospital of Versilia, Lido di Camaiore, Lucca (LU), Italy
- Psychiatry Unit, Hospital of Versilia, Lido di Camaiore, Lucca (Lu), Italy
| | - Sabrina Danti
- Clinical and Health Psychology Unit, Hospital of Pontedera, Pontedera (PI), Italy
| | - Cecilia Carlesi
- Neurology Unit, Hospital of Versilia, Lido di Camaiore, Lucca (LU), Italy
| | - Mario Di Fiorino
- Psychiatry Unit, Hospital of Versilia, Lido di Camaiore, Lucca (Lu), Italy
| |
Collapse
|
45
|
Watson-Scales S, Kalmar B, Lana-Elola E, Gibbins D, La Russa F, Wiseman F, Williamson M, Saccon R, Slender A, Olerinyova A, Mahmood R, Nye E, Cater H, Wells S, Yu YE, Bennett DLH, Greensmith L, Fisher EMC, Tybulewicz VLJ. Analysis of motor dysfunction in Down Syndrome reveals motor neuron degeneration. PLoS Genet 2018; 14:e1007383. [PMID: 29746474 PMCID: PMC5963810 DOI: 10.1371/journal.pgen.1007383] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 05/22/2018] [Accepted: 04/27/2018] [Indexed: 11/23/2022] Open
Abstract
Down Syndrome (DS) is caused by trisomy of chromosome 21 (Hsa21) and results in a spectrum of phenotypes including learning and memory deficits, and motor dysfunction. It has been hypothesized that an additional copy of a few Hsa21 dosage-sensitive genes causes these phenotypes, but this has been challenged by observations that aneuploidy can cause phenotypes by the mass action of large numbers of genes, with undetectable contributions from individual sequences. The motor abnormalities in DS are relatively understudied-the identity of causative dosage-sensitive genes and the mechanism underpinning the phenotypes are unknown. Using a panel of mouse strains with duplications of regions of mouse chromosomes orthologous to Hsa21 we show that increased dosage of small numbers of genes causes locomotor dysfunction and, moreover, that the Dyrk1a gene is required in three copies to cause the phenotype. Furthermore, we show for the first time a new DS phenotype: loss of motor neurons both in mouse models and, importantly, in humans with DS, that may contribute to locomotor dysfunction.
Collapse
Affiliation(s)
| | | | | | | | - Federica La Russa
- Wolfson Centre for Age-Related Diseases, Kings College London, London, United Kingdom
| | | | | | | | - Amy Slender
- The Francis Crick Institute, London, United Kingdom
| | | | | | - Emma Nye
- The Francis Crick Institute, London, United Kingdom
| | - Heather Cater
- MRC Harwell Institute, Harwell Campus, Oxfordshire, United Kingdom
| | - Sara Wells
- MRC Harwell Institute, Harwell Campus, Oxfordshire, United Kingdom
| | - Y. Eugene Yu
- The Children’s Guild Foundation Down Syndrome Research Program, Genetics Program and Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, NY, United States of America
| | - David L. H. Bennett
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | | | | | | |
Collapse
|
46
|
Abstract
Background Monosomy of the X chromosome is the most frequent genetic abnormality in human as it is present in approximately 2% of all conceptions, although 99% of these embryos are spontaneously miscarried. In postnatal life, clinical features of Turner syndrome may include typical dysmorphic stigmata, short stature, sexual infantilism, and renal, cardiac, skeletal, endocrine and metabolic abnormalities. Main text Turner syndrome is due to a partial or total loss of the second sexual chromosome, resulting in the development of highly variable clinical features. This phenotype may not merely be due to genomic imbalance from deleted genes but may also result from additive influences on associated genes within a given gene network, with an altered regulation of gene expression triggered by the absence of the second sex chromosome. Current studies in human and mouse models have demonstrated that this chromosomal abnormality leads to epigenetic changes, including differential DNA methylation in specific groups of downstream target genes in pathways associated with several clinical and metabolic features, mostly on autosomal chromosomes. In this article, we begin exploring the potential involvement of both genetic and epigenetic factors in the origin of X chromosome monosomy. We review the dispute between the meiotic and post-zygotic origins of 45,X monosomy, by mainly analyzing the findings from several studies that compare gene expression of the 45,X monosomy to their euploid and/or 47,XXX trisomic cell counterparts on peripheral blood mononuclear cells, amniotic fluid, human fibroblast cells, and induced pluripotent human cell lines. From these studies, a profile of epigenetic changes seems to emerge in response to chromosomal imbalance. An interesting finding of all these studies is that methylation-based and expression-based pathway analyses are complementary, rather than overlapping, and are correlated with the clinical picture displayed by TS subjects. Conclusions The clarification of these possible causal pathways may have future implications in increasing the life expectancy of these patients and may provide informative targets for early pharmaceutical intervention.
Collapse
Affiliation(s)
- Francisco Álvarez-Nava
- Biological Sciences School, Faculty of Biological Sciences, Central University of Ecuador, Quito, Ecuador
| | - Roberto Lanes
- Pediatric Endocrine Unit, Hospital de Clínicas Caracas, Caracas, Venezuela
| |
Collapse
|
47
|
Richtsmeier JT. A century of development. AMERICAN JOURNAL OF PHYSICAL ANTHROPOLOGY 2018; 165:726-740. [PMID: 29574839 PMCID: PMC6007869 DOI: 10.1002/ajpa.23379] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 12/02/2017] [Accepted: 12/09/2017] [Indexed: 12/13/2022]
Affiliation(s)
- Joan T Richtsmeier
- Department of Anthropology, Pennsylvania State University, University Park, Pennsylvania 16802
| |
Collapse
|
48
|
Systematic Functional Characterization of Human 21st Chromosome Orthologs in Caenorhabditis elegans. G3-GENES GENOMES GENETICS 2018; 8:967-979. [PMID: 29367452 PMCID: PMC5844316 DOI: 10.1534/g3.118.200019] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Individuals with Down syndrome have neurological and muscle impairments due to an additional copy of the human 21st chromosome (HSA21). Only a few of ∼200 HSA21 genes encoding proteins have been linked to specific Down syndrome phenotypes, while the remainder are understudied. To identify poorly characterized HSA21 genes required for nervous system function, we studied behavioral phenotypes caused by loss-of-function mutations in conserved HSA21 orthologs in the nematode Caenorhabditis elegans. We identified 10 HSA21 orthologs that are required for neuromuscular behaviors: cle-1 (COL18A1), cysl-2 (CBS), dnsn-1 (DONSON), eva-1 (EVA1C), mtq-2 (N6ATM1), ncam-1 (NCAM2), pad-2 (POFUT2), pdxk-1 (PDXK), rnt-1 (RUNX1), and unc-26 (SYNJ1). We also found that three of these genes are required for normal release of the neurotransmitter acetylcholine. This includes a known synaptic gene unc-26 (SYNJ1), as well as uncharacterized genes pdxk-1 (PDXK) and mtq-2 (N6ATM1). As the first systematic functional analysis of HSA21 orthologs, this study may serve as a platform to understand genes that underlie phenotypes associated with Down syndrome.
Collapse
|
49
|
Stringer M, Goodlett CR, Roper RJ. Targeting trisomic treatments: optimizing Dyrk1a inhibition to improve Down syndrome deficits. Mol Genet Genomic Med 2017; 5:451-465. [PMID: 28944229 PMCID: PMC5606891 DOI: 10.1002/mgg3.334] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 08/21/2017] [Accepted: 08/24/2017] [Indexed: 12/11/2022] Open
Abstract
Overexpression of Dual-specificity tyrosine-phosphorylated regulated kinase 1A (DYRK1A), located on human chromosome 21, may alter molecular processes linked to developmental deficits in Down syndrome (DS). Trisomic DYRK1A is a rational therapeutic target, and although reductions in Dyrk1a genetic dosage have shown improvements in trisomic mouse models, attempts to reduce Dyrk1a activity by pharmacological mechanisms and correct these DS-associated phenotypes have been largely unsuccessful. Epigallocatechin-3-gallate (EGCG) inhibits DYRK1A activity in vitro and this action has been postulated to account for improvement of some DS-associated phenotypes that have been reported in preclinical studies and clinical trials. However, the beneficial effects of EGCG are inconsistent and there is no direct evidence that any observed improvement actually occurs through Dyrk1a inhibition. Inconclusive outcomes likely reflect a lack of knowledge about the tissue-specific patterns of spatial and temporal overexpression and elevated activity of Dyrk1a that may contribute to emerging DS traits during development. Emerging evidence indicates that Dyrk1a expression varies over the life span in DS mouse models, yet preclinical therapeutic treatments targeting Dyrk1a have largely not considered these developmental changes. Therapies intended to improve DS phenotypes through normalizing trisomic Dyrk1a need to optimize the timing and dose of treatment to match the spatiotemporal patterning of excessive Dyrk1a activity in relevant tissues. This will require more precise identification of developmental periods of vulnerability to enduring adverse effects of elevated Dyrk1a, representing the concurrence of increased Dyrk1a expression together with hypothesized tissue-specific-sensitive periods when Dyrk1a regulates cellular processes that shape the long-term functional properties of the tissue. Future efforts targeting inhibition of trisomic Dyrk1a should identify these putative spatiotemporally specific developmental sensitive periods and determine whether normalizing Dyrk1a activity then can lead to improved outcomes in DS phenotypes.
Collapse
Affiliation(s)
- Megan Stringer
- Department of PsychologyIUPUI402 North Blackford Street, LD 124IndianapolisIndiana46202-3275
| | - Charles R Goodlett
- Department of PsychologyIUPUI402 North Blackford Street, LD 124IndianapolisIndiana46202-3275
| | - Randall J Roper
- Department of BiologyIUPUI723 West Michigan Street SL 306IndianapolisIndiana46202-3275
| |
Collapse
|
50
|
Stringer M, Abeysekera I, Thomas J, LaCombe J, Stancombe K, Stewart RJ, Dria KJ, Wallace JM, Goodlett CR, Roper RJ. Epigallocatechin-3-gallate (EGCG) consumption in the Ts65Dn model of Down syndrome fails to improve behavioral deficits and is detrimental to skeletal phenotypes. Physiol Behav 2017; 177:230-241. [PMID: 28478033 PMCID: PMC5525541 DOI: 10.1016/j.physbeh.2017.05.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 04/15/2017] [Accepted: 05/01/2017] [Indexed: 12/17/2022]
Abstract
Down syndrome (DS) is caused by three copies of human chromosome 21 (Hsa21) and results in phenotypes including intellectual disability and skeletal deficits. Ts65Dn mice have three copies of ~50% of the genes homologous to Hsa21 and display phenotypes associated with DS, including cognitive deficits and skeletal abnormalities. DYRK1A is found in three copies in humans with Trisomy 21 and in Ts65Dn mice, and is involved in a number of critical pathways including neurological development and osteoclastogenesis. Epigallocatechin-3-gallate (EGCG), the main polyphenol in green tea, inhibits Dyrk1a activity. We have previously shown that EGCG treatment (~10mg/kg/day) improves skeletal abnormalities in Ts65Dn mice, yet the same dose, as well as ~20mg/kg/day did not rescue deficits in the Morris water maze spatial learning task (MWM), novel object recognition (NOR) or balance beam task (BB). In contrast, a recent study reported that an EGCG-containing supplement with a dose of 2-3mg per day (~40-60mg/kg/day) improved hippocampal-dependent task deficits in Ts65Dn mice. The current study investigated if an EGCG dosage similar to that study would yield similar improvements in either cognitive or skeletal deficits. Ts65Dn mice and euploid littermates were given EGCG [0.4mg/mL] or a water control, with treatments yielding average daily intakes of ~50mg/kg/day EGCG, and tested on the multivariate concentric square field (MCSF)-which assesses activity, exploratory behavior, risk assessment, risk taking, and shelter seeking-and NOR, BB, and MWM. EGCG treatment failed to improve cognitive deficits; EGCG also produced several detrimental effects on skeleton in both genotypes. In a refined HPLC-based assay, its first application in Ts65Dn mice, EGCG treatment significantly reduced kinase activity in femora but not in the cerebral cortex, cerebellum, or hippocampus. Counter to expectation, 9-week-old Ts65Dn mice exhibited a decrease in Dyrk1a protein levels in Western blot analysis in the cerebellum. The lack of beneficial therapeutic behavioral effects and potentially detrimental skeletal effects of EGCG found in Ts65Dn mice emphasize the importance of identifying dosages of EGCG that reliably improve DS phenotypes and linking those effects to actions of EGCG (or EGCG-containing supplements) in specific targets in brain and bone.
Collapse
Affiliation(s)
- Megan Stringer
- IUPUI, Department of Psychology, 402 North Blackford Street, LD 124, Indianapolis, IN 46202-3275, United States
| | - Irushi Abeysekera
- IUPUI, Department of Biology, 723 West Michigan Street, SL 306, Indianapolis, IN 46202-3275, United States
| | - Jared Thomas
- IUPUI, Department of Biology, 723 West Michigan Street, SL 306, Indianapolis, IN 46202-3275, United States
| | - Jonathan LaCombe
- IUPUI, Department of Biology, 723 West Michigan Street, SL 306, Indianapolis, IN 46202-3275, United States
| | - Kailey Stancombe
- IUPUI, Department of Psychology, 402 North Blackford Street, LD 124, Indianapolis, IN 46202-3275, United States
| | - Robert J Stewart
- IUPUI, Department of Psychology, 402 North Blackford Street, LD 124, Indianapolis, IN 46202-3275, United States
| | - Karl J Dria
- IUPUI, Department of Chemistry and Chemical Biology, 402 North Blackford Street, LD 326, Indianapolis, IN 46202-3275, United States
| | - Joseph M Wallace
- IUPUI, Department of Biomedical Engineering, 723 West Michigan Street, SL 220B, Indianapolis, IN 46202-3275, United States
| | - Charles R Goodlett
- IUPUI, Department of Psychology, 402 North Blackford Street, LD 124, Indianapolis, IN 46202-3275, United States
| | - Randall J Roper
- IUPUI, Department of Biology, 723 West Michigan Street, SL 306, Indianapolis, IN 46202-3275, United States.
| |
Collapse
|