GnRH replacement rescues cognition in Down syndrome

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. Description Intervention for Down syndrome Down syndrome (DS), the result of trisomy of chromosome 21, carries a suite of symptoms including intellectual disability and loss of olfaction. Manfredi-Lozano et al. recognized a similarity between some of the DS symptoms and those seen in patients with a deficiency of gonadotropin-releasing hormone (GnRH) (see the Perspective by Hoffmann). Indeed, analysis of a mouse model of DS showed deficits in GnRH expression. Interventions that restored physiological GnRH levels in the mouse DS model also improved cognitive deficits. In a preliminary clinical trial in patients affected by DS, pulsatile GnRH therapy improved cognition. —PJH In a mouse model and in patients, cognitive symptoms of Down syndrome respond to restoration of gonadotropin-releasing hormone. INTRODUCTION Patients with Down syndrome (DS), a frequent condition (1 per 800 live births) caused by trisomy of chromosome 21, display a variety of characteristics, including cognitive decline due to an early-onset Alzheimer-like disease along with myelination defects in adulthood, impaired olfaction starting before puberty, and subfertility. No viable treatment exists for the cognitive and olfactory deficits seen in DS patients. RATIONALE Gonadotropin-releasing hormone (GnRH), which is secreted in a pulsatile fashion by specialized hypothalamic neurons, is the master molecule that controls reproduction in all mammals. In humans, altered GnRH secretion leads to Kallmann syndrome, which manifests with olfactory defects, gonadal immaturity, and infertility. Hypothalamic GnRH-expressing neurons also project to extrahypothalamic areas, including those involved in intellectual functions. We therefore asked whether the progressive deficits observed in DS bore any temporal correlation to the maturation of the GnRH system; what alterations to this process, if any, could be observed in the brain of animal models of DS; and whether these alterations could be therapeutically reversed in adulthood. RESULTS We first further characterized a trisomic mouse model of DS that triplicates regions analogous to human chromosome 21 (Ts65Dn mice) and displays subfertility and progressive cognitive and olfactory impairments similar to that of DS patients. These nonreproductive neurological symptoms closely paralleled a postpubertal loss of GnRH neurons and fibers in the hypothalamus as well as in extrahypothalamic regions, which is reflected by changes in the levels and pattern of release of the gonadotropin luteinizing hormone (LH) in the blood. The decreased GnRH expression in adult mice was accompanied by an imbalance in a complex network of microRNAs (several of which occur on the trisomic region) and regulatory factors that constitute a “switch” that controls GnRH expression and GnRH neuron maturation in the hypothalamus, starting during the infantile period or “minipuberty.” Indeed, we observed that elements of this switch were dysregulated as far back as the minipubertal period, well before the appearance of cognitive or olfactory deficits. Additionally, this altered expression of microRNAs and transcription factors in the hypothalamus appeared to result in the altered expression of a number of target genes, including several involved in myelination and synaptic transmission, both in the hypothalamus itself and, to a much greater extent, in the hippocampus, as well as in the altered activity of hippocampal neurons. Overexpressing a key microRNA involved in the GnRH developmental switch, miR-200b, in the hypothalamus abolished both the changes in gene expression and the deficits in neuronal activity, olfaction, and cognition in DS mice. Reinduction of miR-200b in adulthood, long after the GnRH switch, increased both the number of GnRH-expressing neurons in the hypothalamus and the proportion of neurons expressing one of its transcriptional activators, Otx2, which is known to control the opening and closing of other critical periods in brain maturation. To verify that these improvements were indeed due to the restoration of GnRH, we used cell therapy with normal hypothalamic neurons as well as chemogenetic and pharmacological interventions to produce GnRH at physiological levels and patterns (i.e., pulsatile secretion) in adult DS mice and found that these treatments all abolished olfactory and cognitive defects in the mice. Finally, based on these results, we performed a pilot study in DS patients to assess the effects of pulsatile GnRH therapy on olfaction, cognition, and brain structure and function. This treatment is safe and is presently used to treat GnRH-deficient conditions like Kallmann syndrome. We found that a 6-month pulsatile GnRH treatment improved both cognitive performance and functional brain connectivity in these patients. CONCLUSION The maintenance of the GnRH system appears to play a developmental role in brain maturation and higher functions. Pulsatile GnRH therapy holds promise to improve cognitive deficits in DS, paving the way for future clinical trials. Pulsatile GnRH therapy improves cognition in DS. DS patients show olfactory and cognitive impairments in addition to intellectual disability and reproductive maturation deficits. GnRH neurons, which control reproduction, also project to brain areas involved in cognition such as the hippocampus. In trisomic Ts65Dn mice, which mimic characteristics of DS patients, GnRH expression progressively disappears. Pulsatile GnRH therapy in DS patients improves brain connectivity and function. IU/liter, international units per liter.

[1]  V. Haroutunian,et al.  FSH blockade improves cognition in mice with Alzheimer’s disease , 2022, Nature.

[2]  D. Conen,et al.  Serum neurofilament light chain for individual prognostication of disease activity in people with multiple sclerosis: a retrospective modelling and validation study , 2022, The Lancet Neurology.

[3]  J. Hipp,et al.  A randomized, double-blind, placebo-controlled phase II trial to explore the effects of a GABAA-α5 NAM (basmisanil) on intellectual disability associated with Down syndrome , 2022, Journal of neurodevelopmental disorders.

[4]  S. Ojeda,et al.  GnRH neurons recruit astrocytes in infancy to facilitate network integration and sexual maturation , 2021, Nature Neuroscience.

[5]  D. Cai,et al.  GnRH pulse frequency and irregularity play a role in male aging , 2021, Nature Aging.

[6]  S. Moenter,et al.  Gonadotropin‐releasing hormone (GnRH) measurements in pituitary portal blood: A history , 2021, Journal of neuroendocrinology.

[7]  C. Phillips,et al.  Restoring statistical validity in group analyses of motion‐corrupted MRI data , 2021, bioRxiv.

[8]  R. Munn,et al.  Signalling pathways contributing to learning and memory deficits in the Ts65Dn mouse model of Down syndrome , 2021, Neuronal signaling.

[9]  M. Culler,et al.  The cryptic gonadotropin-releasing hormone neuronal system of human basal ganglia , 2021, bioRxiv.

[10]  M. Pero-Cebollero,et al.  Complexity Analysis of the Default Mode Network Using Resting-State fMRI in Down Syndrome: Relationships Highlighted by a Neuropsychological Assessment , 2021, Brain sciences.

[11]  Ferath Kherif,et al.  Temporal trajectory of brain tissue property changes induced by electroconvulsive therapy , 2021, NeuroImage.

[12]  Julia P. Owen,et al.  Myelin development in cerebral gray and white matter during adolescence and late childhood , 2020, NeuroImage.

[13]  M. Pero-Cebollero,et al.  Resting‐state default mode network connectivity in young individuals with Down syndrome , 2020, Brain and behavior.

[14]  Mohit Rastogi,et al.  Rescuing Over-activated Microglia Restores Cognitive Performance in Juvenile Animals of the Dp(16) Mouse Model of Down Syndrome , 2020, Neuron.

[15]  P. Chanson,et al.  GnRH stimulation testing and serum inhibin B in males: insufficient specificity for discriminating between congenital hypogonadotropic hypogonadism from constitutional delay of growth and puberty. , 2020, Human reproduction.

[16]  S. Lehmann,et al.  Clinical and biomarker changes of Alzheimer's disease in adults with Down syndrome: a cross-sectional study , 2020, The Lancet.

[17]  Yi Wei Comparative transcriptome analysis of the hippocampus from sleep-deprived and Alzheimer’s disease mice , 2020, Genetics and molecular biology.

[18]  Catherine J. Stoodley,et al.  Hypoplasia of cerebellar afferent networks in Down syndrome revealed by DTI-driven tensor based morphometry , 2020, Scientific Reports.

[19]  S. Antonarakis,et al.  Down syndrome , 2020, Nature Reviews Disease Primers.

[20]  Bin Zhang,et al.  Promoter DNA hypermethylation – Implications for Alzheimer’s disease , 2019, Neuroscience Letters.

[21]  Eleni G. Christodoulou,et al.  Widespread Translational Control of Fibrosis in the Human Heart by RNA-Binding Proteins , 2019, Circulation.

[22]  Nikolaus Weiskopf,et al.  hMRI – A toolbox for quantitative MRI in neuroscience and clinical research , 2019, NeuroImage.

[23]  J. Bischofberger,et al.  Enhanced Dendritic Inhibition and Impaired NMDAR Activation in a Mouse Model of Down Syndrome , 2019, The Journal of Neuroscience.

[24]  T. Arichi,et al.  New approaches to studying early brain development in Down syndrome , 2019, Developmental medicine and child neurology.

[25]  P. Mellon,et al.  Differential CRE Expression in Lhrh-cre and GnRH-cre Alleles and the Impact on Fertility in Otx2-Flox Mice , 2019, Neuroendocrinology.

[26]  E. Head,et al.  Dementia in Down syndrome: unique insights for Alzheimer disease research , 2019, Nature Reviews Neurology.

[27]  Galo Canizares A History , 2018, Thresholds.

[28]  A. Pfefferbaum,et al.  Regional growth trajectories of cortical myelination in adolescents and young adults: longitudinal validation and functional correlates , 2018, Brain Imaging and Behavior.

[29]  Ashley M. Fortress,et al.  Chemogenetic inactivation of the dorsal hippocampus and medial prefrontal cortex, individually and concurrently, impairs object recognition and spatial memory consolidation in female mice , 2018, Neurobiology of Learning and Memory.

[30]  K. Yaffe,et al.  Prevalence of Aging, Dementia, and Multimorbidity in Older Adults With Down Syndrome , 2018, JAMA neurology.

[31]  P. Dutar,et al.  Reinstating plasticity and memory in a tauopathy mouse model with an acetyltransferase activator , 2018, EMBO molecular medicine.

[32]  S. Esposito,et al.  Up-To-Date Review About Minipuberty and Overview on Hypothalamic-Pituitary-Gonadal Axis Activation in Fetal and Neonatal Life , 2018, Front. Endocrinol..

[33]  Antoine Lutti,et al.  Controlling motion artefact levels in MR images by suspending data acquisition during periods of head motion , 2017, bioRxiv.

[34]  Alain Chédotal,et al.  Tridimensional Visualization and Analysis of Early Human Development , 2017, Cell.

[35]  Stylianos E. Antonarakis,et al.  Down syndrome and the complexity of genome dosage imbalance , 2016, Nature Reviews Genetics.

[36]  E. Rugarli,et al.  The Mitochondrial m-AAA Protease Prevents Demyelination and Hair Greying , 2016, PLoS genetics.

[37]  Alain Chédotal,et al.  Development of the neurons controlling fertility in humans: new insights from 3D imaging and transparent fetal brains , 2016, Development.

[38]  J. Buxbaum,et al.  Integrative network analysis of nineteen brain regions identifies molecular signatures and networks underlying selective regional vulnerability to Alzheimer’s disease , 2016, Genome Medicine.

[39]  J. Pujol,et al.  Safety and efficacy of cognitive training plus epigallocatechin-3-gallate in young adults with Down's syndrome (TESDAD): a double-blind, randomised, placebo-controlled, phase 2 trial , 2016, The Lancet Neurology.

[40]  Judith B. Zaugg,et al.  Data-driven hypothesis weighting increases detection power in genome-scale multiple testing , 2016, Nature Methods.

[41]  A. Messina,et al.  A microRNA switch regulates the rise in hypothalamic GnRH production before puberty , 2016, Nature Neuroscience.

[42]  A. Peters,et al.  Down Syndrome Developmental Brain Transcriptome Reveals Defective Oligodendrocyte Differentiation and Myelination , 2016, Neuron.

[43]  C. Zancanaro,et al.  Olfaction in People with Down Syndrome: A Comprehensive Assessment across Four Decades of Age , 2016, PloS one.

[44]  M. Maghnie,et al.  Expert consensus document: European Consensus Statement on congenital hypogonadotropic hypogonadism—pathogenesis, diagnosis and treatment , 2015, Nature Reviews Endocrinology.

[45]  R. Lührmann,et al.  HDAC inhibitor-dependent transcriptome and memory reinstatement in cognitive decline models. , 2015, The Journal of clinical investigation.

[46]  Martin Vingron,et al.  Translational regulation shapes the molecular landscape of complex disease phenotypes , 2015, Nature Communications.

[47]  Ching-Po Lin,et al.  Age‐Related Changes in Resting‐State Networks of A Large Sample Size of Healthy Elderly , 2015, CNS neuroscience & therapeutics.

[48]  A. Contestabile,et al.  Reversing excitatory GABAAR signaling restores synaptic plasticity and memory in a mouse model of Down syndrome , 2015, Nature Medicine.

[49]  J. Pujol,et al.  Anomalous brain functional connectivity contributing to poor adaptive behavior in Down syndrome , 2015, Cortex.

[50]  Frank Bradke,et al.  A simple method for 3D analysis of immunolabeled axonal tracts in a transparent nervous system. , 2014, Cell reports.

[51]  W. Huber,et al.  Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2 , 2014, Genome Biology.

[52]  Y. Bozzi,et al.  Early depolarizing GABA controls critical period plasticity in the rat visual cortex , 2014, Nature Neuroscience.

[53]  Paul Theodor Pyl,et al.  HTSeq—a Python framework to work with high-throughput sequencing data , 2014, bioRxiv.

[54]  Eva Benito,et al.  MicroRNA‐125b induces tau hyperphosphorylation and cognitive deficits in Alzheimer's disease , 2014, The EMBO journal.

[55]  J. Leek svaseq: removing batch effects and other unwanted noise from sequencing data , 2014, bioRxiv.

[56]  E. Rugarli,et al.  Loss of the m‐AAA protease subunit AFG3L2 causes mitochondrial transport defects and tau hyperphosphorylation , 2014, The EMBO journal.

[57]  M. Milanese,et al.  Fluoxetine in adulthood normalizes GABA release and rescues hippocampal synaptic plasticity and spatial memory in a mouse model of Down Syndrome , 2014, Neurobiology of Disease.

[58]  R. Bartesaghi,et al.  Age-related impairment of olfactory bulb neurogenesis in the Ts65Dn mouse model of Down syndrome , 2014, Experimental Neurology.

[59]  Michel Boulouard,et al.  Object recognition test in mice , 2013, Nature Protocols.

[60]  The Lancet Neurology Strengthening connections between Down syndrome and AD , 2013, The Lancet Neurology.

[61]  J. Veldhuis,et al.  Development of a methodology for and assessment of pulsatile luteinizing hormone secretion in juvenile and adult male mice. , 2013, Endocrinology.

[62]  Sarah J. Cohen,et al.  The Rodent Hippocampus Is Essential for Nonspatial Object Memory , 2013, Current Biology.

[63]  P. Dworzynski,et al.  Mutations in FGF17, IL17RD, DUSP6, SPRY4, and FLRT3 are identified in individuals with congenital hypogonadotropic hypogonadism. , 2013, American journal of human genetics.

[64]  D. Cai,et al.  Hypothalamic Programming of Systemic Aging Involving IKKβ/NF-κB and GnRH , 2013, Nature.

[65]  Frank Bradke,et al.  High-resolution imaging of entire organs by 3-dimensional imaging of solvent cleared organs (3DISCO) , 2013, Experimental Neurology.

[66]  Nikolaus Weiskopf,et al.  Quantitative multi-parameter mapping of R1, PD*, MT, and R2* at 3T: a multi-center validation , 2013, Front. Neurosci..

[67]  P. Derambure,et al.  The acute inhibition of rapid eye movement sleep by citalopram may impair spatial learning and passive avoidance in mice , 2013, Journal of Neural Transmission.

[68]  Gerard R. Ridgway,et al.  Symmetric Diffeomorphic Modeling of Longitudinal Structural MRI , 2013, Front. Neurosci..

[69]  A. Prochiantz,et al.  Homeoprotein Signaling in Development, Health, and Disease: A Shaking of Dogmas Offers Challenges and Promises from Bench to Bed , 2013, Pharmacological Reviews.

[70]  M. Dierssen Down syndrome: the brain in trisomic mode , 2012, Nature Reviews Neuroscience.

[71]  Oliver Speck,et al.  Measurement and Correction of Microscopic Head Motion during Magnetic Resonance Imaging of the Brain , 2012, PloS one.

[72]  Frank Bradke,et al.  Three-dimensional imaging of solvent-cleared organs using 3DISCO , 2012, Nature Protocols.

[73]  Wei Li,et al.  RSeQC: quality control of RNA-seq experiments , 2012, Bioinform..

[74]  J. Hardelin,et al.  SEMA3A, a Gene Involved in Axonal Pathfinding, Is Mutated in Patients with Kallmann Syndrome , 2012, PLoS genetics.

[75]  Nikolaus Weiskopf,et al.  High-Resolution Functional MRI at 3 T: 3D/2D Echo-Planar Imaging with Optimized Physiological Noise Correction , 2012, Magnetic resonance in medicine.

[76]  Nathan Cramer,et al.  From Abnormal Hippocampal Synaptic Plasticity in Down Syndrome Mouse Models to Cognitive Disability in Down Syndrome , 2012, Neural plasticity.

[77]  E. Goldson,et al.  Antagonism of NMDA receptors as a potential treatment for Down syndrome: a pilot randomized controlled trial , 2012, Translational Psychiatry.

[78]  Richard L. Doty,et al.  Olfactory dysfunction in Parkinson disease , 2012, Nature Reviews Neurology.

[79]  T. Münch,et al.  Relevance of Exocytotic Glutamate Release from Retinal Glia , 2012, Neuron.

[80]  O. Josephs,et al.  Robust and Fast Whole Brain Mapping of the RF Transmit Field B1 at 7T , 2012, PloS one.

[81]  T. Johnson,et al.  Memantine for dementia in adults older than 40 years with Down's syndrome (MEADOWS): a randomised, double-blind, placebo-controlled trial , 2012, The Lancet.

[82]  K. Gardiner,et al.  Loss of correlations among proteins in brains of the Ts65Dn mouse model of down syndrome. , 2012, Journal of proteome research.

[83]  Frédérique Clément,et al.  DynPeak: An Algorithm for Pulse Detection and Frequency Analysis in Hormonal Time Series , 2011, PloS one.

[84]  S. Bloom,et al.  Sexual Dimorphism of Kisspeptin and Neurokinin B Immunoreactive Neurons in the Infundibular Nucleus of Aged Men and Women , 2011, Front. Endocrin..

[85]  R. Rey,et al.  Early onset of primary hypogonadism revealed by serum anti-Müllerian hormone determination during infancy and childhood in trisomy 21. , 2011, International journal of andrology.

[86]  Yueming Ding,et al.  Molecular characterization of the translocation breakpoints in the Down syndrome mouse model Ts65Dn , 2011, Mammalian Genome.

[87]  D. Donohue,et al.  Dysregulation of Semaphorin7A/β1-integrin signaling leads to defective GnRH-1 cell migration, abnormal gonadal development and altered fertility. , 2011, Human molecular genetics.

[88]  Renata Bartesaghi,et al.  Widespread Proliferation Impairment and Hypocellularity in the Cerebellum of Fetuses with Down Syndrome , 2011, Brain pathology.

[89]  Richard S. Frackowiak,et al.  Regional specificity of MRI contrast parameter changes in normal ageing revealed by voxel-based quantification (VBQ) , 2011, NeuroImage.

[90]  Pedagógia,et al.  Cross Sectional Study , 2019 .

[91]  Anne-Laure Schang,et al.  GnRH receptor gene expression in the developing rat hippocampus: transcriptional regulation and potential roles in neuronal plasticity. , 2011, Endocrinology.

[92]  R. Reeves,et al.  Increased male reproductive success in Ts65Dn “Down syndrome” mice , 2010, Mammalian Genome.

[93]  U. Boehm,et al.  Embryonic gonadotropin-releasing hormone signaling is necessary for maturation of the male reproductive axis , 2010, Proceedings of the National Academy of Sciences.

[94]  J. Finsterbusch,et al.  Optimization and Validation of Methods for Mapping of the Radiofrequency Transmit Field at 3T , 2010, Magnetic resonance in medicine.

[95]  Armen Saghatelyan,et al.  Interneurons Produced in Adulthood Are Required for the Normal Functioning of the Olfactory Bulb Network and for the Execution of Selected Olfactory Behaviors , 2009, The Journal of Neuroscience.

[96]  Y. Hérault,et al.  DYRK1A, a Novel Determinant of the Methionine-Homocysteine Cycle in Different Mouse Models Overexpressing this Down-Syndrome-Associated Kinase , 2009, PloS one.

[97]  O. Marín,et al.  The Embryonic Preoptic Area Is a Novel Source of Cortical GABAergic Interneurons , 2009, The Journal of Neuroscience.

[98]  P. Dechent,et al.  High‐resolution maps of magnetization transfer with inherent correction for RF inhomogeneity and T1 relaxation obtained from 3D FLASH MRI , 2008, Magnetic resonance in medicine.

[99]  Jacqueline N. Crawley,et al.  Unusual Repertoire of Vocalizations in the BTBR T+tf/J Mouse Model of Autism , 2008, PloS one.

[100]  W. Crowley,et al.  The relative role of gonadal sex steroids and gonadotropin-releasing hormone pulse frequency in the regulation of follicle-stimulating hormone secretion in men. , 2008, The Journal of clinical endocrinology and metabolism.

[101]  P. Dechent,et al.  Quantitative FLASH MRI at 3T using a rational approximation of the Ernst equation , 2008, Magnetic resonance in medicine.

[102]  L. Buée,et al.  P1–062: Alzheimer's disease–like tau neuropathology leads to memory deficits and loss of functional synapses in a novel mutated tau transgenic mouse without any motor deficits , 2006, The American journal of pathology.

[103]  Oliver Speck,et al.  Magnetic resonance imaging of freely moving objects: prospective real-time motion correction using an external optical motion tracking system , 2006, NeuroImage.

[104]  J. Carr Stability and change in cognitive ability over the life span: a comparison of populations with and without Down's syndrome. , 2005, Journal of intellectual disability research : JIDR.

[105]  L. Enquist,et al.  Olfactory Inputs to Hypothalamic Neurons Controlling Reproduction and Fertility , 2005, Cell.

[106]  Michael T. McManus,et al.  The RNaseIII enzyme Dicer is required for morphogenesis but not patterning of the vertebrate limb. , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[107]  J. Cummings,et al.  The Montreal Cognitive Assessment, MoCA: A Brief Screening Tool For Mild Cognitive Impairment , 2005, Journal of the American Geriatrics Society.

[108]  M. Freedman,et al.  Learning and memory as a function of age in Down syndrome: A study using animal-based tasks , 2005, Progress in Neuro-Psychopharmacology and Biological Psychiatry.

[109]  A. Delacourte,et al.  Progressive decrease of amyloid precursor protein carboxy terminal fragments (APP‐CTFs), associated with tau pathology stages, in Alzheimer's disease , 2002, Journal of neurochemistry.

[110]  C. Murphy,et al.  Olfactory impairment increases as a function of age in persons with Down syndrome , 2002, Neurobiology of Aging.

[111]  Thomas D. Schmittgen,et al.  Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. , 2001, Methods.

[112]  E. Boncinelli,et al.  The Otx2 homeoprotein regulates expression from the gonadotropin-releasing hormone proximal promoter. , 2000, Molecular endocrinology.

[113]  Yue Feng,et al.  Destabilization and Mislocalization of Myelin Basic Protein mRNAs in quaking Dysmyelination Lacking the QKI RNA-Binding Proteins , 2000, The Journal of Neuroscience.

[114]  G. Koob,et al.  Genetic Differences in Response to Novelty and Spatial Memory Using a Two-Trial Recognition Task in Mice , 2000, Neurobiology of Learning and Memory.

[115]  I. Kola,et al.  Down syndrome and mouse models. , 1998, Current opinion in genetics & development.

[116]  J. Ware,et al.  A 12-Item Short-Form Health Survey: construction of scales and preliminary tests of reliability and validity. , 1996, Medical care.

[117]  R. Kimmel,et al.  Hematopoietic Stem Cell Antigen‐1 (Sca‐1) Expression in Different Lymphoid Tissues of Female Mice Treated With GnRH Agonist , 1995 .

[118]  R. Bronson,et al.  A mouse model for Down syndrome exhibits learning and behaviour deficits , 1995, Nature Genetics.

[119]  E. Courchesne,et al.  Protocols to establish genotype-phenotype correlations in Down syndrome. , 1991, American journal of human genetics.

[120]  E. Knobil The GnRH pulse generator. , 1990, American journal of obstetrics and gynecology.

[121]  J. Haxby,et al.  Dementia in Down's syndrome , 1988, Neurology.

[122]  J. Opitz,et al.  Gonadal function in patients with Down syndrome. , 1987, American journal of medical genetics.

[123]  B. L. Shapiro,et al.  Down syndrome--a disruption of homeostasis. , 1983, American journal of medical genetics.

[124]  A. Hoffman,et al.  Induction of puberty in men by long-term pulsatile administration of low-dose gonadotropin-releasing hormone. , 1982, The New England journal of medicine.

[125]  E. Zimmerman,et al.  Brain grafts reverse hypogonadism of gonadotropin releasing hormone deficiency , 1982, Nature.

[126]  J. Beauvillain,et al.  Immunocytochemical demonstration of LH-RH, somatostatin, and ACTH-like peptide in osmium-postfixed, resin-embedded median eminence. , 1980, The journal of histochemistry and cytochemistry : official journal of the Histochemistry Society.

[127]  W. Cowan,et al.  The connections of the septal region in the rat , 1979, The Journal of comparative neurology.

[128]  T. Plant,et al.  Hypophysial responses to continuous and intermittent delivery of hypopthalamic gonadotropin-releasing hormone. , 1979, Science.

[129]  T. Plant,et al.  Hypophysial responses to continuous and intermittent delivery of hypopthalamic gonadotropin-releasing hormone , 1978 .

[130]  E. Renzi,et al.  The token test: A sensitive test to detect receptive disturbances in aphasics. , 1962, Brain : a journal of neurology.

[131]  Hyun-Jeong Yang,et al.  Brain education-based meditation for patients with hypertension and/or type 2 diabetes , 2019, Medicine.

[132]  Thomas R. Gingeras,et al.  STAR: ultrafast universal RNA-seq aligner , 2013, Bioinform..

[133]  R. D'Hooge,et al.  Progressive age-related cognitive decline in tau mice. , 2013, Journal of Alzheimer's disease : JAD.

[134]  F. Sherriff,et al.  Memantine for dementia. , 2005, The Cochrane database of systematic reviews.

[135]  Kazuhiro Ishii,et al.  [Down syndrome]. , 2003, Ryoikibetsu shokogun shirizu.

[136]  H. Burger,et al.  Induction of ovulation and fertility in amenorrheic women by pulsatile low-dose gonadotropin-releasing hormone , 1984 .

[137]  G. A. Moore,et al.  randomised double blind placebo controlled trial , 2022 .