1
|
Emili M, Stagni F, Russo C, Angelozzi L, Guidi S, Bartesaghi R. Reversal of neurodevelopmental impairment and cognitive enhancement by pharmacological intervention with the polyphenol polydatin in a Down syndrome model. Neuropharmacology 2024; 261:110170. [PMID: 39341334 DOI: 10.1016/j.neuropharm.2024.110170] [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: 04/23/2024] [Revised: 09/16/2024] [Accepted: 09/25/2024] [Indexed: 10/01/2024]
Abstract
Intellectual disability (ID) is the unavoidable hallmark of Down syndrome (DS), a genetic condition due to triplication of chromosome 21. ID in DS is largely attributable to neurogenesis and dendritogenesis alterations taking place in the prenatal/neonatal period, the most critical time window for brain development. There are currently no treatments for ID in DS. Considering the timeline of brain development, treatment aimed at improving the neurological phenotypes of DS should be initiated as early as possible and use safe agents. The goal of this study was to establish whether it is possible to improve DS-linked neurodevelopmental defects through early treatment with polydatin, a natural polyphenol. We used the Ts65Dn mouse model of DS and focused on the hippocampus, a brain region fundamental for long-term memory. We found that in Ts65Dn mice of both sexes treated with polydatin from postnatal (P) day 3 to P15 there was full restoration of neurogenesis, neuron number, and dendritic development. These effects were accompanied by normalization of Cyclin D1 and DSCAM levels, which may account for the rescue of neurogenesis and dendritogenesis, respectively. Importantly, in Ts65Dn mice treated with polydatin from P3 to adolescence (∼P50) there was full restoration of hippocampus-dependent memory, indicating a pro-cognitive outcome of treatment. No adverse effects were observed on the body and brain weight. The efficacy and safety of polydatin in a model of DS prospect the possibility of its use during early life stages for amelioration of DS-linked neurodevelopmental alterations.
Collapse
Affiliation(s)
- Marco Emili
- Department for Life Quality Studies, University of Bologna, Rimini, Italy
| | - Fiorenza Stagni
- Department for Life Quality Studies, University of Bologna, Rimini, Italy
| | - Carla Russo
- Department for Life Quality Studies, University of Bologna, Rimini, Italy
| | - Laura Angelozzi
- Department for Life Quality Studies, University of Bologna, Rimini, Italy
| | - Sandra Guidi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Renata Bartesaghi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.
| |
Collapse
|
2
|
Tallino S, Etebari R, McDonough I, Leon H, Sepulveda I, Winslow W, Bartholomew SK, Perez SE, Mufson EJ, Velazquez R. Assessing the Benefit of Dietary Choline Supplementation Throughout Adulthood in the Ts65Dn Mouse Model of Down Syndrome. Nutrients 2024; 16:4167. [PMID: 39683562 DOI: 10.3390/nu16234167] [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: 11/06/2024] [Revised: 11/21/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND/OBJECTIVES Down syndrome (DS) is the most common cause of early-onset Alzheimer's disease (AD). Dietary choline has been proposed as a modifiable factor to improve the cognitive and pathological outcomes of AD and DS, especially as many do not reach adequate daily intake levels of choline. While lower circulating choline levels correlate with worse pathological measures in AD patients, choline status and intake in DS is widely understudied. Perinatal choline supplementation (Ch+) in the Ts65Dn mouse model of DS protects offspring against AD-relevant pathology and improves cognition. Further, dietary Ch+ in adult AD models also ameliorates pathology and improves cognition. However, dietary Ch+ in adult Ts65Dn mice has not yet been explored; thus, this study aimed to supply Ch+ throughout adulthood to determine the effects on cognition and DS co-morbidities. METHODS We fed trisomic Ts65Dn mice and disomic littermate controls either a choline normal (ChN; 1.1 g/kg) or a Ch+ (5 g/kg) diet from 4.5 to 14 months of age. RESULTS We found that Ch+ in adulthood failed to improve genotype-specific deficits in spatial learning. However, in both genotypes of female mice, Ch+ significantly improved cognitive flexibility in a reverse place preference task in the IntelliCage behavioral phenotyping system. Further, Ch+ significantly reduced weight gain and peripheral inflammation in female mice of both genotypes, and significantly improved glucose metabolism in male mice of both genotypes. CONCLUSIONS Our findings suggest that adulthood choline supplementation benefits behavioral and biological factors important for general well-being in DS and related to AD risk.
Collapse
Affiliation(s)
- Savannah Tallino
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Rachel Etebari
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
| | - Ian McDonough
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
| | - Hector Leon
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Isabella Sepulveda
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Wendy Winslow
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
- Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Samantha K Bartholomew
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Sylvia E Perez
- Barrow Neurological Institute, Phoenix, AZ 85013, USA
- Arizona Alzheimer's Consortium, Phoenix, AZ 85014, USA
| | - Elliott J Mufson
- Barrow Neurological Institute, Phoenix, AZ 85013, USA
- Arizona Alzheimer's Consortium, Phoenix, AZ 85014, USA
| | - Ramon Velazquez
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
- Arizona Alzheimer's Consortium, Phoenix, AZ 85014, USA
| |
Collapse
|
3
|
LaCombe JM, Sloan K, Thomas JR, Blackwell MP, Crawford I, Bishop F, Wallace JM, Roper RJ. Sex-specific trisomic Dyrk1a-related skeletal phenotypes during development in a Down syndrome model. Dis Model Mech 2024; 17:dmm050914. [PMID: 39136051 PMCID: PMC11449447 DOI: 10.1242/dmm.050914] [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: 05/28/2024] [Accepted: 08/04/2024] [Indexed: 08/21/2024] Open
Abstract
Skeletal insufficiency affects all individuals with Down syndrome (DS) or trisomy 21 and may alter bone strength throughout development due to a reduced period of bone formation and early attainment of peak bone mass compared to those in typically developing individuals. Appendicular skeletal deficits also appear in males before females with DS. In femurs of male Ts65Dn DS model mice, cortical deficits were pronounced throughout development, but trabecular deficits and Dyrk1a overexpression were transitory until postnatal day (P) 30, when there were persistent trabecular and cortical deficits and Dyrk1a was trending toward overexpression. Correction of DS-related skeletal deficits by a purported DYRK1A inhibitor or through genetic means beginning at P21 was not effective at P30, but germline normalization of Dyrk1a improved male bone structure by P36. Trabecular and cortical deficits in female Ts65Dn mice were evident at P30 but subsided by P36, typifying periodic developmental skeletal normalizations that progressed to more prominent bone deficiencies. Sex-dependent differences in skeletal deficits with a delayed impact of trisomic Dyrk1a are important to find temporally specific treatment periods for bone and other phenotypes associated with trisomy 21.
Collapse
Affiliation(s)
- Jonathan M LaCombe
- Department of Biology, Indiana University Indianapolis, Indianapolis, IN 46202, USA
- Labcorp Early Development Laboratories, Inc., Greenfield, IN 46140, USA
| | - Kourtney Sloan
- Department of Biology, Indiana University Indianapolis, Indianapolis, IN 46202, USA
| | - Jared R Thomas
- Department of Biology, Indiana University Indianapolis, Indianapolis, IN 46202, USA
| | - Matthew P Blackwell
- Department of Biology, Indiana University Indianapolis, Indianapolis, IN 46202, USA
| | - Isabella Crawford
- Department of Biology, Indiana University Indianapolis, Indianapolis, IN 46202, USA
| | - Flannery Bishop
- Department of Biology, Indiana University Indianapolis, Indianapolis, IN 46202, USA
| | - Joseph M Wallace
- Department of Biomedical Engineering, Purdue University, Indianapolis, IN 46202, USA
| | - Randall J Roper
- Department of Biology, Indiana University Indianapolis, Indianapolis, IN 46202, USA
| |
Collapse
|
4
|
Rastogi M, Bartolucci M, Nanni M, Aloisio M, Vozzi D, Petretto A, Contestabile A, Cancedda L. Integrative multi-omic analysis reveals conserved cell-projection deficits in human Down syndrome brains. Neuron 2024; 112:2503-2523.e10. [PMID: 38810652 DOI: 10.1016/j.neuron.2024.05.002] [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: 01/02/2023] [Revised: 03/17/2024] [Accepted: 05/01/2024] [Indexed: 05/31/2024]
Abstract
Down syndrome (DS) is the most common genetic cause of cognitive disability. However, it is largely unclear how triplication of a small gene subset may impinge on diverse aspects of DS brain physiopathology. Here, we took a multi-omic approach and simultaneously analyzed by RNA-seq and proteomics the expression signatures of two diverse regions of human postmortem DS brains. We found that the overexpression of triplicated genes triggered global expression dysregulation, differentially affecting transcripts, miRNAs, and proteins involved in both known and novel biological candidate pathways. Among the latter, we observed an alteration in RNA splicing, specifically modulating the expression of genes involved in cytoskeleton and axonal dynamics in DS brains. Accordingly, we found an alteration in axonal polarization in neurons from DS human iPSCs and mice. Thus, our study provides an integrated multilayer expression database capable of identifying new potential targets to aid in designing future clinical interventions for DS.
Collapse
Affiliation(s)
- Mohit Rastogi
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, Genova 16163, Italy
| | - Martina Bartolucci
- Core Facilities - Clinical Proteomics and Metabolomics, IRCCS Istituto Giannina Gaslini, Genova 16147, Italy
| | - Marina Nanni
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, Genova 16163, Italy
| | | | - Diego Vozzi
- Central RNA Laboratory, Istituto Italiano di Tecnologia, Genova 16152, Italy
| | - Andrea Petretto
- Core Facilities - Clinical Proteomics and Metabolomics, IRCCS Istituto Giannina Gaslini, Genova 16147, Italy
| | - Andrea Contestabile
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, Genova 16163, Italy.
| | - Laura Cancedda
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, Genova 16163, Italy; Dulbecco Telethon Institute, Rome 00185, Italy.
| |
Collapse
|
5
|
Giallongo S, Ferrigno J, Caltabiano R, Broggi G, Alanazi AM, Distefano A, Tropea E, Tramutola A, Perluigi M, Volti GL, Barone E, Barbagallo IA. Aging exacerbates oxidative stress and liver fibrosis in an animal model of Down Syndrome. Aging (Albany NY) 2024; 16:10203-10215. [PMID: 38942607 PMCID: PMC11236314 DOI: 10.18632/aging.205970] [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: 10/17/2023] [Accepted: 01/11/2024] [Indexed: 06/30/2024]
Abstract
Down Syndrome (DS) is a common genetic disorder characterized by an extra copy of chromosome 21, leading to dysregulation of various metabolic pathways. Oxidative stress in DS is associated with neurodevelopmental defects, neuronal dysfunction, and a dementia onset resembling Alzheimer's disease. Additionally, chronic oxidative stress contributes to cardiovascular diseases and certain cancers prevalent in DS individuals. This study investigates the impact of ageing on oxidative stress and liver fibrosis using a DS murine model (Ts2Cje mice). Our results show that DS mice show increased liver oxidative stress and impaired antioxidant defenses, as evidenced by reduced glutathione levels and increased lipid peroxidation. Therefore, DS liver exhibits an altered inflammatory response and mitochondrial fitness as we showed by assaying the expression of HMOX1, CLPP, and the heat shock proteins Hsp90 and Hsp60. DS liver also displays dysregulated lipid metabolism, indicated by altered expression of PPARα, PPARγ, FATP5, and CTP2. Consistently, these changes might contribute to non-alcoholic fatty liver disease development, a condition characterized by liver fat accumulation. Consistently, histological analysis of DS liver reveals increased fibrosis and steatosis, as showed by Col1a1 increased expression, indicative of potential progression to liver cirrhosis. Therefore, our findings suggest an increased risk of liver pathologies in DS individuals, particularly when combined with the higher prevalence of obesity and metabolic dysfunctions in DS patients. These results shed a light on the liver's role in DS-associated pathologies and suggest potential therapeutic strategies targeting oxidative stress and lipid metabolism to prevent or mitigate liver-related complications in DS individuals.
Collapse
Affiliation(s)
- Sebastiano Giallongo
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania 95124, Italy
| | - Jessica Ferrigno
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania 95124, Italy
| | - Rosario Caltabiano
- Department G.F. Ingrassia, Section of Anatomic Pathology, University of Catania, Catania 95124, Italy
| | - Giuseppe Broggi
- Department G.F. Ingrassia, Section of Anatomic Pathology, University of Catania, Catania 95124, Italy
| | - Amer M Alanazi
- Pharmaceutical Biotechnology Laboratory, Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Alfio Distefano
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania 95124, Italy
| | - Emanuela Tropea
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania 95124, Italy
| | - Antonella Tramutola
- Department of Biochemical Sciences “A. Rossi-Fanelli”, Sapienza University of Rome, Roma, RM 00185, Italy
| | - Marzia Perluigi
- Department of Biochemical Sciences “A. Rossi-Fanelli”, Sapienza University of Rome, Roma, RM 00185, Italy
| | - Giovanni Li Volti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania 95124, Italy
| | - Eugenio Barone
- Department G.F. Ingrassia, Section of Anatomic Pathology, University of Catania, Catania 95124, Italy
| | | |
Collapse
|
6
|
Lanzillotta C, Baniowska MR, Prestia F, Sette C, Nalesso V, Perluigi M, Barone E, Duchon A, Tramutola A, Herault Y, Di Domenico F. Shaping down syndrome brain cognitive and molecular changes due to aging using adult animals from the Ts66Yah murine model. Neurobiol Dis 2024; 196:106523. [PMID: 38705491 DOI: 10.1016/j.nbd.2024.106523] [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: 02/23/2024] [Revised: 04/11/2024] [Accepted: 05/02/2024] [Indexed: 05/07/2024] Open
Abstract
Down syndrome (DS) is the most common condition with intellectual disability and is caused by trisomy of Homo sapiens chromosome 21 (HSA21). The increased dosage of genes on HSA21 is associated with early neurodevelopmental changes and subsequently at adult age with the development of Alzheimer-like cognitive decline. However, the molecular mechanisms promoting brain pathology along aging are still missing. The novel Ts66Yah model represents an evolution of the Ts65Dn, used in characterizing the progression of brain degeneration, and it manifest phenotypes closer to human DS condition. In this study we performed a longitudinal analysis (3-9 months) of adult Ts66Yah mice. Our data support the behavioural alterations occurring in Ts66Yah mice at older age with improvement in the detection of spatial memory defects and also a new anxiety-related phenotype. The evaluation of hippocampal molecular pathways in Ts66Yah mice, as effect of age, demonstrate the aberrant regulation of redox balance, proteostasis, stress response, metabolic pathways, programmed cell death and synaptic plasticity. Intriguingly, the genotype-driven changes observed in those pathways occur early promoting altered brain development and the onset of a condition of premature aging. In turn, aging may account for the subsequent hippocampal deterioration that fall in characteristic neuropathological features. Besides, the analysis of sex influence in the alteration of hippocampal mechanisms demonstrate only a mild effect. Overall, data collected in Ts66Yah provide novel and consolidated insights, concerning trisomy-driven processes that contribute to brain pathology in conjunction with aging. This, in turn, aids in bridging the existing gap in comprehending the intricate nature of DS phenotypes.
Collapse
Affiliation(s)
- Chiara Lanzillotta
- Department of Biochemical Sciences A. Rossi Fanelli, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Monika Rataj Baniowska
- Université de Strasbourg, CNRS, Inserm, Institut de Génétique Biologie Moléculaire et Cellulaire, IGBMC, UMR 7104- UMR-S 1258, F-67400 Illkirch, France
| | - Francesca Prestia
- Department of Biochemical Sciences A. Rossi Fanelli, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Chiara Sette
- Department of Biochemical Sciences A. Rossi Fanelli, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Valérie Nalesso
- Université de Strasbourg, CNRS, Inserm, Institut de Génétique Biologie Moléculaire et Cellulaire, IGBMC, UMR 7104- UMR-S 1258, F-67400 Illkirch, France
| | - Marzia Perluigi
- Department of Biochemical Sciences A. Rossi Fanelli, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Eugenio Barone
- Department of Biochemical Sciences A. Rossi Fanelli, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Arnaud Duchon
- Université de Strasbourg, CNRS, Inserm, Institut de Génétique Biologie Moléculaire et Cellulaire, IGBMC, UMR 7104- UMR-S 1258, F-67400 Illkirch, France
| | - Antonella Tramutola
- Department of Biochemical Sciences A. Rossi Fanelli, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Yann Herault
- Université de Strasbourg, CNRS, Inserm, Institut de Génétique Biologie Moléculaire et Cellulaire, IGBMC, UMR 7104- UMR-S 1258, F-67400 Illkirch, France.
| | - Fabio Di Domenico
- Department of Biochemical Sciences A. Rossi Fanelli, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy.
| |
Collapse
|
7
|
Singh N, Richtsmeier JT, Reeves RH. Comparative analysis of craniofacial shape in two mouse models of Down syndrome: Ts65Dn and TcMAC21. J Anat 2024; 244:1007-1014. [PMID: 38264931 PMCID: PMC11095296 DOI: 10.1111/joa.14012] [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: 07/27/2023] [Revised: 01/03/2024] [Accepted: 01/10/2024] [Indexed: 01/25/2024] Open
Abstract
Mouse models are central to studying and understanding the genotypic-to-phenotypic outcomes of Down syndrome (DS), a complex condition caused by an extra copy of the long arm of human chromosome 21. The recently developed TcMAC21-a transchromosomic mouse strain with comparable gene dosage to human chromosome 21 (Hsa21)-includes more Hsa21 genes than any other model of DS. Recent studies on TcMAC21 have provided valuable insight into the molecular, physiological, and neuroanatomical aspects of the model. However, relatively little is known about the craniofacial phenotype of TcMAC21 mice, particularly as it compares to the widely studied Ts65Dn model. Here we conducted a quantitative study of the cranial morphology of TcMAC21 and Ts65Dn mice and their respective unaffected littermates. Our comparative data comprise forty three-dimensional cranial measurements taken on micro-computed tomography scans of the heads of TcMAC21 and Ts65Dn mice. Our results show that TcMAC21 exhibit similar patterns of craniofacial change to Ts65Dn. However, the DS-specific morphology is more pronounced in Ts65Dn mice. Specifically, Ts65Dn present with more medio-lateral broadening and retraction of the snout compared to TcMAC21. Our findings reveal the complexity of potential gene interaction in the production of craniofacial phenotypes.
Collapse
Affiliation(s)
- Nandini Singh
- California State University, Sacramento, California, USA
| | | | - Roger H Reeves
- Physiology and Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
8
|
LaCombe JM, Sloan K, Thomas JR, Blackwell MP, Crawford I, Wallace JM, Roper RJ. Sex specific emergence of trisomic Dyrk1a-related skeletal phenotypes in the development of a Down syndrome mouse model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.24.595804. [PMID: 38826419 PMCID: PMC11142220 DOI: 10.1101/2024.05.24.595804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Skeletal insufficiency affects all individuals with Down syndrome (DS) or Trisomy 21 (Ts21) and may alter bone strength throughout development due to a reduced period of bone formation and early attainment of peak bone mass compared to typically developing individuals. Appendicular skeletal deficits also appear in males before females with DS. In femurs of male Ts65Dn DS model mice, cortical deficits were pronounced throughout development, but trabecular deficits and Dyrk1a overexpression were transitory until postnatal day (P) 30 when there were persistent trabecular and cortical deficits and Dyrk1a was trending overexpression. Correction of DS-related skeletal deficits by a purported DYRK1A inhibitor or through genetic means beginning at P21 was not effective at P30, but germline normalization of Dyrk1a improved male bone structure by P36. Trabecular and cortical deficits in female Ts65Dn mice were evident at P30 but subsided by P36, typifying periodic developmental skeletal normalizations that progressed to more prominent bone deficiencies. Sex-dependent differences in skeletal deficits with a delayed impact of trisomic Dyrk1a are important to find temporally specific treatment periods for bone and other phenotypes associated with Ts21.
Collapse
Affiliation(s)
- Jonathan M. LaCombe
- Department of Biology, Indiana University-Indianapolis, IN, USA
- Labcorp Early Development Laboratories, Inc., Greenfield, IN, USA
| | - Kourtney Sloan
- Department of Biology, Indiana University-Indianapolis, IN, USA
| | - Jared R. Thomas
- Department of Biology, Indiana University-Indianapolis, IN, USA
| | | | | | - Joseph M. Wallace
- Department of Biomedical Engineering, Purdue University, Indianapolis, IN, USA
| | | |
Collapse
|
9
|
Zhou Z, Bi Y, Zhi C, Chen S, Chen D, Wei Z, Jiang X. Astroglial Activation Is Exacerbated in a Down Syndrome Mouse Model. Neuroscience 2024; 547:88-97. [PMID: 38615829 DOI: 10.1016/j.neuroscience.2024.04.003] [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: 09/25/2023] [Revised: 04/07/2024] [Accepted: 04/09/2024] [Indexed: 04/16/2024]
Abstract
Down syndrome (DS), also known as trisomy 21, is one of the most common chromosomal disorders associated with intellectual disability. Mouse models are valuable for mechanistic and therapeutic intervention studies. The purpose of this study was to investigate astroglial anomalies in Dp16, a widely used DS mouse model. Brain sections were prepared from one-month-old Dp16 mice and their littermates, immunostained with an anti-GFAP or anti-S100B antibody, and imaged to reconstruct astroglial morphology in three dimensions. No significant difference in the number of astrocytes was found in either the hippocampal CA1 region or cortex between Dp16 and WT mice. However, the average astroglial volume in Dp16 was significantly (P < 0.05) greater than that in WT, suggesting the astroglial activation. Reanalysis of the single-nucleus RNA sequencing data indicated that the genes differentially expressed between WT and Dp16 astrocytes were associated with synapse organization and neuronal projection. In contrast, in vitro cultured neonatal astrocytes did not exhibit significant morphological changes. The expression of Gfap in in vitro cultured Dp16 astrocytes was not increased as it was in in vivo hippocampal tissue. However, after treatment with lipopolysaccharides, the inflammatory response gene IFNβ increased significantly more in Dp16 astrocytes than in WT astrocytes. Overall, our results showed that the increase in astrogliogenesis in DS was not apparent in the early life of Dp16 mice, while astrocyte activation, which may be partly caused by increased responses to inflammatory stimulation, was significant. The inflammatory response of astrocytes might be a potential therapeutic target for DS intellectual disability.
Collapse
Affiliation(s)
- Zuolin Zhou
- Birth Defects and Rare Diseases Research Center, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, Zhejiang, China
| | - Yanhua Bi
- Birth Defects and Rare Diseases Research Center, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, Zhejiang, China
| | - Chunchun Zhi
- Birth Defects and Rare Diseases Research Center, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, Zhejiang, China
| | - Siqi Chen
- Birth Defects and Rare Diseases Research Center, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, Zhejiang, China
| | - Die Chen
- Birth Defects and Rare Diseases Research Center, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, Zhejiang, China
| | - Zhen Wei
- Laboratory Animal Center, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Xiaoling Jiang
- Birth Defects and Rare Diseases Research Center, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, Zhejiang, China.
| |
Collapse
|
10
|
Fatemi SH, Otte ED, Folsom TD, Eschenlauer AC, Roper RJ, Aman JW, Thuras PD. Early Chronic Fluoxetine Treatment of Ts65Dn Mice Rescues Synaptic Vesicular Deficits and Prevents Aberrant Proteomic Alterations. Genes (Basel) 2024; 15:452. [PMID: 38674386 PMCID: PMC11049293 DOI: 10.3390/genes15040452] [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: 02/29/2024] [Revised: 03/25/2024] [Accepted: 03/26/2024] [Indexed: 04/28/2024] Open
Abstract
Down syndrome (DS) is the most common form of inherited intellectual disability caused by trisomy of chromosome 21, presenting with intellectual impairment, craniofacial abnormalities, cardiac defects, and gastrointestinal disorders. The Ts65Dn mouse model replicates many abnormalities of DS. We hypothesized that investigation of the cerebral cortex of fluoxetine-treated trisomic mice may provide proteomic signatures that identify therapeutic targets for DS. Subcellular fractionation of synaptosomes from cerebral cortices of age- and brain-area-matched samples from fluoxetine-treated vs. water-treated trisomic and euploid male mice were subjected to HPLC-tandem mass spectrometry. Analysis of the data revealed enrichment of trisomic risk genes that participate in regulation of synaptic vesicular traffic, pre-synaptic and post-synaptic development, and mitochondrial energy pathways during early brain development. Proteomic analysis of trisomic synaptic fractions revealed significant downregulation of proteins involved in synaptic vesicular traffic, including vesicular endocytosis (CLTA, CLTB, CLTC), synaptic assembly and maturation (EXOC1, EXOC3, EXOC8), anterograde axonal transport (EXOC1), neurotransmitter transport to PSD (SACM1L), endosomal-lysosomal acidification (ROGDI, DMXL2), and synaptic signaling (NRXN1, HIP1, ITSN1, YWHAG). Additionally, trisomic proteomes revealed upregulation of several trafficking proteins, involved in vesicular exocytosis (Rab5B), synapse elimination (UBE3A), scission of endocytosis (DBN1), transport of ER in dendritic spines (MYO5A), presynaptic activity-dependent bulk endocytosis (FMR1), and NMDA receptor activity (GRIN2A). Chronic fluoxetine treatment of Ts65Dn mice rescued synaptic vesicular abnormalities and prevented abnormal proteomic changes in adult Ts65Dn mice, pointing to therapeutic targets for potential treatment of DS.
Collapse
Affiliation(s)
- S. Hossein Fatemi
- Department of Psychiatry and Behavioral Sciences, University of Minnesota Medical School, Minneapolis, MN 55455, USA;
| | - Elysabeth D. Otte
- Department of Biology, Indiana University, Indianapolis, IN 46202, USA;
| | - Timothy D. Folsom
- Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN 55455, USA;
| | - Arthur C. Eschenlauer
- Minnesota Supercomputing Institute, University of Minnesota, Minneapolis, MN 55455, USA;
| | - Randall J. Roper
- Department of Biology, Indiana University-Purdue University, Indianapolis, IN 46202, USA;
| | - Justin W. Aman
- Department of Psychiatry and Behavioral Sciences, University of Minnesota Medical School, Minneapolis, MN 55455, USA;
| | - Paul D. Thuras
- Department of Psychiatry and Behavioral Sciences, University of Minnesota Medical School and VA Health Care System, One Veterans Drive, Minneapolis, MN 55417, USA
| |
Collapse
|
11
|
Staurenghi E, Testa G, Leoni V, Cecci R, Floro L, Giannelli S, Barone E, Perluigi M, Leonarduzzi G, Sottero B, Gamba P. Altered Brain Cholesterol Machinery in a Down Syndrome Mouse Model: A Possible Common Feature with Alzheimer's Disease. Antioxidants (Basel) 2024; 13:435. [PMID: 38671883 PMCID: PMC11047305 DOI: 10.3390/antiox13040435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 03/29/2024] [Accepted: 03/30/2024] [Indexed: 04/28/2024] Open
Abstract
Down syndrome (DS) is a complex chromosomal disorder considered as a genetically determined form of Alzheimer's disease (AD). Maintenance of brain cholesterol homeostasis is essential for brain functioning and development, and its dysregulation is associated with AD neuroinflammation and oxidative damage. Brain cholesterol imbalances also likely occur in DS, concurring with the precocious AD-like neurodegeneration. In this pilot study, we analyzed, in the brain of the Ts2Cje (Ts2) mouse model of DS, the expression of genes encoding key enzymes involved in cholesterol metabolism and of the levels of cholesterol and its main precursors and products of its metabolism (i.e., oxysterols). The results showed, in Ts2 mice compared to euploid mice, the downregulation of the transcription of the genes encoding the enzymes 3-hydroxy-3-methylglutaryl-CoA reductase and 24-dehydrocholesterol reductase, the latter originally recognized as an indicator of AD, and the consequent reduction in total cholesterol levels. Moreover, the expression of genes encoding enzymes responsible for brain cholesterol oxidation and the amounts of the resulting oxysterols were modified in Ts2 mouse brains, and the levels of cholesterol autoxidation products were increased, suggesting an exacerbation of cerebral oxidative stress. We also observed an enhanced inflammatory response in Ts2 mice, underlined by the upregulation of the transcription of the genes encoding for α-interferon and interleukin-6, two cytokines whose synthesis is increased in the brains of AD patients. Overall, these results suggest that DS and AD brains share cholesterol cycle derangements and altered oxysterol levels, which may contribute to the oxidative and inflammatory events involved in both diseases.
Collapse
Affiliation(s)
- Erica Staurenghi
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, 10043 Orbassano, Italy; (E.S.); (R.C.); (L.F.); (S.G.); (G.L.); (B.S.); (P.G.)
| | - Gabriella Testa
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, 10043 Orbassano, Italy; (E.S.); (R.C.); (L.F.); (S.G.); (G.L.); (B.S.); (P.G.)
| | - Valerio Leoni
- Laboratory of Clinical Pathology, Hospital Pio XI of Desio, ASST-Brianza and Department of Medicine and Surgery, University of Milano-Bicocca, 20832 Desio, Italy;
| | - Rebecca Cecci
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, 10043 Orbassano, Italy; (E.S.); (R.C.); (L.F.); (S.G.); (G.L.); (B.S.); (P.G.)
| | - Lucrezia Floro
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, 10043 Orbassano, Italy; (E.S.); (R.C.); (L.F.); (S.G.); (G.L.); (B.S.); (P.G.)
| | - Serena Giannelli
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, 10043 Orbassano, Italy; (E.S.); (R.C.); (L.F.); (S.G.); (G.L.); (B.S.); (P.G.)
| | - Eugenio Barone
- Department of Biochemical Sciences “A. Rossi-Fanelli”, Sapienza University, 00185 Roma, Italy; (E.B.); (M.P.)
| | - Marzia Perluigi
- Department of Biochemical Sciences “A. Rossi-Fanelli”, Sapienza University, 00185 Roma, Italy; (E.B.); (M.P.)
| | - Gabriella Leonarduzzi
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, 10043 Orbassano, Italy; (E.S.); (R.C.); (L.F.); (S.G.); (G.L.); (B.S.); (P.G.)
| | - Barbara Sottero
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, 10043 Orbassano, Italy; (E.S.); (R.C.); (L.F.); (S.G.); (G.L.); (B.S.); (P.G.)
| | - Paola Gamba
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, 10043 Orbassano, Italy; (E.S.); (R.C.); (L.F.); (S.G.); (G.L.); (B.S.); (P.G.)
| |
Collapse
|
12
|
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
|
13
|
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
|
14
|
Hawley LE, Stringer M, Deal AJ, Folz A, Goodlett CR, Roper RJ. Sex-specific developmental alterations in DYRK1A expression in the brain of a Down syndrome mouse model. Neurobiol Dis 2024; 190:106359. [PMID: 37992782 PMCID: PMC10843801 DOI: 10.1016/j.nbd.2023.106359] [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: 09/19/2023] [Revised: 11/02/2023] [Accepted: 11/18/2023] [Indexed: 11/24/2023] Open
Abstract
Aberrant neurodevelopment in Down syndrome (DS)-caused by triplication of human chromosome 21-is commonly attributed to gene dosage imbalance, linking overexpression of trisomic genes with disrupted developmental processes, with DYRK1A particularly implicated. We hypothesized that regional brain DYRK1A protein overexpression in trisomic mice varies over development in sex-specific patterns that may be distinct from Dyrk1a transcription, and reduction of Dyrk1a copy number from 3 to 2 in otherwise trisomic mice reduces DYRK1A, independent of other trisomic genes. DYRK1A overexpression varied with age, sex, and brain region, with peak overexpression on postnatal day (P) 6 in both sexes. Sex-dependent differences were also evident from P15-P24. Reducing Dyrk1a copy number confirmed that these differences depended on Dyrk1a gene dosage and not other trisomic genes. Trisomic Dyrk1a mRNA and protein expression were not highly correlated. Sex-specific patterns of DYRK1A overexpression during trisomic neurodevelopment may provide mechanistic targets for therapeutic intervention in DS.
Collapse
Affiliation(s)
- Laura E Hawley
- Department of Biology, Indiana University - Purdue University Indianapolis, 723 W. Michigan Street, SL306, Indianapolis, IN, 46202, USA
| | - Megan Stringer
- Department of Psychology, Indiana University - Purdue University Indianapolis, 402 N. Blackford Street, LD124, Indianapolis, IN, 46202, USA
| | - Abigail J Deal
- Department of Biology, Indiana University - Purdue University Indianapolis, 723 W. Michigan Street, SL306, Indianapolis, IN, 46202, USA
| | - Andrew Folz
- Department of Biology, Indiana University - Purdue University Indianapolis, 723 W. Michigan Street, SL306, Indianapolis, IN, 46202, USA
| | - Charles R Goodlett
- Department of Psychology, Indiana University - Purdue University Indianapolis, 402 N. Blackford Street, LD124, Indianapolis, IN, 46202, USA
| | - Randall J Roper
- Department of Biology, Indiana University - Purdue University Indianapolis, 723 W. Michigan Street, SL306, Indianapolis, IN, 46202, USA.
| |
Collapse
|
15
|
Lisgaras CP, Scharfman HE. High Frequency Oscillations (>250Hz) Outnumber Interictal Spikes in Preclinical Studies of Alzheimer's Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.30.564797. [PMID: 37961135 PMCID: PMC10634943 DOI: 10.1101/2023.10.30.564797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Interictal spikes (IIS) and seizures are well-documented in Alzheimer's disease (AD). IIS typically outnumber seizures, supporting their role as a prominent EEG biomarker in AD. In preclinical models, we showed that high frequency oscillations (HFOs>250Hz) also occur, but it is currently unknown how HFOs compare to IIS. Therefore, we asked whether the incidence of HFOs and IIS differed and if they are differentially affected by behavioral state. We used three mouse lines that simulate aspects of AD: Tg2576, presenilin 2 knockout, and Ts65Dn mice. We recorded and quantified HFOs and IIS in the hippocampus during wakefulness, slow-wave sleep, and rapid eye movement sleep. In all three mouse lines, HFOs were more frequent than IIS. High numbers of HFOs correlated with fewer IIS, suggesting for the first time possible competing dynamics among them in AD. Notably, HFOs occurred in more behavioral states than IIS. In summary, HFOs were the most abundant EEG abnormality when compared to IIS, and occurred in all behavioral states, suggesting they are a better biomarker than IIS. These findings pertained to three mouse lines, which is important because they simulate different aspects of AD. We also show that HFOs may inhibit IIS. SHORT SUMMARY Interictal spikes (IIS) and seizures are common in Alzheimer's disease (AD). IIS are more frequent than seizures and occur during earlier disease stages. In preclinical models, we showed that high frequency oscillations (HFOs>250Hz) occur, but a comparison between IIS and HFOs is lacking. Here we used 3 mouse lines with AD features and local field potential recordings to quantify IIS and HFOs. We found that HFOs outnumbered IIS and that their total numbers were inversely correlated with IIS. HFOs occurred during more behavioral states than IIS. Therefore, HFOs were the most abundant EEG abnormality, and this was generalizable across 3 types of preclinical AD.
Collapse
|
16
|
Cisterna B, Boschi F, Lacavalla MA, Vattemi GNA, Zancanaro C, Malatesta M. Physical training promotes remodeling of the skeletal muscle extracellular matrix: An ultrastructural study in a murine model of Down syndrome. Microsc Res Tech 2023; 86:1517-1528. [PMID: 37381675 DOI: 10.1002/jemt.24379] [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: 04/04/2023] [Revised: 06/15/2023] [Accepted: 06/16/2023] [Indexed: 06/30/2023]
Abstract
Down syndrome (DS) is a genetically based disease caused by triplication of chromosome 21. DS is characterized by multi-systemic premature aging associated with deficit in motor coordination, balance, and postural control. Using a morphological, morphometrical, and immunocytochemical ultrastructural approach, this study investigated in vastus lateralis muscle of Ts65Dn mouse, a murine model of DS, the effect of an adapted physical training on the extracellular matrix (ECM) characteristics and whether the forecasted exercise-induced ECM remodeling impacts on sarcomere organization. Morphometry demonstrated thicker basement membrane and larger collagen bundles with larger interfibrillar spacing as well as irregularly arrayed myofibrils and lower telethonin density on Z-lines in trisomic versus euploid sedentary mice. In agreement with the multi-systemic premature aging described in DS, these ECM alterations were similar to those previously observed in skeletal muscle of aged mice. Adapted physical training induced remodeling of ECM in both trisomic and euploid mice, that is, enlargement of the collagen bundles associated with hypertrophy of collagen fibrils and reduction of the interfibrillar spacing. A re-alignment of the myofibrils and a higher telethonin density on Z-line was found in trisomic mice. Altogether, our findings suggest that physical training is an effective tool in limiting/counteracting the trisomy-associated musculoskeletal structural anomalies. The current findings constitute a solid experimental background for further study investigating the possible positive effect of physical training on skeletal muscle performance. RESEARCH HIGHLIGHTS: Vastus lateralis muscle of trisomic mice shows aging-like alterations of extracellular matrix. Training promotes extracellular matrix remodeling. Training may be an effective tool to counteract trisomy-associated alterations of skeletal muscle.
Collapse
Affiliation(s)
- Barbara Cisterna
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Federico Boschi
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | - Maria Assunta Lacavalla
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | | | - Carlo Zancanaro
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Manuela Malatesta
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| |
Collapse
|
17
|
Lisgaras CP, Scharfman HE. Interictal spikes in Alzheimer's disease: Preclinical evidence for dominance of the dentate gyrus and cholinergic control by the medial septum. Neurobiol Dis 2023; 187:106294. [PMID: 37714307 PMCID: PMC10617404 DOI: 10.1016/j.nbd.2023.106294] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 09/11/2023] [Accepted: 09/13/2023] [Indexed: 09/17/2023] Open
Abstract
Interictal spikes (IIS) are a common type of abnormal electrical activity in Alzheimer's disease (AD) and preclinical models. The brain regions where IIS are largest are not known but are important because such data would suggest sites that contribute to IIS generation. Because hippocampus and cortex exhibit altered excitability in AD models, we asked which areas dominate the activity during IIS along the cortical-CA1-dentate gyrus (DG) dorso-ventral axis. Because medial septal (MS) cholinergic neurons are overactive when IIS typically occur, we also tested the novel hypothesis that silencing the MS cholinergic neurons selectively would reduce IIS. We used mice that simulate aspects of AD: Tg2576 mice, presenilin 2 (PS2) knockout mice and Ts65Dn mice. To selectively silence MS cholinergic neurons, Tg2576 mice were bred with choline-acetyltransferase (ChAT)-Cre mice and offspring were injected in the MS with AAV encoding inhibitory designer receptors exclusively activated by designer drugs (DREADDs). We recorded local field potentials along the cortical-CA1-DG axis using silicon probes during wakefulness, slow-wave sleep (SWS) and rapid eye movement (REM) sleep. We detected IIS in all transgenic or knockout mice but not age-matched controls. IIS were detectable throughout the cortical-CA1-DG axis and occurred primarily during REM sleep. In all 3 mouse lines, IIS amplitudes were significantly greater in the DG granule cell layer vs. CA1 pyramidal layer or overlying cortex. Current source density analysis showed robust and early current sources in the DG, and additional sources in CA1 and the cortex also. Selective chemogenetic silencing of MS cholinergic neurons significantly reduced IIS rate during REM sleep without affecting the overall duration, number of REM bouts, latency to REM sleep, or theta power during REM. Notably, two control interventions showed no effects. Consistent maximal amplitude and strong current sources of IIS in the DG suggest that the DG is remarkably active during IIS. In addition, selectively reducing MS cholinergic tone, at times when MS is hyperactive, could be a new strategy to reduce IIS in AD.
Collapse
Affiliation(s)
- Christos Panagiotis Lisgaras
- Departments of Child & Adolescent Psychiatry, Neuroscience & Physiology, and Psychiatry, and the Neuroscience Institute New York University Langone Health, 550 First Ave., New York, NY 10016, United States of America; Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric Research, New York State Office of Mental Health, 140 Old Orangeburg Road, Bldg. 35, Orangeburg, NY 10962, United States of America.
| | - Helen E Scharfman
- Departments of Child & Adolescent Psychiatry, Neuroscience & Physiology, and Psychiatry, and the Neuroscience Institute New York University Langone Health, 550 First Ave., New York, NY 10016, United States of America; Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric Research, New York State Office of Mental Health, 140 Old Orangeburg Road, Bldg. 35, Orangeburg, NY 10962, United States of America
| |
Collapse
|
18
|
DeRuisseau LR, Receno CN, Cunningham C, Bates ML, Goodell M, Liang C, Eassa B, Pascolla J, DeRuisseau KC. Breathing and Oxygen Carrying Capacity in Ts65Dn and Down Syndrome. FUNCTION 2023; 4:zqad058. [PMID: 37954975 PMCID: PMC10634617 DOI: 10.1093/function/zqad058] [Citation(s) in RCA: 2] [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: 08/11/2023] [Revised: 09/29/2023] [Accepted: 10/03/2023] [Indexed: 11/14/2023] Open
Abstract
Individuals with Down syndrome (Ds) are at increased risk of respiratory infection, aspiration pneumonia, and apnea. The Ts65Dn mouse is a commonly used model of Ds, but there have been no formal investigations of awake breathing and respiratory muscle function in these mice. We hypothesized that breathing would be impaired in Ts65Dn vs. wild-type (WT), and would be mediated by both neural and muscular inputs. Baseline minute ventilation was not different at 3, 6, or 12 mo of age. However, VT/Ti, a marker of the neural drive to breathe, was lower in Ts65Dn vs. WT and central apneas were more prevalent. The response to breathing hypoxia was not different, but the response to hypercapnia was attenuated, revealing a difference in carbon dioxide sensing, and/or motor output in Ts65Dn. Oxygen desaturations were present in room air, demonstrating that ventilation may not be sufficient to maintain adequate oxygen saturation in Ts65Dn. We observed no differences in arterial PO2 or PCO2, but Ts65Dn had lower hemoglobin and hematocrit. A retrospective medical record review of 52,346 Ds and 52,346 controls confirmed an elevated relative risk of anemia in Ds. We also performed eupneic in-vivo electromyography and in-vitro muscle function and histological fiber typing of the diaphragm, and found no difference between strains. Overall, conscious respiration is impaired in Ts65Dn, is mediated by neural mechanisms, and results in reduced hemoglobin saturation. Oxygen carrying capacity is reduced in Ts65Dn vs. WT, and we demonstrate that individuals with Ds are also at increased risk of anemia.
Collapse
Affiliation(s)
- Lara R DeRuisseau
- Department of Basic Sciences, University of Health Sciences and Pharmacy, St. Louis, MO 63110, USA
| | - Candace N Receno
- Department of Exercise Science and Athletic Training, Ithaca College, Ithaca, NY 14850, USA
| | - Caitlin Cunningham
- Department of Statistics, Mathematics and Computer Science, Le Moyne College, Syracuse, NY 13214, USA
| | - Melissa L Bates
- Departments of Health and Human Physiology, Internal Medicine, and the Stead Family Department of Pediatrics, University of Iowa, Iowa City, IA 52242, USA
| | - Morgan Goodell
- Lake Erie College of Osteopathic Medicine, Elmira, NY 14901, USA
| | - Chen Liang
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY 14642,USA
| | - Brianna Eassa
- Department of Biological Sciences, Le Moyne College, Syracuse, NY 13214, USA
| | - Jessica Pascolla
- Department of Basic Sciences, University of Health Sciences and Pharmacy, St. Louis, MO 63110, USA
| | - Keith C DeRuisseau
- Department of Basic Sciences, University of Health Sciences and Pharmacy, St. Louis, MO 63110, USA
| |
Collapse
|
19
|
Zhou Z, Zhi C, Chen D, Cai Z, Jiang X. Single-nucleus RNA sequencing reveals cell type-specific transcriptome alterations of Down syndrome hippocampus using the Dp16 mouse model. Genes Genomics 2023; 45:1305-1315. [PMID: 37548883 DOI: 10.1007/s13258-023-01433-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 07/19/2023] [Indexed: 08/08/2023]
Abstract
BACKGROUND Down syndrome (DS), the most frequently occurring human chromosomal disorder, is caused by trisomy 21. The exact molecular effects of trisomy on certain cell populations in the brain remain poorly understood. OBJECTIVE The purpose of this study was to investigate the effects of trisomy on the transcriptomes of various types of neurons and nonneuronal cells in the hippocampus. METHODS A total of 8993 nuclei from the WT and 6445 nuclei from the Dp16 hippocampus were analyzed by single-nucleus RNA sequencing (snRNA-seq). Cell clustering was achieved by the Seurat program. RESULTS Hippocampal cells were grouped into multiple neuronal and nonneuronal populations. Only a limited number of trisomic genes were upregulated (q < 0.001) over 1.25-fold in a specific type of hippocampal cell. Specifically, deregulation of genes associated with synaptic signaling and organization was observed in multiple cell populations, including excitatory neurons, oligodendrocytes, and microglia. This observation suggests the potential importance of synapse deficits in DS. Interestingly, GO annotation of the upregulated genes suggested potential activation of the immune system by hippocampal excitatory neurons. Fewer trisomic genes were altered in nonneuronal cells than in neurons. Notably, microglial transcriptome analysis revealed significantly (q < 0.001) increased expression of C1qb and C1qc, which suggested potential involvement of complement-mediated synapse loss mediated by microglia in DS. CONCLUSION The trisomy-related hippocampal deficits should be driven by a small amount, not all, of the trisomic genes in a specific type of cell. Our work may help to narrow down both the molecular and cellular targets for future gene therapies in DS.
Collapse
Affiliation(s)
- Zuolin Zhou
- The Children's Hospital, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, Hangzhou, 310051, Zhejiang, China
| | - Chunchun Zhi
- The Children's Hospital, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, Hangzhou, 310051, Zhejiang, China
| | - Die Chen
- The Children's Hospital, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, Hangzhou, 310051, Zhejiang, China
| | - Zhaowei Cai
- Laboratory Animal Research Center, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Xiaoling Jiang
- The Children's Hospital, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, Hangzhou, 310051, Zhejiang, China.
| |
Collapse
|
20
|
Thomazeau A, Lassalle O, Manzoni OJ. Glutamatergic synaptic deficits in the prefrontal cortex of the Ts65Dn mouse model for Down syndrome. Front Neurosci 2023; 17:1171797. [PMID: 37841687 PMCID: PMC10569174 DOI: 10.3389/fnins.2023.1171797] [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: 02/22/2023] [Accepted: 09/11/2023] [Indexed: 10/17/2023] Open
Abstract
Down syndrome (DS), the most prevalent cause of intellectual disability, stems from a chromosomal anomaly resulting in an entire or partial extra copy of chromosome 21. This leads to intellectual disability and a range of associated symptoms. While there has been considerable research focused on the Ts65Dn mouse model of DS, particularly in the context of the hippocampus, the synaptic underpinnings of prefrontal cortex (PFC) dysfunction in DS, including deficits in working memory, remain largely uncharted territory. In a previous study featuring mBACtgDyrk1a mice, which manifest overexpression of the Dyrk1a gene, a known candidate gene linked to intellectual disability and microcephaly in DS, we documented adverse effects on spine density, alterations in the molecular composition of synapses, and the presence of synaptic plasticity deficits within the PFC. The current study aimed to enrich our understanding of the roles of different genes in DS by studying Ts65Dn mice, which overexpress several genes including Dyrk1a, to compare with our previous work on mBACtgDyrk1a mice. Through ex-vivo electrophysiological experiments, including patch-clamp and extracellular field potential recordings, we identified alterations in the intrinsic properties of PFC layer V/VI pyramidal neurons in Ts65Dn male mice. Additionally, we observed changes in the synaptic plasticity range. Notably, long-term depression was absent in Ts65Dn mice, while synaptic or pharmacological long-term potentiation remained fully expressed in these mice. These findings provide valuable insights into the intricate synaptic mechanisms contributing to PFC dysfunction in DS, shedding light on potential therapeutic avenues for addressing the neurocognitive symptoms associated with this condition.
Collapse
Affiliation(s)
- Aurore Thomazeau
- Côte d’Azur, CNRS UMR7275, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
- Université Aix-Marseille, Marseille, France
- INMED, INSERM U1249, Marseille, France
| | - Olivier Lassalle
- Université Aix-Marseille, Marseille, France
- INMED, INSERM U1249, Marseille, France
| | - Olivier J. Manzoni
- Université Aix-Marseille, Marseille, France
- INMED, INSERM U1249, Marseille, France
| |
Collapse
|
21
|
Shao LR, Gao F, Chinnasamy V, Kazuki Y, Oshimura M, Reeves RH, Stafstrom CE. Increased propensity for infantile spasms and altered neocortical excitation-inhibition balance in a mouse model of down syndrome carrying human chromosome 21. Neurobiol Dis 2023; 184:106198. [PMID: 37315904 DOI: 10.1016/j.nbd.2023.106198] [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: 02/28/2023] [Revised: 05/30/2023] [Accepted: 06/11/2023] [Indexed: 06/16/2023] Open
Abstract
Children with Down syndrome (DS, trisomy of chromosome 21) have an increased risk of infantile spasms (IS). As an epileptic encephalopathy, IS may further impair cognitive function and exacerbate neurodevelopmental delays already present in children with DS. To investigate the pathophysiology of IS in DS, we induced IS-like epileptic spasms in a genetic mouse model of DS that carries human chromosome 21q, TcMAC21, the animal model most closely representing gene dosage imbalance in DS. Repetitive extensor/flexor spasms were induced by the GABAB receptor agonist γ-butyrolactone (GBL) and occurred predominantly in young TcMAC21 mice (85%) but also in some euploid mice (25%). During GBL application, background electroencephalographic (EEG) amplitude was reduced, and rhythmic, sharp-and-slow wave activity or high-amplitude burst (epileptiform) events emerged in both TcMAC21 and euploid mice. Spasms occurred only during EEG bursts, but not every burst was accompanied by a spasm. Electrophysiological experiments revealed that basic membrane properties (resting membrane potential, input resistance, action-potential threshold and amplitude, rheobase, input-output relationship) of layer V pyramidal neurons were not different between TcMAC21 mice and euploid controls. However, excitatory postsynaptic currents (EPSCs) evoked at various intensities were significantly larger in TcMAC21 mice than euploid controls, while inhibitory postsynaptic currents (IPSCs) were similar between the two groups, resulting in an increased excitation-inhibition (E-I) ratio. These data show that behavioral spasms with epileptic EEG activity can be induced in young TcMAC21 DS mice, providing proof-of-concept evidence for increased IS susceptibility in these DS mice. Our findings also show that basic membrane properties are similar in TcMAC21 and euploid mice, while the neocortical E-I balance is altered to favor increased excitation in TcMAC21 mice, which may predispose to IS generation.
Collapse
Affiliation(s)
- Li-Rong Shao
- Division of Pediatric Neurology, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Feng Gao
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Viveka Chinnasamy
- Division of Pediatric Neurology, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yasuhiro Kazuki
- Chromosome Engineering Research Center, Tottori University, Tottori, Japan
| | - Mistuo Oshimura
- Chromosome Engineering Research Center, Tottori University, Tottori, Japan; Trans Chromosomics, Inc., Tottori, Japan
| | - Roger H Reeves
- Chromosome Engineering Research Center, Tottori University, Tottori, Japan; Department of Human Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Carl E Stafstrom
- Division of Pediatric Neurology, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
22
|
Perelli L, Carbone F, Zhang L, Huang JK, Le C, Khan H, Citron F, Del Poggetto E, Gutschner T, Tomihara H, Soeung M, Minelli R, Srinivasan S, Peoples M, Lam TNA, Lundgren S, Xia R, Zhu C, Mohamed AMT, Zhang J, Sircar K, Sgambato A, Gao J, Jonasch E, Draetta GF, Futreal A, Bakouny Z, Van Allen EM, Choueiri T, Signoretti S, Msaouel P, Litchfield K, Turajlic S, Wang L, Chen YB, Di Natale RG, Hakimi AA, Giuliani V, Heffernan TP, Viale A, Bristow CA, Tannir NM, Carugo A, Genovese G. Interferon signaling promotes tolerance to chromosomal instability during metastatic evolution in renal cancer. NATURE CANCER 2023; 4:984-1000. [PMID: 37365326 PMCID: PMC10368532 DOI: 10.1038/s43018-023-00584-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 05/18/2023] [Indexed: 06/28/2023]
Abstract
Molecular routes to metastatic dissemination are critical determinants of aggressive cancers. Through in vivo CRISPR-Cas9 genome editing, we generated somatic mosaic genetically engineered models that faithfully recapitulate metastatic renal tumors. Disruption of 9p21 locus is an evolutionary driver to systemic disease through the rapid acquisition of complex karyotypes in cancer cells. Cross-species analysis revealed that recurrent patterns of copy number variations, including 21q loss and dysregulation of the interferon pathway, are major drivers of metastatic potential. In vitro and in vivo genomic engineering, leveraging loss-of-function studies, along with a model of partial trisomy of chromosome 21q, demonstrated a dosage-dependent effect of the interferon receptor genes cluster as an adaptive mechanism to deleterious chromosomal instability in metastatic progression. This work provides critical knowledge on drivers of renal cell carcinoma progression and defines the primary role of interferon signaling in constraining the propagation of aneuploid clones in cancer evolution.
Collapse
Affiliation(s)
- Luigi Perelli
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Federica Carbone
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Nerviano Medical Sciences, NMS Group Spa, Milan, Italy
| | - Li Zhang
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Justin K Huang
- TRACTION platform, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Courtney Le
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hania Khan
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Francesca Citron
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Edoardo Del Poggetto
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tony Gutschner
- Junior Research Group 'RNA Biology and Pathogenesis', Medical Faculty, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Hideo Tomihara
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Melinda Soeung
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rosalba Minelli
- TRACTION platform, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sanjana Srinivasan
- TRACTION platform, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael Peoples
- TRACTION platform, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Truong Nguyen Anh Lam
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sebastian Lundgren
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ruohan Xia
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Cihui Zhu
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alaa M T Mohamed
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jianhua Zhang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kanishka Sircar
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alessandro Sgambato
- Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - JianJun Gao
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Eric Jonasch
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Giulio F Draetta
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Andrew Futreal
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ziad Bakouny
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Eliezer M Van Allen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Toni Choueiri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Sabina Signoretti
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Pavlos Msaouel
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA
| | | | | | - Linghua Wang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ying Bei Chen
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Renzo G Di Natale
- Department of Urology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - A Ari Hakimi
- Department of Urology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Virginia Giuliani
- TRACTION platform, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Timothy P Heffernan
- TRACTION platform, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Andrea Viale
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Christopher A Bristow
- TRACTION platform, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nizar M Tannir
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alessandro Carugo
- TRACTION platform, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Biology, IRBM S.p.A., Rome, Italy.
| | - Giannicola Genovese
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- TRACTION platform, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
23
|
Guedj F, Kane E, Bishop LA, Pennings JLA, Herault Y, Bianchi DW. The Impact of Mmu17 Non-Hsa21 Orthologous Genes in the Ts65Dn Mouse Model of Down Syndrome: The Gold Standard Refuted. Biol Psychiatry 2023; 94:84-97. [PMID: 37074246 PMCID: PMC10330375 DOI: 10.1016/j.biopsych.2023.02.012] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 02/07/2023] [Accepted: 02/08/2023] [Indexed: 04/20/2023]
Abstract
BACKGROUND Despite successful preclinical treatment studies to improve neurocognition in the Ts65Dn mouse model of Down syndrome, translation to humans has failed. This raises questions about the appropriateness of the Ts65Dn mouse as the gold standard. We used the novel Ts66Yah mouse that carries an extra chromosome and the identical segmental Mmu16 trisomy as Ts65Dn without the Mmu17 non-Hsa21 orthologous region. METHODS Forebrains from embryonic day 18.5 Ts66Yah and Ts65Dn mice, along with euploid littermate controls, were used for gene expression and pathway analyses. Behavioral experiments were performed in neonatal and adult mice. Because male Ts66Yah mice are fertile, parent-of-origin transmission of the extra chromosome was studied. RESULTS Forty-five protein-coding genes mapped to the Ts65Dn Mmu17 non-Hsa21 orthologous region; 71%-82% are expressed during forebrain development. Several of these genes are uniquely overexpressed in Ts65Dn embryonic forebrain, producing major differences in dysregulated genes and pathways. Despite these differences, the primary Mmu16 trisomic effects were highly conserved in both models, resulting in commonly dysregulated disomic genes and pathways. Delays in motor development, communication, and olfactory spatial memory were present in Ts66Yah but more pronounced in Ts65Dn neonates. Adult Ts66Yah mice showed milder working memory deficits and sex-specific effects in exploratory behavior and spatial hippocampal memory, while long-term memory was preserved. CONCLUSIONS Our findings suggest that triplication of the non-Hsa21 orthologous Mmu17 genes significantly contributes to the phenotype of the Ts65Dn mouse and may explain why preclinical trials that used this model have unsuccessfully translated to human therapies.
Collapse
Affiliation(s)
- Faycal Guedj
- Prenatal Genomics and Fetal Therapy Section, Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - Elise Kane
- Prenatal Genomics and Fetal Therapy Section, Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - Lauren A Bishop
- Prenatal Genomics and Fetal Therapy Section, Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - Jeroen L A Pennings
- Center for Health Protection, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Yann Herault
- Université de Strasbourg, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Department of Translational Medicine and Neurogenetics, Strasbourg, France
| | - Diana W Bianchi
- Prenatal Genomics and Fetal Therapy Section, Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland; Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
24
|
Xing Z, Li Y, Cortes-Gomez E, Jiang X, Gao S, Pao A, Shan J, Song Y, Perez A, Yu T, Highsmith MR, Boadu F, Conroy JM, Singh PK, Bakin AV, Cheng J, Duan Z, Wang J, Liu S, Tycko B, Yu YE. Dissection of a Down syndrome-associated trisomy to separate the gene dosage-dependent and -independent effects of an extra chromosome. Hum Mol Genet 2023; 32:2205-2218. [PMID: 37014740 PMCID: PMC10281752 DOI: 10.1093/hmg/ddad056] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 02/13/2023] [Accepted: 03/27/2023] [Indexed: 04/05/2023] Open
Abstract
As an aneuploidy, trisomy is associated with mammalian embryonic and postnatal abnormalities. Understanding the underlying mechanisms involved in mutant phenotypes is broadly important and may lead to new strategies to treat clinical manifestations in individuals with trisomies, such as trisomy 21 [Down syndrome (DS)]. Although increased gene dosage effects because of a trisomy may account for the mutant phenotypes, there is also the possibility that phenotypic consequences of a trisomy can arise because of the presence of a freely segregating extra chromosome with its own centromere, i.e. a 'free trisomy' independent of gene dosage effects. Presently, there are no reports of attempts to functionally separate these two types of effects in mammals. To fill this gap, here we describe a strategy that employed two new mouse models of DS, Ts65Dn;Df(17)2Yey/+ and Dp(16)1Yey/Df(16)8Yey. Both models carry triplications of the same 103 human chromosome 21 gene orthologs; however, only Ts65Dn;Df(17)2Yey/+ mice carry a free trisomy. Comparison of these models revealed the gene dosage-independent impacts of an extra chromosome at the phenotypic and molecular levels for the first time. They are reflected by impairments of Ts65Dn;Df(17)2Yey/+ males in T-maze tests when compared with Dp(16)1Yey/Df(16)8Yey males. Results from the transcriptomic analysis suggest the extra chromosome plays a major role in trisomy-associated expression alterations of disomic genes beyond gene dosage effects. This model system can now be used to deepen our mechanistic understanding of this common human aneuploidy and obtain new insights into the effects of free trisomies in other human diseases such as cancers.
Collapse
Affiliation(s)
- Zhuo Xing
- The Children’s Guild Foundation Down Syndrome Research Program, Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Yichen Li
- The Children’s Guild Foundation Down Syndrome Research Program, Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Eduardo Cortes-Gomez
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Xiaoling Jiang
- The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Shuang Gao
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Bioinformatics, OmniSeq Inc., Buffalo, NY, USA
| | - Annie Pao
- The Children’s Guild Foundation Down Syndrome Research Program, Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Jidong Shan
- Molecular Cytogenetics Core, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Yinghui Song
- Molecular Cytogenetics Core, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Amanda Perez
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Tao Yu
- The Children’s Guild Foundation Down Syndrome Research Program, Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Max R Highsmith
- Department of Electric Engineering and Computer Science, University of Missouri, Columbia, MO, USA
| | - Frimpong Boadu
- Department of Electric Engineering and Computer Science, University of Missouri, Columbia, MO, USA
| | - Jeffrey M Conroy
- Research and Development, OmniSeq Inc., Buffalo, NY, USA
- Research Support Services, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Prashant K Singh
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Andrei V Bakin
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Jianlin Cheng
- Department of Electric Engineering and Computer Science, University of Missouri, Columbia, MO, USA
| | - Zhijun Duan
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
- Division of Hematology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Jianmin Wang
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Song Liu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Benjamin Tycko
- Hackensack-Meridian Health Center for Discovery and Innovation, Nutley, NJ, USA
- John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Y Eugene Yu
- The Children’s Guild Foundation Down Syndrome Research Program, Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Genetics, Genomics and Bioinformatics Program, State University of New York at Buffalo, Buffalo, NY, USA
| |
Collapse
|
25
|
Sarver DC, Xu C, Rodriguez S, Aja S, Jaffe AE, Gao FJ, Delannoy M, Periasamy M, Kazuki Y, Oshimura M, Reeves RH, Wong GW. Hypermetabolism in mice carrying a near-complete human chromosome 21. eLife 2023; 12:e86023. [PMID: 37249575 PMCID: PMC10229126 DOI: 10.7554/elife.86023] [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: 01/07/2023] [Accepted: 05/07/2023] [Indexed: 05/31/2023] Open
Abstract
The consequences of aneuploidy have traditionally been studied in cell and animal models in which the extrachromosomal DNA is from the same species. Here, we explore a fundamental question concerning the impact of aneuploidy on systemic metabolism using a non-mosaic transchromosomic mouse model (TcMAC21) carrying a near-complete human chromosome 21. Independent of diets and housing temperatures, TcMAC21 mice consume more calories, are hyperactive and hypermetabolic, remain consistently lean and profoundly insulin sensitive, and have a higher body temperature. The hypermetabolism and elevated thermogenesis are likely due to a combination of increased activity level and sarcolipin overexpression in the skeletal muscle, resulting in futile sarco(endo)plasmic reticulum Ca2+ ATPase (SERCA) activity and energy dissipation. Mitochondrial respiration is also markedly increased in skeletal muscle to meet the high ATP demand created by the futile cycle and hyperactivity. This serendipitous discovery provides proof-of-concept that sarcolipin-mediated thermogenesis via uncoupling of the SERCA pump can be harnessed to promote energy expenditure and metabolic health.
Collapse
Affiliation(s)
- Dylan C Sarver
- Department of Physiology, Johns Hopkins University School of MedicineBaltimoreUnited States
- Center for Metabolism and Obesity Research, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Cheng Xu
- Department of Physiology, Johns Hopkins University School of MedicineBaltimoreUnited States
- Center for Metabolism and Obesity Research, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Susana Rodriguez
- Department of Physiology, Johns Hopkins University School of MedicineBaltimoreUnited States
- Center for Metabolism and Obesity Research, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Susan Aja
- Center for Metabolism and Obesity Research, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Andrew E Jaffe
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Mental Health, Johns Hopkins Bloomberg School of Public HealthBaltimoreUnited States
- The Lieber Institute for Brain DevelopmentBaltimoreUnited States
- Center for Computational Biology, Johns Hopkins UniversityBaltimoreUnited States
- Department of Genetic Medicine, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public HealthBaltimoreUnited States
| | - Feng J Gao
- Department of Physiology, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Michael Delannoy
- Department of Cell Biology, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Muthu Periasamy
- Department of Physiology and Cell Biology, The Ohio State UniversityColumbusUnited States
- Burnett School of Biomedical Sciences, College of Medicine, University of Central FloridaOrlandoUnited States
| | - Yasuhiro Kazuki
- Division of Genome and Cellular Functions, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine, Tottori UniversityTottoriJapan
- Chromosome Engineering Research Center, Tottori UniversityTottoriJapan
| | - Mitsuo Oshimura
- Chromosome Engineering Research Center, Tottori UniversityTottoriJapan
| | - Roger H Reeves
- Department of Physiology, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Genetic Medicine, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - G William Wong
- Department of Physiology, Johns Hopkins University School of MedicineBaltimoreUnited States
- Center for Metabolism and Obesity Research, Johns Hopkins University School of MedicineBaltimoreUnited States
| |
Collapse
|
26
|
Inguscio CR, Lacavalla MA, Cisterna B, Zancanaro C, Malatesta M. Physical Training Chronically Stimulates the Motor Neuron Cell Nucleus in the Ts65Dn Mouse, a Model of Down Syndrome. Cells 2023; 12:1488. [PMID: 37296609 PMCID: PMC10252427 DOI: 10.3390/cells12111488] [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: 04/20/2023] [Revised: 05/20/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
Down syndrome (DS) is a genetically-based disease based on the trisomy of chromosome 21 (Hsa21). DS is characterized by intellectual disability in association with several pathological traits among which early aging and altered motor coordination are prominent. Physical training or passive exercise were found to be useful in counteracting motor impairment in DS subjects. In this study we used the Ts65Dn mouse, a widely accepted animal model of DS, to investigate the ultrastructural architecture of the medullary motor neuron cell nucleus taken as marker of the cell functional state. Using transmission electron microscopy, ultrastructural morphometry, and immunocytochemistry we carried out a detailed investigation of possible trisomy-related alteration(s) of nuclear constituents, which are known to vary their amount and distribution as a function of nuclear activity, as well as the effect of adapted physical training upon them. Results demonstrated that trisomy per se affects nuclear constituents to a limited extent; however, adapted physical training is able to chronically stimulate pre-mRNA transcription and processing activity in motor neuron nuclei of trisomic mice, although to a lesser extent than in their euploid mates. These findings are a step towards understanding the mechanisms underlying the positive effect of physical activity in DS.
Collapse
Affiliation(s)
| | | | | | - Carlo Zancanaro
- Department of Neurosciences, Biomedicine and Movement Sciences, Anatomy and Histology Section, University of Verona, Strada Le Grazie 8, I-37134 Verona, Italy; (C.R.I.); (M.A.L.); (B.C.); (M.M.)
| | | |
Collapse
|
27
|
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
|
28
|
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
|
29
|
Sarver DC, Xu C, Rodriguez S, Aja S, Jaffe AE, Gao FJ, Delannoy M, Periasamy M, Kazuki Y, Oshimura M, Reeves RH, Wong GW. Hypermetabolism in mice carrying a near complete human chromosome 21. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.30.526183. [PMID: 36778465 PMCID: PMC9915508 DOI: 10.1101/2023.01.30.526183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The consequences of aneuploidy have traditionally been studied in cell and animal models in which the extrachromosomal DNA is from the same species. Here, we explore a fundamental question concerning the impact of aneuploidy on systemic metabolism using a non-mosaic transchromosomic mouse model (TcMAC21) carrying a near complete human chromosome 21. Independent of diets and housing temperatures, TcMAC21 mice consume more calories, are hyperactive and hypermetabolic, remain consistently lean and profoundly insulin sensitive, and have a higher body temperature. The hypermetabolism and elevated thermogenesis are due to sarcolipin overexpression in the skeletal muscle, resulting in futile sarco(endo)plasmic reticulum Ca 2+ ATPase (SERCA) activity and energy dissipation. Mitochondrial respiration is also markedly increased in skeletal muscle to meet the high ATP demand created by the futile cycle. This serendipitous discovery provides proof-of-concept that sarcolipin-mediated thermogenesis via uncoupling of the SERCA pump can be harnessed to promote energy expenditure and metabolic health.
Collapse
Affiliation(s)
- Dylan C. Sarver
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA,Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Cheng Xu
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA,Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Susana Rodriguez
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA,Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Susan Aja
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, 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
| | - Feng J. Gao
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Michael Delannoy
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Muthu Periasamy
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA.,Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Yasuhiro Kazuki
- Division of Genome and Cellular Functions, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine, Tottori University, Tottori, Japan,Chromosome Engineering Research Center, Tottori University, Tottori, Japan
| | - Mitsuo Oshimura
- Chromosome Engineering Research Center, Tottori University, Tottori, Japan
| | - Roger H. Reeves
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, 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, Maryland, USA,Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA,Correspondence:
| |
Collapse
|
30
|
Savardi A, Patricelli Malizia A, De Vivo M, Cancedda L, Borgogno M. Preclinical Development of the Na-K-2Cl Co-transporter-1 (NKCC1) Inhibitor ARN23746 for the Treatment of Neurodevelopmental Disorders. ACS Pharmacol Transl Sci 2023; 6:1-11. [PMID: 36654749 PMCID: PMC9841778 DOI: 10.1021/acsptsci.2c00197] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Indexed: 01/06/2023]
Abstract
Alterations in the expression of the Cl- importer Na-K-2Cl co-transporter-1 (NKCC1) and the exporter K-Cl co-transporter 2 (KCC2) lead to impaired intracellular chloride concentration in neurons and imbalanced excitation/inhibition in the brain. These alterations have been observed in several neurological disorders (e.g., Down syndrome and autism). Recently, we have reported the discovery of the selective NKCC1 inhibitor "compound ARN23746" for the treatment of Down syndrome and autism in mouse models. Here, we report on an extensive preclinical characterization of ARN23746 toward its development as a clinical candidate. ARN23746 shows an overall excellent metabolism profile and good brain penetration. Moreover, ARN23746 is effective in rescuing cognitive impairment in Down syndrome mice upon per os administration, in line with oral treatment of neurodevelopmental disorders. Notably, ARN23746 does not present signs of toxicity or diuresis even if administered up to 50 times the effective dose. These results further support ARN23746 as a solid candidate for clinical trial-enabling studies.
Collapse
Affiliation(s)
| | | | - Marco De Vivo
- IAMA
Therapeutics, via Filippo
Turati 2/9, 16128 Genoa, Italy
- Molecular
Modeling & Drug Discovery Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Laura Cancedda
- IAMA
Therapeutics, via Filippo
Turati 2/9, 16128 Genoa, Italy
- Brain
Development & Disease Laboratory, Istituto
Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Marco Borgogno
- IAMA
Therapeutics, via Filippo
Turati 2/9, 16128 Genoa, Italy
| |
Collapse
|
31
|
Intranasal Administration of KYCCSRK Peptide Rescues Brain Insulin Signaling Activation and Reduces Alzheimer's Disease-like Neuropathology in a Mouse Model for Down Syndrome. Antioxidants (Basel) 2023; 12:antiox12010111. [PMID: 36670973 PMCID: PMC9854894 DOI: 10.3390/antiox12010111] [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: 12/19/2022] [Revised: 12/29/2022] [Accepted: 12/30/2022] [Indexed: 01/03/2023] Open
Abstract
Down syndrome (DS) is the most frequent genetic cause of intellectual disability and is strongly associated with Alzheimer's disease (AD). Brain insulin resistance greatly contributes to AD development in the general population and previous studies from our group showed an early accumulation of insulin resistance markers in DS brain, already in childhood, and even before AD onset. Here we tested the effects promoted in Ts2Cje mice by the intranasal administration of the KYCCSRK peptide known to foster insulin signaling activation by directly interacting and activating the insulin receptor (IR) and the AKT protein. Therefore, the KYCCSRK peptide might represent a promising molecule to overcome insulin resistance. Our results show that KYCCSRK rescued insulin signaling activation, increased mitochondrial complexes levels (OXPHOS) and reduced oxidative stress levels in the brain of Ts2Cje mice. Moreover, we uncovered novel characteristics of the KYCCSRK peptide, including its efficacy in reducing DYRK1A (triplicated in DS) and BACE1 protein levels, which resulted in reduced AD-like neuropathology in Ts2Cje mice. Finally, the peptide elicited neuroprotective effects by ameliorating synaptic plasticity mechanisms that are altered in DS due to the imbalance between inhibitory vs. excitatory currents. Overall, our results represent a step forward in searching for new molecules useful to reduce intellectual disability and counteract AD development in DS.
Collapse
|
32
|
Fulton SL, Wenderski W, Lepack AE, Eagle AL, Fanutza T, Bastle RM, Ramakrishnan A, Hays EC, Neal A, Bendl J, Farrelly LA, Al-Kachak A, Lyu Y, Cetin B, Chan JC, Tran TN, Neve RL, Roper RJ, Brennand KJ, Roussos P, Schimenti JC, Friedman AK, Shen L, Blitzer RD, Robison AJ, Crabtree GR, Maze I. Rescue of deficits by Brwd1 copy number restoration in the Ts65Dn mouse model of Down syndrome. Nat Commun 2022; 13:6384. [PMID: 36289231 PMCID: PMC9606253 DOI: 10.1038/s41467-022-34200-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/17/2022] [Indexed: 12/25/2022] Open
Abstract
With an incidence of ~1 in 800 births, Down syndrome (DS) is the most common chromosomal condition linked to intellectual disability worldwide. While the genetic basis of DS has been identified as a triplication of chromosome 21 (HSA21), the genes encoded from HSA21 that directly contribute to cognitive deficits remain incompletely understood. Here, we found that the HSA21-encoded chromatin effector, BRWD1, was upregulated in neurons derived from iPS cells from an individual with Down syndrome and brain of trisomic mice. We showed that selective copy number restoration of Brwd1 in trisomic animals rescued deficits in hippocampal LTP, cognition and gene expression. We demonstrated that Brwd1 tightly binds the BAF chromatin remodeling complex, and that increased Brwd1 expression promotes BAF genomic mistargeting. Importantly, Brwd1 renormalization rescued aberrant BAF localization, along with associated changes in chromatin accessibility and gene expression. These findings establish BRWD1 as a key epigenomic mediator of normal neurodevelopment and an important contributor to DS-related phenotypes.
Collapse
Affiliation(s)
- Sasha L Fulton
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Wendy Wenderski
- Department of Pathology, Stanford Medical School, Palo Alto, CA, 94305, USA
- Department of Genetics, Stanford Medical School, Palo Alto, CA, 94305, USA
- Department of Developmental Biology, Stanford Medical School, Palo Alto, CA, 94305, USA
- Howard Hughes Medical Institute, Stanford University, Palo Alto, CA, 94305, USA
| | - Ashley E Lepack
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Andrew L Eagle
- Department of Physiology, Michigan State University, East Lansing, MI, 48824, USA
| | - Tomas Fanutza
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Ryan M Bastle
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Aarthi Ramakrishnan
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Emma C Hays
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Arianna Neal
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Jaroslav Bendl
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Center for Disease Neuroepigenomics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Lorna A Farrelly
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Amni Al-Kachak
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Yang Lyu
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Bulent Cetin
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Jennifer C Chan
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Tina N Tran
- Department of Biomedical Sciences, Cornell University, Ithaca, NY, 14853, USA
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, 14853, USA
| | - Rachael L Neve
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Randall J Roper
- Department of Biology, Indiana University-Purdue University, Indianapolis, IN, 46202, USA
| | - Kristen J Brennand
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Departments of Psychiatry and Genetics, Wu Tsai Institute, Yale School of Medicine, New Haven, CT, 065109, USA
| | - Panos Roussos
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Center for Disease Neuroepigenomics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- J.J. Peters Veterans Affairs Hospital, Bronx, NY, 10468, USA
| | - John C Schimenti
- Department of Biomedical Sciences, Cornell University, Ithaca, NY, 14853, USA
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, 14853, USA
| | - Allyson K Friedman
- Department of Biological Sciences, City University of New York-Hunter College, New York, NY, 10065, USA
| | - Li Shen
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Robert D Blitzer
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Alfred J Robison
- Department of Physiology, Michigan State University, East Lansing, MI, 48824, USA
| | - Gerald R Crabtree
- Department of Pathology, Stanford Medical School, Palo Alto, CA, 94305, USA
- Department of Genetics, Stanford Medical School, Palo Alto, CA, 94305, USA
- Department of Developmental Biology, Stanford Medical School, Palo Alto, CA, 94305, USA
- Howard Hughes Medical Institute, Stanford University, Palo Alto, CA, 94305, USA
| | - Ian Maze
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Howard Hughes Medical Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
33
|
Llambrich S, González-Colom R, Wouters J, Roldán J, Salassa S, Wouters K, Van Bulck V, Sharpe J, Callaerts-Vegh Z, Vande Velde G, Martínez-Abadías N. Green Tea Catechins Modulate Skeletal Development with Effects Dependent on Dose, Time, and Structure in a down Syndrome Mouse Model. Nutrients 2022; 14:nu14194167. [PMID: 36235819 PMCID: PMC9572077 DOI: 10.3390/nu14194167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/30/2022] [Accepted: 10/03/2022] [Indexed: 11/26/2022] Open
Abstract
Altered skeletal development in Down syndrome (DS) results in a brachycephalic skull, flattened face, shorter mandibular ramus, shorter limbs, and reduced bone mineral density (BMD). Our previous study showed that low doses of green tea extract enriched in epigallocatechin-3-gallate (GTE-EGCG), administered continuously from embryonic day 9 to postnatal day 29, reduced facial dysmorphologies in the Ts65Dn (TS) mouse model of DS, but high doses could exacerbate them. Here, we extended the analyses to other skeletal structures and systematically evaluated the effects of high and low doses of GTE-EGCG treatment over postnatal development in wild-type (WT) and TS mice using in vivo µCT and geometric morphometrics. TS mice developed shorter and wider faces, skulls, and mandibles, together with shorter and narrower humerus and scapula, and reduced BMD dynamically over time. Besides facial morphology, GTE-EGCG did not rescue any other skeletal phenotype in TS treated mice. In WT mice, GTE-EGCG significantly altered the shape of the skull and mandible, reduced the length and width of the long bones, and lowered the BMD. The disparate effects of GTE-EGCG depended on the dose, developmental timepoint, and anatomical structure analyzed, emphasizing the complex nature of DS and the need to further investigate the simultaneous effects of GTE-EGCG supplementation.
Collapse
Affiliation(s)
- Sergi Llambrich
- Biomedical MRI, Department of Imaging and Pathology, University of Leuven (KU Leuven), 3000 Leuven, Belgium
| | - Rubèn González-Colom
- Departament de Biologia Evolutiva, Ecologia i Ciències Ambientals (BEECA), Facultat de Biologia, Universitat de Barcelona (UB), 08028 Barcelona, Spain
| | - Jens Wouters
- Biomedical MRI, Department of Imaging and Pathology, University of Leuven (KU Leuven), 3000 Leuven, Belgium
| | - Jorge Roldán
- Departament de Biologia Evolutiva, Ecologia i Ciències Ambientals (BEECA), Facultat de Biologia, Universitat de Barcelona (UB), 08028 Barcelona, Spain
| | - Sara Salassa
- Departament de Biologia Evolutiva, Ecologia i Ciències Ambientals (BEECA), Facultat de Biologia, Universitat de Barcelona (UB), 08028 Barcelona, Spain
| | - Kaat Wouters
- Laboratory of Biological Psychology, University of Leuven (KU Leuven), 3000 Leuven, Belgium
| | - Vicky Van Bulck
- Laboratory of Biological Psychology, University of Leuven (KU Leuven), 3000 Leuven, Belgium
| | - James Sharpe
- Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology (BIST), 08003 Barcelona, Spain
- Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), 08003 Barcelona, Spain
- EMBL Barcelona, European Molecular Biology Laboratory, 08003 Barcelona, Spain
| | | | - Greetje Vande Velde
- Biomedical MRI, Department of Imaging and Pathology, University of Leuven (KU Leuven), 3000 Leuven, Belgium
- Correspondence: (G.V.V.); (N.M.-A.); Tel.: +32-16330924 (G.V.V.); +34-934034564 (N.M.-A.)
| | - Neus Martínez-Abadías
- Departament de Biologia Evolutiva, Ecologia i Ciències Ambientals (BEECA), Facultat de Biologia, Universitat de Barcelona (UB), 08028 Barcelona, Spain
- Correspondence: (G.V.V.); (N.M.-A.); Tel.: +32-16330924 (G.V.V.); +34-934034564 (N.M.-A.)
| |
Collapse
|
34
|
Manfredi-Lozano M, Leysen V, Adamo M, Paiva I, Rovera R, Pignat JM, Timzoura FE, Candlish M, Eddarkaoui S, Malone SA, Silva MSB, Trova S, Imbernon M, Decoster L, Cotellessa L, Tena-Sempere M, Claret M, Paoloni-Giacobino A, Plassard D, Paccou E, Vionnet N, Acierno J, Maceski AM, Lutti A, Pfrieger F, Rasika S, Santoni F, Boehm U, Ciofi P, Buée L, Haddjeri N, Boutillier AL, Kuhle J, Messina A, Draganski B, Giacobini P, Pitteloud N, Prevot V. GnRH replacement rescues cognition in Down syndrome. Science 2022; 377:eabq4515. [PMID: 36048943 PMCID: PMC7613827 DOI: 10.1126/science.abq4515] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
At the present time, no viable treatment exists for cognitive and olfactory deficits in Down syndrome (DS). We show in a DS model (Ts65Dn mice) that these progressive nonreproductive neurological symptoms closely parallel a postpubertal decrease in hypothalamic as well as extrahypothalamic expression of a master molecule that controls reproduction-gonadotropin-releasing hormone (GnRH)-and appear related to an imbalance in a microRNA-gene network known to regulate GnRH neuron maturation together with altered hippocampal synaptic transmission. Epigenetic, cellular, chemogenetic, and pharmacological interventions that restore physiological GnRH levels abolish olfactory and cognitive defects in Ts65Dn mice, whereas pulsatile GnRH therapy improves cognition and brain connectivity in adult DS patients. GnRH thus plays a crucial role in olfaction and cognition, and pulsatile GnRH therapy holds promise to improve cognitive deficits in DS.
Collapse
Affiliation(s)
- Maria Manfredi-Lozano
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, LabexDistAlz, Lille, France
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 days for health, EGID, Lille, France
| | - Valerie Leysen
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, LabexDistAlz, Lille, France
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 days for health, EGID, Lille, France
| | - Michela Adamo
- Department of Endocrinology, Diabetology, and Metabolism, Lausanne University Hospital, 1011 Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Lausanne 1005, Switzerland
| | - Isabel Paiva
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), UMR 7364, Université de Strasbourg-CNRS, Strasbourg, France
| | - Renaud Rovera
- Univ. Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, Bron 69500, France
| | - Jean-Michel Pignat
- Department of Clinical Neurosciences, Neurorehabilitation Unit, University Hospital CHUV, Lausanne, Switzerland
| | - Fatima Ezzahra Timzoura
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, LabexDistAlz, Lille, France
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 days for health, EGID, Lille, France
| | - Michael Candlish
- Experimental Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, 66421, Homburg, Germany
| | - Sabiha Eddarkaoui
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, LabexDistAlz, Lille, France
| | - Samuel A. Malone
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, LabexDistAlz, Lille, France
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 days for health, EGID, Lille, France
| | - Mauro S. B. Silva
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, LabexDistAlz, Lille, France
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 days for health, EGID, Lille, France
| | - Sara Trova
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, LabexDistAlz, Lille, France
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 days for health, EGID, Lille, France
| | - Monica Imbernon
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, LabexDistAlz, Lille, France
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 days for health, EGID, Lille, France
| | - Laurine Decoster
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, LabexDistAlz, Lille, France
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 days for health, EGID, Lille, France
| | - Ludovica Cotellessa
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, LabexDistAlz, Lille, France
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 days for health, EGID, Lille, France
| | - Manuel Tena-Sempere
- Univ. Cordoba, IMIBC/HURS, CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Cordoba, Spain
| | - Marc Claret
- Neuronal Control of Metabolism Laboratory, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 08036 Barcelona, Spain
| | - Ariane Paoloni-Giacobino
- Department of Genetic Medicine, University Hospitals of Geneva, 4 rue Gabrielle-Perret-Gentil, 1211, Genève 14, Switzerland
| | - Damien Plassard
- CNRS UMR 7104, INSERM U1258, GenomEast Platform, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, Illkirch, France
| | - Emmanuelle Paccou
- Department of Endocrinology, Diabetology, and Metabolism, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Nathalie Vionnet
- Department of Endocrinology, Diabetology, and Metabolism, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - James Acierno
- Department of Endocrinology, Diabetology, and Metabolism, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Aleksandra Maleska Maceski
- Neurologic Clinic and Polyclinic, MS Centre and Research Centre for Clinical Neuroimmunology and Neuroscience Basel; University Hospital Basel, University of Basel, Basel Switzerland
| | - Antoine Lutti
- Laboratory for Research in Neuroimaging LREN, Centre for Research in Neurosciences, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Frank Pfrieger
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, 67000 Strasbourg, France
| | - S. Rasika
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, LabexDistAlz, Lille, France
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 days for health, EGID, Lille, France
| | - Federico Santoni
- Faculty of Biology and Medicine, University of Lausanne, Lausanne 1005, Switzerland
| | - Ulrich Boehm
- Experimental Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, 66421, Homburg, Germany
| | - Philippe Ciofi
- Univ. Bordeaux, Inserm, U1215, Neurocentre Magendie, Bordeaux, France
| | - Luc Buée
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, LabexDistAlz, Lille, France
| | - Nasser Haddjeri
- Univ. Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, Bron 69500, France
| | - Anne-Laurence Boutillier
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), UMR 7364, Université de Strasbourg-CNRS, Strasbourg, France
| | - Jens Kuhle
- Neurologic Clinic and Polyclinic, MS Centre and Research Centre for Clinical Neuroimmunology and Neuroscience Basel; University Hospital Basel, University of Basel, Basel Switzerland
| | - Andrea Messina
- Department of Endocrinology, Diabetology, and Metabolism, Lausanne University Hospital, 1011 Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Lausanne 1005, Switzerland
| | - Bogdan Draganski
- Laboratory for Research in Neuroimaging LREN, Centre for Research in Neurosciences, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Switzerland
- Neurology Department, Max-Planck-Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Paolo Giacobini
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, LabexDistAlz, Lille, France
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 days for health, EGID, Lille, France
| | - Nelly Pitteloud
- Department of Endocrinology, Diabetology, and Metabolism, Lausanne University Hospital, 1011 Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Lausanne 1005, Switzerland
| | - Vincent Prevot
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, LabexDistAlz, Lille, France
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 days for health, EGID, Lille, France
| |
Collapse
|
35
|
Hawley LE, Prochaska F, Stringer M, Goodlett CR, Roper RJ. Sexually dimorphic DYRK1A overexpression on postnatal day 15 in the Ts65Dn mouse model of Down syndrome: Effects of pharmacological targeting on behavioral phenotypes. Pharmacol Biochem Behav 2022; 217:173404. [DOI: 10.1016/j.pbb.2022.173404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 05/09/2022] [Accepted: 05/10/2022] [Indexed: 10/18/2022]
|
36
|
Tallino S, Winslow W, Bartholomew SK, Velazquez R. Temporal and brain region-specific elevations of soluble Amyloid-β 40-42 in the Ts65Dn mouse model of Down syndrome and Alzheimer's disease. Aging Cell 2022; 21:e13590. [PMID: 35290711 PMCID: PMC9009111 DOI: 10.1111/acel.13590] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/13/2022] [Accepted: 03/01/2022] [Indexed: 12/12/2022] Open
Abstract
Down syndrome (DS) is a leading cause of intellectual disability that also results in hallmark Alzheimer's disease (AD) pathologies such as amyloid beta (Aβ) plaques and hyperphosphorylated tau. The Ts65Dn mouse model is commonly used to study DS, as trisomic Ts65Dn mice carry 2/3 of the triplicated gene homologues as occur in human DS. The Ts65Dn strain also allows investigation of mechanisms common to DS and AD pathology, with many of these triplicated genes implicated in AD; for example, trisomic Ts65Dn mice overproduce amyloid precursor protein (APP), which is then processed into soluble Aβ40-42 fragments. Notably, Ts65Dn mice show alterations to the basal forebrain, which parallels the loss of function in this region observed in DS and AD patients early on in disease progression. However, a complete picture of soluble Aβ40-42 accumulation in a region-, age-, and sex-specific manner has not yet been characterized in the Ts65Dn model. Here, we show that trisomic mice accumulate soluble Aβ40-42 in the basal forebrain, frontal cortex, hippocampus, and cerebellum in an age-specific manner, with elevation in the frontal cortex and hippocampus as early as 4 months of age. Furthermore, we detected sex differences in accumulation of Aβ40-42 within the basal forebrain, with females having significantly higher Aβ40-42 at 7-8 months of age. Lastly, we show that APP expression in the basal forebrain and hippocampus inversely correlates with Aβ40-42 levels. This spatial and temporal characterization of soluble Aβ40-42 in the Ts65Dn model allows for further exploration of the role soluble Aβ plays in the progression of other AD-like pathologies in these key brain regions.
Collapse
Affiliation(s)
- Savannah Tallino
- Arizona State University‐Banner Neurodegenerative Disease Research Center at the Biodesign Institute Arizona State University Tempe Arizona USA
- School of Life Sciences Arizona State University Tempe Arizona USA
| | - Wendy Winslow
- Arizona State University‐Banner Neurodegenerative Disease Research Center at the Biodesign Institute Arizona State University Tempe Arizona USA
| | - Samantha K. Bartholomew
- Arizona State University‐Banner Neurodegenerative Disease Research Center at the Biodesign Institute Arizona State University Tempe Arizona USA
| | - Ramon Velazquez
- Arizona State University‐Banner Neurodegenerative Disease Research Center at the Biodesign Institute Arizona State University Tempe Arizona USA
- School of Life Sciences Arizona State University Tempe Arizona USA
- Arizona Alzheimer’s Consortium Phoenix Arizona USA
| |
Collapse
|
37
|
Pagnotta S, Tramutola A, Barone E, Di Domenico F, Pittalà V, Salerno L, Folgiero V, Caforio M, Locatelli F, Petrini S, Butterfield DA, Perluigi M. CAPE and its synthetic derivative VP961 restore BACH1/NRF2 axis in Down Syndrome. Free Radic Biol Med 2022; 183:1-13. [PMID: 35283228 DOI: 10.1016/j.freeradbiomed.2022.03.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/17/2022] [Accepted: 03/08/2022] [Indexed: 12/19/2022]
Abstract
The cells possess several mechanisms to counteract the over-production of reactive oxygen species (ROS) and reactive nitrogen species (RNS), including enzymes such as superoxide dismutase, catalase and glutathione peroxidase. Moreover, an important sensor involved in the anti-oxidant response is KEAP1-NRF2-ARE signaling complex. Under oxidative stress (OS), the transcription factor NRF2 can dissociate from the KEAP1-complex in the cytosol and translocate into the nucleus to promote the transcriptional activation of anti-oxidant genes, such as heme oxygenase 1 and NADPH quinone oxidoreductase. Within this context, the activation of NRF2 response is further regulated by BACH1, a transcription repressor, that compete with the KEAP1-NRF2-ARE complex. In this work, we focused on the role of BACH1/NRF2 ratio in the regulation of the anti-oxidant response, proposing their antithetical relation as a valuable target for a therapeutic strategy to test drugs able to exert neuroprotective effects, notably in aging and neurodegenerative diseases. Among these, Down syndrome (DS) is a complex genetic disorder characterized by BACH1 gene triplication that likely results in the impairment of NRF2 causing increased OS. Our results revealed that BACH1 overexpression alters the BACH1/NRF2 ratio in the nucleus and disturbs the induction of antioxidant response genes ultimately resulting in the accumulation of oxidative damage both in Ts2Cje mice (a mouse model of DS) and human DS lymphoblastoid cell lines (LCLs). Based on this evidence, we tested Caffeic Acid Phenethyl Ester (CAPE) and the synthetic analogue VP961, which have been proven to modulate NRF2 activity. We showed that CAPE and VP961 administration to DS LCLs was able to promote NRF2 nuclear translocation, which resulted in the amelioration of antioxidant response. Overall, our study supports the hypothesis that BACH1 triplication in DS subjects is implicated in the alteration of redox homeostasis and therapeutic strategies to overcome this effect are under investigation in our laboratory.
Collapse
Affiliation(s)
- Sara Pagnotta
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Laboratory affiliiated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Antonella Tramutola
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Laboratory affiliiated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Eugenio Barone
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Laboratory affiliiated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Fabio Di Domenico
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Laboratory affiliiated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Valeria Pittalà
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
| | - Loredana Salerno
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
| | - Valentina Folgiero
- Department of Pediatric Hematology/Oncology and of Cell and Gene Therapy, Bambino Gesù Children's Hospital, Rome, Italy
| | - Matteo Caforio
- Department of Pediatric Hematology/Oncology and of Cell and Gene Therapy, Bambino Gesù Children's Hospital, Rome, Italy
| | - Franco Locatelli
- Department of Pediatric Hematology/Oncology and of Cell and Gene Therapy, Bambino Gesù Children's Hospital, Rome, Italy; Department of Pediatrics, Sapienza University of Rome, Rome, Italy
| | - Stefania Petrini
- Confocal Microscopy Core Facility, Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - D Allan Butterfield
- Department of Chemistry and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Marzia Perluigi
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Laboratory affiliiated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy.
| |
Collapse
|
38
|
Cisterna B, Bontempi P, Sobolev AP, Costanzo M, Malatesta M, Zancanaro C. Quantitative magnetic resonance characterization of the effect of physical training on skeletal muscle of the Ts65Dn mice, a model of Down syndrome. Quant Imaging Med Surg 2022; 12:2066-2074. [PMID: 35284271 PMCID: PMC8899935 DOI: 10.21037/qims-21-729] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 11/12/2021] [Indexed: 11/11/2024]
Abstract
Down syndrome (DS) is characterized by muscle hypotonia and low muscle strength associated with motor dysfunction. Elucidation of the determinants of muscle weakness in DS would be relevant for therapeutic approaches aimed at treating/mitigating a physical disability with a strong impact on the quality of life in persons with DS. The Ts65Dn mice is a recognized mouse model of DS, with trisomic mice presenting gross motor and muscle phenotypes. The aim of this work was to assess the effect of physical exercise, a well-known tool to improve skeletal muscle condition, in the hindlimbs of trisomic and euploid male mice using quantitative magnetic resonance imaging (MRI). Magnetic resonance spectroscopy (MRS) metabolomics and histological fiber typing were used to further characterize the post-exercise muscle. Quantitative MRI showed not significantly different amounts of skeletal muscle in proximal hindlimbs in trisomic and euploid mice both at baseline and after physical exercise (P>0.05). Similar results were obtained for hindlimbs subfascia adipose tissue, and subcutaneous adipose tissue (P>0.05). MRS showed lower amounts of exercise-related metabolites (valine, isoleucine, leucine) in euploid vs. trisomic mice after exercise (P≤0.05). The percentage of slow-twitch fibers was similar in the two genotypes (P>0.05). We conclude that in DS adapted physical exercise (one month of training) does not induce quantitative changes in skeletal muscle or fiber type composition therein; however, the metabolic response of skeletal muscle to exercise may be affected by trisomy. These findings prompt further research investigating the role of physical exercise as a cue to clarify the mechanisms of the muscular deficit found in DS.
Collapse
Affiliation(s)
- Barbara Cisterna
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Pietro Bontempi
- Department of Computer Science, University of Verona, Verona, Italy
| | | | - Manuela Costanzo
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Manuela Malatesta
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Carlo Zancanaro
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| |
Collapse
|
39
|
Jamal R, LaCombe J, Patel R, Blackwell M, Thomas JR, Sloan K, Wallace JM, Roper RJ. Increased dosage and treatment time of Epigallocatechin-3-gallate (EGCG) negatively affects skeletal parameters in normal mice and Down syndrome mouse models. PLoS One 2022; 17:e0264254. [PMID: 35196359 PMCID: PMC8865638 DOI: 10.1371/journal.pone.0264254] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 02/07/2022] [Indexed: 12/15/2022] Open
Abstract
Bone abnormalities affect all individuals with Down syndrome (DS) and are linked to abnormal expression of DYRK1A, a gene found in three copies in people with DS and Ts65Dn DS model mice. Previous work in Ts65Dn male mice demonstrated that both genetic normalization of Dyrk1a and treatment with ~9 mg/kg/day Epigallocatechin-3-gallate (EGCG), the main polyphenol found in green tea and putative DYRK1A inhibitor, improved some skeletal deficits. Because EGCG treatment improved mostly trabecular skeletal deficits, we hypothesized that increasing EGCG treatment dosage and length of administration would positively affect both trabecular and cortical bone in Ts65Dn mice. Treatment of individuals with DS with green tea extract (GTE) containing EGCG also showed some weight loss in individuals with DS, and we hypothesized that weights would be affected in Ts65Dn mice after EGCG treatment. Treatment with ~20 mg/kg/day EGCG for seven weeks showed no improvements in male Ts65Dn trabecular bone and only limited improvements in cortical measures. Comparing skeletal analyses after ~20mg/kg/day EGCG treatment with previously published treatments with ~9, 50, and 200 mg/kg/day EGCG showed that increased dosage and treatment time increased cortical structural deficits leading to weaker appendicular bones in male mice. Weight was not affected by treatment in mice, except for those given a high dose of EGCG by oral gavage. These data indicate that high doses of EGCG, similar to those reported in some treatment studies of DS and other disorders, may impair long bone structure and strength. Skeletal phenotypes should be monitored when high doses of EGCG are administered therapeutically.
Collapse
Affiliation(s)
- Raza Jamal
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, United States of America
| | - Jonathan LaCombe
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, United States of America
| | - Roshni Patel
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, United States of America
| | - Matthew Blackwell
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, United States of America
| | - Jared R. Thomas
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, United States of America
| | - Kourtney Sloan
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, United States of America
| | - Joseph M. Wallace
- Department of Biomedical Engineering, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, United States of America
| | - Randall J. Roper
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, United States of America
| |
Collapse
|
40
|
Llambrich S, González R, Albaigès J, Wouters J, Marain F, Himmelreich U, Sharpe J, Dierssen M, Gsell W, Martínez-Abadías N, Vande Velde G. Multimodal in vivo Imaging of the Integrated Postnatal Development of Brain and Skull and Its Co-modulation With Neurodevelopment in a Down Syndrome Mouse Model. Front Med (Lausanne) 2022; 9:815739. [PMID: 35223915 PMCID: PMC8874331 DOI: 10.3389/fmed.2022.815739] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 01/14/2022] [Indexed: 11/13/2022] Open
Abstract
The brain and skeletal systems are intimately integrated during development through common molecular pathways. This is evidenced by genetic disorders where brain and skull dysmorphologies are associated. However, the mechanisms underlying neural and skeletal interactions are poorly understood. Using the Ts65Dn mouse model of Down syndrome (DS) as a case example, we performed the first longitudinal assessment of brain, skull and neurobehavioral development to determine alterations in the coordinated morphogenesis of brain and skull. We optimized a multimodal protocol combining in vivo micro-computed tomography (μCT) and magnetic resonance imaging (μMRI) with morphometric analyses and neurodevelopmental tests to longitudinally monitor the different systems' development trajectories during the first postnatal weeks. We also explored the impact of a perinatal treatment with green tea extracts enriched in epigallocatechin-3-gallate (GTE-EGCG), which can modulate cognition, brain and craniofacial development in DS. Our analyses quantified alterations associated with DS, with skull dysmorphologies appearing before brain anomalies, reduced integration and delayed acquisition of neurodevelopmental traits. Perinatal GTE-EGCG induced disparate effects and disrupted the magnitude of integration and covariation patterns between brain and skull. Our results exemplify how a longitudinal research approach evaluating the development of multiple systems can reveal the effect of morphological integration modulating the response of pathological phenotypes to treatment, furthering our understanding of complex genetic disorders.
Collapse
Affiliation(s)
- Sergi Llambrich
- Biomedical MRI, Department of Imaging and Pathology, KU Leuven, Flanders, Belgium
| | - Rubèn González
- Grup de Recerca en Antropologia Biológica (GREAB), Department of Biologia Evolutiva, Ecologia i Ciències Ambientals (BEECA), Universitat de Barcelona, Barcelona, Spain
- Center for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
| | - Julia Albaigès
- Center for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Raras (CIBERER), Barcelona, Spain
| | - Jens Wouters
- Biomedical MRI, Department of Imaging and Pathology, KU Leuven, Flanders, Belgium
| | - Fopke Marain
- Biomedical MRI, Department of Imaging and Pathology, KU Leuven, Flanders, Belgium
| | - Uwe Himmelreich
- Biomedical MRI, Department of Imaging and Pathology, KU Leuven, Flanders, Belgium
| | - James Sharpe
- Center for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain
- European Molecular Biology Laboratory (EMBL) Barcelona, European Molecular Biology Laboratory, Barcelona, Spain
| | - Mara Dierssen
- Center for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Raras (CIBERER), Barcelona, Spain
| | - Willy Gsell
- Biomedical MRI, Department of Imaging and Pathology, KU Leuven, Flanders, Belgium
| | - Neus Martínez-Abadías
- Grup de Recerca en Antropologia Biológica (GREAB), Department of Biologia Evolutiva, Ecologia i Ciències Ambientals (BEECA), Universitat de Barcelona, Barcelona, Spain
- Center for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
- European Molecular Biology Laboratory (EMBL) Barcelona, European Molecular Biology Laboratory, Barcelona, Spain
- *Correspondence: Neus Martínez-Abadías
| | - Greetje Vande Velde
- Biomedical MRI, Department of Imaging and Pathology, KU Leuven, Flanders, Belgium
- Greetje Vande Velde
| |
Collapse
|
41
|
Kazuki Y, Gao FJ, Yamakawa M, Hirabayashi M, Kazuki K, Kajitani N, Miyagawa-Tomita S, Abe S, Sanbo M, Hara H, Kuniishi H, Ichisaka S, Hata Y, Koshima M, Takayama H, Takehara S, Nakayama Y, Hiratsuka M, Iida Y, Matsukura S, Noda N, Li Y, Moyer AJ, Cheng B, Singh N, Richtsmeier JT, Oshimura M, Reeves RH. A transchromosomic rat model with human chromosome 21 shows robust Down syndrome features. Am J Hum Genet 2022; 109:328-344. [PMID: 35077668 DOI: 10.1016/j.ajhg.2021.12.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 12/21/2021] [Indexed: 12/31/2022] Open
Abstract
Progress in earlier detection and clinical management has increased life expectancy and quality of life in people with Down syndrome (DS). However, no drug has been approved to help individuals with DS live independently and fully. Although rat models could support more robust physiological, behavioral, and toxicology analysis than mouse models during preclinical validation, no DS rat model is available as a result of technical challenges. We developed a transchromosomic rat model of DS, TcHSA21rat, which contains a freely segregating, EGFP-inserted, human chromosome 21 (HSA21) with >93% of its protein-coding genes. RNA-seq of neonatal forebrains demonstrates that TcHSA21rat expresses HSA21 genes and has an imbalance in global gene expression. Using EGFP as a marker for trisomic cells, flow cytometry analyses of peripheral blood cells from 361 adult TcHSA21rat animals show that 81% of animals retain HSA21 in >80% of cells, the criterion for a "Down syndrome karyotype" in people. TcHSA21rat exhibits learning and memory deficits and shows increased anxiety and hyperactivity. TcHSA21rat recapitulates well-characterized DS brain morphology, including smaller brain volume and reduced cerebellar size. In addition, the rat model shows reduced cerebellar foliation, which is not observed in DS mouse models. Moreover, TcHSA21rat exhibits anomalies in craniofacial morphology, heart development, husbandry, and stature. TcHSA21rat is a robust DS animal model that can facilitate DS basic research and provide a unique tool for preclinical validation to accelerate DS drug development.
Collapse
|
42
|
Mouse models of aneuploidy to understand chromosome disorders. Mamm Genome 2021; 33:157-168. [PMID: 34719726 PMCID: PMC8913467 DOI: 10.1007/s00335-021-09930-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 10/20/2021] [Indexed: 12/04/2022]
Abstract
An organism or cell carrying a number of chromosomes that is not a multiple of the haploid count is in a state of aneuploidy. This condition results in significant changes in the level of expression of genes that are gained or lost from the aneuploid chromosome(s) and most cases in humans are not compatible with life. However, a few aneuploidies can lead to live births, typically associated with deleterious phenotypes. We do not understand why phenotypes arise from aneuploid syndromes in humans. Animal models have the potential to provide great insight, but less than a handful of mouse models of aneuploidy have been made, and no ideal system exists in which to study the effects of aneuploidy per se versus those of raised gene dosage. Here, we give an overview of human aneuploid syndromes, the effects on physiology of having an altered number of chromosomes and we present the currently available mouse models of aneuploidy, focusing on models of trisomy 21 (which causes Down syndrome) because this is the most common, and therefore, the most studied autosomal aneuploidy. Finally, we discuss the potential role of carrying an extra chromosome on aneuploid phenotypes, independent of changes in gene dosage, and methods by which this could be investigated further.
Collapse
|
43
|
Cisterna B, Malatesta M, Zancanaro C, Boschi F. A computational approach to quantitatively define sarcomere dimensions and arrangement in skeletal muscle. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2021; 211:106437. [PMID: 34624632 DOI: 10.1016/j.cmpb.2021.106437] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 09/18/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND AND OBJECTIVE The skeletal muscle is composed of integrated tissues mainly composed of myofibers i.e., long, cylindrical syncytia, whose cytoplasm is mostly occupied by parallel myofibrils. In section, each myofibril is organized in serially end-to-end arranged sarcomeres connected by Z lines. In muscle disorders, these structural and functional units can undergo structural alterations in terms of Z-line and sarcomere lengths, as well as lateral alignment of Z-line among adjacent myofibrils. In this view, objectifying alterations of the myofibril and sarcomere architecture would provide a solid foundation for qualitative observations. In this work, specific quantitative parameters characterizing the sarcomere and myofibril arrangement were defined using a computerized analysis of ultrastructural images. METHODS computerized analysis was carried out on transmission electron microscopy pictures of the murine vastus lateralis muscle. Samples from both euploid (control) and trisomic (showing myofiber alterations) Ts65Dn mice were used. Two routines were written in MATLAB to measure specific structural parameters on sarcomeres and myofibrils. The output included the Z-line, M-line, and sarcomere lengths, the Aspect Ratio (AsR) and Curviness (Cur) sarcomere shape parameters, myofibril axis (α angle), and the H parameter (evaluation of sequence of Z-lines of adjacent myofibrils). RESULTS Both routines worked well in control (euploid) skeletal muscle yielding consistent quantitative data of sarcomere and myofibril structural organization. In comparison with euploid, trisomic muscle showed statistically significant lower Z-line length, similar M-line length, and statistically significant lower sarcomere length. Both AsR and Cur were statistically significantly lower in trisomic muscle, suggesting the sarcomere is barrel-shaped in the latter. The angle (α) distribution showed that the sarcomere axes are almost parallel in euploid muscle, while a large variability occurs in trisomic tissue. The mean value of H was significantly higher in trisomic versus euploid muscle indicating that Z-lines are not perfectly aligned in trisomic muscle. CONCLUSIONS Our procedure allowed us to accurately extract and quantify sarcomere and myofibril parameters from the high-resolution electron micrographs thereby yielding an effective tool to quantitatively define trisomy-associated muscle alterations. These results pave the way to future objective quantification of skeletal muscle changes in pathological conditions. SHORT ABSTRACT The skeletal muscle is composed of integrated tissues mainly composed of myofibers i.e., long, cylindrical syncytia, whose cytoplasm is mostly occupied by parallel myofibrils organized in serially end-to-end arranged sarcomeres. Several pieces of evidence have highlighted that in muscle disorders and diseases the sarcomere structure may be altered. Therefore, objectifying alterations of the myofibril and sarcomere architecture would provide a solid foundation for qualitative observations. A computerized analysis was carried out on transmission electron microscopy images of euploid (control) and trisomic (showing myofiber alterations) skeletal muscle. Two routines were written in MATLAB to measure nine sarcomere and myofibril structural parameters. Our computational method confirmed and expanded on previous qualitative ultrastructural findings defining several trisomy-associated skeletal muscle alterations. The proposed procedure is a potentially useful tool to quantitatively define skeletal muscle changes in pathological conditions involving the sarcomere.
Collapse
Affiliation(s)
- Barbara Cisterna
- Anatomy and Histology Section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, Verona 37134, Italy
| | - Manuela Malatesta
- Anatomy and Histology Section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, Verona 37134, Italy
| | - Carlo Zancanaro
- Anatomy and Histology Section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, Verona 37134, Italy
| | - Federico Boschi
- Department of Computer Science, University of Verona, Strada Le Grazie 15, Verona 37134, Italy.
| |
Collapse
|
44
|
Thomas JR, Sloan K, Cave K, Wallace JM, Roper RJ. Skeletal Deficits in Male and Female down Syndrome Model Mice Arise Independent of Normalized Dyrk1a Expression in Osteoblasts. Genes (Basel) 2021; 12:1729. [PMID: 34828335 PMCID: PMC8624983 DOI: 10.3390/genes12111729] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/25/2021] [Accepted: 10/26/2021] [Indexed: 01/11/2023] Open
Abstract
Trisomy 21 (Ts21) causes alterations in skeletal development resulting in decreased bone mass, shortened stature and weaker bones in individuals with Down syndrome (DS). There is a sexual dimorphism in bone mineral density (BMD) deficits associated with DS with males displaying earlier deficits than females. The relationships between causative trisomic genes, cellular mechanisms, and influence of sex in DS skeletal abnormalities remain unknown. One hypothesis is that the low bone turnover phenotype observed in DS results from attenuated osteoblast function, contributing to impaired trabecular architecture, altered cortical geometry, and decreased mineralization. DYRK1A, found in three copies in humans with DS, Ts65Dn, and Dp1Tyb DS model mice, has been implicated in the development of postnatal skeletal phenotypes associated with DS. Reduced copy number of Dyrk1a to euploid levels from conception in an otherwise trisomic Ts65Dn mice resulted in a rescue of appendicular bone deficits, suggesting DYRK1A contributes to skeletal development and homeostasis. We hypothesized that reduction of Dyrk1a copy number in trisomic osteoblasts would improve cellular function and resultant skeletal structural anomalies in trisomic mice. Female mice with a floxed Dyrk1a gene (Ts65Dn,Dyrk1afl/wt) were mated with male Osx-Cre+ (expressed in osteoblasts beginning around E13.5) mice, resulting in reduced Dyrk1a copy number in mature osteoblasts in Ts65Dn,Dyrk1a+/+/Osx-Cre P42 male and female trisomic and euploid mice, compared with littermate controls. Male and female Ts65Dn,Dyrk1a+/+/+ (3 copies of DYRK1A in osteoblasts) and Ts65Dn,Dyrk1a+/+/Osx-Cre (2 copies of Dyrk1a in osteoblasts) displayed similar defects in both trabecular architecture and cortical geometry, with no improvements with reduced Dyrk1a in osteoblasts. This suggests that trisomic DYRK1A does not affect osteoblast function in a cell-autonomous manner at or before P42. Although male Dp1Tyb and Ts65Dn mice exhibit similar skeletal deficits at P42 in both trabecular and cortical bone compartments between euploid and trisomic mice, female Ts65Dn mice exhibit significant cortical and trabecular deficits at P42, in contrast to an absence of genotype effect in female Dp1Tyb mice in trabecular bone. Taken together, these data suggest skeletal deficits in DS mouse models and are sex and age dependent, and influenced by strain effects, but are not solely caused by the overexpression of Dyrk1a in osteoblasts. Identifying molecular and cellular mechanisms, disrupted by gene dosage imbalance, that are involved in the development of skeletal phenotypes associated with DS could help to design therapies to rescue skeletal deficiencies seen in DS.
Collapse
Affiliation(s)
- Jared R. Thomas
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA; (J.R.T.); (K.S.); (K.C.)
| | - Kourtney Sloan
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA; (J.R.T.); (K.S.); (K.C.)
| | - Kelsey Cave
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA; (J.R.T.); (K.S.); (K.C.)
| | - 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; (J.R.T.); (K.S.); (K.C.)
| |
Collapse
|
45
|
Bartolucci ML, Berteotti C, Alvente S, Bastianini S, Guidi S, Lo Martire V, Matteoli G, Silvani A, Stagni F, Bosi M, Alessandri-Bonetti G, Bartesaghi R, Zoccoli G. Obstructive sleep apneas naturally occur in mice during REM sleep and are highly prevalent in a mouse model of Down syndrome. Neurobiol Dis 2021; 159:105508. [PMID: 34509609 DOI: 10.1016/j.nbd.2021.105508] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 08/02/2021] [Accepted: 09/08/2021] [Indexed: 11/16/2022] Open
Abstract
STUDY OBJECTIVES The use of mouse models in sleep apnea study is limited by the belief that central (CSA) but not obstructive sleep apneas (OSA) occur in rodents. We aimed to develop a protocol to investigate the presence of OSAs in wild-type mice and, then, to apply it to a validated model of Down syndrome (Ts65Dn), a human pathology characterized by a high incidence of OSAs. METHODS In a pilot study, nine C57BL/6J wild-type mice were implanted with electrodes for electroencephalography (EEG), neck electromyography (nEMG), and diaphragmatic activity (DIA), and then placed in a whole-body-plethysmographic (WBP) chamber for 8 h during the rest (light) phase to simultaneously record sleep and breathing activity. CSA and OSA were discriminated on the basis of WBP and DIA signals recorded simultaneously. The same protocol was then applied to 12 Ts65Dn mice and 14 euploid controls. RESULTS OSAs represented about half of the apneic events recorded during rapid-eye-movement-sleep (REMS) in each experimental group, while the majority of CSAs were found during non-rapid eye movement sleep. Compared with euploid controls, Ts65Dn mice had a similar total occurrence rate of apneic events during sleep, but a significantly higher occurrence rate of OSAs during REMS, and a significantly lower occurrence rate of CSAs during NREMS. CONCLUSIONS Mice physiologically exhibit both CSAs and OSAs. The latter appear almost exclusively during REMS, and are highly prevalent in Ts65Dn. Mice may, thus, represent a useful model to accelerate the understanding of the pathophysiology and genetics of sleep-disordered breathing and to help the development of new therapies.
Collapse
Affiliation(s)
- Maria Lavinia Bartolucci
- Section of Orthodontics, Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum - University of Bologna, Bologna, Italy; PRISM Lab, Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - Chiara Berteotti
- PRISM Lab, Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - Sara Alvente
- PRISM Lab, Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - Stefano Bastianini
- PRISM Lab, Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - Sandra Guidi
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - Viviana Lo Martire
- PRISM Lab, Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - Gabriele Matteoli
- PRISM Lab, Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - Alessandro Silvani
- PRISM Lab, Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - Fiorenza Stagni
- Department for Life Quality Studies, University of Bologna, Rimini, Italy
| | - Marcello Bosi
- Sleep Disorder Center, Villa Igea-Ospedali Privati Forlì, Forlì, Italy
| | - Giulio Alessandri-Bonetti
- Section of Orthodontics, Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - Renata Bartesaghi
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - Giovanna Zoccoli
- PRISM Lab, Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum - University of Bologna, Bologna, Italy.
| |
Collapse
|
46
|
Adams AD, Hoffmann V, Koehly L, Guedj F, Bianchi DW. Novel insights from fetal and placental phenotyping in 3 mouse models of Down syndrome. Am J Obstet Gynecol 2021; 225:296.e1-296.e13. [PMID: 33766516 PMCID: PMC8429205 DOI: 10.1016/j.ajog.2021.03.019] [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: 10/14/2020] [Revised: 03/09/2021] [Accepted: 03/11/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND In human fetuses with Down syndrome, placental pathology, structural anomalies and growth restriction are present. There is currently a significant lack of information regarding the early life span in mouse models of Down syndrome. OBJECTIVE The objective of this study was to examine embryonic day 18.5 and placental phenotype in the 3 most common mouse models of Down syndrome (Ts65Dn, Dp(16)1/Yey, Ts1Cje). Based on prenatal and placental phenotyping in 3 mouse models of Down syndrome, we hypothesized that one or more of them would have a similar phenotype to human fetuses with trisomy 21, which would make it the most suitable for in utero treatment studies. STUDY DESIGN Here, C57BL6J/6 female mice were mated to Dp(16)1/Yey and Ts1Cje male mice and Ts65Dn female mice to C57BL/B6Eic3Sn.BLiAF1/J male mice. At embryonic day 18.5, dams were euthanized. Embryos and placentas were examined blindly for weight and size. Embryos were characterized as euploid or trisomic, male or female by polymerase chain reaction. A subset of embryos (34 euploid and 34 trisomic) were examined for malformations. RESULTS The Ts65Dn mouse model showed the largest differences in fetal growth, brain development, and placental development when comparing euploid and trisomic embryos. For the Dp(16)1/Yey mouse model, genotype did not impact fetal growth, but there were differences in brain and placental development. For the Ts1Cje mouse model, no significant association was found between genotype and fetal growth, brain development, or placental development. Euploid mouse embryos had no congenital anomalies; however, 1 mouse embryo died. Hepatic necrosis was seen in 6 of 12 Dp(16)1/Yey (50%) and 1 of 12 Ts1Cje (8%) mouse embryos; hepatic congestion or inflammation was observed in 3 of 10 Ts65Dn mouse embryos (30%). Renal pelvis dilation was seen in 5 of 12 Dp(16)1/Yey (42%), 5 of 10 Ts65Dn (50%), and 3 of 12 Ts1Cje (25%) mouse embryos. In addition, 1 Ts65Dn mouse embryo and 1 Dp(16)1/Yey mouse embryo had an aortic outflow abnormality. Furthermore, 2 Ts1Cje mouse embryos had ventricular septal defects. Ts65Dn mouse placentas had increased spongiotrophoblast necrosis. CONCLUSION Fetal and placental growth showed varying trends across strains. Congenital anomalies were primarily seen in trisomic embryos. The presence of liver abnormalities in all 3 mouse models of Down syndrome (10 of 34 cases) is a novel finding. Renal pelvis dilation was also common (13 of 34 cases). Future research will examine human autopsy material to determine if these findings are relevant to infants with Down syndrome. Differences in placental histology were also observed among strains.
Collapse
Affiliation(s)
- April D Adams
- Section on Prenatal Genomics and Fetal Therapy, Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD.
| | - Victoria Hoffmann
- Division of Veterinary Resources, Office of the Director, National Institutes of Health, Bethesda, MD
| | - Laura Koehly
- Social Network Methods Section, Social and Behavioral Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD
| | - Faycal Guedj
- Section on Prenatal Genomics and Fetal Therapy, Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD
| | - Diana W Bianchi
- Section on Prenatal Genomics and Fetal Therapy, Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD; Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD
| |
Collapse
|
47
|
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
|
48
|
Borgogno M, Savardi A, Manigrasso J, Turci A, Portioli C, Ottonello G, Bertozzi SM, Armirotti A, Contestabile A, Cancedda L, De Vivo M. Design, Synthesis, In Vitro and In Vivo Characterization of Selective NKCC1 Inhibitors for the Treatment of Core Symptoms in Down Syndrome. J Med Chem 2021; 64:10203-10229. [PMID: 34137257 PMCID: PMC8311653 DOI: 10.1021/acs.jmedchem.1c00603] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Intracellular chloride concentration [Cl-]i is defective in several neurological disorders. In neurons, [Cl-]i is mainly regulated by the action of the Na+-K+-Cl- importer NKCC1 and the K+-Cl- exporter KCC2. Recently, we have reported the discovery of ARN23746 as the lead candidate of a novel class of selective inhibitors of NKCC1. Importantly, ARN23746 is able to rescue core symptoms of Down syndrome (DS) and autism in mouse models. Here, we describe the discovery and extensive characterization of this chemical class of selective NKCC1 inhibitors, with focus on ARN23746 and other promising derivatives. In particular, we present compound 40 (ARN24092) as a backup/follow-up lead with in vivo efficacy in a mouse model of DS. These results further strengthen the potential of this new class of compounds for the treatment of core symptoms of brain disorders characterized by the defective NKCC1/KCC2 expression ratio.
Collapse
Affiliation(s)
- Marco Borgogno
- Molecular Modeling and Drug Discovery Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Annalisa Savardi
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy.,Dulbecco Telethon Institute, 38123 Rome, Italy
| | - Jacopo Manigrasso
- Molecular Modeling and Drug Discovery Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Alessandra Turci
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy.,Università degli Studi di Genova, via Balbi, 5, 16126 Genoa, Italy
| | - Corinne Portioli
- Molecular Modeling and Drug Discovery Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy.,Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Giuliana Ottonello
- Analytical Chemistry Facility, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Sine Mandrup Bertozzi
- Analytical Chemistry Facility, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Andrea Armirotti
- Analytical Chemistry Facility, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Andrea Contestabile
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Laura Cancedda
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy.,Dulbecco Telethon Institute, 38123 Rome, Italy
| | - Marco De Vivo
- Molecular Modeling and Drug Discovery Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| |
Collapse
|
49
|
The flavonoid 7,8-DHF fosters prenatal brain proliferation potency in a mouse model of Down syndrome. Sci Rep 2021; 11:6300. [PMID: 33737521 PMCID: PMC7973813 DOI: 10.1038/s41598-021-85284-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 02/24/2021] [Indexed: 12/17/2022] Open
Abstract
Neurogenesis impairment is a key determinant of intellectual disability in Down syndrome (DS), a genetic pathology due to triplication of chromosome 21. Since neurogenesis ceases after birth, apart in the hippocampus and olfactory bulb, the only means to tackle the problem of neurogenesis impairment in DS at its root is to intervene during gestation. A few studies in DS mouse models show that this is possible, although the drugs used may raise caveats in terms of safety. We previously found that neonatal treatment with 7,8-dihydroxyflavone (7,8-DHF), a flavonoid present in plants, restores hippocampal neurogenesis in the Ts65Dn model of DS. The goal of the current study was to establish whether prenatal treatment with 7,8-DHF improves/restores overall brain proliferation potency. Pregnant Ts65Dn females received 7,8-DHF from embryonic day 10 until delivery. On postnatal day 2 (P2) the pups were injected with BrdU and were killed after either 2 h or 52–60 days (P52–60). Evaluation of the number of proliferating (BrdU+) cells in various forebrain neurogenic niches of P2 mice showed that in treated Ts65Dn mice proliferation potency was improved or even restored in most of the examined regions, including the hippocampus. Quantification of the surviving BrdU+ cells in the dentate gyrus of P52–60 mice showed no difference between treated and untreated Ts65Dn mice. At P52–60, however, treated Ts65Dn mice exhibited a larger number of granule cells in comparison with their untreated counterparts, although their number did not reach that of euploid mice. Results show that 7,8-DHF has a widespread impact on prenatal proliferation potency in Ts65Dn mice and exerts mild long-term effects. It remains to be established whether treatment extending into the neonatal period can lead to an improvement in brain development that is retained in adulthood.
Collapse
|
50
|
Toussaint N, Redhead Y, Vidal-García M, Lo Vercio L, Liu W, Fisher EMC, Hallgrímsson B, Tybulewicz VLJ, Schnabel JA, Green JBA. A landmark-free morphometrics pipeline for high-resolution phenotyping: application to a mouse model of Down syndrome. Development 2021; 148:dev188631. [PMID: 33712441 PMCID: PMC7969589 DOI: 10.1242/dev.188631] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 02/01/2021] [Indexed: 12/20/2022]
Abstract
Characterising phenotypes often requires quantification of anatomical shape. Quantitative shape comparison (morphometrics) traditionally uses manually located landmarks and is limited by landmark number and operator accuracy. Here, we apply a landmark-free method to characterise the craniofacial skeletal phenotype of the Dp1Tyb mouse model of Down syndrome and a population of the Diversity Outbred (DO) mouse model, comparing it with a landmark-based approach. We identified cranial dysmorphologies in Dp1Tyb mice, especially smaller size and brachycephaly (front-back shortening), homologous to the human phenotype. Shape variation in the DO mice was partly attributable to allometry (size-dependent shape variation) and sexual dimorphism. The landmark-free method performed as well as, or better than, the landmark-based method but was less labour-intensive, required less user training and, uniquely, enabled fine mapping of local differences as planar expansion or shrinkage. Its higher resolution pinpointed reductions in interior mid-snout structures and occipital bones in both the models that were not otherwise apparent. We propose that this landmark-free pipeline could make morphometrics widely accessible beyond its traditional niches in zoology and palaeontology, especially in characterising developmental mutant phenotypes.
Collapse
Affiliation(s)
- Nicolas Toussaint
- School of Biomedical Engineering and Imaging Sciences, King's College London, UK
| | - Yushi Redhead
- Centre for Craniofacial Biology & Regeneration, King's College London, UK
- The Francis Crick Institute, London NW1 1AT, UK
| | - Marta Vidal-García
- Department of Cell Biology & Anatomy, University of Calgary, Calgary AB T2N 4N1, Canada
| | - Lucas Lo Vercio
- Department of Cell Biology & Anatomy, University of Calgary, Calgary AB T2N 4N1, Canada
| | - Wei Liu
- Department of Cell Biology & Anatomy, University of Calgary, Calgary AB T2N 4N1, Canada
| | - Elizabeth M C Fisher
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Benedikt Hallgrímsson
- Department of Cell Biology & Anatomy, University of Calgary, Calgary AB T2N 4N1, Canada
| | - Victor L J Tybulewicz
- The Francis Crick Institute, London NW1 1AT, UK
- Department of Immunology & Inflammation, Imperial College London, London W12 0NN, UK
| | - Julia A Schnabel
- School of Biomedical Engineering and Imaging Sciences, King's College London, UK
| | - Jeremy B A Green
- Centre for Craniofacial Biology & Regeneration, King's College London, UK
| |
Collapse
|