Down syndrome: Neurobiological alterations and therapeutic targets

Down syndrome: Neurobiological alterations and therapeutic targets

Accepted Manuscript Title: Down syndrome: neurobiological alterations and therapeutic targets Authors: Rosa Anna Vacca, Sweta Bawari, Daniela Valenti,...

1MB Sizes 0 Downloads 54 Views

Accepted Manuscript Title: Down syndrome: neurobiological alterations and therapeutic targets Authors: Rosa Anna Vacca, Sweta Bawari, Daniela Valenti, Devesh Tewari, Seyed Fazel Nabavi, Samira Shirooie, Archana N. Sah, Mariateresa Volpicella, Nady Braidy, Seyed Mohammad Nabavi PII: DOI: Reference:

S0149-7634(18)30802-9 https://doi.org/10.1016/j.neubiorev.2019.01.001 NBR 3313

To appear in: Received date: Revised date: Accepted date:

17 October 2018 2 January 2019 2 January 2019

Please cite this article as: Vacca RA, Bawari S, Valenti D, Tewari D, Fazel Nabavi S, Shirooie S, Sah AN, Volpicella M, Braidy N, Nabavi SM, Down syndrome: neurobiological alterations and therapeutic targets, Neuroscience and Biobehavioral Reviews (2019), https://doi.org/10.1016/j.neubiorev.2019.01.001 This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Down syndrome: neurobiological alterations and therapeutic targets

Rosa Anna Vacca1*, Sweta Bawari2, Daniela Valenti1, Devesh Tewari2, Seyed Fazel Nabavi3, Samira Shirooie4, Archana N. Sah2, Mariateresa Volpicella5, Nady Braidy6, Seyed Mohammad

1

IP T

Nabavi3

Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Council of

2

SC R

Research, Bari, Italy;

Department of Pharmaceutical Sciences, Faculty of Technology, Kumaun University, Nainital,

India;

Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran,

U

3

Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Kermanshah University of

A

4

N

Iran;

M

Medical Sciences, Iran;

Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy;

6

Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Australia.

EP

*Correspondence at:

TE D

5

CC

Rosa Anna Vacca, Institute of Biomembranes, Bioenergetics and Molecular Biothecnology, IBIOM-CNR, Via Amendola 165/A, 70126 Bari, Italy;

A

e-mail: [email protected]

Highlights  Down syndrome is both a neurodevelopment and neurodegenerative disorder  Genetic and metabolic alterations involved in Down syndrome neuropathology are discussed  Drugs targeting altered genes and metabolic pathways for therapeutic challenge in DS are discussed 1

Abstract Down syndrome (DS) is a genetic disease that occurs due to an aneuploidy of human chromosome 21. Trisomy of chromosome 21 is a primary genetic cause of developmental abnormalities leading to cognitive and learning deficits. Impairments in GABAergic transmission, noradrenergic neuronal

IP T

loss, anomalous glutamatergic transmission and N-methyl-D-aspartate receptor signalling, mitochondrial dysfunction, increased oxidative stress and inflammation, differentially expressed

SC R

microRNAs, increased expression of crucial chromosome 21 genes, and DNA hyper-methylation and hyperactive homocysteine trans-sulfuration pathway, are common incongruities that have been reported in DS and might contribute to cognitive impairment and intellectual disability. This review

U

provides an update on metabolic and neurobiological alterations in DS. It also provides an overview

N

of the currently available pharmacological therapies that may influence and/or reverse these

M

A

alterations in DS.

TE D

Key words: chromosome 21 trisomy; neurodevelopment disease; neurotransmission; neurogenesis; mitochondrial dysfunction; neurodegeneration

EP

Abbreviations:

CC

Aβ, beta-amyloid; AD, Alzheimer’s disease; AMPK, 5' AMP-activated protein kinase; APP, amyloid precursor protein; BAX, BCL2-Associated X Protein; CAT, catalase; CBS, cystathionine

A

beta-synthase; CNS, central nervous system; CoQ10, Coenzyme Q10; Drp1, dynamin-related protein 1; DS, Down syndrome; DSCR, Down syndrome critical region; DYRK1A, dual-specificity tyrosine (Y)-phosphorylation regulated kinase 1A; EGCG, epigallocatechin-3-gallate; GABA, γaminobutyric acid; GIs, γ-secretase inhibitors; GPCR, G-protein coupled receptor; GPX, glutathione peroxidase; Hsa21, human chromosome 21; iPSCs, induced pluripotent stem cells; LTD, long-term depression; LTP, long-term potentiation; Mecp2, methyl-CpG binding protein 2; 2

miRNAs, microRNAs; Mnf2, mitofusin 2; MRC, mitochondrial respiratory chain; mTOR, mammalian target of rapamycin; NAM, negative allosteric modulator; NFAT, nuclear factor of activated T-cells; NKCC1, Na-K-Clcotransporter 1; NMDA, N-methyl-D- aspartate; NPCs, neural progenitor cells; NRIP1, nuclear receptor interacting protein 1; Opa1, optic atrophy 1; PKA, protein kinase; A; PiB, C-labeled Pittsburgh compound B; RCAN1, regulator of calcineurin 1; RIP140,

IP T

receptor-interacting protein 140; ROS, reactive oxygen species; SOD, superoxide dismutase; TSP-

SC R

1, thrombospondin 1.

Table of Contents 1. Introduction

Down syndrome

A

2.1. Impairments in neurogenesis

N

U

2. Alterations in metabolic and signalling pathways critical for the neuropathology of

M

2.1.1. Genetic deregulation

TE D

2.1.2. Epigenetic deregulation 2.1.3. Mitochondrial dysfunction 2.2. Impairments in neurotransmission

EP

2.1.1. GABAergic transmission

CC

2.1.2. Glutamatergic transmission 2.1.3. Other synaptic transmissions

A

2.3. Impairments in neuroplasticity 2.3.1. Astrocyte-mediated synaptic dysfunction 2.4 Neurodegeneration

3. Selected pharmacological interventions and their targets in Down syndrome 3.1.

Bumetanide and Basmisanil RG1662

3.2.

Fluorexetine 3

3.3.

Memantine

3.4.

Donepezil and β- and γ-secretase modulators

3.5.

Coenzyme Q10, metformin and melatonin

3.6

Naturally occurring phytochemicals

3.7.

Other dietary supplements

IP T

4. Concluding remarks and future prospective 5. Acknowledgments

A

CC

EP

TE D

M

A

N

U

SC R

6. References

4

1. Introduction Down syndrome (DS) is a genetic disorder that develops as a consequence of an aneuploidy of human chromosome 21 (Hsa21) (Antonarakis et al., 2004; Letourneau et al., 2014; Opitz and Gilbert-Barness, 1990; Ruparelia et al., 2010). The most frequent form of DS is a result of full Hsa21 trisomy, which is an outcome of the inability of Hsa21 to segregate during meiosis in a

IP T

developing ovum or, to a lesser extent, in sperm, culminating in an extra copy of the entire Hsa21 in all cell types. The mosaic form is rare and occurs in 3-4 % of DS population, in which some cells

SC R

within a single tissue type exhibit a normal karyotype while others exhibit a Hsa21 trisomy (Antonarakis, 2017; Asim et al., 2015; Rachidi and Lopes, 2008; Reeves et al., 2001; Sherman et al., 2007). The occurrence of partial Hsa21 trisomy leading to DS phenotype is extremely rare

N

U

(Pelleri et al., 2016).

A

The incidence of DS is estimated to be 1/750-800 live new-borns, but the risk of Hsa21 non-

M

disjunction increases with advanced maternal age (Loane et al., 2013; McKenzie et al., 2016;

TE D

Rudolf et al., 2017). Differences in the use of prenatal screening and pregnancy termination have led to a wide variation in live birth prevalence between countries (Morice et al., 2008; Rudolf et al., 2017). A recent epidemiological study on the prevalence of major birth defects in the live birth

EP

population in the United States between 2004 and 2006 reported that the estimated annual number

CC

of infants born each year with DS was 5,657 (Kirby, 2017). In Europe, a twenty-year study (19902009) showed that the total number of cases of infants born with DS was about 14,000 or about 700

A

per year (Loane et al., 2013). In comparison, approximately 21,000 infants are born with DS in India every year (Verma, 2000; Verma and Bijarnia, 2002). However, a prospective cohort-based study from an Indian tertiary health care centre between 2005 and 2010 reported a higher incidence of mortality (13 %, of which 80.3 % in children less than 2 years of age). These discrepancies may be due to poor family conditions and reporting errors from medical and allied healthcare professionals (Nahar et al., 2013). 5

While trisomy 21, as the genetic cause of DS, was first reported by Lejeune, Gautier and Turpin nearly 60 years ago (Lejeune et al., 1959), there has been a concerted effort to identify the pathogenic mechanism/s through which the trisomy 21 induces the clinical phenotype. Currently,

IP T

the genotype/phenotype correlation in DS remains unclear.

DS is considered the most prominent condition associated with neurodevelopmental abnormalities

SC R

which determine delay or failure in the acquisition of motor skills, speaking and reading with shortterm memory impairment and learning difficulties (for refs see the reviews (Roizen and Patterson, 2003; Sherman et al., 2007). The syndrome is characterized by neuropathological changes occurring

U

in the foetal and neonatal life that lead to alterations in brain development. Indeed, the most striking

N

hallmarks of DS phenotype are impairments of brain development and intellectual disability, as

A

well as craniofacial defects (Kazemi et al., 2016). Additionally, the brains from people with DS

M

have structural and functional abnormalities with developmental alterations in morphogenesis, such

TE D

as reduction in brain volume (including cerebral grey and white matter and cerebellum), and histogenesis, including cortical dysgenesis, delayed myelination, lower neuronal density and abnormal synaptic plasticity (Rachidi and Lopes, 2011). Almost all DS population develop in adult

EP

life neuropathological features leading to early ageing, senile dementia and neurological alterations

CC

consistent with the Alzheimer’s disease (AD) phenotype, including extracellular plaques and intracellular tangles (Head et al., 2016; Zis and Strydom, 2018). For these reasons, DS is often

A

considered a neurodegenerative disorder.

Apart from intellectual disabilities, a wide range of typical traits can be recognized in DS population that are associated with numerous comorbidities with a high phenotypic variation, but occurring with a higher frequency with respect to the euploid population. The most frequent DSassociated diseases are congenital cardiac defects, occurring in almost 50% of babies with DS 6

(Diamandopoulos and Green, 2018). This is followed by acute lymphoblastic leukaemia, occurring in children with DS that are aged less than 5 years old with a frequency of 1/300 and an incidence 40.7 times greater than that in individuals without DS at the same age (Chisholm, 2018). As well, gastro-intestinal diseases such as Hirschprung disease, affects about 2% of babies born with DS and constitutes about 12% of all cases of gastro-intestinal congenital malformations (Asim et al., 2015;

IP T

Holmes, 2014). Other common medical conditions in DS population are otolaryngologic and periodontal diseases, visual impairments, obesity, obstructive sleep apnea, increased susceptibility

SC R

to seizures, and respiratory diseases (Carfì et al., 2014; Pueschel, 1990; Roizen and Patterson, 2003). DS is also correlated with male infertility (Stefanidis et al., 2011). In addition, people with DS are highly vulnerable to auto-inflammatory diseases such as celiac disease, thyroiditis and

U

alopecia indicative of a chronic deregulation of the immune system (Sullivan et al., 2017). As well,

N

behavioural and psychological problems including attention deficit hyperactivity and autism

A

spectrum disorders (Davis et al., 2018; Määttä et al., 2006) in children, and neuropsychiatric

M

symptoms in adults, have been reported in people with DS (Dekker et al., 2018).

TE D

The last few decades have seen a tremendous increase in the life expectancy of patients with DS reaching an average age of 60 years (Bittles and Glasson, 2004; Carfì et al., 2014). This reflects the advancement in the field of medical research and simultaneously poses considerable challenges to

EP

the healthcare system in terms of catering to the plethora of phenotypic characteristics and other life

CC

threatening comorbidities that accompany DS in adulthood.

A

Intellectual disability invariably associated with DS, still remains a major challenge to manage by families and healthcare professionals involved in the care of people with DS. Every DS individual presents cognitive and learning deficits in a degree ranging from mild to severe. For example, in a DS population, analysis of 53 females (mean age of 35 years) and of 76 males (mean age of 29 years) resulted in intellectual disability scores of 19% mild, 30% moderate 33% severe and 18% profound, with a sex difference being females with better cognitive abilities and speech production 7

compared with males (Määttä et al., 2006). Of note, enriched environment and specific educational methods were shown to improve cognitive development and intellectual disability scores and biological response as suggested by results obtained in behavioural studies in both mouse models of DS, and DS children (Engevik et al., 2016; Martínez-Cué et al., 2005). However, adults with DS have a high risk for progressive cognitive decline and loss of acquired abilities (see the review

IP T

(Krinsky-McHale and Silverman, 2013).

SC R

Many theories have been proposed to explain DS-related abnormalities in the central nervous system (CNS), including the gene dosage effect, the amplified developmental instability, and the critical region hypotheses (Krinsky-McHale and Silverman, 2013). However, the case of

U

monozygotic twins with trisomy of Hsa21 but with discordant phenotypes (Grynberg et al., 2007),

N

and the case of subjects with DS phenotype but carrying a partial 21 trisomy of a very restricted

A

region coding for not already known genes (34 kb on distal 21q22) (Pelleri et al., 2016), have

M

highlighted the likely involvement of other mechanisms to explain the wide phenotypic variation

TE D

occurring in subjects with DS. For instance, epigenetic histone modification and DNA methylation, as well as microRNA regulation of gene expression have also been proposed to play a causal role in the aetiology of DS (Mentis, 2016). Transcriptome maps of human endothelial progenitor cells with

EP

Hsa21 trisomy obtained by massive–scale RNA-sequencing analysis have revealed up or down-

CC

regulation of transcripts outside the Hsa21 (Costa et al., 2011). More recently, a meta-analysis, comparing transcript expression levels and profiles of several human tissues and cells with trisomy

A

21 with their corresponding diploid, has confirmed the gene dosage hypothesis with 3:2 DS/normal ratio for Hsa21 genes, and listed genes mapped on other chromosomes differentially expressed (Pelleri et al., 2018). Mitochondrial dysfunction due to impairment of key regulatory processes (see the review Valenti et al., 2018), leading to a lower ATP production and deficits in total brain energy, as well as overproduction of reactive oxygen species (ROS) (Valenti et al., 2017, 2016, 2011, 2010), are also 8

believed to be involved in the pathogenesis of DS and in DS-related cognitive impairment. Free radical species, produced by dysfunctional mitochondria, can induce a progressive accumulation of oxidative damage in mitochondrial DNA, and endogenous defence mechanisms against oxidative stress are known to be impaired in DS (Druzhyna et al., 1998). This can in turn lead to early cellular aging and neurodegeneration. It is well established, that the efficiency of mitochondrial

IP T

bioenergetics and dynamics is fundamental for the neurobiological mechanisms underlying intellectual development (for refs see (Khacho and Slack, 2018; Valenti et al., 2014). Aberrant

SC R

mitochondrial energy metabolism and oxidative stress could result in the increased susceptibility of individuals with DS to a large spectrum of diseases such as AD, cardiomyopathy and autism spectrum disorders (Helguera et al., 2013).

Coskun et al. (2017) tested the hypothesis that

U

peripheral cells from elderly subjects with DS show dementia-specific and disease-specific

N

metabolic features. Using lymphoblastic-cell-lines derived from individuals with DS and DS-with-

A

dementia, the study showed that DS cells exhibited a slower growth rate under minimum feeding

M

and reduced expression of the autophagy marker LC3-II. Taken together, these findings underscore

TE D

the close relationship between metabolic dysfunction and impaired autophagy in DS (Coskun et al., 2017).

EP

Several mouse models have been generated in order to study genotype/phenotype correlations in DS and in particular, neurobiological alterations in DS brain (see Table 1). Ts65Dn was the first viable

CC

trisomy model (Reeves et al., 1995) and was widely used as a DS animal model given that it recapitulates the main phenotypic feature of DS ((Reeves et al., 1995). This model possesses a

A

major portion of mouse chromosome 16 (MMU16) carrying some of Hsa21 orthologues at its distal end translocated to the centromeric region of mouse chromosome 17 (MMU17). However, it should be mentioned that Ts65Dn, as well as other mouse models, is not trisomic for all Hsa21 orthologues, and is also trisomic for a number of non-Hsa21 orthologues. Therefore, the use of transgenic and trans-chromosomic mice as models to study DS are vigorously debated. Therefore, 9

other model systems, such as stem cell models have also been proposed to study alterations in neuronal development that occur in DS. For example, induced pluripotent stem cells (iPSCs), obtained from skin or blood cells can be genetically "reprogrammed" to assume a stem cell-like state via specific differentiation protocols, to first generate neural progenitor cells (NPCs), and then human neurons in vitro (Scudellari, 2016). These cells are considered an important tool to

IP T

recapitulate neurodevelopmental stages (Hibaoui et al., 2014) or to reproduce early stages of AD

SC R

type pathology in DS (Dashinimaev et al., 2017).

In this review we report an overview of selected preclinical and clinical studies on both mouse models of DS and humans. A comprehensive search for integrative reviews and original research

U

articles published up to December 2018 on relevant aspects of neurobiological alterations and

N

therapeutic targets in DS using the PubMed/Medline, ISI and clinicaltrials.gov online databases was

A

conducted. The current review begins with a description of critical Hsa21 genes and their targets, as

M

well as epigenetic DNA and protein modifications that are likely to be involved in DS phenotype.

TE D

We then discuss how alterations in regulatory signalling and metabolic pathways are critically involved in neurobiological abnormalities associated with DS. Finally, the therapeutic efficacy of selected pharmacological strategies, including naturally occurring phytochemicals to attenuate the

CC

EP

aberrant metabolic pathways associated with DS are also discussed.

2. Alterations in metabolic and signalling pathways critical for the neuropathology of Down

A

syndrome

The neuropathogenesis of DS occurs as a result of a combination of several factors leading to alterations in neuronal es including neurogenesis, neurotransmission and neuroplasticity. Mitochondrial dysfunction, oxidative stress and neuroinflammation, altered glucose metabolism, impairment of homocysteine metabolism, and altered proteostasis and secondary messenger

10

signalling pathways (Butterfield and Perluigi, 2018), are indicated as possible contributors in the impairments of both CNS formation and degeneration (Figure 1).

2.1. Impairments in neurogenesis During foetal development, neural stem cells proliferate and differentiate into neurons in a process

IP T

called neurogenesis (Kitamura et al., 2009). Notably, in the hippocampus, neurogenesis occurs postnatally and continues throughout life (called adult neurogenesis) as an adaptive response to

SC R

regulate brain plasticity and memory (Spalding et al., 2013). Reduced neurogenesis is considered among the major neurodevelopmental defects leading to cognitive disability in DS. Results obtained using brains of individuals with DS, DS-derived iPSCs, and NPCs from the hippocampus of DS

U

mouse models as experimental systems, showed a decline in proliferation potency, impaired

N

neuronal maturation, and neural cell death, which occurred concurrently with alterations to

A

neurogenesis in DS (Gimeno et al., 2014; Hibaoui et al., 2014). Data obtained from immunostaining

M

experiments using cell proliferation protein markers have shown a reduction in proliferation

TE D

potency in the hippocampal dentate gyrus and neocortical germinal matrix in brains of foetuses with DS starting in the early gestational age (17-23 weeks) (Contestabile et al., 2007; Guidi et al., 2008). The dentate gyrus has a key role in cognition and memory acting as a pre-processor of incoming

EP

information, which is subsequently processed in the granule cells and pyramidal neurons of the

CC

hippocampal CA3 (Miyata et al., 2017). A reduction in cell proliferation was also shown in vitro both in neurospheres obtained from the frontal cortex of DS foetuses (Lu et al., 2012) and in NPCs

A

obtained from the dentate gyrus of the hippocampus of Ts65Dn mouse model (Valenti et al., 2016). The reduced proliferation potency of neural precursor cells in the DS brain is accompanied by impairments in neuro-differentiation. Immunohistochemical quantification of the number of mature neurons (NeuN-positive cells) and astrocytes (GFAP-positive cells) in both the DG of hippocampus of Ts65Dn mice and hippocampal and para-hippocampal regions of foetuses with Hsa21 trisomy, showed a reduction in the number of neurons and an increase in the number of astrocytes with 11

respect to diploidy (Guidi et al., 2008). Consistently, analysis of iPSCs derived from monozygotic twins discordant for trisomy 21 showed that DS iPSCs exhibit a reduced number of neurons and a shift from neuronal to astroglial and oligodendroglial differentiation. Additionally, neurons also displayed reduced length of neurites (Hibaoui et al., 2014). The reduction in both neural proliferation and differentiation in several brain regions indicates that the impairment in

IP T

neurogenesis is an early event in DS.

SC R

Several mechanisms involved in defective neuronal proliferation and differentiation in DS have been evaluated using mouse and cellular models of DS (Stagni et al., 2018). These include overexpression of some Hsa21 gene products, as well as epigenetic alterations. Herein, we report

U

evidence supporting also the crucial role played by mitochondrial dysfunction in the deregulation of

M

2.1.1. Genetic deregulation

A

N

neurogenesis in DS.

TE D

The ‘gene-dosage effect’ hypothesis i.e. the 1.5-fold increase of Hsa21 genes, due to an extra copy of Hsa21, has been first suggested to be responsible for the metabolic alterations leading neurological and cognitive impairments in people with DS (Delabar et al., 1993). Importantly,

EP

genes on Hsa21 encode many transcription factors or regulatory proteins that induce a secondary

CC

genome-wide transcriptional deregulation, resulting in down-or up-regulation of crucial genes on both Hsa21 and other chromosomes involved in impaired neurogenesis in DS (Rachidi and Lopes,

A

2011).

The dual-specificity tyrosine (Y)-phosphorylation regulated kinase 1A (DYRK1A) is thought to be a crucial Hsa21 gene strongly implicated in various DS phenotypes as demonstrated in TgDYRK1A transgenic mice over-expressing DYRK1A (Altafaj et al., 2001). Indeed, hyperactivity of DYRK1A protein is known to be involved in the derangement of excitatory-inhibitory balance, generation of 12

neurodevelopmental abnormalities and other behavioural phenotypes of DS associated with cognitive deficits (Ruiz-Mejias et al., 2016). DYRK1A is also associated with regulation of synaptic plasticity and memory consolidation. Amplified gene dosage resulted in defects of brain morphogenesis, low levels of BDNF and mnemonic deficits in DS mice (Guedj et al., 2009). Overexpression of DYRK1A has also been found in the brain of foetuses and adults with DS

IP T

(Lockstone et al., 2007) and in DS iPSCs-derived NPCs (Hibaoui et al., 2014). DYRK1A is present in both the nucleus and cytoplasm of mammalian cells (Figure 2), and appears to have a crucial role

SC R

during neurogenesis and CNS development. Genome-wide analysis of DYRK1A-associated loci reveals that in the nucleus this kinase is recruited to proximal promoters of growth-related genes, actively transcribed by RNA polymerase II, involved in different cellular processes such as

U

translation, RNA processing and cell cycle (Di Vona et al., 2015). In addition, DYRK1A

N

phosphorylates multiple targets in both the nucleus and the cytoplasm, including among others

A

cyclin L2 and cycline D1 (both are involved in cell cycle regulation and neurogenesis), α-synuclein

M

(presynaptic regulation), dynamin 1 (which regulates the endocytosis and apoptotic signalling),

TE D

APP, Tau (both involved in AD), synaptojanin 1 (for synaptic transmission), and the cAMP response element binding (CREB), (which regulates diverse cellular responses), as extensively reviewed in (Becker et al., 2014; Duchon and Herault, 2016; Stagni et al., 2018). In particular, it has

EP

been reported that DYRK1A phosphorylates cyclin D1 at Thr286 inducing its degradation, which in

CC

turn determines alterations in cell cycle and negative regulation of neural progenitor cell proliferation (Nakano-Kobayashi et al., 2017). An increase in the DYRK1A gene dosage also

A

induces elongation of both G1 and S phases. This correlated with a transient increase in the population of intermediate progenitor neurons and overall reduction in the total neuronal output of the postnatal brain (Smith and Calegari, 2015). It is well established that DYRK1A also phosphorylates p53, which leads to induction of p53 target genes and impaired G1/G0-S phase transition, resulting in a reduced proliferation (Park et al., 2010).

13

Inhibition of DYRK1A activity by the naturally occurring polyphenol, epigallocatechin-3-gallate (EGCG) ameliorated cognitive behavioural phenotypes and excitatory-inhibitory equilibrium in a DS mouse model (de la Torre and Dierssen, 2012; Guedj et al., 2009; Souchet et al., 2015), highlighting the involvement of DYRK1A gene over-dosage in the regulation of neurogenesis and cognitive impairment in DS (Stagni et al., 2015). A synthetic compound 3-(4-fluorophenyl)-5-(3,4which

is

a

fluoric

derivative

of

3,5-

IP T

dihydroxyphenyl)-1H-pyrrolo[2,3-b]pyridine,

di(polyhydroxyaryl)-7-azaindole named F-DANDY, has been reported to selectively inhibit

SC R

DYRK1A both in vitro and in vivo (in Ts65Dn mice), improving learning and memory in Ts65Dn mice (Neumann et al., 2018). Leucettine 41, a synthetic congener of Leucettamine B alkaloid derivative, has also been shown to normalize overexpressed DYRK1A in Ts65Dn, Tg(Dyrk1a) and

U

Dp1Yey mouse models of DS, by selectively inhibiting DYRK1A, thus improving cognitive

N

function (Nguyen et al., 2018). As well, a synthetic compound named ALGERNON (altered

A

generation of neurons) was found to inhibit DYRK1A in cultured neural stem cells from Ts65Dn

M

mice as well as in vivo in Ts65Dn mice, thereby enhancing proliferation in neural stem cells and

TE D

restoring cognitive function in Ts65Dn mice (Nakano-Kobayashi et al., 2017).

The regulator of calcineurin 1 (RCAN1), also known as DS critical region 1 protein (DSCR1), is an

EP

endogenous inhibitor of the calcineurin phosphatase and is overexpressed in DS (Fuentes et al.,

CC

2000). It has been reported to play an important role in DS neurodevelopmental deficits impairing neurotrophin trafficking and inducing alterations in the development of the sympathetic nervous

A

system (Patel et al., 2015), in Dp(16)1Yey/+ mice, a mouse model of DS trisomic solely for the human 21q11-q22.3 synthetic region (Table 1) (T. Yu et al., 2010). In TgRCAN1 mice overexpressing the sole RCAN1 gene, the density of dendritic spines on hippocampal pyramidal neurons was reduced and accompanied by a failure to maintain long-term potentiation, strongly suggesting that RCAN1 plays an important role in brain development and its up-regulation likely contributes to the neural deficits associated with DS (Martin et al., 2012). Studies in RCAN1- and 14

DYRK1A–overexpressing mice have shown that DYRK1A, which phosphorylates RCAN1 at Ser112 and Thr192 residues, acts synergistically with RCAN1 to regulate the phosphorylation levels of the Nuclear factor of activated T-cells (NFAT). This transcription factor, after translocation into the nucleus, up-regulates the expression of interleukin 2, with a consequent stimulation of growth and differentiation of T cell response (Arron et al., 2006; Jung et al., 2011).

IP T

NFAT also regulates proliferation and differentiation of neural precursor cells; it has been reported that NFAT activation reduces the percentage of cells in G0/1 phase of the cell cycle and causes cell

SC R

cycle elongation (Serrano-Pérez et al., 2015). Altogether these studies have suggested an important role of DYRK1A/RCAN1/NFAT signalling pathways in the regulation of NPC proliferation (Stagni

U

et al., 2018).

N

Less studied regarding deficit of neurogenesis in DS is the Hsa21-encoded receptor-interacting

A

protein 140 (RIP140), whose murine orthologous protein is named nuclear receptor interacting

M

protein 1 (NRIP1). NRIP1, through the interaction of nuclear receptor factors, regulates gene

TE D

expression and controls several metabolic processes in mouse (Fritah et al., 2010). Increased expression of the RIP140 protein has been demonstrated in human hippocampus of people with DS exhibiting strong cognitive disabilities (Gardiner, 2006). Knock-out of NRIP1 in mice results in

EP

learning and memory deficits, as well as an increased stress response, suggesting an involvement of

CC

this Hsa21 gene in cognitive function (Duclot et al., 2012). Recently Zhao and co-workers have reported that RIP140 participates in the differentiation of stem cells into neuronal progenitor cells

A

through a mechanism mediated by the ERK1/2 pathway (Zhao et al., 2017). Thus, ERK1/2 signalling may provide the molecular basis by which RIP140 participates in neural differentiation.

Cystathionine beta synthase (CBS) is another Hsa21-encoded crucial enzyme that might be involved in altered neurogenesis in DS. It is involved in homocysteine metabolism (Chen et al., 2010). Homocysteine is a mediator of two crucial biochemical processes; activated methyl cycle, 15

and trans-sulfuration pathway (Medina et al., 2001). Its significance can be deduced from the fact that alterations in homocysteine levels is implicated in various diseases including neurological disorders (Blom and Smulders, 2011; Miller, 2003; Schalinske and Smazal, 2012), AD (Chen et al., 2010), and congenital abnormalities, such as neural tube defects, cleft palate and congenital heart

IP T

disease (Blom and Smulders, 2011; Škovierová et al., 2016).

Numerous studies have reported significantly diminished plasma homocysteine levels in individuals

SC R

with DS due to excessive CBS-mediated hyperactivity of homocysteine trans-sulfuration pathway (Gane, B and Bhat, V, 2014; Obeid et al., 2012; Pogribna et al., 2001). The hyperactive transsulfuration pathway also accounts for a decline in tetrahydrofolate generation from 5-

U

methyltetrahydrofolate resulting in a deficit of functional folate, which is indispensable for de novo

N

synthesis of RNA and DNA (Gane, B and Bhat, V, 2014; Pogribna et al., 2001; Song et al., 2015).

A

Interestingly, another study showed how CBS impairment plays a crucial role in both

M

neurodevelopmental abnormalities and early neurodegeneration in DS. In that study, analysis of

TE D

CBS-positive neurons and astrocytes in cerebelli from foetuses and adults with DS showed a high level of CBS in granular cell layers during foetal development which correlated to an abnormal number of inhibitory neurons; increased CBS in the adult DS brain have been related to AD-like

EP

dementia (Kanaumi et al., 2006). A study carried out on Ts1Yah mouse model to evaluate the effect

CC

on DS etiology of the trisomy of genes located in a subtelomeric portion of Hsa21 (namely Abcg1– U2af1), containing CBS, reported that overexpression of the hippocampal CBS gene was associated

A

with improved spatial memory and enhanced long term potentiation (Pereira et al., 2009). Another study, carried out on Ms2Yah mouse model to determine the effect of the monosomy of the genes in the same Abcg1-U2af1 region, reported more improved effects on long-term potentiation and memory (Sahun et al., 2014).

16

As well, several microRNA (miRNA) are also overexpressed in some brain regions in people with DS and are proposed to contribute to neurodevelopmental deficits. It is well established that miRNAs are 18-24 nucleotide long non-coding RNAs and post-transcriptional regulators of gene expression acting by either destabilizing mRNAs or by repressing translation, thus regulating various physiological processes, such as development, proliferation, differentiation and apoptosis

IP T

(Cannell et al., 2008; Catalanotto et al., 2016; Kamhieh-Milz et al., 2014). Hsa21 harbours at least 29 miRNAs, but only five, namely miR-99a, let-7c, miR-125b-2, miR-155, and miR-802 have been

SC R

thoroughly studied and found implicated in DS pathogenesis, as reviewed in (Brás et al., 2018; Mentis, 2016; Sanchez-Mut et al., 2016). Interestingly, miR-155 and miR-802 directly downregulate the epigenetic modulator methyl-CpG binding protein 2 (Mecp2) (Bofill-De Ros et al.,

U

2015), and are implicated in another neurodevelopmental disorder, known as Rett syndrome (De

N

Filippis et al., 2015; Liyanage and Rastegar, 2014). Overexpression of miR-155 in the brain of

A

people with DS also results in the down-regulation of SNX27, a key regulatory protein assuring the

M

glutamate receptor recycling whose loss contributes to synaptic dysfunction in DS (Wang et al.,

TE D

2013). Similarly, differentially expressed miRNAs, specifically miR-138 along with its target zeste homolog 2 (EZH2), were also reported in DS foetal hippocampus, indicating their role in

EP

neurological manifestations of DS (Shi et al., 2016).

CC

2.1.2 Epigenetic deregulation Over-expression of many Hsa21 genes causes dysregulation of some epigenetic mechanisms in DS

A

including DNA methylation, histone modification and chromatin remodelling, and are reported to play a crucial role in learning and memory defects in DS (for a comprehensive review see (Dekker et al., 2014). DNA methylation serves as a major epigenetic modulator of gene expression (Crider et al., 2012; Moore et al., 2013). Incongruity in DNA methylation is an important causal factor in the development of neural tube defects (Blom and Smulders, 2011; Škovierová et al., 2016), and neurological (Moore et al., 2013) disorders. DNA methylation influences multiple aspects of 17

neurogenesis from stem cell maintenance and proliferation, followed by neuronal differentiation and maturation, and synaptogenesis; mutations in proteins involved in the establishment, maintenance and removal of DNA methylation impact brain development and functions (Jobe and Zhao, 2017).

IP T

Anomalous DNA methylation is a common occurrence in trisomy 21. Hyper-methylation of DNA is association with upregulation of DNA methyltransferase (DNMT) genes DNMT3B and DNMT3L

SC R

and downregulation of TET2 and TET3 demethylation genes have also been reported from a study using skin fibroblasts isolated from DS monozygotic twins (Sailani et al., 2015). Several other genes such as TCF7, SH3BP2, CD3Z, NOD2 TMEM131, EIF4E, SUMO3, CPT1B, NPDC1 and

U

PLD6 from blood-derived leukocytes and T-lymphocytes of DS individuals exhibited differential

N

methylation (Kerkel et al., 2010). Jin and co-workers proposed a role of epigenetic mechanisms in

A

the plethora of phenotypic outcomes of trisomy 21. They also showed the presence of global DNA

M

hyper-methylation in placental villi cells of DS foetuses and leukocytes from DS adults (Jin et al.,

TE D

2013). Hyper-methylated lymphocytic DNA has also been reported in children with DS (Pogribna et al., 2001). As previously discussed, it should be highlighted that repression of the demethylase Mecp2, secondary to trisomic overexpression of Hsa21-derived miRNAs miR-155 and miR802

EP

(Bofill-De Ros et al., 2015), may partly contribute to the hyper-methylation and neurochemical

CC

alterations observed in the brains of DS individuals. hypo-methylation of mitochondrial DNA has also been reported in DS due to a mitochondrial methyl imbalance linked to a reduction in S-

A

adenosyl-methionine synthesis (Infantino et al., 2011).

Histone deacetylation and acetylation are other epigenetic mechanisms influenced by some Hsa21 genes. DYRK1A, for example, activates SIRT1 phosphorylation and promotes histone deacetylation (Guo et al., 2010); on the other hand, DYRK1A interacts with NRSF/REST affecting chromatin re-

18

modeling and deregulates the levels of gene clusters essential for neural differentiation leading to neuronal alterations in DS (Lepagnol-Bestel et al., 2009).

2.1.3. Mitochondrial dysfunction Mitochondrial metabolism strongly influences the distinct developmental steps of adult

IP T

neurogenesis (Beckervordersandforth, 2017). Several recent studies reported that activation of mitochondrial biogenesis and function induces a metabolic shift from glycolysis to oxidative

SC R

phosphorylation, which mediates the cell fate decision to start the neurogenesis process inducing neural stem cells to generate actively proliferating intermediate progenitor cells which differentiate into mature neurons. Disturbances in mitochondrial function and signalling lead to changes in the

U

cell fate and negatively affect the neurogenesis and neuroplasticity processes (for refs see the

A

N

reviews Arrázola et al., 2018; Beckervordersandforth, 2017; Khacho et al., 2019).

M

In this light, we have already reviewed and discussed how deficits in neural energy due to

TE D

mitochondrial dysfunction is critical for the aetiology of DS (Valenti et al., 2018, 2014). Mitochondrial dysfunction is an early event in DS and is even considered to be responsible for nondisjunctional disabilities of chromosomes during meiosis in embryos that results in aneuploidies

EP

like DS (Coskun and Busciglio, 2012; Eichenlaub-Ritter et al., 2011; Schon et al., 2000). The

CC

presence of significantly high ROS and disrupted mitochondrial network has been reported in DS human fibroblasts as well as mouse embryonic fibroblasts (Zamponi et al., 2018).

A

Oligonucleotide microarray analysis profiling of the transcriptome of Hsa21 genes of DS foetal hearts showed downregulation of mitochondrial genes specifically those encoding for mitochondrial enzymes (Conti et al., 2007). Similar downregulation of mitochondrial genes along with mitochondrial morphological alterations and respiratory activity aberrations were also observed in DS foetal fibroblasts and neural progenitor cells (Izzo et al., 2017b; Piccoli et al., 2013; Valenti et al., 2017). Accordingly, a recent systematic proteome and proteostasis profiling study in human DS 19

fibroblasts revealed significant downregulation of the mitochondrial proteome, which strongly concurs to the DS phenotype (Liu et al., 2017). However, one study proposed that downregulation of mitochondrial function could be an adaptive mechanism to prevent ROS generation and subsequent cellular damage (Helguera et al., 2013).

IP T

Mitochondrial phenotype in DS is characterized by reduced efficiency to produce ATP through oxidative phosphorylation, decreased respiratory capacity, impaired ability to generate

SC R

mitochondrial membrane potential, as well as alterations in mitochondrial structure and dynamics (see the review (Valenti et al., 2018). The molecular bases for mitochondrial dysfunction and cell energy deficiency in DS have been well studied. An overview of the Hsa21 genes and key

U

regulatory signalling pathways involved in mitochondrial dysfunctions in DS have been recently

N

reviewed (Izzo et al., 2018; Valenti et al., 2018). In particular, down-regulation of PGC-

A

1α/Sirt1/AMPK axis is involved in the impairment of mitochondrial biogenesis (Piccoli et al., 2013;

M

Valenti et al., 2016). Alterations to the expression of dynamin-1-like protein (Drp1), mitofusin 2

TE D

(Mnf2) and optic atrophy 1 (Opa1) has been associated with alterations in mitochondrial structure and network (Izzo et al., 2017b; Valenti et al., 2017). In addition, a decrease in cAMP levels and protein kinase A (PKA)-mediated phosphorylation of the mitochondrial respiratory chain (MRC) in

EP

DS lead to a reduced catalytic efficiency of MRC complex I and ATP synthase with a consequent

CC

decline in ATP production, reduced cellular energy, an increase in ROS produced by the mitochondria and oxidative stress (Valenti et al., 2011, 2010). Overexpression of RCAN1 in TG

A

mice has been shown to promote mitochondrial dysfunction (Wong et al., 2015) and causes DS-like immune dysfunction as shown by Martin and co-workers (Martin et al., 2013). In addition, the miRNAs let-7c and miR-155, have been reported to target genes involved in the regulation of mitochondrial function (Izzo et al., 2017a). Finally, overexpression of the Hsa21-encoded Ets-2 induces translocation of cytochrome c by mitochondria and induction of mitochondrial death pathway leading to degeneration of DS cortical neurons (Helguera, 2005). 20

Repair mechanisms against oxidative stress mediated damage to mitochondrial DNA are also known to be compromised in DS (Druzhyna et al., 1998). Recently, alteration in mitochondrial dynamics and bioenergetics due to deregulation of Drp1 expression and activity has also been shown to impair hippocampal neurogenesis in Ts65Dn mice (Valenti et al., 2017) and the rescue of

IP T

mitochondrial functions restores normal proliferation of NPCs from the hippocampus of Ts65Dn mice (Valenti et al., 2016). This data confirms the hypothesis of Beckervordersandforth that

SC R

mitochondrial metabolism strongly regulates adult neurogenesis (Beckervordersandforth, 2017).

2.2. Impairments in neurotransmission

U

The interplay of both inhibitory and excitatory neurotransmitters plays a major role in cognition and

N

neurodevelopmental processes (Cohen Kadosh et al., 2015). Derangement in this excitatory-

A

inhibitory equilibrium has long been implicated in DS-associated neuronal impairments (Créau,

M

2012). Herein we review and discuss the molecular mechanisms of the changes in the main

TE D

neurotransmission processes in DS (see Figure 2).

2.2.1. GABAergic transmission

EP

The main inhibitory neurotransmitter in humans, γ-aminobutyric acid (GABA), acts by two

CC

receptors families, GABAARs and GABABRs. GABAARs are ligand-gated chloride channels composed of a combination of several subunit subtypes (Olsen and Sieghart, 2009). GABABR is a

A

class of G-protein coupled receptor (GPCR) that associates with a subset of G-proteins controlling specific ion channels (Padgett and Slesinger, 2010) GABABR is composed by three different subunits reported to modulate both synaptic transmission and postsynaptic responses by a direct interaction of Ca2+ and K+ channels, respectively (Pinard et al., 2010).

21

In postnatal development or in axonal compartments, GABAAR activation induces membrane depolarization due to a higher intracellular Cl- concentration (Zorrilla de San Martin et al., 2018). In adult neurons, GABAAR becomes hyperpolarized due to a change in the Cl- balance to lower intracellular Cl- concentration and exerts an inhibitory function (see the review (Herbison and Moenter, 2011). Interestingly, recent studies have shown a reversal of function of GABA AR

IP T

signalling in the hippocampus of Ts65Dn mice which was found to be excitatory rather than inhibitory, generated by an inversion in the direction of Cl- currents due to an increase in the

SC R

expression and activity of the Cl- cotransporter NKCC1 (Contestabile et al., 2017; Deidda et al., 2015).

U

Numerous studies on GABAergic transmission in DS have been performed in mouse models and in

N

Ts65Dn mice where GABA transmission in neurons was found to be increased. Alterations in the

A

KCNJ6 gene, which encodes the subunit 2 of the GIRK channel (GIRK2/Kir3.2), and maps to

M

Hsa21, provided the first direct association between DS and GABAergic transmission (Hattori et

TE D

al., 2000; Ohira et al., 1997). An extra KCNJ6 copy led to increased mRNA and protein expression of GIRK2 in the hippocampus, cortex and midbrain of Ts65Dn mice (Harashima et al., 2006). Subsequently, GABAB/GIRK currents were shown to increase in cultured primary hippocampal

EP

neurons (Best et al., 2007), CA1 pyramidal neurons and dentate gyrus granule cells from acute

CC

slices of the Ts65Dn hippocampus (Best et al., 2012; Kleschevnikov et al., 2012). This increase in neuronal GABAB/GIRK signalling in Ts65Dn mice is likely to enhance GIRK-mediated shunting

A

inhibition, and reduces excitatory PSPs, and alter neuronal passive properties which is likely to affect neuronal excitability and plasticity (Koyrakh, 2005; Lüscher et al., 1997).

Similarly, genetic restoration of GIRK2 to disomy in Ts65Dn mice attenuated elevations in GABAB-triggered currents in CA1 pyramidal neurons (Joshi et al., 2016) and dentate gyrus granule cells (Best et al., 2012; Kleschevnikov et al., 2012). 22

Association of GABAergic over-inhibition and cognitive and learning deficits in DS was also demonstrated in a study carried out by Fernandez et al., 2007. The study showed improvement in cognition and long term potentiation (LTP) of Ts65Dn mice on chronic treatment with the GABAA antagonists Picrotoxin (4 weeks) and Pentylenetetrazole (1 year), respectively) (Fernandez et al., 2007). Similar improvements in learning and memory of Ts65Dn mice was also reported following

IP T

administration of selective inverse agonists for α5- GABAA receptor subtype (Braudeau et al., 2011).

SC R

Of note, a discrepancy between the increase in GABA transmission in mouse models and a reduction in GABA concentrations in human foetal brains with DS (Whittle et al., 2007) and in brains of children with DS as measured by magnetic resonance imaging (Śmigielska-Kuzia et al.,

N

U

2010) has been previously reported .

A

GABAergic dysfunction disrupts the optimal excitatory/inhibitory synaptic balance in DS

M

depolarizing GABA transmission and leading to impairments in synaptic plasticity and learning and

TE D

memory deficits in DS (Deidda et al., 2014; Fernandez et al., 2007; Souchet et al., 2015; Zorrilla de San Martin et al., 2018). In line with these findings, enhanced excitability and reduced GABA inhibition has also been reported in cerebellar granule cells from Ts65Dn mice (Das et al., 2013;

EP

Szemes et al., 2013; Usowicz and Garden, 2012). Treatment with the α5-containing GABAAR

CC

negative modulator RO4938581, but not bumetanide, ameliorated hyperactivity in Ts65Dn mice (Deidda et al., 2015; Martinez-Cue et al., 2013). This suggests that additional mechanisms apart

A

from altered GABA signalling may be associated with the hyperactive phenotypes in Ts65Dn mice. For example, treatment with the GluN2B-selective antagonist Ro25–6981 (Hanson et al., 2013) or EGCG, an inhibitor of the DS triplicated kinase DYRK1A (Xie et al., 2008), the monoacylglycerol lipase inhibitor JZL184 (Lysenko et al., 2014), the neuro-hormone Melatonin (Corrales et al., 2013), the Sonic Hedgehog agonist SAG1.1 (Das et al., 2013), and the selective serotonin reuptake inhibitor fluoxetine (Begenisic et al., 2014; Bianchi et al., 2010; Guidi et al., 2014), have each been 23

shown to restore CA3-CA1 LTP and/or behavioural performances in vulnerable Ts65Dn mice. GABAergic synaptic transmission has also been shown to be modulated by the Akt-mTOR pathway, which has been reported to be hyper-activated in the hippocampus of Ts1Cje mice, a murine model of DS, leading to an increase in local dendritic translation and synaptic plasticity

IP T

impairment (Troca-Marín et al., 2014).

2.2.2. Glutamatergic transmission

SC R

Apart from alterations in GABAergic signalling, anomalous glutamatergic transmission and Nmethyl-D- aspartate receptor (NMDAR) signalling have been reported in the pathobiology of DS. For example, studies have reported delayed development and reduced production of excitatory

U

glutamatergic neurons in the cortex of Ts65Dn mice (Chakrabarti et al., 2010, 2007; Guidi et al.,

N

2014; Tyler and Haydar, 2013). As well, a lower density of glutamatergic synapses was observed

A

using electron microscopy in the cortex and hippocampus of Ts65Dn mice (Ayberk Kurt et al.,

M

2004; Chakrabarti et al., 2007; García-Cerro et al., 2014; Guidi et al., 2013; Kurt et al., 2000; Rueda

Weick et al., 2013).

TE D

et al., 2010; Stagni et al., 2013), and in human DS iPSCs-derived neurons (Hibaoui et al., 2014; Despite these findings, electrophysiological studies that investigate the

EP

involvement of glutamatergic signalling in trisomic mice are nascent in the current literature.

CC

Impaired glutamatergic transmission has been proposed to account for behavioural disability and learning and memory deficits in both individuals with DS and animal models (Boada et al., 2012;

A

Kaur et al., 2014). This has been shown in the Ts2 mouse model for DS, having in triplicate a short Hsa21 region containing APP (Brault et al., 2015), which exhibited a shortage in the levels of glutamate in the hippocampus (Kaur et al., 2014). The reduced glutamate levels resulted in decreased efficiency in nest building, object finding and entering alternate arms of a Y-maze (Kaur et al., 2014). A failure in systemic glutamate uptake, which has been reported to be independent of age has been demonstrated in platelets and fibroblasts from DS individuals (Begni et al., 2003). 24

Deficits in glutamate levels in the hippocampal region (Reynolds and Warner, 1988) and reduction of glutamate, norepinephrine and serotonin levels have also been demonstrated in para-hippocampal gyrus of post-mortem tissue samples from the human DS brain (Risser et al., 1997). This disparity in inherent glutamate uptake in DS is attributed to oxidative stress, as a result of mitochondrial dysfunction, and has also been correlated to Hsa21 gene products and overexpression of the Hsa21-

IP T

encoded proteins APP and superoxide dismutase 1 (SOD1), which are known to further exacerbate

SC R

neurodegeneration and intellectual disability in people with DS (Begni et al., 2003).

2.2.3. Other synaptic transmissions

DS is also associated with progressive noradrenergic neuronal loss in locus coeruleus. Locus

U

coeruleus is built by noradrenergic neurons located in the brainstem, in close proximity to the fourth

N

ventricle (Aston-Jones et al., 1994). It directs noradrenergic transmission to the majority of brain

A

regions involved in cognitive function (Phillips et al., 2016). Noradrenergic loss has been shown to

M

propagate cholinergic degeneration, neuroinflammation and memory loss in Ts65Dn mice

TE D

(Lockrow et al., 2011). Restoration of noradrenergic neurotransmission using a prodrug for norepinephrine or a β-adrenergic receptor agonist, reversed cognitive dysfunction in Ts65Dn mouse model of DS (Salehi et al., 2009). Phillips and colleague demonstrated that low levels of

EP

noradrenaline and impairment of noradrenergic transmission correlated with the occurrence of

CC

neurodegeneration and dementia in adults with DS. Indeed, detection of low levels of noradrenaline has been found in the cortical and subcortical regions of the postmortem brain of DS people with

A

dementia, and in particular, the occurrence of low serum levels of the noradrenaline metabolite 3methoxy-4-hydroxy phenylglycol (MHPG), increased the risk of developing dementia in DS adults by ten-fold (Phillips et al., 2016).

A number of studies have demonstrated disturbances in the cholinergic system in DS, and this may be likely due to alteration in acetylcholine metabolism with possibly significant relationships to 25

AD-like symptoms in DS adult, since impaired acetylcholine metabolism has been reported in the AD brain (Carvajal and Inestrosa, 2011). Reduction in the cholinergic neurotransmitters choline acetyltransferase has also been reported in cortical and sub-cortical regions of DS adult brain tissues when evaluated in frozen post mortem brain tissues of people with DS (Godridge et al., 1987). In Ts65Dn mice, basal forebrain cholinergic neurons degenerate with age and may contribute to

IP T

memory impairments (Chang and Gold, 2008; Granholm et al., 2000). Additionally, choline acetyltransferase activity was increased in the cortex and hippocampus and may likely compensate

SC R

for the reduction in the number of cholinergic neurons (Contestabile et al., 2006).

It has been well established that the serotonergic system is impaired in DS (Bar-Peled et al., 1991;

U

Whittle et al., 2007). The neuromodulator serotonin (5-hydroxytryptamine, 5-HT) is central for

N

regulation of a wide range of physiological processes in the CNS such as neurogenesis, neuronal

A

morphology and synaptogenesis (see the review (Daubert and Condron, 2010), and has been

M

implicated in the regulation of mood, cognitive processes and mediating the release of hormones

TE D

(Hoyer et al., 1994). Thirteen diverse 5‐HT receptors have been identified, twelve of which belong to the GPCR superfamily (Van Oekelen et al., 2003). In particular, analysis of the serotonin 5HT1A receptor in DS brains showed that its levels increase during early development but decreases

EP

below normal levels at the birth (Bar-Peled et al., 1991). In addition, reduced serotonin levels are

CC

present in the para-hippocampal gyrus and frontal cortical pole of adults with DS (Risser et al., 1997). Given that impaired serotonin uptake can lead to a reduction in the number of neurons in the

A

adult brain (Whitaker-Azmitia, 2001), Bianchi and co-authors discussed how alterations in the serotonergic systems, occurring during early life in DS, could contribute to impaired neurogenesis in DS brain and how it may be corrected (Bianchi et al., 2010). London and co-authors suggested that DYRK1A overexpression might be associated with the modification of monoamine neurotransmitters including serotonin; indeed, the transgenic mice for the DYRK1A gene 26

(mBacTGDYRK1A) showed a dramatic reduction of the serotonin contents in several brain areas especially in the hypothalamus (London et al., 2018).

Additionally, a histaminergic deficit has been proposed to contribute to cognitive impairment in DS.

IP T

More specifically, a reduction of histamine-releasing factor (HRF) in the brain of people with DS has been correlated to a decrease in histamine levels associated with cognitive decline and AD-like

SC R

dementia in DS (Kim et al., 2001).

2.3. Impairments in neuroplasticity

U

Neuroplasticity is an intrinsic adaptive ability of the nervous system in response to the changing

N

milieu, continuous neural activity, learning, and experience (Demarin and Morović, 2014). This

A

adaptation can range from functional modulations, such as alterations in neurochemical

M

transmission, modulation of dendritic functions, long-term potentiation, or structural adaptations (Demarin and Morović, 2014; Ruge et al., 2012) that include processes from synaptogenesis and

TE D

neurogenesis (Demarin and Morović, 2014; Fuchs and Flügge, 2014) to the revival and reformation of the existing synapses and signalling network (Mundkur, 2005). Basically, neuroplasticity

EP

accounts for learning and memory, capability of the nervous system and is also responsible for regaining lost abilities of the damaged neuronal system through structural and functional

CC

rehabilitation (Kleim and Jones, 2008).

A

As has been elaborated in this review, DS is a manifestation of abnormalities in neurotransmission, synaptic and neurogenesis impairments. However, neuroplasticity has been also reported to be aberrant in DS (Cramer and Galdzicki, 2012; Dierssen et al., 2003; García-Vallejo et al., 2018; Gardiner et al., 2010). Impaired neuroplasticity in DS is associated with cognitive deficits, and various structural and functional incongruence (Cramer and Galdzicki, 2012), such as impaired 27

dendritic maturation and reduced dendritic length (Takashima et al., 1994), morphological aberration and deficits in the number of dendritic spines (Marin-Padilla, 1976; Purpura, 1975; Suetsugu and Mehraein, 1980; Weitzdoerfer et al., 2001) and reduced synaptic contacts (Rueda et al., 2012). Therefore, improving neuroplasticity seems to be an additional strategy to correcting cognitive impairments in DS. This is because neuroplasticity can reform as well as replenish

IP T

neuronal structures, thus re-establishing the neuronal communication network (García-Vallejo et al.,

SC R

2018; Kays et al., 2012).

Neuronal and synaptic plasticity are known to be modulated by experience-dependent learning. Therefore, experience-dependent cognitive training could prove to be helpful in ameliorating

U

cognitive and intellectual disabilities in DS (Bartesaghi et al., 2015). Cognitive training along with

N

EGCG treatment has been shown to improve memory and adaptive behaviour in DS subjects in a

A

phase II clinical trial (ClinicalTrials.gov Identifier: NCT01699711) (de la Torre et al., 2016). As

M

well, providing more spacious and amiable housing conditions to Ts65Dn mice along with EGCG

TE D

treatment improved cognitive functions via increases in dendritic spine density of cornu ammonis 1, and restoration of the balance of inhibitory and excitatory synaptic markers of the cornu ammonis 1

EP

and dentate gyrus back to normal ranges (Catuara-Solarz et al., 2016).

DYRK1A gene also plays a pivotal role in modulation of brain plasticity and is known to be

CC

influenced by environmental stimulation through visual-spatial training (Bartesaghi et al., 2015; Pons-Espinal et al., 2013). A study conducted in the year 2009-2010 showed the efficacy of Wii

A

gaming technology (that creates virtual reality) in improving sensorimotor functioning in children with DS. Wii gaming aimed at relaying extensive sensorimotor neuronal activity in DS children through mirror mechanism thus targeting experience dependent modulation of neuroplasticity (Wuang et al., 2011).

28

Alterations in plasticity have also been observed in various animal models of DS, such as Ts65Dn and Ts1Cje mouse models. In the Ts1Cje mouse model, the trisomic region of MMU16 is translocated to MMU12 rendering it partially trisomic (Davisson, 2005) (Table 1). Both Ts65Dn and Ts1Cje mouse models exhibit abnormality in structure of pyramidal neurons and dendritic spines, as well as impairment of synaptic plasticity. Ts65Dn mice also display impairment of

IP T

experience-dependent neuroplasticity (Dierssen, 2012). The partially trisomic ts1Rhr mouse model (Gupta et al., 2016), displays structural aberrance in dendritic spines and reduced spine density in

SC R

facial dentate. It also exhibits impairment of synaptic plasticity and long-term potentiation in the hippocampus. This mouse model displays morphological aberrations in dendritic spines as well as impairments in neurogenesis. As well, the Kcnj6 transgenic mouse model exhibits functional

U

alterations in excitatory synaptic plasticity and long-term depression (Dierssen, 2012). Another

N

transgenic mouse model, the DYRK1A bacterial artificial chromosome transgenic mouse model

A

(mBACtgDyrk1a) which overexpresses DYRK1A, exhibits enhancement in spine density in the

M

pyramidal neurons of prefrontal cortex (Thomazeau et al., 2014). Bidirectional alteration in

TE D

plasticity with impairment in both NMDAR-mediated LTP and endocannabinoid mediated long term depression (LTD) in the hippocampus is also a prominent feature of mBACtgDyrk1a (Ahn et al., 2006; Thomazeau et al., 2014). It also exhibits increases in expression of proteins associated

EP

with the inhibitory GABAergic pathway and decreases in the expression of proteins of the

CC

excitatory glutaminergic pathway (Souchet et al., 2014).

A

2.3.1. Astrocyte-mediated synaptic dysfunction

Astrocytes play a critical role in the regulation of synaptic plasticity and memory formation (Adamsky and Goshen, 2018; Chung et al., 2015). DS astrocytes have been recognized to have a direct involvement in the reduced synaptic density and abnormal dendritic spine structure and function (Torres et al., 2018). Indeed, during brain development, astrocytes secrete some factors able to induce the formation of functional synapses between neurons (Clarke and Barres, 2013). 29

The astrocyte-neuron interaction is mediated by direct cell-to-cell connection as well as by a complex group of astrocyte-produced molecules, namely “astrocyte secretome” that dynamically changes according to neuronal activity and astrocyte activation (Clarke and Barres, 2013; Kıray et al., 2016). Both neurons and astrocytes in DS show a marked reduction in the thrombospondin 1 (TSP-1) protein expression, an astrocyte-secreted protein having a powerful effect on the

IP T

modulation of both spine number and morphology when evaluated in post mortem foetal brain cell cultures (Garcia et al., 2010; Torres et al., 2018). Depletion of TSP-1 from normal astrocytes

SC R

induced severe changes in spine morphology, while reinstatement of TSP-1 levels rescued the deficit in astrocyte-mediated spine and synaptic morphology. As well, iPSC-derived astrocytes have been shown to have a negative impact on synaptogenesis in DS, due to hyper-activation of the

U

mTOR pathway in neurons, and possibly contributing to DS neuropathogenesis (Araujo et al.,

M

2.4. Neurodegeneration

A

N

2017).

TE D

Neurodegeneration and the high risk to develop amyloid neuropathology with AD-like symptoms are part of the neurobiological alterations occurring in individuals with DS in adult age. Several neuroimaging researches have long showed that adult DS persons display typical brain alterations

EP

occurring in AD (Beacher et al., 2009; Lin et al., 2016; Sabbagh et al., 2015; Teipel and Hampel,

CC

2006). In a study carried out on both AD and DS subjects with dementia, high similarity was reported in the neuropsychological profiles of the two disease groups (Dick et al., 2016). In a recent

A

study, evaluation of brain 3D anatomical images and cognitive status of people with DS with an age of 40 years or older revealed a relationship between cognitive deterioration and severity of specific anatomical changes, such as ‘volume reduction in the substantia innominata region of the basal forebrain, hippocampus, lateral temporal cortex and left arcuate fasciculus’ (Pujol et al., 2018) which indicate early progression of cognitive impairment toward AD in adults with DS. In the next

30

section, we report data that disclose crucial mechanisms and pathways involved in early neurodegeneration in DS.

2.4.1. Amyloid pathway APP gene product encoded by Hsa21 represents a prominent component of the amyloid cascade

IP T

hypothesis of early manifestation of AD in the DS population. APP gene expression in the DS brain was found to be 1.6 fold higher than in the euploid brain and thought to be responsible of the

SC R

accumulation of the plaques containing fibrillar β-amyloid (Aβ) peptide (Lee et al., 2017; Mao et al., 2003). The neurotoxicity of Aβ has been attributed to microglia activation and increased oxidative stress production (Zana et al., 2007). Alteration of the main pathological hallmarks of AD Aβ-containing

plaques,

and

neurofibrillary

tangles

U

(including

(NFT)

containing

N

hyperphosphorylated tau protein) are similar in DS and AD (Rafii et al., 2015; Tamaoka, 1998;

A

Tapiola et al., 2001). Aβ appears to have a similar pattern in DS and sporadic AD and the C-labeled

M

Pittsburgh compound B (PiB), used to image Aβ plaques in the brains, showing early striatal

TE D

binding, which is similar to that observed in familial AD. Similarly, tangles appear to occur early in the hippocampus in DS as well in AD (see the review (Head et al., 2016). Production of ROS and oxidative stress, as well as energy depletion by mitochondrial dysfunction are involved in

EP

intracellular accumulation of Aβ and the pathogenesis of neurodegeneration in DS (Busciglio et al.,

CC

2002; Paola et al., 2000). As well, activation of the mTOR signalling pathway linked to tau hyperphosphorylation and Aβ generation have been recently shown to correlate with APP triplication and

A

neurodegeneration in DS (Di Domenico et al., 2018). Interestingly, alterations in protein quality control mechanisms, such as ubiquitin-proteasome and autophagy, are believed to induce accumulation of Aβ peptides and hyper-phosphorylated tau proteins in the DS brain, and has been proposed to be directly or indirectly involved in early neurodegeneration in DS (Di Domenico et al., 2013; Granese et al., 2013; Nabavi et al., 2018).

31

2.4.2. Oxidative stress, mitochondrial dysfunction and neural cell death Enhanced production of ROS is an attribute that is evident in DS since the embryonic stage (Busciglio and Yankner, 1995; Perluigi and Butterfield, 2012), resulting in apoptotic neuronal death and intellectual disability (Busciglio and Yankner, 1995). However, there are a number of in vitro and in vivo studies that support a link between the accumulation of oxidative stress and neuronal

IP T

cell death, considering that neurons are particularly vulnerable to damage and neurodegeneration (for refs see (Barone et al., 2017). ROS, which include volatile hydrogen peroxide, superoxide

SC R

anions, and hydroxyl radicals are produced under physiological conditions by aerobic respiration and several catabolic and anabolic processes (Halliwell, 1991). Recently, we discussed how mitochondrial dysfunction, due to Hsa21 gene overexpression-dependent deregulation of signalling

U

pathways controlling mitochondrial functions, is central in the etiological mechanisms leading to

N

oxidative stress, cognitive decline, early aging and AD-like dementia in DS (Valenti et al., 2018).

A

Thus, mitochondrial dysfunction is not only involved in neurodevelopmental impairment (as

M

discussed in the paragraph 2.2.3) but also in neurodegenerative processes, leading to cognitive

TE D

decline in DS. The mitochondrial respiratory chain and in particular, complexes I and III, are the major source of production of the superoxide anion, generated by electron leakage during electron transport, that is converted into H2O2 and O2 by the mitochondrial-MnSOD (Raha and Robinson,

EP

2000). Impaired mitochondrial respiratory chain and disruption in oxidative phosphorylation can

CC

lead to a decrease in mitochondrial ATP production and an increase in ROS (Alfadda and Sallam,

A

2012).

We have previously demonstrated that dysfunctional mitochondrial complex I in DS cells overproduces ROS in DS human skin fibroblast cell lines (Valenti et al., 2011). Under physiological conditions, ROS are quickly removed before they can cause cellular dysfunction. However, DS cells are not able to efficiently scavenge ROS overproduction and consequently are more susceptible to oxidative damage. Indeed, the elevated intracellular levels of ROS in DS have long 32

been proposed to occur as a direct consequence of overexpression of the SOD1, which plays a crucial role in the metabolism of superoxide radicals and scavenging of ROS (Créau, 2012), and whose overexpression has been indicated in the formation and accumulation of oxidative stress in DS (Rodríguez-Sureda et al., 2015). Hydrogen peroxide generated as a result of superoxide radical metabolism by SOD is further converted to water by catalase (CAT) and glutathione peroxidase

IP T

(GPX). Therefore, an imbalance in SOD/GPX and CAT ratio exacerbates cytosolic ROS accumulation (de Haan et al., 1995). An imbalance in SOD/GPX ratio has been reported in all DS

SC R

tissues (Barone et al., 2017; de Haan et al., 1995; Perluigi and Butterfield, 2012).

Alterations to iron metabolism for deregulation of redox homeostasis in DS has also been recently

U

proposed (Barone et al., 2017). A redox proteomic analysis of brains of people with DS under the

N

age of 40 compared with age-matched controls, revealed accumulation of protein carbonyls (Di

A

Domenico et al., 2013), suggesting that both an increase in oxidative stress and reduced clearance

M

of oxidized proteins, possibly due to an impairment in proteostasis network (Aivazidis et al., 2017),

TE D

may contribute to early neurodegeneration in DS.

3. Proposed pharmacological intervention and their targets in Down syndrome

EP

Various pharmacotherapies have been proposed in DS, mainly for improving cognitive behaviour.

CC

Many drugs that have shown to be effective in mouse models of DS have prompted clinical trials in young adults or children with Hsa21 trisomy. Some clinical studies have reported significant

A

adverse effects that have diminished their translational impact or clinical efficacy (de la Torre and Dierssen, 2012). Herein, we will present and discuss drugs already approved for human use and being tested in clinical trials as promising therapeutic strategies for DS (see Figure 2 and Table 2). Their underlying targets and mechanisms of action in relation to DS and potential pitfalls for their use in human, if any, will also be evaluated.

33

3.1. Bumetanide and basmisanil (RG1662) The Na-K-Cl cotransporter 1 (NKCC1) has been reported to be upregulated in the hippocampus of Ts65Dn mouse model of DS and considered to be responsible for aberrant GABA AR signalling, which is found to be excitatory rather than inhibitory (see paragraph 2.1.1). This incongruity in GABAAR signalling is believed to be responsible for the impairment of LTP and LTD. LTP and

IP T

LTD are characterized by the strengthening or weakening (respectively) of synaptic efficiency, following specific stimulation protocols and provides information on learning and memory

SC R

efficiency (Contestabile et al., 2017). Bumetanide, NKCC1 inhibitor, capable of reversing GABAAR signalling, has been shown to be beneficial in improving hippocampal synaptic plasticity and enhancing memory in the Ts65Dn mouse model (Contestabile et al., 2017; Deidda et al., 2015).

U

Targeting GABA transmission, and in particular Cl- cotransporters seems to be very effective in

N

improving cognitive deficits in DS. However, the bumetanide effect is not maintained after the

A

treatment is ceased (Deidda et al., 2015), thus presuming a lifelong treatment with this drug.

M

Bumetanide is an FDA-approved diuretic drug for the treatment of acute pathologies. A limitation

TE D

for the long-term use of bumetanide to correct cognitive impairment in DS is that chronic consumption of this diuretic could have adverse effects on renal function. RG1662 or Basmisanil is a negative allosteric modulator of α5-GABAAR, developed by Hoffmann-

EP

La Roche Ltd (RO4938581) (Mohler, 2012; Potier et al., 2014). It was found to be efficacious in

CC

rescuing cognitive and hippocampal synaptic deficits in the Ts65Dn mouse model (Martinez-Cue et al., 2013). Braudeau and colleagues (2011) reported that α5-selective GABAAR inverse agonists are

A

able to restore cognitive function in a DS mouse model. These results assisted the hypothesis that altering the GABAergic-mediated balance between inhibitory and excitatory neurotransmission can competently alleviate cognitive impairments in DS in preclinical models (Braudeau et al., 2011).

Despite the promising results obtained in pre-clinical trials, the phase II multicentre, double blind clinical trial of RG1662 (a derivative of RO4938581) in DS subjects namely CLEMATIS 34

(ClinicalTrials.gov identifier: NCT02024789) did not obtain any significant outcomes to ameliorate cognitive disabilities in DS population as reported by a La Roche statement (from the web site: https://ds-int.org/node/3547).

3.2. Fluoxetine

IP T

Fluoxetine is a selective serotonin reuptake inhibitor generally used as an antidepressant in adults but has also been reported to modulate the immune system (Di Rosso et al., 2016). It has been

SC R

shown to stimulate survival as well as neurogenesis in the dentate gyrus of the Ts65Dn mouse model (Bianchi et al., 2010) and to restore functional connectivity of the synaptic network of the hippocampus in Ts65Dn mice (Stagni et al., 2013). Fluoxetine successfully produced favourable

U

morphological changes in dendritic architecture of the granule cells of Ts65Dn mice during the

N

earliest phases of development resulting in proper maturation of granule neurons which is

A

considered to be a key factor to rescue neurogenesis and neurodevelopmental defects in DS mouse

M

model (Guidi et al., 2013). In the granule and subgranular cell layer of dentate gyrus of Ts65Dn

TE D

mice, fluoxetine treatment promoted proliferation and survival of these hippocampal cells, thus resulting in enhanced neurogenesis (Clark et al., 2006). In addition, prolonged oral treatment with fluoxetine in adult Ts65Dn mice has been shown to regularize GABA release, improving the spatial

EP

memory skills and recovering hippocampal synaptic plasticity (Begenisic et al., 2014). On a

CC

positive note, treatment with fluoxetine in the perinatal period resulted in long-term improvements on cognitive impairment and prevented early signs of AD-like pathology in adult Ts65Dn mice

A

(Stagni et al., 2015). Beneficial effect in neuronal precursor proliferation potency and dendritic development were also reported in 2-day-old Ts65Dn mice (Guidi et al., 2014). Fluoxetine has been proposed as a potential prenatal pharmacotherapy able to improve foetal hippocampal neurogenesis and improper brain formation (Kuehn, 2016). However, many adverse effects have been reported following long-term treatment of this drug (Riediger et al., 2017), which could limit the continual use of fluoxetine during infancy and adolescence. 35

3.3. Memantine Memantine is a non-competitive NMDA receptor antagonist proposed to improve glutamatergic transmission to ameliorate memory, behaviour and cognitive impairment (Costa et al., 2008; Rueda et al., 2010). An increase in brain derived neurotrophic factors has also been reported in the

IP T

hippocampus and frontal cortex of Ts65Dn mice following treatment with memantine (Lockrow et al., 2011). A pharmacokinetic study inTs65Dn mice revealed that memantine can transfer to the

SC R

offspring through placenta and maternal milk (Victorino et al., 2017). The memory enhancing effect of memantine is thought to be a consequence of its NMDA receptor antagonism mediated blocking of excessive calcium influx (Lipton, 2004) which is known to mediate disparities in neuronal

U

signalling involved in memory and cognition (Parsons et al., 2007). As well, memantine has been

N

shown to alleviate long-term depression in the hippocampus of Ts65Dn mice which has been

A

proposed to be another possible mechanism for its memory enhancing effects in DS (Scott-McKean

M

and Costa, 2011). Despite the promising outcomes obtained for cognition and memory

TE D

enhancement in DS mouse models, memantine seems to fail to re-capacitate cognition and enhance memory in DS adults. Indeed, after a randomised double-blind placebo-controlled trial with adults (>40 years) with DS, with and without dementia (ISRCTN registry identifier: ISRCTN47562898),

EP

Hanney and colleagues concluded that there were non-significant differences between treated and

CC

placebo groups and that memantine, although well-tolerated, is not an effective treatment to correct dementia in DS (Hanney et al., 2012). On the contrary, Boada and colleagues reported beneficial

A

effects of memantine on cognition in a pilot randomized double blinded trial with young adult with DS (between the ages of 18 and 32 year (ClinicalTrials.gov identifier: NCT01112683). Although they found no significant differences between the memantine treated and placebo group in paired learning and pattern recognition memory, which were the two main primary outcomes of the trial, they found a significant improvement in the memantine group in one of subset memory outcomes

36

and only infrequent and mild adverse events (Boada et al., 2012), suggesting the importance to explore other biochemical biomarkers in future clinical studies with this drug.

3.4. Donepezil, β- and γ-secretase modulators and Aβ immunotherapies for cognitive decline Donepezil is an acetylcholinesterase inhibitor currently used to improve neurobehavioral symptoms

IP T

for elderly patients with AD by enhancing the cholinergic transmission (Birks and Harvey, 2003). This drug has been shown to be effective and safe in adults with DS for AD-like dementia,

SC R

improving global and specific mental functions of DS patients with severe cognitive impairment, as demonstrated in a randomized, double blind, placebo-controlled trial (Kondoh et al., 2011). Recently, it has been reported in the case of an adolescent with severe behavioural disturbances for

U

which donepezil treatment resulted in the alleviation of symptoms and total recovery of the patient

N

to his previous psychosocial levels (Tamasaki et al., 2016). In an attempt to demonstrate the

A

efficacy of donepezil on cognitive improvements in children, a randomized, double-blind, placebo-

M

controlled multicentre study has been performed with children with DS aged 10-17. That study

TE D

failed to demonstrate any benefit for donepezil versus placebo although the drug appeared to be well tolerated (Kishnani et al., 2010).

EP

β- and γ-secretase modulators could be considered good candidate for improvement of cognitive

CC

decline due to their action in targeting the formation of cytotoxic Aβ. The formation of Aβ takes place following the cleavage of APP through two aspartic proteases such as β-secretase and γ-

A

secretase (Birks and Harvey, 2003). APP is cleaved at the β-site by β-secretase and is therefore known as the β-site APP cleaving enzyme 1 (BACE 1) (Vassar et al., 1999). A potential strategy to target Aβ plaques in DS could be inhibition of BACE1. However, the homologous BACE2 protein does not share a relationship in the cleavage of APP in humans (Ballard et al., 2016), but may have some protective effect (Sun et al., 2006). Some preclinical studies have showed evidence that the quantity of amyloid protein in the brain, cerebrospinal fluid and plasma can be reduce by β37

secretase inhibitors (Chen et al., 2015; Ghosh et al., 2012; Netzer et al., 2010). However, some other scientists believe that no inhibitors can decrease the amount of Aβ in the human brain, plasma or cerebrospinal fluid (CSF) (Varghese, 2010).

γ-Secretase is a composed by four dissimilar proteins: nicastrin (Nct), presenilin (PS), presenilin-

IP T

enhancer 2 (Pen-2) and anterior pharynx-defective 1 (Aph-1), that are present in a similar (1:1:1:1) stoichiometry (Sato et al., 2007). γ-Secretase is an aspartyl protease belonging to the intra-

SC R

membrane cleaving protease family (I-CLiPs) which includes rhomboid proteases, signal peptide peptidases, and site- 2 peptidases (Bursavich et al., 2016). Semagacestat and avagacesat are two γsecretase inhibitors (GIs) already used in phase II clinical trials for AD. It was found that

U

semagacestat did not improve cognitive decline in AD, and exacerbated functional ability at the

N

higher doses (Doody et al., 2013). Moreover, significant adverse GI effects have been previously

A

reported (Coric et al., 2012; Doody et al., 2013). Although the FDA-approved GIs cannot be used as

M

therapy in DS due to significant adverse effects, recently Stagni and co-worker tested the GI

TE D

ELND006 in Ts65Dn perinatal period providing the proof of principle of the efficacy of an early inhibition of γ-secretase to improve brain development in DS conditions (Stagni et al., 2017).

EP

Aβ immunotherapies as a strategy to reduce Aβ overload in the brain by using synthetic peptides or

CC

monoclonal antibodies, have been developed and currently trialled in AD, and represent a promising approach to modify the course of the disease (Mo et al., 2017). Interestingly, evaluation

A

of a vaccine against Aβ (namely ACI-24) is also under investigation by Rafii MS group in a DS clinical trial (ClinicalTrials.gov Identifier: NCT02738450) with the primary outcome of safety and the secondary outcome of reducing neurobehavioral and neurologic manifestations of AD in adults with DS.

3.5. Coenzyme Q10, metformin and melatonin 38

Coenzyme Q10 (CoQ10) is a crucial cofactor of the inner mitochondrial membrane involved in mitochondrial respiratory chain and free radical scavenging. Numerous studies have highlighted its antioxidant potential in preventing oxidative DNA damage and its other neuroprotective effects (Hernández-Camacho et al., 2018; Negida et al., 2016; Yang et al., 2016). Significantly reduced platelet and lymphocytic CoQ10 content and low plasma levels of CoQ10 have been reported in

IP T

people with DS in paediatric age and was found to have a proportional relationship to the intelligence quotient of DS children (Zaki et al., 2017). CoQ10 treatment showed to lower the

SC R

oxidative stress status in children with DS (Miles et al., 2007), however, in a randomized, phase II clinical trial (ClinicalTrials.gov Identifier: NCT00891917), the authors reported no efficacy regarding the primary outcomes e.g. language skills, expressive language ability, and speech In another randomized double-blinded trial in DS children, prolonged CoQ10

U

articulation.

N

administration for 20 weeks produced an increase in plasma CoQ10 levels and induced protection

M

A

against DNA oxidation by modulating DNA repair mechanisms (Tiano et al., 2012).

TE D

Metformin, an FDA-approved drug used for the treatment of type 2 diabetes, has also been recently proposed as a potential therapeutic strategy in DS. Metformin was shown to promote neurogenesis in rodent and human neural precursors and enhance spatial memory formation in normal adult mice

EP

(Wang et al., 2012), and has been shown to be a pharmacological activator of PGC-1α with a

CC

consequent ability to counteract impairment of mitochondrial network and correct mitochondrial dysfunction in fibroblasts with Hsa21 trisomy (Izzo et al., 2017b). However, prior to the clinical

A

translation of metformin as a chronic therapy in DS, the relationship between dosage and effect must be considered, given that the drug has never been used in children.

Melatonin (N-acetyl-5-methoxytryptamine) is a neurohormone mainly secreted from the pineal gland (Tan et al., 2010). It has a wide range of regulatory and protective effects, such as synchronizing circadian rhythm, protecting against oxidative stress, regulating energy metabolism, 39

modulating the immune system, and postponing the ageing process (Singh and Jadhav, 2014). In a pilot study, the comparison between children with DS and age-matched controls in the melatonin plasma levels, showed a reduction in the plasma concentration of melatonin in people with DS, resulting in an increased oxidative risk for DS children (Uberos et al., 2010). Furthermore, treatment of adult Ts65Dn mice with melatonin induced antioxidant and anti-ageing effects in the

IP T

hippocampus (Parisotto et al., 2016). However, an additional study in the same DS mouse model has shown that pre- and post-natal melatonin administration, induced only a partial rescue of brain

SC R

oxidative stress and cognitive behaviours (Corrales et al., 2017). Recently, the neuroprotective and anti neuro-inflammatory effects of melatonin have been suggested, given its action in modulating mitochondria integrity and function in neurons in aging (Idowu et al., 2017; Reiter et al., 2018). No

A

3.6. Naturally-occurring phytochemicals

N

U

studies on the effect and safety of melatonin in DS population have been reported to date.

M

Polyphenols are naturally occurring phytochemicals produced as secondary metabolites in plants in

TE D

response to various stress conditions, such as environmental stress or injury. They are mainly present in vegetables, fruits, and cereals (Vacca et al., 2016; Valenti et al., 2017). Consumption of a polyphenol-enriched diet has demonstrated protective effects against a large number of ailments for

EP

examples cardiovascular diseases, neurodegenerative diseases, carcinomas and diabetes (Fantini et

CC

al., 2015; Grootaert et al., 2015; Vacca et al., 2016). Evidence are present for polyphenol-mediated modulation of lipid oxidation and signalling pathways regulating mitochondrial functions, redox

A

homeostasis and homocysteine metabolism. In addition, polyphenols modulate activity and expression of important Hsa21 genes involved in DS pathogenesis, such as DYRK1A, RCAN1 and the amyloid precursor protein (APP) as well as miRNAs, such as miR-155 (for refs see (Vacca et al., 2016; Valenti et al., 2018). Therefore, these natural integrators may have potential effects in preventing or managing some DS clinical manifestations. However, the assessment of the dose and

40

effect relationship as strategies adequate to improve bioavailability and delivery are not yet completely established for the translation of polyphenols to the clinical use as therapeutics in DS.

Two polyphenols, EGCG and resveratrol, have been proposed as possible natural diet integrators for the management of DS. EGCG is the major polyphenol present in Camellia sinensisis belonging

IP T

to the flavonoid subclass. Specifically, it is a gallic acid esterified catechin and a potent antioxidant (Banerjee and Chatterjee, 2015; Chowdhury et al., 2016). EGCG has been suggested as a potential

SC R

drug candidate for treatment and management of some clinical features of DS due to its multimodal capability of targeting many signalling pathways responsible for the regulation of neural health and function (Vacca et al., 2016). The most studied action of EGCG in DS is its capacity to inhibit

U

DYRK1A (Park et al., 2009; Yin et al., 2017). the hyperactive protein kinase in DS which affects

N

many downstream pathways linked to neurogenesis and neuroplasticity (see the paragraph 2.1.1).

A

Treatment with EGCG of both Ts65Dn mice and young adults with DS have proved to normalize

M

DYRK1A activity and rescue brain plasticity and partially improve some cognitive functions linked

TE D

to memory and learning (De la Torre et al., 2014).

A study by Catuara-Solarz co-workers reported that treatment of environmental enrichment and

EP

45% EGCG containing green tea extract enhanced the corticohippocampal-dependent learning and

CC

memory. The cognitive improvements were escorted by rescue of cornu ammonis 1 dendritic spine density and a stabilization of the amount of excitatory and inhibitory synaptic markers in dentate

A

gyrus and cornu ammonis 1 (Catuara-Solarz et al., 2016).

In vitro studies on neurons, fibroblasts and limphoblastoid cell with Hsa21 trisomy have revealed other targets of EGCG beyond DYRK1A. EGCG has been shown to be a potent scavenger of ROS produced by mitochondria and potent regulator of mitochondrial homoeostasis (Schroeder et al., 2009). Other beneficial effects of EGCG include activation of mitochondrial biogenesis and 41

bioenergetics trough the targeting of Sirt1/AMPK/PGC1-α pathways (Sutherland et al., 2005; Valenti et al., 2018, 2016, 2013). Protection against β-amyloid-induced mitochondrial apoptosis (Zhang et al., 2017) and glutamate cytotoxicity (J. Yu et al., 2010) was also found. Modulation of cAMP levels by EGCG has also been reported, and an increase in the activity of cAMP-dependent PKA has been shown to increase the NADH: ubiquinone oxidoreductase subunit S4 (NDUFS4)

IP T

phosphorylation in T21 human cells (Valenti et al., 2013).

SC R

However, some questions should be resolved prior to the introduction of EGCG as dietetic integration in DS. These include elucidating the most appropriate mode of delivery and types of green tea extracts (that are commercially available) instead of pure EGCG, the dose-response

U

relationship, and the combination of EGCG with other nutraceuticals in order to improve its

N

bioavailability. At this regards some contradicting studies have been reported. For instance, Souchet

A

and co-workers evaluated the effect of an EGCG extract namely POL-60, containing a mix of green

M

tea polyphenols (27% EGCG, and 42% other catechins, 8 % caffeine and 1% sucrose) in

TE D

mBACtgDyrk1a, a mouse model overexpressing DYRK1A, and in Ts65Dn. POL60 was able to rescue components of glutamatergic and GABAergic pathways in the hippocampus and cortex but not in the cerebellum, and reversed brain molecular alterations that disrupt excitatory/inhibitory

EP

balance. Moreover, it was also found that components of GABAergic pathway were partially

CC

rescued by other extracts such as decaffeinated green tea extract containing 45% EGCG (Souchet et

A

al., 2015).

However, reports are also available which suggest that even using a pure stabilized EGCG in the concentrations which produced therapeutic effects on skeletal phenotypes failed to improve cognitive phenotypes (Stringer et al., 2015). The authors concluded that reliable effect is dependent on the dosage of EGCG. Another recent study by the same group showed that the inhibitory effect of EGCG on DYK1A kinase activity was tissue-dependent. Additionally, lack of advantageous 42

therapeutic behavioural effects and potentially detrimental skeletal effects of high EGCG dose in Ts65Dn mice accentuated the importance of EGCG dosage identification (Stringer et al., 2017).

It should also consider that administration of EGCG in the neonatal period rescues many trisomyassociated brain alterations. However, neonatal treatment did not elicit durable effects on the

IP T

physiology of the hippocampus in adulthood (Stagni et al., 2016), thus suggesting long-term consumption of this polyphenol as a pharmacological tool in DS. It should be highlight that EGCG

SC R

has been tested in a double-blind phase II clinical trial with young adults with DS (Table 2). The study reported EGCG benefits in potentiating cognitive training and improving visual recognition memory (de la Torre et al., 2016). EGCG treatment in early childhood could further enhance the

U

effects on cognitive behaviour. In a case study we demonstrated that 6 month-treatment of a 10-

N

years old DS child with a combination of EGCG and fish oil omega-3, which has been shown to

A

improve EGCG bioavailability and synergize its effectiveness (Giunta et al., 2010), improves

M

mitochondrial functions, attention and concentration and has no adverse effects (Vacca and Valenti,

TE D

2015). Thus, the combination of EGCG with other nutraceutics could enhance its efficacy.

Another interesting polyphenol in DS is trans-resveratrol, which belongs to the stilbenoid class. We

EP

previously reported that trans-resveratrol improves mitochondrial biogenesis and bioenergetics

CC

deficits in hippocampal progenitor cells from Ts65Dn mice by targeting PGC-1α/Sirt1/AMPK axis (Valenti et al., 2016). Moreover, trans-resveratrol downregulates chromosome 21-encoded miR-155

A

and plays a key role in the regulation of mitochondrial biogenesis (Latruffe et al., 2015; QuinonesLombrana and Blanco, 2015). Therefore, miR-155 normalization through resveratrol could serve as a potential approach for the prevention of mitochondrial biogenesis impairment and other metabolic changes that occur in DS. However, the poor bioavailability of resveratrol raises a question mark for its efficacy in vivo. Trans-resveratrol is indeed poorly absorbed and rapidly metabolized into metabolites (i.e. glucuronide and sulfate derivatives) that are unstable and subject to rapid urinary 43

clearance (Goldberg et al., 2003). The natural glucoside of resveratrol, the polydatin (transresveratrol-3-O-glucoside), a polyphenol belonging to the chemical family of stilbenoids, present in the plant Polygonum cuspidatum, has the capability to easily cross cell membranes by an active mechanism using glucose carriers and is resistant to enzymatic metabolism. During first pass metabolism, the glucose molecule of polydatin is cleaved off by cellular β-glucosidase, converting

IP T

polydatin into trans-resveratrol where it maximizes tissue concentration (Du et al., 2013). Thus, polydatin could have much higher capacity than resveratrol to be used as a natural drug in DS and

SC R

investigations are currently underway in our laboratory to test its efficacy in cells with Hsa21 trisomy and animal models with DS. Indeed, polydatin exhibits many pharmacological activities similar to those reported for resveratrol, including cardioprotection, neuroprotection, antioxidative,

U

anti-inflammatory and multiple-organ protection (Ravagnan et al., 2013; Sies, 2010), and has been

N

approved by the FDA for human use and tested in preclinical and clinical trials for other diseases

M

A

(Cremon et al., 2017; Gao et al., 2016).

TE D

Other polyphenols, such as hydroxytyrosol, quercetin and curcumin have also been found to play an important antioxidant role and can modulate several signalling pathways regulating mitochondrial function and ROS homeostasis that have been found to be deregulated in DS (Vacca et al., 2016).

CC

EP

However, studies testing the effects of these natural molecules on people with DS remain nascent.

3.7. Other dietary supplements

A

Evidence for the deficiency of some micronutrients such as vitamins, amino acids and enzymes have been widely described in DS. Low glutathione concentrations and irregular folate metabolism have been reported in DS (Salemi et al., 2015). Deficiency of folate and vitamin B12 is considered to represent a significant risk factor for children with DS (Obermann-Borst et al., 2011; Saghazadeh et al., 2017; Sukla et al., 2015). Similar results were obtained in clinical studies of DS population in Egypt and Italy (Licastro et al., 2006; Meguid et al., 2001). Malabsorption of vitamin B12 without 44

proteinuria was also reported in a child with DS (Cartlidge and Curnock, 1986). Folate is a vital B vitamin which is found in meat, fruits, green vegetables, and beans that provide one-carbon molecules for DNA synthesis, and methylation of proteins (Bernstein et al., 2007; Patterson, 2008). The role of the folate pathway in chromosome 21 non-disjunction was first described by James and

IP T

co-workers (James et al., 1999).

It has been hypothesized that folate has the ability to lower homocysteine levels, and a folate rich

SC R

diet may decrease the incidence for having chromosome non-disjunction, the incidence of children with trisomy (Patterson, 2008). However clinical trials in early infancy of children with DS with folinic acid supplementation alone (ClinicalTrials.gov Identifier: NCT00294593) or in combination

U

with vitamins and antioxidants (ClinicalTrials.gov Identifier: NCT00378456) failed to improve

A

N

cognitive and psychomotor development (Blehaut et al., 2010; Ellis et al., 2008) (see Table 2).

M

Supplementation with α-tocopherol at a dose of 400 IU/day for 4 months in the diets of DS children

TE D

may decrease oxidative DNA damage (Mustafa Nachvak et al., 2014). α-Tocopherol also decreased lipid peroxidation levels in the Ts65Dn mice model and improved hippocampal dentate gyrus hypo cellularity. Early stage supplementation of α-tocopherol has been reported to improve cognitive

EP

deficits in Ts65Dn mouse model of DS (Shichiri et al., 2011). However, at higher dosage it may

CC

enhance mortality (Miller et al., 2005).

A

In children with DS, normal Mg2+, Ca2+ and Zn2+ levels were found (Józefczuk et al., 2017). However, in another meta-analysis lower blood levels of Ca2+, Zn2+ and selenium was reported (Saghazadeh et al., 2017). Similar results have been reported from a study conducted on 16 DS subjects (Kadrabová et al., 1996). As well, a reduction in vitamin D, which plays an important role for calcium absorption and immune response (Christakos and DeLuca, 2011), has been described in children and adolescents with DS, in particular in the presence of obesity and autoimmune diseases 45

(Stagi et al., 2015). In the case of hypovitaminosis D, the need for cholecalciferol (Vitamin D3) supplementation has been discussed (Stagi et al., 2015).

Significant differences in amino acids levels have also been found in the DS individuals. A decrease in the plasma concentration of serine and an elevation of plasma concentration of lysine was found

IP T

correlated with premature ageing (Mircher et al., 1997). Several neurotransmitter amino acids have been reported to be altered in the brain of people with DS. Glutamate shortfall is observed in the

SC R

hippocampus of DS individuals, and reduced level of GABA is also found in the temporal cortex and hippocampus in patients with neocortical neurofibrillary tangles (Reynolds and Warner, 1988). Glutathione is a potent antioxidant that eliminates free radicals. Studies have revealed that children

U

with DS have low levels of glutathione which is mainly due to overexpression of the SOD1 gene

A

N

(Wang et al., 2003).

M

Many clinical trials have been conducted in DS population using nutrients as pharmacological

TE D

agents such as zinc, selenium, megavitamin/mineral preparations, vitamin A, vitamin B6 and its precursors, vasopressin, whose results have been previously reviewed (Ani et al., 2000). However, only few randomized trials have been conducted and none of these trials have shown any

EP

improvement in the clinical outcomes. Nevertheless, analysis of anonymous questionnaires has

CC

identified inappropriate use of supplementation with antioxidants, vitamins and fatty acids. These nutraceuticals are very often started in the early infancy, and without any objective quantification

A

on neurodevelopmental progress, and 20% of which are not monitored by pediatricians for potential adverse effects (Lewanda et al., 2018).

4. Concluding remarks and future prospective Gene mapping and development of numerous models for DS have greatly expanded horizons in understanding anomalous metabolic functions and pathways that mark DS (Table 1). In this review, 46

we have shown a wide picture of how Hsa21 trisomy can lead to alterations of signalling pathways and epigenetic mechanisms as well as impairment of crucial cellular metabolic pathways and bioenergetics. Altogether, these alterations lead to DS neuropathology (Figure 1). Attempts are being made for further elucidation of the underlying pathogenicity of DS, as well as for the

neurological and biochemical pathways present in DS (Figure 2 and Table 2).

IP T

development and testing of drugs from synthetic and natural origin that could target aberrant

SC R

Various potential therapeutic agents that may applicable for DS are already available on the market. For instance, fluoxetine, bumetanide and metformin, already used for other diseases, are not however, currently under clinical investigations for DS, and further experimental work is required

U

prior to discovering a safe and effective drug to improve DS-related intellectual disability. Natural

N

products and dietary supplements may also be advantageous for the prevention and management of

A

neurological disorders associated with DS. Polyphenols such as EGCG and resveratrol are quite

M

beneficial for the treatment of symptoms associated with DS given their multi-targets action.

TE D

However, additional clinical evidence is necessary and rigorous studies are required to explore the pharmacokinetics of these natural products before they can be used effectively in the clinic.

EP

We believe that mitochondria should be crucial pharmacological targets to maintain neural energy

CC

and redox homeostasis, fundamental for all neurological processes, and suggest that combinations of therapeutic agents may better improve the complex neurobiological alterations in DS. Moreover,

A

future clinical trials should be more refined to include specific and objective outcomes to obtain definitive findings on the efficacy of a treatment in DS. On the other hand, it should be considered that differential drug responses might occur in DS population due to disparity in comorbidities associated to DS (Hefti and Blanco, 2017).

47

Apart from pharmacological strategies for the treatment of DS, epigenomic therapies targeting epigenetic marks have gained considerable attraction to ameliorate DS-related phenotypes. Epidrugs such as methyltransferase inhibitors and histone deacetylase inhibitors, are already used to treat cancer (Campbell and Tummino, 2014) and may be useful to target neurological or specific epigenetic pathways in DS (Mentis, 2016). More recently, epigenomic engineering by the CRISPR-

IP T

Cas 9 system for the Hsa21 editing in DS has been proposed as an innovative and future strategy for

SC R

DS treatment (Mentis, 2016).

We would like to conclude this review with a quote from Prof Jerome Lejeune, the scientist that discovered sixty years ago (in the 1959) the genetic cause of DS regarding the prospects of a cure

U

for DS: “We will find a way. It is impossible not to. It is a much less difficult intellectual effort than

A

Acknowledgments

M

5.

N

sending a man on the moon”.

TE D

We are particularly grateful to parents of DS children that very kindly contributed to support in part this study in particular to “Associazione A.M.A.R. Down-Onlus”. The authors wish to thank Dr.

A

CC

EP

Laura Marra for manuscript editing.

48

6. References Adamsky, A., Goshen, I., 2018. Astrocytes in Memory Function: Pioneering Findings and Future Directions. Neuroscience 370, 14–26. https://doi.org/10.1016/j.neuroscience.2017.05.033 Ahn, K.-J., Jeong, H.K., Choi, H.-S., Ryoo, S.-R., Kim, Y.J., Goo, J.-S., Choi, S.-Y., Han, J.-S., Ha, I., Song, W.-J., 2006. DYRK1A BAC transgenic mice show altered synaptic plasticity with learning and memory defects. Neurobiology of disease 22, 463–472. https://doi.org/10.1016/j.nbd.2005.12.006

IP T

Aivazidis, S., Coughlan, C.M., Rauniyar, A.K., Jiang, H., Liggett, L.A., Maclean, K.N., Roede, J.R., 2017. The burden of trisomy 21 disrupts the proteostasis network in Down syndrome. PLOS ONE 12, e0176307. https://doi.org/10.1371/journal.pone.0176307

SC R

Alfadda, A.A., Sallam, R.M., 2012. Reactive Oxygen Species in Health and Disease. Journal of Biomedicine and Biotechnology 2012, 1–14. https://doi.org/10.1155/2012/936486

N

U

Altafaj, X., Dierssen, M., Baamonde, C., Martí, E., Visa, J., Guimerà, J., Oset, M., González, J.R., Flórez, J., Fillat, C., Estivill, X., 2001. Neurodevelopmental delay, motor abnormalities and cognitive deficits in transgenic mice overexpressing Dyrk1A (minibrain), a murine model of Down’s syndrome. Human Molecular Genetics 10, 1915–1923.

M

A

Ani, C., Grantham-McGregor, S., Muller, D., 2000. Nutritional supplementation in Down syndrome: theoretical considerations and current status. Developmental Medicine and Child Neurology 42, 207–213.

TE D

Antonarakis, S.E., 2017. Down syndrome and the complexity of genome dosage imbalance. Nature Reviews Genetics 18, 147–163. https://doi.org/10.1038/nrg.2016.154 Antonarakis, S.E., Lyle, R., Dermitzakis, E.T., Reymond, A., Deutsch, S., 2004. Chromosome 21 and down syndrome: from genomics to pathophysiology. Nature reviews. Genetics 5, 725–738. https://doi.org/10.1038/nrg1448

CC

EP

Araujo, B.H.S., Kaid, C., De Souza, J.S., Gomes da Silva, S., Goulart, E., Caires, L.C.J., Musso, C.M., Torres, L.B., Ferrasa, A., Herai, R., Zatz, M., Okamoto, O.K., Cavalheiro, E.A., 2017. Down Syndrome iPSC-Derived Astrocytes Impair Neuronal Synaptogenesis and the mTOR Pathway In Vitro. Molecular Neurobiology. https://doi.org/10.1007/s12035-017-0818-6

A

Arrázola, M.S., Andraini, T., Szelechowski, M., Mouledous, L., Arnauné-Pelloquin, L., Davezac, N., Belenguer, P., Rampon, C., Miquel, M.-C., 2018. Mitochondria in Developmental and Adult Neurogenesis. Neurotoxicity Research. https://doi.org/10.1007/s12640-018-9942-y Arron, J.R., Winslow, M.M., Polleri, A., Chang, C.-P., Wu, H., Gao, X., Neilson, J.R., Chen, L., Heit, J.J., Kim, S.K., Yamasaki, N., Miyakawa, T., Francke, U., Graef, I.A., Crabtree, G.R., 2006. NFAT dysregulation by increased dosage of DSCR1 and DYRK1A on chromosome 21. Nature 441, 595–600. https://doi.org/10.1038/nature04678 Asim, A., Kumar, A., Muthuswamy, S., Jain, S., Agarwal, S., 2015. “Down syndrome: an insight of the disease.” Journal of Biomedical Science 22. https://doi.org/10.1186/s12929-015-0138-y 49

Aston-Jones, G., Rajkowski, J., Kubiak, P., Alexinsky, T., 1994. Locus coeruleus neurons in monkey are selectively activated by attended cues in a vigilance task. The Journal of Neuroscience: The Official Journal of the Society for Neuroscience 14, 4467–4480. Ayberk Kurt, M., Ilker Kafa, M., Dierssen, M., Ceri Davies, D., 2004. Deficits of neuronal density in CA1 and synaptic density in the dentate gyrus, CA3 and CA1, in a mouse model of Down syndrome. Brain Research 1022, 101–109. https://doi.org/10.1016/j.brainres.2004.06.075

IP T

Ballard, C., Mobley, W., Hardy, J., Williams, G., Corbett, A., 2016. Dementia in Down’s syndrome. The Lancet. Neurology 15, 622–636. https://doi.org/10.1016/S1474-4422(16)00063-6

SC R

Banerjee, S., Chatterjee, J., 2015. Efficient extraction strategies of tea (Camellia sinensis) biomolecules. Journal of food science and technology 52, 3158–3168. https://doi.org/10.1007/s13197-014-1487-3

Barone, E., Arena, A., Head, E., Butterfield, D.A., Perluigi, M., 2017. Disturbance of redox homeostasis in Down Syndrome: Role of iron dysmetabolism. Free radical biology & medicine. https://doi.org/10.1016/j.freeradbiomed.2017.07.009

N

U

Bar-Peled, O., Gross-Isseroff, R., Ben-Hur, H., Hoskins, I., Groner, Y., Biegon, A., 1991. Fetal human brain exhibits a prenatal peak in the density of serotonin 5-HT1A receptors. Neurosci. Lett. 127, 173–176.

M

A

Bartesaghi, R., Haydar, T.F., Delabar, J.M., Dierssen, M., Martinez-Cue, C., Bianchi, D.W., 2015. New Perspectives for the Rescue of Cognitive Disability in Down Syndrome. Journal of Neuroscience. https://doi.org/10.1523/JNEUROSCI.2775-15.2015

TE D

Beacher, F., Daly, E., Simmons, A., Prasher, V., Morris, R., Robinson, C., Lovestone, S., Murphy, K., Murphy, D.G.M., 2009. Alzheimer’s disease and Down’s syndrome: an in vivo MRI study. Psychol Med 39, 675–684. https://doi.org/10.1017/S0033291708004054

EP

Becker, W., Soppa, U., Tejedor, F.J., 2014. DYRK1A: a potential drug target for multiple Down syndrome neuropathologies. CNS & neurological disorders drug targets 13, 26–33.

CC

Beckervordersandforth, R., 2017. Mitochondrial Metabolism-Mediated Regulation of Adult Neurogenesis. Brain Plasticity 3, 73–87. https://doi.org/10.3233/BPL-170044

A

Begenisic, T., Baroncelli, L., Sansevero, G., Milanese, M., Bonifacino, T., Bonanno, G., Cioni, G., Maffei, L., Sale, A., 2014. Fluoxetine in adulthood normalizes GABA release and rescues hippocampal synaptic plasticity and spatial memory in a mouse model of Down Syndrome. Neurobiology of Disease 63, 12–19. https://doi.org/10.1016/j.nbd.2013.11.010 Begni, B., Brighina, L., Fumagalli, L., Andreoni, S., Castelli, E., Francesconi, C., Del Bo, R., Bresolin, N., Ferrarese, C., 2003. Altered glutamate uptake in peripheral tissues from Down syndrome patients. Neuroscience letters 343, 73–76. Bernstein, B.E., Meissner, A., Lander, E.S., 2007. The mammalian epigenome. Cell 128, 669–681. https://doi.org/10.1016/j.cell.2007.01.033 50

Best, T.K., Cramer, N.P., Chakrabarti, L., Haydar, T.F., Galdzicki, Z., 2012. Dysfunctional hippocampal inhibition in the Ts65Dn mouse model of Down syndrome. Experimental Neurology 233, 749–757. https://doi.org/10.1016/j.expneurol.2011.11.033 Best, T.K., Siarey, R.J., Galdzicki, Z., 2007. Ts65Dn, a Mouse Model of Down Syndrome, Exhibits Increased GABA B -Induced Potassium Current. Journal of Neurophysiology 97, 892–900. https://doi.org/10.1152/jn.00626.2006

IP T

Bianchi, P., Ciani, E., Guidi, S., Trazzi, S., Felice, D., Grossi, G., Fernandez, M., Giuliani, A., Calza, L., Bartesaghi, R., 2010. Early Pharmacotherapy Restores Neurogenesis and Cognitive Performance in the Ts65Dn Mouse Model for Down Syndrome. Journal of Neuroscience 30, 8769– 8779. https://doi.org/10.1523/JNEUROSCI.0534-10.2010

SC R

Birks, J., Harvey, R., 2003. Donepezil for dementia due to Alzheimer’s disease, in: The Cochrane Collaboration (Ed.), Cochrane Database of Systematic Reviews. John Wiley & Sons, Ltd, Chichester, UK. https://doi.org/10.1002/14651858.CD001190

U

Bittles, A.H., Glasson, E.J., 2004. Clinical, social, and ethical implications of changing life expectancy in Down syndrome. Developmental Medicine & Child Neurology 46, 282–286. https://doi.org/10.1111/j.1469-8749.2004.tb00483.x

M

A

N

Blehaut, H., Mircher, C., Ravel, A., Conte, M., de Portzamparc, V., Poret, G., Huon de Kermadec, F., Rethore, M.-O., Sturtz, F.G., 2010. Effect of Leucovorin (Folinic Acid) on the Developmental Quotient of Children with Down’s Syndrome (Trisomy 21) and Influence of Thyroid Status. PLoS ONE 5, e8394. https://doi.org/10.1371/journal.pone.0008394

TE D

Blom, H.J., Smulders, Y., 2011. Overview of homocysteine and folate metabolism. With special references to cardiovascular disease and neural tube defects. Journal of inherited metabolic disease 34, 75–81. https://doi.org/10.1007/s10545-010-9177-4

EP

Boada, R., Hutaff-Lee, C., Schrader, A., Weitzenkamp, D., Benke, T.A., Goldson, E.J., Costa, A.C.S., 2012. Antagonism of NMDA receptors as a potential treatment for Down syndrome: a pilot randomized controlled trial. Translational psychiatry 2, e141. https://doi.org/10.1038/tp.2012.66

CC

Bofill-De Ros, X., Santos, M., Vila-Casadesús, M., Villanueva, E., Andreu, N., Dierssen, M., Fillat, C., 2015. Genome-wide miR-155 and miR-802 target gene identification in the hippocampus of Ts65Dn Down syndrome mouse model by miRNA sponges. BMC genomics 16, 907. https://doi.org/10.1186/s12864-015-2160-6

A

Brás, A., Rodrigues, A., Gomes, B., Rueff, J., 2018. Down syndrome and microRNAs (Review). Biomedical Reports 8, 11–16. https://doi.org/10.3892/br.2017.1019 Braudeau, J., Delatour, B., Duchon, A., Pereira, P.L., Dauphinot, L., de Chaumont, F., Olivo-Marin, J.-C., Dodd, R.H., Hérault, Y., Potier, M.-C., 2011. Specific targeting of the GABA-A receptor α5 subtype by a selective inverse agonist restores cognitive deficits in Down syndrome mice. Journal of psychopharmacology (Oxford, England) 25, 1030–1042. https://doi.org/10.1177/0269881111405366 51

Brault, V., Duchon, A., Romestaing, C., Sahun, I., Pothion, S., Karout, M., Borel, C., Dembele, D., Bizot, J.-C., Messaddeq, N., Sharp, A.J., Roussel, D., Antonarakis, S.E., Dierssen, M., Hérault, Y., 2015. Opposite phenotypes of muscle strength and locomotor function in mouse models of partial trisomy and monosomy 21 for the proximal Hspa13-App region. PLoS Genet. 11, e1005062. https://doi.org/10.1371/journal.pgen.1005062 Bursavich, M.G., Harrison, B.A., Blain, J.-F., 2016. Gamma Secretase Modulators: New Alzheimer’s Drugs on the Horizon? Journal of Medicinal Chemistry 59, 7389–7409. https://doi.org/10.1021/acs.jmedchem.5b01960

SC R

IP T

Busciglio, J., Pelsman, A., Wong, C., Pigino, G., Yuan, M., Mori, H., Yankner, B.A., 2002. Altered metabolism of the amyloid beta precursor protein is associated with mitochondrial dysfunction in Down’s syndrome. Neuron 33, 677–688. Busciglio, J., Yankner, B.A., 1995. Apoptosis and increased generation of reactive oxygen species in Down’s syndrome neurons in vitro. Nature 378, 776–779. https://doi.org/10.1038/378776a0

U

Butterfield, D.A., Perluigi, M., 2018. Down syndrome: From development to adult life to Alzheimer disease. Free Radical Biology and Medicine 114, 1–2. https://doi.org/10.1016/j.freeradbiomed.2017.10.374

M

A

N

Campbell, R.M., Tummino, P.J., 2014. Cancer epigenetics drug discovery and development: the challenge of hitting the mark. Journal of Clinical Investigation 124, 64–69. https://doi.org/10.1172/JCI71605

TE D

Cannell, I.G., Kong, Y.W., Bushell, M., 2008. How do microRNAs regulate gene expression? Biochemical Society Transactions 36, 1224–1231. https://doi.org/10.1042/BST0361224 Carfì, A., Antocicco, M., Brandi, V., Cipriani, C., Fiore, F., Mascia, D., Settanni, S., Vetrano, D.L., Bernabei, R., Onder, G., 2014. Characteristics of Adults with Down Syndrome: Prevalence of AgeRelated Conditions. Frontiers in Medicine 1, 51. https://doi.org/10.3389/fmed.2014.00051

EP

Cartlidge, P.H., Curnock, D.A., 1986. Specific malabsorption of vitamin B12 in Down’s syndrome. Archives of Disease in Childhood 61, 514–515.

CC

Carvajal, F.J., Inestrosa, N.C., 2011. Interactions of AChE with A? Aggregates in Alzheimer?s Brain: Therapeutic Relevance of IDN 5706. Frontiers in Molecular Neuroscience 4. https://doi.org/10.3389/fnmol.2011.00019

A

Catalanotto, C., Cogoni, C., Zardo, G., 2016. MicroRNA in Control of Gene Expression: An Overview of Nuclear Functions. International journal of molecular sciences 17. https://doi.org/10.3390/ijms17101712 Catuara-Solarz, S., Espinosa-Carrasco, J., Erb, I., Langohr, K., Gonzalez, J.R., Notredame, C., Dierssen, M., 2016. Combined Treatment With Environmental Enrichment and (-)Epigallocatechin-3-Gallate Ameliorates Learning Deficits and Hippocampal Alterations in a Mouse Model of Down Syndrome. eNeuro 3, ENEURO.0103-16.2016. https://doi.org/10.1523/ENEURO.0103-16.2016 52

Chakrabarti, L., Best, T.K., Cramer, N.P., Carney, R.S.E., Isaac, J.T.R., Galdzicki, Z., Haydar, T.F., 2010. Olig1 and Olig2 triplication causes developmental brain defects in Down syndrome. Nature Neuroscience 13, 927–934. https://doi.org/10.1038/nn.2600 Chakrabarti, L., Galdzicki, Z., Haydar, T.F., 2007. Defects in Embryonic Neurogenesis and Initial Synapse Formation in the Forebrain of the Ts65Dn Mouse Model of Down Syndrome. Journal of Neuroscience 27, 11483–11495. https://doi.org/10.1523/JNEUROSCI.3406-07.2007

IP T

Chang, Q., Gold, P.E., 2008. Age-related changes in memory and in acetylcholine functions in the hippocampus in the Ts65Dn mouse, a model of Down syndrome. Neurobiol Learn Mem 89, 167– 177. https://doi.org/10.1016/j.nlm.2007.05.007

N

U

SC R

Chen, J.J., Liu, Q., Yuan, C., Gore, V., Lopez, P., Ma, V., Amegadzie, A., Qian, W., Judd, T.C., Minatti, A.E., Brown, J., Cheng, Y., Xue, M., Zhong, W., Dineen, T.A., Epstein, O., Human, J., Kreiman, C., Marx, I., Weiss, M.M., Hitchcock, S.A., Powers, T.S., Chen, K., Wen, P.H., Whittington, D.A., Cheng, A.C., Bartberger, M.D., Hickman, D., Werner, J.A., Vargas, H.M., Everds, N.E., Vonderfecht, S.L., Dunn, R.T., Wood, S., Fremeau, R.T., White, R.D., Patel, V.F., 2015. Development of 2-aminooxazoline 3-azaxanthenes as orally efficacious β-secretase inhibitors for the potential treatment of Alzheimer’s disease. Bioorganic & Medicinal Chemistry Letters 25, 767–774. https://doi.org/10.1016/j.bmcl.2014.12.092

M

A

Chen, N.C., Yang, F., Capecci, L.M., Gu, Z., Schafer, A.I., Durante, W., Yang, X.-F., Wang, H., 2010. Regulation of homocysteine metabolism and methylation in human and mouse tissues. FASEB journal : official publication of the Federation of American Societies for Experimental Biology 24, 2804–2817. https://doi.org/10.1096/fj.09-143651

TE D

Chisholm, K.M., 2018. Down syndrome. Atlas of Genetics and Cytogenetics in Oncology and Haematology. https://doi.org/10.4267/2042/68892

EP

Chowdhury, A., Sarkar, J., Chakraborti, T., Pramanik, P.K., Chakraborti, S., 2016. Protective role of epigallocatechin-3-gallate in health and disease: A perspective. Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie 78, 50–59. https://doi.org/10.1016/j.biopha.2015.12.013

CC

Christakos, S., DeLuca, H.F., 2011. Minireview: Vitamin D: is there a role in extraskeletal health? Endocrinology 152, 2930–2936. https://doi.org/10.1210/en.2011-0243

A

Chung, W.-S., Allen, N.J., Eroglu, C., 2015. Astrocytes Control Synapse Formation, Function, and Elimination. Cold Spring Harbor Perspectives in Biology 7, a020370. https://doi.org/10.1101/cshperspect.a020370 Clark, S., Schwalbe, J., Stasko, M.R., Yarowsky, P.J., Costa, A.C.S., 2006. Fluoxetine rescues deficient neurogenesis in hippocampus of the Ts65Dn mouse model for Down syndrome. Experimental neurology 200, 256–261. https://doi.org/10.1016/j.expneurol.2006.02.005 Clarke, L.E., Barres, B.A., 2013. Emerging roles of astrocytes in neural circuit development. Nat. Rev. Neurosci. 14, 311–321. https://doi.org/10.1038/nrn3484 Cohen Kadosh, K., Krause, B., King, A.J., Near, J., Cohen Kadosh, R., 2015. Linking GABA and 53

glutamate levels to cognitive skill acquisition during development. Human brain mapping 36, 4334–4345. https://doi.org/10.1002/hbm.22921 Contestabile, A., Fila, T., Ceccarelli, C., Bonasoni, P., Bonapace, L., Santini, D., Bartesaghi, R., Ciani, E., 2007. Cell cycle alteration and decreased cell proliferation in the hippocampal dentate gyrus and in the neocortical germinal matrix of fetuses with Down syndrome and in Ts65Dn mice. Hippocampus 17, 665–678. https://doi.org/10.1002/hipo.20308

IP T

Contestabile, A., Magara, S., Cancedda, L., 2017. The GABAergic Hypothesis for Cognitive Disabilities in Down Syndrome. Frontiers in cellular neuroscience 11, 54. https://doi.org/10.3389/fncel.2017.00054

SC R

Contestabile, A., Fila, T., Bartesaghi, R., Contestabile, Antonio, Ciani, E., 2006. Choline acetyltransferase activity at different ages in brain of Ts65Dn mice, an animal model for Down’s syndrome and related neurodegenerative diseases. J. Neurochem. 97, 515–526. https://doi.org/10.1111/j.1471-4159.2006.03769.x

N

U

Conti, A., Fabbrini, F., D’Agostino, P., Negri, R., Greco, D., Genesio, R., D’Armiento, M., Olla, C., Paladini, D., Zannini, M., Nitsch, L., 2007. Altered expression of mitochondrial and extracellular matrix genes in the heart of human fetuses with chromosome 21 trisomy. BMC genomics 8, 268. https://doi.org/10.1186/1471-2164-8-268

TE D

M

A

Coric, V., van Dyck, C.H., Salloway, S., Andreasen, N., Brody, M., Richter, R.W., Soininen, H., Thein, S., Shiovitz, T., Pilcher, G., Colby, S., Rollin, L., Dockens, R., Pachai, C., Portelius, E., Andreasson, U., Blennow, K., Soares, H., Albright, C., Feldman, H.H., Berman, R.M., 2012. Safety and tolerability of the gamma-secretase inhibitor avagacestat in a phase 2 study of mild to moderate Alzheimer disease. Archives of neurology 69, 1430–1440. https://doi.org/10.1001/archneurol.2012.2194

EP

Corrales, A., Martínez, P., García, S., Vidal, V., García, E., Flórez, J., Sanchez-Barceló, E.J., Martínez-Cué, C., Rueda, N., 2013. Long-term oral administration of melatonin improves spatial learning and memory and protects against cholinergic degeneration in middle-aged Ts65Dn mice, a model of Down syndrome. J. Pineal Res. 54, 346–358. https://doi.org/10.1111/jpi.12037

A

CC

Corrales, A., Parisotto, E.B., Vidal, V., García-Cerro, S., Lantigua, S., Diego, M., Wilhem Filho, D., Sanchez-Barceló, E.J., Martínez-Cué, C., Rueda, N., 2017. Pre- and post-natal melatonin administration partially regulates brain oxidative stress but does not improve cognitive or histological alterations in the Ts65Dn mouse model of Down syndrome. Behavioural Brain Research 334, 142–154. https://doi.org/10.1016/j.bbr.2017.07.022 Coskun, P., Helguera, P., Nemati, Z., Bohannan, R.C., Thomas, J., Samuel, S.E., Argueta, J., Doran, E., Wallace, D.C., Lott, I.T., Busciglio, J., 2017. Metabolic and Growth Rate Alterations in Lymphoblastic Cell Lines Discriminate Between Down Syndrome and Alzheimer’s Disease. Journal of Alzheimer’s disease : JAD 55, 737–748. https://doi.org/10.3233/JAD-160278 Coskun, P.E., Busciglio, J., 2012. Oxidative Stress and Mitochondrial Dysfunction in Down’s Syndrome: Relevance to Aging and Dementia. Current gerontology and geriatrics research 2012, 383170. https://doi.org/10.1155/2012/383170 54

Costa, A.C.S., Scott-McKean, J.J., Stasko, M.R., 2008. Acute injections of the NMDA receptor antagonist memantine rescue performance deficits of the Ts65Dn mouse model of Down syndrome on a fear conditioning test. Neuropsychopharmacology : official publication of the American College of Neuropsychopharmacology 33, 1624–1632. https://doi.org/10.1038/sj.npp.1301535

IP T

Costa, V., Angelini, C., D’Apice, L., Mutarelli, M., Casamassimi, A., Sommese, L., Gallo, M.A., Aprile, M., Esposito, R., Leone, L., Donizetti, A., Crispi, S., Rienzo, M., Sarubbi, B., Calabrò, R., Picardi, M., Salvatore, P., Infante, T., De Berardinis, P., Napoli, C., Ciccodicola, A., 2011. Massive-Scale RNA-Seq Analysis of Non Ribosomal Transcriptome in Human Trisomy 21. PLoS ONE 6, e18493. https://doi.org/10.1371/journal.pone.0018493

SC R

Cramer, N., Galdzicki, Z., 2012a. From abnormal hippocampal synaptic plasticity in down syndrome mouse models to cognitive disability in down syndrome. Neural plasticity 2012, 101542. https://doi.org/10.1155/2012/101542

U

Cramer, N., Galdzicki, Z., 2012b. From abnormal hippocampal synaptic plasticity in down syndrome mouse models to cognitive disability in down syndrome. Neural Plasticity. https://doi.org/10.1155/2012/101542

N

Créau, N., 2012. Molecular and Cellular Alterations in Down Syndrome: Toward the Identification of Targets for Therapeutics. Neural Plasticity 2012, 1–14. https://doi.org/10.1155/2012/171639

TE D

M

A

Cremon, C., Stanghellini, V., Barbaro, M.R., Cogliandro, R.F., Bellacosa, L., Santos, J., Vicario, M., Pigrau, M., Alonso Cotoner, C., Lobo, B., Azpiroz, F., Bruley des Varannes, S., Neunlist, M., DeFilippis, D., Iuvone, T., Petrosino, S., Di Marzo, V., Barbara, G., 2017. Randomised clinical trial: the analgesic properties of dietary supplementation with palmitoylethanolamide and polydatin in irritable bowel syndrome. Alimentary Pharmacology & Therapeutics 45, 909–922. https://doi.org/10.1111/apt.13958

EP

Crider, K.S., Yang, T.P., Berry, R.J., Bailey, L.B., 2012. Folate and DNA methylation: a review of molecular mechanisms and the evidence for folate’s role. Advances in nutrition (Bethesda, Md.) 3, 21–38. https://doi.org/10.3945/an.111.000992

CC

Das, I., Park, J.-M., Shin, J.H., Jeon, S.K., Lorenzi, H., Linden, D.J., Worley, P.F., Reeves, R.H., 2013. Hedgehog Agonist Therapy Corrects Structural and Cognitive Deficits in a Down Syndrome Mouse Model. Science Translational Medicine 5, 201ra120-201ra120. https://doi.org/10.1126/scitranslmed.3005983

A

Dashinimaev, E.B., Artyuhov, A.S., Bolshakov, A.P., Vorotelyak, E.A., Vasiliev, A.V., 2017. Neurons Derived from Induced Pluripotent Stem Cells of Patients with Down Syndrome Reproduce Early Stages of Alzheimer’s Disease Type Pathology in vitro. Journal of Alzheimer’s Disease 56, 835–847. https://doi.org/10.3233/JAD-160945 Daubert, E.A., Condron, B.G., 2010. Serotonin: a regulator of neuronal morphology and circuitry. Trends in Neurosciences 33, 424–434. https://doi.org/10.1016/j.tins.2010.05.005 Davis, M.A.C., Spriggs, A., Rodgers, A., Campbell, J., 2018. The Effects of a Peer-Delivered Social Skills Intervention for Adults with Comorbid Down Syndrome and Autism Spectrum 55

Disorder. Journal of Autism and Developmental Disorders 48, 1869–1885. https://doi.org/10.1007/s10803-017-3437-1 Davisson, M.T., 2005. Mouse models of Down syndrome. Drug Discovery Today: Disease Models. https://doi.org/10.1016/j.ddmod.2005.05.007

IP T

De Filippis, B., Valenti, D., Chiodi, V., Ferrante, A., de Bari, L., Fiorentini, C., Domenici, M.R., Ricceri, L., Vacca, R.A., Fabbri, A., Laviola, G., 2015. Modulation of Rho GTPases rescues brain mitochondrial dysfunction, cognitive deficits and aberrant synaptic plasticity in female mice modeling Rett syndrome. European Neuropsychopharmacology 25, 889–901. https://doi.org/10.1016/j.euroneuro.2015.03.012

SC R

de Haan, J.B., Cristiano, F., Iannello, R.C., Kola, I., 1995. Cu/Zn-superoxide dismutase and glutathione peroxidase during aging. Biochemistry and molecular biology international 35, 1281– 1297.

A

N

U

de la Torre, R., de Sola, S., Hernandez, G., Farre, M., Pujol, J., Rodriguez, J., Espadaler, J.M., Langohr, K., Cuenca-Royo, A., Principe, A., Xicota, L., Janel, N., Catuara-Solarz, S., SanchezBenavides, G., Blehaut, H., Duenas-Espin, I., Del Hoyo, L., Benejam, B., Blanco-Hinojo, L., Videla, S., Fito, M., Delabar, J.M., Dierssen, M., 2016. 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. The Lancet. Neurology 15, 801–810. https://doi.org/10.1016/S1474-4422(16)30034-5

TE D

M

De la Torre, R., De Sola, S., Pons, M., Duchon, A., de Lagran, M.M., Farré, M., Fitó, M., Benejam, B., Langohr, K., Rodriguez, J., Pujadas, M., Bizot, J.C., Cuenca, A., Janel, N., Catuara, S., Covas, M.I., Blehaut, H., Herault, Y., Delabar, J.M., Dierssen, M., 2014. Epigallocatechin-3-gallate, a DYRK1A inhibitor, rescues cognitive deficits in Down syndrome mouse models and in humans. Mol Nutr Food Res 58, 278–288. https://doi.org/10.1002/mnfr.201300325

EP

de la Torre, R., Dierssen, M., 2012. Therapeutic approaches in the improvement of cognitive performance in Down syndrome, in: Progress in Brain Research. Elsevier, pp. 1–14. https://doi.org/10.1016/B978-0-444-54299-1.00001-7

CC

Deidda, G., Bozarth, I.F., Cancedda, L., 2014. Modulation of GABAergic transmission in development and neurodevelopmental disorders: investigating physiology and pathology to gain therapeutic perspectives. Frontiers in Cellular Neuroscience 8. https://doi.org/10.3389/fncel.2014.00119

A

Deidda, G., Parrini, M., Naskar, S., Bozarth, I.F., Contestabile, A., Cancedda, L., 2015. Reversing excitatory GABAAR signaling restores synaptic plasticity and memory in a mouse model of Down syndrome. Nature medicine 21, 318–326. https://doi.org/10.1038/nm.3827 Dekker, A.D., De Deyn, P.P., Rots, M.G., 2014. Epigenetics: the neglected key to minimize learning and memory deficits in Down syndrome. Neurosci Biobehav Rev 45, 72–84. https://doi.org/10.1016/j.neubiorev.2014.05.004 Dekker, A.D., Sacco, S., Carfi, A., Benejam, B., Vermeiren, Y., Beugelsdijk, G., Schippers, M., 56

Hassefras, L., Eleveld, J., Grefelman, S., Fopma, R., Bomer-Veenboer, M., Boti, M., Oosterling, G.D.E., Scholten, E., Tollenaere, M., Checkley, L., Strydom, A., Van Goethem, G., Onder, G., Blesa, R., zu Eulenburg, C., Coppus, A.M.W., Rebillat, A.-S., Fortea, J., De Deyn, P.P., 2018. The Behavioral and Psychological Symptoms of Dementia in Down Syndrome (BPSD-DS) Scale: Comprehensive Assessment of Psychopathology in Down Syndrome. Journal of Alzheimer’s Disease 63, 797–819. https://doi.org/10.3233/JAD-170920

IP T

Delabar, J.M., Theophile, D., Rahmani, Z., Chettouh, Z., Blouin, J.L., Prieur, M., Noel, B., Sinet, P.M., 1993. Molecular mapping of twenty-four features of Down syndrome on chromosome 21. European journal of human genetics: EJHG 1, 114–124. Demarin, V., Morović, S., 2014. Neuroplasticity. Periodicum biologorum 116, 209–211.

U

SC R

Di Domenico, F., Coccia, R., Cocciolo, A., Murphy, M.P., Cenini, G., Head, E., Butterfield, D.A., Giorgi, A., Schinina, M.E., Mancuso, C., Cini, C., Perluigi, M., 2013. Impairment of proteostasis network in Down syndrome prior to the development of Alzheimer’s disease neuropathology: Redox proteomics analysis of human brain. Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease 1832, 1249–1259. https://doi.org/10.1016/j.bbadis.2013.04.013

A

N

Di Domenico, F., Tramutola, A., Foppoli, C., Head, E., Perluigi, M., Butterfield, D.A., 2018. mTOR in Down syndrome: Role in Aß and tau neuropathology and transition to Alzheimer diseaselike dementia. Free Radical Biology and Medicine 114, 94–101. https://doi.org/10.1016/j.freeradbiomed.2017.08.009

TE D

M

Di Rosso, M.E., Palumbo, M.L., Genaro, A.M., 2016. Immunomodulatory effects of fluoxetine: A new potential pharmacological action for a classic antidepressant drug? Pharmacological Research 109, 101–107. https://doi.org/10.1016/j.phrs.2015.11.021 Diamandopoulos, K., Green, J., 2018. Down syndrome: An integrative review. Journal of Neonatal Nursing 24, 235–241. https://doi.org/10.1016/j.jnn.2018.01.001

EP

Dick, M.B., Doran, E., Phelan, M., Lott, I.T., 2016. Cognitive Profiles on the Severe Impairment Battery Are Similar in Alzheimer Disease and Down Syndrome With Dementia. Alzheimer disease and associated disorders 30, 251–257. https://doi.org/10.1097/WAD.0000000000000132

CC

Dierssen, M., 2012. Down syndrome: The brain in trisomic mode. Nature Reviews Neuroscience. https://doi.org/10.1038/nrn3314

A

Dierssen, M., Benavides-Piccione, R., Martínez-Cué, C., Estivill, X., Flórez, J., Elston, G.N., DeFelipe, J., 2003. Alterations of neocortical pyramidal cell phenotype in the Ts65Dn mouse model of Down syndrome: effects of environmental enrichment. Cerebral cortex (New York, N.Y. : 1991). http://dx.doi.org/10.1093/cercor/13.7.758 Di Vona, C., Bezdan, D., Islam, A.B.M.M.K., Salichs, E., López-Bigas, N., Ossowski, S., de la Luna, S., 2015. Chromatin-wide Profiling of DYRK1A Reveals a Role as a Gene-Specific RNA Polymerase II CTD Kinase. Molecular Cell 57, 506–520. https://doi.org/10.1016/j.molcel.2014.12.026 57

Doody, R.S., Raman, R., Farlow, M., Iwatsubo, T., Vellas, B., Joffe, S., Kieburtz, K., He, F., Sun, X., Thomas, R.G., Aisen, P.S., Siemers, E., Sethuraman, G., Mohs, R., 2013. A phase 3 trial of semagacestat for treatment of Alzheimer’s disease. The New England journal of medicine 369, 341–350. https://doi.org/10.1056/NEJMoa1210951 Druzhyna, N., Nair, R.G., LeDoux, S.P., Wilson, G.L., 1998. Defective repair of oxidative damage in mitochondrial DNA in Down’s syndrome. Mutation research 409, 81–89.

IP T

Du, Q.-H., Peng, C., Zhang, H., 2013. Polydatin: A review of pharmacology and pharmacokinetics. Pharmaceutical Biology 51, 1347–1354. https://doi.org/10.3109/13880209.2013.792849

SC R

Duchon, A., Herault, Y., 2016. DYRK1A, a Dosage-Sensitive Gene Involved in Neurodevelopmental Disorders, Is a Target for Drug Development in Down Syndrome. Frontiers in Behavioral Neuroscience 10. https://doi.org/10.3389/fnbeh.2016.00104 Duclot, F., Lapierre, M., Fritsch, S., White, R., Parker, M.G., Maurice, T., Cavaillès, V., 2012. Cognitive impairments in adult mice with constitutive inactivation of RIP140 gene expression. Genes, Brain and Behavior 11, 69–78. https://doi.org/10.1111/j.1601-183X.2011.00731.x

A

N

U

Eichenlaub-Ritter, U., Wieczorek, M., Luke, S., Seidel, T., 2011. Age related changes in mitochondrial function and new approaches to study redox regulation in mammalian oocytes in response to age or maturation conditions. Mitochondrion 11, 783–796. https://doi.org/10.1016/j.mito.2010.08.011

TE D

M

Ellis, J.M., Tan, H.K., Gilbert, R.E., Muller, D.P.R., Henley, W., Moy, R., Pumphrey, R., Ani, C., Davies, S., Edwards, V., Green, H., Salt, A., Logan, S., 2008. Supplementation with antioxidants and folinic acid for children with Down’s syndrome: randomised controlled trial. BMJ 336, 594– 597. https://doi.org/10.1136/bmj.39465.544028.AE Engevik, L.I., Næss, K.-A.B., Hagtvet, B.E., 2016. Cognitive stimulation of pupils with Down syndrome: A study of inferential talk during book-sharing. Research in Developmental Disabilities 55, 287–300. https://doi.org/10.1016/j.ridd.2016.05.004

CC

EP

Fantini, M., Benvenuto, M., Masuelli, L., Frajese, G.V., Tresoldi, I., Modesti, A., Bei, R., 2015. In vitro and in vivo antitumoral effects of combinations of polyphenols, or polyphenols and anticancer drugs: perspectives on cancer treatment. International journal of molecular sciences 16, 9236–9282. https://doi.org/10.3390/ijms16059236

A

Fernandez, F., Morishita, W., Zuniga, E., Nguyen, J., Blank, M., Malenka, R.C., Garner, C.C., 2007. Pharmacotherapy for cognitive impairment in a mouse model of Down syndrome. Nature neuroscience 10, 411–413. https://doi.org/10.1038/nn1860 Fritah, A., Christian, M., Parker, M.G., 2010. The metabolic coregulator RIP140: an update. American Journal of Physiology-Endocrinology and Metabolism 299, E335–E340. https://doi.org/10.1152/ajpendo.00243.2010 Fuchs, E., Flügge, G., 2014. Adult neuroplasticity: More than 40 years of research. Neural Plasticity. https://doi.org/10.1155/2014/541870 58

Fuentes, J.J., Genescà, L., Kingsbury, T.J., Cunningham, K.W., Pérez-Riba, M., Estivill, X., de la Luna, S., 2000. DSCR1, overexpressed in Down syndrome, is an inhibitor of calcineurin-mediated signaling pathways. Human Molecular Genetics 9, 1681–1690. Gane, B, Bhat, V, 2014. Folate and Homocysteine metabolism in Indian children with Down syndrome. Curr. Pediatr.

IP T

Gao, Y., Chen, T., Lei, Xianghui, Li, Y., Dai, X., Cao, Y., Ding, Q., Lei, Xiabao, Li, T., Lin, X., 2016. Neuroprotective effects of polydatin against mitochondrial-dependent apoptosis in the rat cerebral cortex following ischemia/reperfusion injury. Molecular Medicine Reports 14, 5481–5488. https://doi.org/10.3892/mmr.2016.5936

SC R

Garcia, O., Torres, M., Helguera, P., Coskun, P., Busciglio, J., 2010. A role for thrombospondin-1 deficits in astrocyte-mediated spine and synaptic pathology in down’s syndrome. PLoS ONE. https://doi.org/10.1371/journal.pone.0014200

U

García-Cerro, S., Martínez, P., Vidal, V., Corrales, A., Flórez, J., Vidal, R., Rueda, N., Arbonés, M.L., Martínez-Cué, C., 2014. Overexpression of Dyrk1A is implicated in several cognitive, electrophysiological and neuromorphological alterations found in a mouse model of Down syndrome. PLoS ONE 9, e106572. https://doi.org/10.1371/journal.pone.0106572

M

A

N

García-Vallejo, F., Ortiz, A.R.R., Gómez, C.A., Ospina, M.S., Villegas, J.C.M., Gómez, A.S., Soto, J.M.S., 2018. Functional Neurogenomics: A New Approach to Study Cognitive Disability in Down Syndrome Brain, in: Advances in Research on Down Syndrome. InTech.

TE D

Gardiner, K., 2006. Transcriptional dysregulation in Down syndrome: predictions for altered protein complex stoichiometries and post-translational modifications, and consequences for learning/behavior genes ELK, CREB, and the estrogen and glucocorticoid receptors. Behav. Genet. 36, 439–453. https://doi.org/10.1007/s10519-006-9051-1

EP

Gardiner, K., Herault, Y., Lott, I.T., Antonarakis, S.E., Reeves, R.H., Dierssen, M., 2010. Down Syndrome: From Understanding the Neurobiology to Therapy. Journal of Neuroscience. https://doi.org/10.1523/JNEUROSCI.3728-10.2010

CC

Ghosh, A.K., Brindisi, M., Tang, J., 2012. Developing β-secretase inhibitors for treatment of Alzheimer’s disease: β-Secretase inhibitor drugs. Journal of Neurochemistry 120, 71–83. https://doi.org/10.1111/j.1471-4159.2011.07476.x

A

Gimeno, A., García-Giménez, J.L., Audí, L., Toran, N., Andaluz, P., Dasí, F., Viña, J., Pallardó, F. V., 2014. Decreased cell proliferation and higher oxidative stress in fibroblasts from down syndrome fetuses. Preliminary study. Biochimica et Biophysica Acta - Molecular Basis of Disease. https://doi.org/10.1016/j.bbadis.2013.10.014 Giunta, B., Hou, H., Zhu, Y., Salemi, J., Ruscin, A., Shytle, R.D., Tan, J., 2010. Fish oil enhances anti-amyloidogenic properties of green tea EGCG in Tg2576 mice. Neuroscience Letters 471, 134– 138. https://doi.org/10.1016/j.neulet.2010.01.026 Godridge, H., Reynolds, G.P., Czudek, C., Calcutt, N.A., Benton, M., 1987. Alzheimer-like 59

neurotransmitter deficits in adult Down’s syndrome brain tissue. Journal of neurology, neurosurgery, and psychiatry 50, 775–778. Goldberg, D.M., Yan, J., Soleas, G.J., 2003. Absorption of three wine-related polyphenols in three different matrices by healthy subjects. Clinical Biochemistry 36, 79–87.

IP T

Granese, B., Scala, I., Spatuzza, C., Valentino, A., Coletta, M., Vacca, R.A., De Luca, P., Andria, G., 2013. Validation of microarray data in human lymphoblasts shows a role of the ubiquitinproteasome system and NF-kB in the pathogenesis of Down syndrome. BMC medical genomics 6, 24. https://doi.org/10.1186/1755-8794-6-24

SC R

Granholm, A.C., Sanders, L.A., Crnic, L.S., 2000. Loss of cholinergic phenotype in basal forebrain coincides with cognitive decline in a mouse model of Down’s syndrome. Exp. Neurol. 161, 647– 663. https://doi.org/10.1006/exnr.1999.7289 Grootaert, C., Kamiloglu, S., Capanoglu, E., Van Camp, J., 2015. Cell Systems to Investigate the Impact of Polyphenols on Cardiovascular Health. Nutrients 7, 9229–9255. https://doi.org/10.3390/nu7115462

N

U

Grynberg, M., Graesslin, O., Teyssedre, J., Quereux, C., Gaillard, D., Carré-Pigeon, F., 2007. Prenatal diagnosis in monozygotic twins with Down syndrome who had different phenotypes. Prenatal Diagnosis 27, 552–554. https://doi.org/10.1002/pd.1711

M

A

Guedj, F., Sébrié, C., Rivals, I., Ledru, A., Paly, E., Bizot, J.C., Smith, D., Rubin, E., Gillet, B., Arbones, M., Delabar, J.M., 2009. Green tea polyphenols rescue of brain defects induced by overexpression of DYRK1A. PLoS ONE 4, e4606. https://doi.org/10.1371/journal.pone.0004606

TE D

Guidi, S., Bonasoni, P., Ceccarelli, C., Santini, D., Gualtieri, F., Ciani, E., Bartesaghi, R., 2008. Neurogenesis impairment and increased cell death reduce total neuron number in the hippocampal region of fetuses with Down syndrome. Brain Pathol. 18, 180–197. https://doi.org/10.1111/j.17503639.2007.00113.x

CC

EP

Guidi, S., Stagni, F., Bianchi, P., Ciani, E., Giacomini, A., De Franceschi, M., Moldrich, R., Kurniawan, N., Mardon, K., Giuliani, A., Calzà, L., Bartesaghi, R., 2014. Prenatal pharmacotherapy rescues brain development in a Down’s syndrome mouse model. Brain 137, 380–401. https://doi.org/10.1093/brain/awt340

A

Guidi, S., Stagni, F., Bianchi, P., Ciani, E., Ragazzi, E., Trazzi, S., Grossi, G., Mangano, C., Calza, L., Bartesaghi, R., 2013. Early pharmacotherapy with fluoxetine rescues dendritic pathology in the Ts65Dn mouse model of down syndrome. Brain pathology (Zurich, Switzerland) 23, 129–143. https://doi.org/10.1111/j.1750-3639.2012.00624.x Guo, X., Williams, J.G., Schug, T.T., Li, X., 2010. DYRK1A and DYRK3 Promote Cell Survival through Phosphorylation and Activation of SIRT1. Journal of Biological Chemistry 285, 13223– 13232. https://doi.org/10.1074/jbc.M110.102574 Gupta, M., Dhanasekaran, A.R., Gardiner, K.J., 2016. Mouse models of Down syndrome: gene content and consequences. Mammalian Genome. https://doi.org/10.1007/s00335-016-9661-8 60

Halliwell, B., 1991. Reactive oxygen species in living systems: source, biochemistry, and role in human disease. Am. J. Med. 91, 14S-22S. Hanney, M., Prasher, V., Williams, N., Jones, E.L., Aarsland, D., Corbett, A., Lawrence, D., Yu, L.-M., Tyrer, S., Francis, P.T., Johnson, T., Bullock, R., Ballard, C., 2012. Memantine for dementia in adults older than 40 years with Down’s syndrome (MEADOWS): a randomised, double-blind, placebo-controlled trial. Lancet (London, England) 379, 528–536. https://doi.org/10.1016/S01406736(11)61676-0

SC R

IP T

Hanson, J.E., Weber, M., Meilandt, W.J., Wu, T., Luu, T., Deng, L., Shamloo, M., Sheng, M., Scearce-Levie, K., Zhou, Q., 2013. GluN2B Antagonism Affects Interneurons and Leads to Immediate and Persistent Changes in Synaptic Plasticity, Oscillations and Behavior. Neuropsychopharmacology 38, 1221–1233. https://doi.org/10.1038/npp.2013.19

U

Harashima, C., Jacobowitz, D.M., Witta, J., Borke, R.C., Best, T.K., Siarey, R.J., Galdzicki, Z., 2006. Abnormal expression of the G-protein-activated inwardly rectifying potassium channel 2 (GIRK2) in hippocampus, frontal cortex, and substantia nigra of Ts65Dn mouse: A model of Down syndrome. The Journal of Comparative Neurology 494, 815–833. https://doi.org/10.1002/cne.20844

TE D

M

A

N

Hattori, M., Fujiyama, A., Taylor, T.D., Watanabe, H., Yada, T., Park, H.-S., Toyoda, A., Ishii, K., Totoki, Y., Choi, D.-K., Soeda, E., Ohki, M., Takagi, T., Sakaki, Y., Taudien, S., Blechschmidt, K., Polley, A., Menzel, U., Delabar, J., Kumpf, K., Lehmann, R., Patterson, D., Reichwald, K., Rump, A., Schillhabel, M., Schudy, A., Zimmermann, W., Rosenthal, A., Kudoh, J., Shibuya, K., Kawasaki, K., Asakawa, S., Shintani, A., Sasaki, T., Nagamine, K., Mitsuyama, S., Antonarakis, S.E., Minoshima, S., Shimizu, N., Nordsiek, G., Hornischer, K., Brandt, P., Scharfe, M., Schön, O., Desario, A., Reichelt, J., Kauer, G., Blöcker, H., Ramser, J., Beck, A., Klages, S., Hennig, S., Riesselmann, L., Dagand, E., Haaf, T., Wehrmeyer, S., Borzym, K., Gardiner, K., Nizetic, D., Francis, F., Lehrach, H., Reinhardt, R., Yaspo, M.-L., 2000. The DNA sequence of human chromosome 21. Nature 405, 311–319. https://doi.org/10.1038/35012518

EP

Head, E., Lott, I.T., Wilcock, D.M., Lemere, C.A., 2016. Aging in Down syndrome and the Development of Alzheimer’s disease Neuropathology. Current Alzheimer research 13, 18–29.

CC

Helguera, P., 2005. ets-2 Promotes the Activation of a Mitochondrial Death Pathway in Down’s Syndrome Neurons. Journal of Neuroscience 25, 2295–2303. https://doi.org/10.1523/JNEUROSCI.5107-04.2005

A

Helguera, P., Seiglie, J., Rodriguez, J., Hanna, M., Helguera, G., Busciglio, J., 2013. Adaptive downregulation of mitochondrial function in down syndrome. Cell metabolism 17, 132–140. https://doi.org/10.1016/j.cmet.2012.12.005 Herbison, A.E., Moenter, S.M., 2011. Depolarising and Hyperpolarising Actions of GABAA Receptor Activation on Gonadotrophin-Releasing Hormone Neurones: Towards an Emerging Consensus: GABAA modulation of GnRH neurones. Journal of Neuroendocrinology 23, 557–569. https://doi.org/10.1111/j.1365-2826.2011.02145.x Hernández-Camacho, J.D., Bernier, M., López-Lluch, G., Navas, P., 2018. Coenzyme Q10 Supplementation in Aging and Disease. Frontiers in Physiology 9. 61

https://doi.org/10.3389/fphys.2018.00044 Hibaoui, Y., Grad, I., Letourneau, A., Sailani, M.R., Dahoun, S., Santoni, F.A., Gimelli, S., Guipponi, M., Pelte, M.F., Béna, F., Antonarakis, S.E., Feki, A., 2014. Modelling and rescuing neurodevelopmental defect of Down syndrome using induced pluripotent stem cells from monozygotic twins discordant for trisomy 21. EMBO Molecular Medicine 6, 259–277. https://doi.org/10.1002/emmm.201302848

IP T

Holmes, G., 2014. Gastrointestinal disorders in Down syndrome. Gastroenterol Hepatol Bed Bench 7, 6–8.

SC R

Hoyer, D., Clarke, D.E., Fozard, J.R., Hartig, P.R., Martin, G.R., Mylecharane, E.J., Saxena, P.R., Humphrey, P.P., 1994. International Union of Pharmacology classification of receptors for 5hydroxytryptamine (Serotonin). Pharmacol. Rev. 46, 157–203. Idowu, A.J., Kumar, S.L., Yidong, B., Russel, R., 2017. Melatonin modulates neuronal mitochondria function during normal ageing in mice. Nigerian Journal of Physiological Sciences: Official Publication of the Physiological Society of Nigeria 32, 145–152.

A

N

U

Infantino, V., Castegna, A., Iacobazzi, F., Spera, I., Scala, I., Andria, G., Iacobazzi, V., 2011. Impairment of methyl cycle affects mitochondrial methyl availability and glutathione level in Down’s syndrome. Molecular Genetics and Metabolism 102, 378–382. https://doi.org/10.1016/j.ymgme.2010.11.166

TE D

M

Izzo, A., Manco, R., Cristofaro, T. de, Bonfiglio, F., Cicatiello, R., Mollo, N., Martino, M. De, Genesio, R., Zannini, M., Conti, A., Nitsch, L., 2017a. Overexpression of Chromosome 21 miRNAs May Affect Mitochondrial Function in the Hearts of Down Syndrome Fetuses. International Journal of Genomics 2017, 1–10. https://doi.org/10.1155/2017/8737649

EP

Izzo, A., Mollo, N., Nitti, M., Paladino, S., Calì, G., Genesio, R., Bonfiglio, F., Cicatiello, R., Barbato, M., Sarnataro, V., Conti, A., Nitsch, L., 2018. Mitochondrial dysfunction in down syndrome: molecular mechanisms and therapeutic targets. Molecular Medicine 24. https://doi.org/10.1186/s10020-018-0004-y

A

CC

Izzo, A., Nitti, M., Mollo, N., Paladino, S., Procaccini, C., Faicchia, D., Calì, G., Genesio, R., Bonfiglio, F., Cicatiello, R., Polishchuk, E., Polishchuk, R., Pinton, P., Matarese, G., Conti, A., Nitsch, L., 2017b. Metformin restores the mitochondrial network and reverses mitochondrial dysfunction in Down syndrome cells. Hum. Mol. Genet. 26, 1056–1069. https://doi.org/10.1093/hmg/ddx016 James, S.J., Pogribna, M., Pogribny, I.P., Melnyk, S., Hine, R.J., Gibson, J.B., Yi, P., Tafoya, D.L., Swenson, D.H., Wilson, V.L., Gaylor, D.W., 1999. Abnormal folate metabolism and mutation in the methylenetetrahydrofolate reductase gene may be maternal risk factors for Down syndrome. The American journal of clinical nutrition 70, 495–501. Jin, S., Lee, Y.K., Lim, Y.C., Zheng, Z., Lin, X.M., Ng, D.P.Y., Holbrook, J.D., Law, H.Y., Kwek, K.Y.C., Yeo, G.S.H., Ding, C., 2013. Global DNA hypermethylation in down syndrome placenta. PLoS genetics 9, e1003515. https://doi.org/10.1371/journal.pgen.1003515 62

Jobe, E.M., Zhao, X., 2017. DNA Methylation and Adult Neurogenesis. Brain Plasticity 3, 5–26. https://doi.org/10.3233/BPL-160034 Joshi, K., Shen, L., Michaeli, A., Salter, M., Thibault-Messier, G., Hashmi, S., Eubanks, J.H., Cortez, M.A., Snead, O.C., 2016. Infantile spasms in down syndrome: Rescue by knockdown of the GIRK2 channel: GIRK2. Annals of Neurology 80, 511–521. https://doi.org/10.1002/ana.24749

IP T

Józefczuk, J., Kasprzycka, W., Czarnecki, R., Graczyk, A., Józefczuk, P., Krzysztof, M., Lampart, U., Mrozowska-Ząbek, E., Surdy, W., Kwiatkowska-Graczyk, R., 2017. Bioelements in hair of children with selected neurological disorders. Acta Biochimica Polonica 64, 279–285. https://doi.org/10.18388/abp.2016_1380

SC R

Jung, M.-S., Park, J.-H., Ryu, Y.S., Choi, S.-H., Yoon, S.-H., Kwen, M.-Y., Oh, J.Y., Song, W.-J., Chung, S.-H., 2011. Regulation of RCAN1 Protein Activity by Dyrk1A Protein-mediated Phosphorylation. Journal of Biological Chemistry 286, 40401–40412. https://doi.org/10.1074/jbc.M111.253971

U

Kadrabová, J., Madáriĉ, A., Šustrová, M., Ginter, E., 1996. Changed serum trace element profile in Down’s syndrome. Biological Trace Element Research 54, 201–206. https://doi.org/10.1007/BF02784431

M

A

N

Kamhieh-Milz, J., Moftah, R.F.H., Bal, G., Futschik, M., Sterzer, V., Khorramshahi, O., Burow, M., Thiel, G., Stuke-Sontheimer, A., Chaoui, R., Kamhieh-Milz, S., Salama, A., 2014. Differentially expressed microRNAs in maternal plasma for the noninvasive prenatal diagnosis of Down syndrome (trisomy 21). BioMed research international 2014, 402475. https://doi.org/10.1155/2014/402475

TE D

Kanaumi, T., Ichinohe, A., Kimura, H., Iwasaki, H., Hirose, S., Takashima, S., 2006. Development and Aging Expression of Cystathionine-Beta Synthase in the Temporal Lobe and Cerebellum of Down Syndrome Patients. Neuroembryology and Aging 4, 202–207. https://doi.org/10.1159/000118930

CC

EP

Kaur, G., Sharma, A., Xu, W., Gerum, S., Alldred, M.J., Subbanna, S., Basavarajappa, B.S., Pawlik, M., Ohno, M., Ginsberg, S.D., Wilson, D.A., Guilfoyle, D.N., Levy, E., 2014. Glutamatergic Transmission Aberration: A Major Cause of Behavioral Deficits in a Murine Model of Down’s Syndrome. Journal of Neuroscience 34, 5099–5106. https://doi.org/10.1523/JNEUROSCI.5338-13.2014

A

Kava, M.P., Tullu, M.S., Muranjan, M.N., Girisha, K.., 2004. Down syndrome: Archives of Medical Research 35, 31–35. https://doi.org/10.1016/j.arcmed.2003.06.005 Kays, J.L., Hurley, R. a, Taber, K.H., 2012. The Dynamic Brain: Neuroplasticity and Mental Health. The Journal of Neuropsychiatry and Clinical Neurosciences. https://doi.org/10.1176/appi.neuropsych.12050109 Kazemi, M., Salehi, M., Kheirollahi, M., 2016. Down Syndrome: Current Status, Challenges and Future Perspectives. International Journal of Molecular and Cellular Medicine 5, 125–133. 63

Kerkel, K., Schupf, N., Hatta, K., Pang, D., Salas, M., Kratz, A., Minden, M., Murty, V., Zigman, W.B., Mayeux, R.P., Jenkins, E.C., Torkamani, A., Schork, N.J., Silverman, W., Croy, B.A., Tycko, B., 2010. Altered DNA methylation in leukocytes with trisomy 21. PLoS genetics 6, e1001212. https://doi.org/10.1371/journal.pgen.1001212 Khacho, M., Slack, R.S., 2018. Mitochondrial dynamics in the regulation of neurogenesis: From development to the adult brain: Mitochondrial Dynamics and Brain Neurogenesis. Developmental Dynamics 247, 47–53. https://doi.org/10.1002/dvdy.24538

IP T

Kim, S.H., Cairns, N., Fountoulakisc, M., Lubec, G., 2001. Decreased brain histamine-releasing factor protein in patients with Down syndrome and Alzheimer’s disease. Neuroscience Letters 300, 41–44.

SC R

Kirby, R.S., 2017. The prevalence of selected major birth defects in the United States. Seminars in Perinatology 41, 338–344. https://doi.org/10.1053/j.semperi.2017.07.004

U

Kishnani, P.S., Heller, J.H., Spiridigliozzi, G.A., Lott, I., Escobar, L., Richardson, S., Zhang, R., McRae, T., 2010. Donepezil for treatment of cognitive dysfunction in children with Down syndrome aged 10-17. American Journal of Medical Genetics Part A 152A, 3028–3035. https://doi.org/10.1002/ajmg.a.33730

A

N

Kitamura, T., Saitoh, Y., Takashima, N., Murayama, A., Niibori, Y., Ageta, H., Sekiguchi, M., Sugiyama, H., Inokuchi, K., 2009. Adult Neurogenesis Modulates the Hippocampus-Dependent Period of Associative Fear Memory. Cell 139, 814–827. https://doi.org/10.1016/j.cell.2009.10.020

TE D

M

Kıray, H., Lindsay, S.L., Hosseinzadeh, S., Barnett, S.C., 2016. The multifaceted role of astrocytes in regulating myelination. Experimental Neurology 283, 541–549. https://doi.org/10.1016/j.expneurol.2016.03.009 Kleim, J.A., Jones, T.A., 2008. Principles of Experience-Dependent Neural Plasticity: Implications for Rehabilitation After Brain Damage. Journal of Speech Language and Hearing Research. https://doi.org/10.1044/1092-4388(2008/018)

CC

EP

Kleschevnikov, A.M., Belichenko, P.V., Gall, J., George, L., Nosheny, R., Maloney, M.T., Salehi, A., Mobley, W.C., 2012. Increased efficiency of the GABAA and GABAB receptor-mediated neurotransmission in the Ts65Dn mouse model of Down syndrome. Neurobiol. Dis. 45, 683–691. https://doi.org/10.1016/j.nbd.2011.10.009

A

Kondoh, T., Kanno, A., Itoh, H., Nakashima, M., Honda, R., Kojima, M., Noguchi, M., Nakane, H., Nozaki, H., Sasaki, H., Nagai, T., Kosaki, R., Kakee, N., Okuyama, T., Fukuda, M., Ikeda, M., Shibata, Y., Moriuchi, H., 2011. Donepezil significantly improves abilities in daily lives of female Down syndrome patients with severe cognitive impairment: a 24-week randomized, double-blind, placebo-controlled trial. Int J Psychiatry Med 41, 71–89. https://doi.org/10.2190/PM.41.1.g Koyrakh, L., 2005. Molecular and Cellular Diversity of Neuronal G-Protein-Gated Potassium Channels. Journal of Neuroscience 25, 11468–11478. https://doi.org/10.1523/JNEUROSCI.348405.2005 64

Krinsky-McHale, S.J., Silverman, W., 2013. Dementia and mild cognitive impairment in adults with intellectual disability: Issues of diagnosis: Dementia and Mild Cognitive Impairment. Developmental Disabilities Research Reviews 18, 31–42. https://doi.org/10.1002/ddrr.1126 Kuehn, B.M., 2016. Treating trisomies: Prenatal Down’s syndrome therapies explored in mice. Nature Medicine 22, 6–7. https://doi.org/10.1038/nm0116-6

IP T

Kurt, M.A., Davies, D.C., Kidd, M., Dierssen, M., Flórez, J., 2000. Synaptic deficit in the temporal cortex of partial trisomy 16 (Ts65Dn) mice. Brain Research 858, 191–197. https://doi.org/10.1016/S0006-8993(00)01984-3

SC R

Latruffe, N., Lancon, A., Frazzi, R., Aires, V., Delmas, D., Michaille, J.-J., Djouadi, F., Bastin, J., Cherkaoui-Malki, M., 2015. Exploring new ways of regulation by resveratrol involving miRNAs, with emphasis on inflammation. Annals of the New York Academy of Sciences 1348, 97–106. https://doi.org/10.1111/nyas.12819

U

Lee, J.H., Lee, A.J., Dang, L.-H., Pang, D., Kisselev, S., Krinsky-McHale, S.J., Zigman, W.B., Luchsinger, J.A., Silverman, W., Tycko, B., Clark, L.N., Schupf, N., 2017. Candidate gene analysis for Alzheimer’s disease in adults with Down syndrome. Neurobiology of Aging 56, 150–158. https://doi.org/10.1016/j.neurobiolaging.2017.04.018

A

N

Lejeune, J., Gauthier, M., Turpin, R., 1959. [Human chromosomes in tissue cultures]. C. R. Hebd. Seances Acad. Sci. 248, 602–603.

TE D

M

Lepagnol-Bestel, A.-M., Zvara, A., Maussion, G., Quignon, F., Ngimbous, B., Ramoz, N., Imbeaud, S., Loe-Mie, Y., Benihoud, K., Agier, N., Salin, P.A., Cardona, A., Khung-Savatovsky, S., Kallunki, P., Delabar, J.-M., Puskas, L.G., Delacroix, H., Aggerbeck, L., Delezoide, A.-L., Delattre, O., Gorwood, P., Moalic, J.-M., Simonneau, M., 2009. DYRK1A interacts with the REST/NRSF-SWI/SNF chromatin remodelling complex to deregulate gene clusters involved in the neuronal phenotypic traits of Down syndrome. Human Molecular Genetics 18, 1405–1414. https://doi.org/10.1093/hmg/ddp047

CC

EP

Letourneau, A., Santoni, F.A., Bonilla, X., Sailani, M.R., Gonzalez, D., Kind, J., Chevalier, C., Thurman, R., Sandstrom, R.S., Hibaoui, Y., Garieri, M., Popadin, K., Falconnet, E., Gagnebin, M., Gehrig, C., Vannier, A., Guipponi, M., Farinelli, L., Robyr, D., Migliavacca, E., Borel, C., Deutsch, S., Feki, A., Stamatoyannopoulos, J.A., Herault, Y., van Steensel, B., Guigo, R., Antonarakis, S.E., 2014. Domains of genome-wide gene expression dysregulation in Down’s syndrome. Nature 508, 345–350. https://doi.org/10.1038/nature13200

A

Lewanda, A.F., Gallegos, M.F., Summar, M., 2018. Patterns of Dietary Supplement Use in Children with Down Syndrome. The Journal of Pediatrics. https://doi.org/10.1016/j.jpeds.2018.05.022 Licastro, F., Marocchi, A., Penco, S., Porcellini, E., Lio, D., Dogliotti, G., Corsi, M.M., 2006. Does Down’s syndrome support the homocysteine theory of atherogenesis?: Experience in elderly subjects with trisomy 21. Archives of gerontology and geriatrics 43, 381–387. Lin, A.-L., Powell, D., Caban-Holt, A., Jicha, G., Robertson, W., Gold, B.T., Davis, R., Abner, E., 65

Wilcock, D.M., Schmitt, F.A., Head, E., 2016. 1 H-MRS metabolites in adults with Down syndrome: Effects of dementia. NeuroImage: Clinical 11, 728–735. https://doi.org/10.1016/j.nicl.2016.06.001 Lipton, S.A., 2004. Paradigm shift in NMDA receptor antagonist drug development: molecular mechanism of uncompetitive inhibition by memantine in the treatment of Alzheimer’s disease and other neurologic disorders. J. Alzheimers Dis. 6, S61-74.

IP T

Liu, Y., Borel, C., Li, L., Müller, T., Williams, E.G., Germain, P.-L., Buljan, M., Sajic, T., Boersema, P.J., Shao, W., Faini, M., Testa, G., Beyer, A., Antonarakis, S.E., Aebersold, R., 2017. Systematic proteome and proteostasis profiling in human Trisomy 21 fibroblast cells. Nature Communications 8. https://doi.org/10.1038/s41467-017-01422-6

SC R

Liyanage, V.R.B., Rastegar, M., 2014. Rett Syndrome and MeCP2. NeuroMolecular Medicine 16, 231–264. https://doi.org/10.1007/s12017-014-8295-9

A

N

U

Loane, M., Morris, J.K., Addor, M.-C., Arriola, L., Budd, J., Doray, B., Garne, E., Gatt, M., Haeusler, M., Khoshnood, B., Klungsoyr Melve, K., Latos-Bielenska, A., McDonnell, B., Mullaney, C., O’Mahony, M., Queisser-Wahrendorf, A., Rankin, J., Rissmann, A., Rounding, C., Salvador, J., Tucker, D., Wellesley, D., Yevtushok, L., Dolk, H., 2013. Twenty-year trends in the prevalence of Down syndrome and other trisomies in Europe: impact of maternal age and prenatal screening. European journal of human genetics : EJHG 21, 27–33. https://doi.org/10.1038/ejhg.2012.94

TE D

M

Lockrow, J., Boger, H., Gerhardt, G., Aston-Jones, G., Bachman, D., Granholm, A.-C., 2011. A noradrenergic lesion exacerbates neurodegeneration in a Down syndrome mouse model. Journal of Alzheimer’s disease : JAD 23, 471–489. https://doi.org/10.3233/JAD-2010-101218 Lockstone, H.E., Harris, L.W., Swatton, J.E., Wayland, M.T., Holland, A.J., Bahn, S., 2007. Gene expression profiling in the adult Down syndrome brain. Genomics 90, 647–660. https://doi.org/10.1016/j.ygeno.2007.08.005

CC

EP

London, J., Rouch, C., Bui, L.C., Assayag, E., Souchet, B., Daubigney, F., Medjaoui, H., Luquet, S., Magnan, C., Delabar, J.M., Dairou, J., Janel, N., 2018. Overexpression of the DYRK1A Gene (Dual-Specificity Tyrosine Phosphorylation-Regulated Kinase 1A) Induces Alterations of the Serotoninergic and Dopaminergic Processing in Murine Brain Tissues. Molecular Neurobiology 55, 3822–3831. https://doi.org/10.1007/s12035-017-0591-6

A

Lu, J., Lian, G., Zhou, H., Esposito, G., Steardo, L., Delli-Bovi, L.C., Hecht, J.L., Lu, Q.R., Sheen, V., 2012. OLIG2 over-expression impairs proliferation of human Down syndrome neural progenitors. Hum. Mol. Genet. 21, 2330–2340. https://doi.org/10.1093/hmg/dds052 Lüscher, C., Jan, L.Y., Stoffel, M., Malenka, R.C., Nicoll, R.A., 1997. G Protein-Coupled Inwardly Rectifying K+ Channels (GIRKs) Mediate Postsynaptic but Not Presynaptic Transmitter Actions in Hippocampal Neurons. Neuron 19, 687–695. https://doi.org/10.1016/S0896-6273(00)80381-5 Lysenko, L.V., Kim, J., Henry, C., Tyrtyshnaia, A., Kohnz, R.A., Madamba, F., Simon, G.M., Kleschevnikova, N.E., Nomura, D.K., Ezekowitz, R.. A.B., Kleschevnikov, A.M., 2014. 66

Monoacylglycerol Lipase Inhibitor JZL184 Improves Behavior and Neural Properties in Ts65Dn Mice, a Model of Down Syndrome. PLoS ONE 9, e114521. https://doi.org/10.1371/journal.pone.0114521 Määttä, T., Tervo-Määttä, T., Taanila, A., Kaski, M., Iivanainen, M., 2006. Mental health, behaviour and intellectual abilities of people with Down syndrome. Down Syndrome Research and Practice 11, 37–43. https://doi.org/10.3104/reports.313

IP T

Mao, R., Zielke, C.L., Zielke, H.R., Pevsner, J., 2003. Global up-regulation of chromosome 21 gene expression in the developing Down syndrome brain. Genomics 81, 457–467. Marin‐Padilla, M., 1976. Pyramidal cell abnormalities in the motor cortex of a child with Down’s

SC R

syndrome. A Golgi study. Journal of Comparative Neurology 167, 63–81.

U

Martin, K.R., Corlett, A., Dubach, D., Mustafa, T., Coleman, H.A., Parkington, H.C., Merson, T.D., Bourne, J.A., Porta, S., Arbonés, M.L., Finkelstein, D.I., Pritchard, M.A., 2012. Over-expression of RCAN1 causes Down syndrome-like hippocampal deficits that alter learning and memory. Human Molecular Genetics 21, 3025–3041. https://doi.org/10.1093/hmg/dds134

A

N

Martin, K.R., Layton, D., Seach, N., Corlett, A., Barallobre, M.J., Arbonés, M.L., Boyd, R.L., Scott, B., Pritchard, M.A., 2013. Upregulation of RCAN1 causes Down syndrome-like immune dysfunction. Journal of Medical Genetics 50, 444–454. https://doi.org/10.1136/jmedgenet-2013101522

TE D

M

Martinez-Cue, C., Martinez, P., Rueda, N., Vidal, R., Garcia, S., Vidal, V., Corrales, A., Montero, J.A., Pazos, A., Florez, J., Gasser, R., Thomas, A.W., Honer, M., Knoflach, F., Trejo, J.L., Wettstein, J.G., Hernandez, M.-C., 2013. Reducing GABAA 5 Receptor-Mediated Inhibition Rescues Functional and Neuromorphological Deficits in a Mouse Model of Down Syndrome. Journal of Neuroscience 33, 3953–3966. https://doi.org/10.1523/JNEUROSCI.1203-12.2013

EP

Martínez-Cué, C., Rueda, N., García, E., Davisson, M.T., Schmidt, C., Flórez, J., 2005. Behavioral, cognitive and biochemical responses to different environmental conditions in male Ts65Dn mice, a model of Down syndrome. Behav. Brain Res. 163, 174–185. https://doi.org/10.1016/j.bbr.2005.04.016

A

CC

McKenzie, K., Milton, M., Smith, G., Ouellette-Kuntz, H., 2016. Systematic Review of the Prevalence and Incidence of Intellectual Disabilities: Current Trends and Issues. Current Developmental Disorders Reports 3, 104–115. https://doi.org/10.1007/s40474-016-0085-7 Medina, M., Urdiales, J.L., Amores-Sánchez, M.I., 2001. Roles of homocysteine in cell metabolism: old and new functions. European Journal of Biochemistry 268, 3871–3882. Meguid, N.A., Kholoussi, N.M., Afifi, H.H., 2001. Evaluation of superoxide dismutase and glutathione peroxidase enzymes and their cofactors in egyptian children with down’s syndrome. Biological Trace Element Research 81, 21–28. https://doi.org/10.1385/BTER:81:1:21 Mentis, A.F., 2016. Epigenomic engineering for Down syndrome. Neuroscience & Biobehavioral Reviews 71, 323–327. https://doi.org/10.1016/j.neubiorev.2016.09.012 67

Miles, M. V, Patterson, B.J., Chalfonte-Evans, M.L., Horn, P.S., Hickey, F.J., Schapiro, M.B., Steele, P.E., Tang, P.H., Hotze, S.L., 2007. Coenzyme Q10 (ubiquinol-10) supplementation improves oxidative imbalance in children with trisomy 21. Pediatric neurology 37, 398–403. https://doi.org/10.1016/j.pediatrneurol.2007.08.003 Miller, A.L., 2003. The methionine-homocysteine cycle and its effects on cognitive diseases. Alternative medicine review : a journal of clinical therapeutic 8, 7–19.

IP T

Miller, E.R. 3rd, Pastor-Barriuso, R., Dalal, D., Riemersma, R.A., Appel, L.J., Guallar, E., 2005. Meta-analysis: high-dosage vitamin E supplementation may increase all-cause mortality. Annals of internal medicine 142, 37–46.

SC R

Mircher, C., Salabelle, A., Peeters, M.A., Rabier, D., Parvy, P., Kamoun, P., Lejeune, J., 1997. [Variation of amino acids in relation to age in Down syndrome subjects]. Archives de pediatrie : organe officiel de la Societe francaise de pediatrie 4, 1093–1099.

U

Miyata, M., Maruo, T., Kaito, A., Wang, S., Yamamoto, H., Fujiwara, T., Mizoguchi, A., Mandai, K., Takai, Y., 2017. Roles of afadin in the formation of the cellular architecture of the mouse hippocampus and dentate gyrus. Molecular and Cellular Neuroscience 79, 34–44. https://doi.org/10.1016/j.mcn.2016.12.007

M

A

N

Mo, J.-J., Li, J., Yang, Z., Liu, Z., Feng, J.-S., 2017. Efficacy and safety of anti-amyloid- β immunotherapy for Alzheimer’s disease: a systematic review and network meta-analysis. Annals of Clinical and Translational Neurology 4, 931–942. https://doi.org/10.1002/acn3.469

TE D

Mohler, H., 2012. Cognitive enhancement by pharmacological and behavioral interventions: the murine Down syndrome model. Biochemical pharmacology 84, 994–999. https://doi.org/10.1016/j.bcp.2012.06.028 Moore, L.D., Le, T., Fan, G., 2013. DNA methylation and its basic function. Neuropsychopharmacology : official publication of the American College of Neuropsychopharmacology 38, 23–38. https://doi.org/10.1038/npp.2012.112

CC

EP

Morice, E., Andreae, L.C., Cooke, S.F., Vanes, L., Fisher, E.M.C., Tybulewicz, V.L.J., Bliss, T.V.P., 2008. Preservation of long-term memory and synaptic plasticity despite short-term impairments in the Tc1 mouse model of Down syndrome. Learning & Memory 15, 492–500. https://doi.org/10.1101/lm.969608

A

Mundkur, N., 2005. Neuroplasticity in children, in: Indian Journal of Pediatrics. https://doi.org/10.1007/BF02731115 Mustafa Nachvak, S., Reza Neyestani, T., Ali Mahboob, S., Sabour, S., Ali Keshawarz, S., Speakman, J.R., 2014. alpha-Tocopherol supplementation reduces biomarkers of oxidative stress in children with Down syndrome: a randomized controlled trial. European journal of clinical nutrition 68, 1119–1123. https://doi.org/10.1038/ejcn.2014.97 Nabavi, S.F., Sureda, A., Dehpour, A.R., Shirooie, S., Silva, A.S., Devi, K.P., Ahmed, T., Ishaq, N., Hashim, R., Sobarzo-Sánchez, E., Daglia, M., Braidy, N., Volpicella, M., Vacca, R.A., Nabavi, 68

S.M., 2018. Regulation of autophagy by polyphenols: Paving the road for treatment of neurodegeneration. Biotechnology Advances 36(6), 1768-1778. https://doi.org/10.1016/j.biotechadv.2017.12.001 Nahar, R., Kotecha, U., Puri, R.D., Pandey, R.M., Verma, I.C., 2013. Survival Analysis of Down Syndrome Cohort in a Tertiary Health Care Center in India. The Indian Journal of Pediatrics 80, 118–123. https://doi.org/10.1007/s12098-012-0836-3

IP T

Nakano-Kobayashi, A., Awaya, T., Kii, I., Sumida, Y., Okuno, Y., Yoshida, S., Sumida, T., Inoue, H., Hosoya, T., Hagiwara, M., 2017. Prenatal neurogenesis induction therapy normalizes brain structure and function in Down syndrome mice. Proceedings of the National Academy of Sciences of the United States of America 114, 10268–10273. https://doi.org/10.1073/pnas.1704143114

SC R

Negida, A., Menshawy, A., El Ashal, G., Elfouly, Y., Hani, Y., Hegazy, Y., El Ghonimy, S., Fouda, S., Rashad, Y., 2016. Coenzyme Q10 for Patients with Parkinson’s Disease: A Systematic Review and Meta-Analysis. CNS & neurological disorders drug targets 15, 45–53.

U

Netzer, W.J., Powell, C., Nong, Y., Blundell, J., Wong, L., Duff, K., Flajolet, M., Greengard, P., 2010. Lowering beta-amyloid levels rescues learning and memory in a Down syndrome mouse model. PloS one 5, e10943. https://doi.org/10.1371/journal.pone.0010943

M

A

N

Neumann, F., Gourdain, S., Albac, C., Dekker, A.D., Bui, L.C., Dairou, J., Schmitz-Afonso, I., Hue, N., Rodrigues-Lima, F., Delabar, J.M., Potier, M.-C., Le Caer, J.-P., Touboul, D., Delatour, B., Cariou, K., Dodd, R.H., 2018. DYRK1A inhibition and cognitive rescue in a Down syndrome mouse model are induced by new fluoro-DANDY derivatives. Scientific reports 8, 2859. https://doi.org/10.1038/s41598-018-20984-z

EP

TE D

Nguyen, T.L., Duchon, A., Manousopoulou, A., Loaec, N., Villiers, B., Pani, G., Karatas, M., Mechling, A.E., Harsan, L.-A., Limanton, E., Bazureau, J.-P., Carreaux, F., Garbis, S.D., Meijer, L., Herault, Y., 2018. Correction of cognitive deficits in mouse models of Down syndrome by a pharmacological inhibitor of DYRK1A. Disease models & mechanisms 11. https://doi.org/10.1242/dmm.035634

CC

Obeid, R., Hartmuth, K., Herrmann, W., Gortner, L., Rohrer, T.R., Geisel, J., Reed, M.C., Nijhout, H.F., 2012. Blood biomarkers of methylation in Down syndrome and metabolic simulations using a mathematical model. Molecular nutrition & food research 56, 1582–1589. https://doi.org/10.1002/mnfr.201200162

A

Obermann-Borst, S.A., van Driel, L.M.J.W., Helbing, W.A., de Jonge, R., Wildhagen, M.F., Steegers, E.A.P., Steegers-Theunissen, R.P.M., 2011. Congenital heart defects and biomarkers of methylation in children: a case-control study: CONGENITAL HEART DEFECTS AND BIOMARKERS OF METHYLATION. European Journal of Clinical Investigation 41, 143–150. https://doi.org/10.1111/j.1365-2362.2010.02388.x Ohira, M., Seki, N., Nagase, T., Suzuki, E., Nomura, N., Ohara, O., Hattori, M., Sakaki, Y., Eki, T., Murakami, Y., Saito, T., Ichikawa, H., Ohki, M., 1997. Gene identification in 1.6-Mb region of the Down syndrome region on chromosome 21. Genome Research 7, 47–58. https://doi.org/10.1101/gr.7.1.47 69

Olsen, R.W., Sieghart, W., 2009. GABAA receptors: Subtypes provide diversity of function and pharmacology. Neuropharmacology 56, 141–148. https://doi.org/10.1016/j.neuropharm.2008.07.045 Opitz, J.M., Gilbert-Barness, E.F., 1990. Reflections on the pathogenesis of Down syndrome. American journal of medical genetics. Supplement 7, 38–51.

IP T

Padgett, C.L., Slesinger, P.A., 2010. GABAB Receptor Coupling to G-proteins and Ion Channels, in: Advances in Pharmacology. Elsevier, pp. 123–147. https://doi.org/10.1016/S10543589(10)58006-2

SC R

Paola, D., Domenicotti, C., Nitti, M., Vitali, A., Borghi, R., Cottalasso, D., Zaccheo, D., Odetti, P., Strocchi, P., Marinari, U.M., Tabaton, M., Pronzato, M.A., 2000. Oxidative Stress Induces Increase in Intracellular Amyloid β-Protein Production and Selective Activation of βI and βII PKCs in NT2 Cells. Biochemical and Biophysical Research Communications 268, 642–646. https://doi.org/10.1006/bbrc.2000.2164

N

U

Parisotto, E.B., Vidal, V., García-Cerro, S., Lantigua, S., Wilhelm Filho, D., Sanchez-Barceló, E.J., Martínez-Cué, C., Rueda, N., 2016. Chronic Melatonin Administration Reduced Oxidative Damage and Cellular Senescence in the Hippocampus of a Mouse Model of Down Syndrome. Neurochemical Research 41, 2904–2913. https://doi.org/10.1007/s11064-016-2008-8

M

A

Park, J., Oh, Y., Yoo, L., Jung, M.-S., Song, W.-J., Lee, S.-H., Seo, H., Chung, K.C., 2010. Dyrk1A Phosphorylates p53 and Inhibits Proliferation of Embryonic Neuronal Cells. Journal of Biological Chemistry 285, 31895–31906. https://doi.org/10.1074/jbc.M110.147520

TE D

Park, J., Song, W.-J., Chung, K.C., 2009. Function and regulation of Dyrk1A: towards understanding Down syndrome. Cellular and molecular life sciences: CMLS 66, 3235–3240. https://doi.org/10.1007/s00018-009-0123-2

EP

Parsons, C.G., Stöffler, A., Danysz, W., 2007. Memantine: a NMDA receptor antagonist that improves memory by restoration of homeostasis in the glutamatergic system - too little activation is bad, too much is even worse. Neuropharmacology. https://doi.org/10.1016/j.neuropharm.2007.07.013

A

CC

Patel, A., Yamashita, N., Ascaño, M., Bodmer, D., Boehm, E., Bodkin-Clarke, C., Ryu, Y.K., Kuruvilla, R., 2015. RCAN1 links impaired neurotrophin trafficking to aberrant development of the sympathetic nervous system in Down syndrome. Nature Communications 6. https://doi.org/10.1038/ncomms10119 Patterson, D., 2008. Folate metabolism and the risk of Down syndrome. Down’s syndrome, research and practice : the journal of the Sarah Duffen Centre 12, 93–97. https://doi.org/10.3104/updates.2051 Pelleri, M.C., Cattani, C., Vitale, L., Antonaros, F., Strippoli, P., Locatelli, C., Cocchi, G., Piovesan, A., Caracausi, M., 2018. Integrated Quantitative Transcriptome Maps of Human Trisomy 21 Tissues and Cells. Frontiers in Genetics 9. https://doi.org/10.3389/fgene.2018.00125 70

Pelleri, M.C., Cicchini, E., Locatelli, C., Vitale, L., Caracausi, M., Piovesan, A., Rocca, A., Poletti, G., Seri, M., Strippoli, P., Cocchi, G., 2016. Systematic reanalysis of partial trisomy 21 cases with or without Down syndrome suggests a small region on 21q22.13 as critical to the phenotype. Human Molecular Genetics 25, 2525–2538. https://doi.org/10.1093/hmg/ddw116

IP T

Pereira, P.L., Magnol, L., Sahun, I., Brault, V., Duchon, A., Prandini, P., Gruart, A., Bizot, J.-C., Chadefaux-Vekemans, B., Deutsch, S., Trovero, F., Delgado-Garcia, J.M., Antonarakis, S.E., Dierssen, M., Herault, Y., 2009. A new mouse model for the trisomy of the Abcg1-U2af1 region reveals the complexity of the combinatorial genetic code of down syndrome. Human molecular genetics 18, 4756–4769. https://doi.org/10.1093/hmg/ddp438

SC R

Perluigi, M., Butterfield, D.A., 2012. Oxidative Stress and Down Syndrome: A Route toward Alzheimer-Like Dementia. Current gerontology and geriatrics research 2012, 724904. https://doi.org/10.1155/2012/724904

U

Phillips, C., Fahimi, A., Das, D., Mojabi, F.S., Ponnusamy, R., Salehi, A., 2016. Noradrenergic System in Down Syndrome and Alzheimer’s Disease A Target for Therapy. Curr Alzheimer Res 13, 68–83.

M

A

N

Piccoli, C., Izzo, A., Scrima, R., Bonfiglio, F., Manco, R., Negri, R., Quarato, G., Cela, O., Ripoli, M., Prisco, M., Gentile, F., Cali, G., Pinton, P., Conti, A., Nitsch, L., Capitanio, N., 2013. Chronic pro-oxidative state and mitochondrial dysfunctions are more pronounced in fibroblasts from Down syndrome foeti with congenital heart defects. Human molecular genetics 22, 1218–1232. https://doi.org/10.1093/hmg/dds529

TE D

Pinard, A., Seddik, R., Bettler, B., 2010. GABAB Receptors: Physiological Functions and Mechanisms of Diversity, in: Advances in Pharmacology. Elsevier, pp. 231–255. Pogribna, M., Melnyk, S., Pogribny, I., Chango, A., Yi, P., James, S.J., 2001. Homocysteine Metabolism in Children with Down Syndrome: In Vitro Modulation. The American Journal of Human Genetics 69, 88–95. https://doi.org/10.1086/321262

CC

EP

Pons-Espinal, M., Martinez de Lagran, M., Dierssen, M., 2013. Environmental enrichment rescues DYRK1A activity and hippocampal adult neurogenesis in TgDyrk1A. Neurobiology of Disease. https://doi.org/10.1016/j.nbd.2013.08.008

A

Potier, M.-C., Braudeau, J., Dauphinot, L., Delatour, B., 2014. Reducing GABAergic inhibition restores cognitive functions in a mouse model of Down syndrome. CNS & neurological disorders drug targets 13, 8–15. Pueschel, S.M., 1990. Clinical aspects of Down syndrome from infancy to adulthood. American journal of medical genetics. Supplement 7, 52–56. Pujol, J., Fenoll, R., Ribas-Vidal, N., Martínez-Vilavella, G., Blanco-Hinojo, L., García-Alba, J., Deus, J., Novell, R., Esteba-Castillo, S., 2018. A longitudinal study of brain anatomy changes preceding dementia in Down syndrome. NeuroImage: Clinical 18, 160–166. https://doi.org/10.1016/j.nicl.2018.01.024 71

PURPURA, D.P., 1975. Normal and aberrant neuronal development in the cerebral cortex of human fetus and young infant, in: Brain Mechanisms in Mental Retardation. Elsevier, pp. 141–169. Quinones-Lombrana, A., Blanco, J.G., 2015. Chromosome 21-derived hsa-miR-155-5p regulates mitochondrial biogenesis by targeting Mitochondrial Transcription Factor A (TFAM). Biochimica et biophysica acta 1852, 1420–1427. https://doi.org/10.1016/j.bbadis.2015.04.004

IP T

Rachidi, M., Lopes, C., 2011. Mental Retardation and Human Chromosome 21 Gene Overdosage: From Functional Genomics and Molecular Mechanisms Towards Prevention and Treatment of the Neuropathogenesis of Down Syndrome, in: Clelland, J.D. (Ed.), Genomics, Proteomics, and the Nervous System. Springer New York, New York, NY, pp. 21–86. https://doi.org/10.1007/978-14419-7197-5_2

SC R

Rachidi, M., Lopes, C., 2008. Mental retardation and associated neurological dysfunctions in Down syndrome: a consequence of dysregulation in critical chromosome 21 genes and associated molecular pathways. European journal of paediatric neurology : EJPN : official journal of the European Paediatric Neurology Society 12, 168–182. https://doi.org/10.1016/j.ejpn.2007.08.010

A

N

U

Rafii, M.S., Wishnek, H., Brewer, J.B., Donohue, M.C., Ness, S., Mobley, W.C., Aisen, P.S., Rissman, R.A., 2015. The down syndrome biomarker initiative (DSBI) pilot: proof of concept for deep phenotyping of Alzheimer’s disease biomarkers in down syndrome. Frontiers in behavioral neuroscience 9, 239. https://doi.org/10.3389/fnbeh.2015.00239

M

Raha, S., Robinson, B.H., 2000. Mitochondria, oxygen free radicals, disease and ageing. Trends Biochem. Sci. 25, 502–508.

TE D

Ravagnan, G., De Filippis, A., Cartenì, M., De Maria, S., Cozza, V., Petrazzuolo, M., Tufano, M.A., Donnarumma, G., 2013. Polydatin, A Natural Precursor of Resveratrol, Induces β-Defensin Production and Reduces Inflammatory Response. Inflammation 36, 26–34. https://doi.org/10.1007/s10753-012-9516-8

EP

Reeves, R.H., Baxter, L.L., Richtsmeier, J.T., 2001. Too much of a good thing: mechanisms of gene action in Down syndrome. Trends in genetics : TIG 17, 83–88.

CC

Reeves, R.H., Irving, N.G., Moran, T.H., Wohn, A., Kitt, C., Sisodia, S.S., Schmidt, C., Bronson, R.T., Davisson, M.T., 1995. A mouse model for Down syndrome exhibits learning and behaviour deficits. Nature Genetics 11, 177–184. https://doi.org/10.1038/ng1095-177

A

Reiter, R., Tan, D., Rosales-Corral, S., Galano, A., Zhou, X., Xu, B., 2018. Mitochondria: Central Organelles for Melatonin′s Antioxidant and Anti-Aging Actions. Molecules 23, 509. https://doi.org/10.3390/molecules23020509 Reynolds, G.P., Warner, C.E.J., 1988. Amino acid neurotransmitter deficits in adult Down’s syndrome brain tissue. Neuroscience Letters 94, 224–227. https://doi.org/10.1016/03043940(88)90299-6 Riediger, C., Schuster, T., Barlinn, K., Maier, S., Weitz, J., Siepmann, T., 2017. Adverse Effects of Antidepressants for Chronic Pain: A Systematic Review and Meta-analysis. Frontiers in Neurology 72

8. https://doi.org/10.3389/fneur.2017.00307 Risser, D., Lubec, G., Cairns, N., Herrera-Marschitz, M., 1997. Excitatory amino acids and monoamines in parahippocampal gyrus and frontal cortical pole of adults with Down syndrome. Life sciences 60, 1231–1237. Rodríguez-Sureda, V., Vilches, Á., Sánchez, O., Audí, L., Domínguez, C., 2015. Intracellular oxidant activity, antioxidant enzyme defense system, and cell senescence in fibroblasts with trisomy 21. Oxidative Medicine and Cellular Longevity 2015, 509241. https://doi.org/10.1155/2015/509241

IP T

Roizen, N.J., Patterson, D., 2003. Down’s syndrome. Lancet (London, England) 361, 1281–1289. https://doi.org/10.1016/S0140-6736(03)12987-X

SC R

Rudolf, G., Tul, N., Verdenik, I., Volk, M., Brezigar, A., Kokalj Vokac, N., Jersin, N., Prosenc, B., Premru Srsen, T., Peterlin, B., 2017. Impact of prenatal screening on the prevalence of Down syndrome in Slovenia. PloS one 12, e0180348. https://doi.org/10.1371/journal.pone.0180348

U

Rueda, N., Flórez, J., Martínez-Cué, C., 2012. Mouse models of down syndrome as a tool to unravel the causes of mental disabilities. Neural Plasticity. https://doi.org/10.1155/2012/584071

A

N

Rueda, N., Llorens-Martín, M., Flórez, J., Valdizán, E., Banerjee, P., Trejo, J.L., Martínez-Cué, C., 2010. Memantine Normalizes Several Phenotypic Features in the Ts65Dn Mouse Model of Down Syndrome. Journal of Alzheimer’s Disease 21, 277–290. https://doi.org/10.3233/JAD-2010-100240

M

Ruge, D., Liou, L.-M., Hoad, D., 2012. Improving the Potential of Neuroplasticity. Journal of Neuroscience. https://doi.org/10.1523/JNEUROSCI.0430-12.2012

TE D

Ruiz-Mejias, M., Martinez de Lagran, M., Mattia, M., Castano-Prat, P., Perez-Mendez, L., CiriaSuarez, L., Gener, T., Sancristobal, B., García-Ojalvo, J., Gruart, A., Delgado-García, J.M., Sanchez-Vives, M. V., Dierssen, M., 2016. Overexpression of Dyrk1A , a Down Syndrome Candidate, Decreases Excitability and Impairs Gamma Oscillations in the Prefrontal Cortex. The Journal of Neuroscience 36, 3648–3659. https://doi.org/10.1523/JNEUROSCI.2517-15.2016

CC

EP

Ruparelia, A., Wiseman, F., Sheppard, O., Tybulewicz, V.L.J., Fisher, E.M.C., 2010. Down syndrome and the molecular pathogenesis resulting from trisomy of human chromosome 21. Journal of biomedical research 24, 87–99. https://doi.org/10.1016/S1674-8301(10)60016-4

A

Sabbagh, M.N., Chen, K., Rogers, J., Fleisher, A.S., Liebsack, C., Bandy, D., Belden, C., Protas, H., Thiyyagura, P., Liu, X., Roontiva, A., Luo, J., Jacobson, S., Malek-Ahmadi, M., Powell, J., Reiman, E.M., 2015. Florbetapir PET, FDG PET, and MRI in Down syndrome individuals with and without Alzheimer’s dementia. Alzheimers Dement 11, 994–1004. https://doi.org/10.1016/j.jalz.2015.01.006 Saghazadeh, A., Mahmoudi, M., Dehghani Ashkezari, A., Oliaie Rezaie, N., Rezaei, N., 2017. Systematic review and meta-analysis shows a specific micronutrient profile in people with Down Syndrome: Lower blood calcium, selenium and zinc, higher red blood cell copper and zinc, and higher salivary calcium and sodium. PloS one 12, e0175437. https://doi.org/10.1371/journal.pone.0175437 73

Sahun, I., Marechal, D., Pereira, P.L., Nalesso, V., Gruart, A., Garcia, J.M.D., Antonarakis, S.E., Dierssen, M., Herault, Y., 2014. Cognition and hippocampal plasticity in the mouse is altered by monosomy of a genomic region implicated in Down syndrome. Genetics 197, 899–912. https://doi.org/10.1534/genetics.114.165241 Sailani, M.R., Santoni, F.A., Letourneau, A., Borel, C., Makrythanasis, P., Hibaoui, Y., Popadin, K., Bonilla, X., Guipponi, M., Gehrig, C., Vannier, A., Carre-Pigeon, F., Feki, A., Nizetic, D., Antonarakis, S.E., 2015. DNA-Methylation Patterns in Trisomy 21 Using Cells from Monozygotic Twins. PloS one 10, e0135555. https://doi.org/10.1371/journal.pone.0135555

SC R

IP T

Salehi, A., Faizi, M., Colas, D., Valletta, J., Laguna, J., Takimoto-Kimura, R., Kleschevnikov, A., Wagner, S.L., Aisen, P., Shamloo, M., Mobley, W.C., 2009. Restoration of norepinephrinemodulated contextual memory in a mouse model of Down syndrome. Science translational medicine 1, 7ra17. https://doi.org/10.1126/scitranslmed.3000258

U

Salemi, M., Barone, C., Romano, C., Salluzzo, M.G., Giambirtone, M., Morale, M.C., Calogero, A.E., Grillo, L., Bosco, P., Romano, C., 2015. A peculiar VNTR in the cystathionine beta-synthase gene is a risk factor for Down Syndrome. Cellular and molecular biology (Noisy-le-Grand, France) 61, 49–51.

M

A

N

Sanchez-Mut, J. V, Heyn, H., Vidal, E., Moran, S., Sayols, S., Delgado-Morales, R., Schultz, M.D., Ansoleaga, B., Garcia-Esparcia, P., Pons-Espinal, M., de Lagran, M.M., Dopazo, J., Rabano, A., Avila, J., Dierssen, M., Lott, I., Ferrer, I., Ecker, J.R., Esteller, M., 2016. Human DNA methylomes of neurodegenerative diseases show common epigenomic patterns. Translational Psychiatry 6, e718–e718. https://doi.org/10.1038/tp.2015.214

TE D

Sato, T., Diehl, T.S., Narayanan, S., Funamoto, S., Ihara, Y., De Strooper, B., Steiner, H., Haass, C., Wolfe, M.S., 2007. Active gamma-secretase complexes contain only one of each component. The Journal of biological chemistry 282, 33985–33993. https://doi.org/10.1074/jbc.M705248200

EP

Schalinske, K.L., Smazal, A.L., 2012. Homocysteine imbalance: a pathological metabolic marker. Advances in nutrition (Bethesda, Md.) 3, 755–762. https://doi.org/10.3945/an.112.002758

CC

Schon, E.A., Kim, S.H., Ferreira, J.C., Magalhaes, P., Grace, M., Warburton, D., Gross, S.J., 2000. Chromosomal non-disjunction in human oocytes: is there a mitochondrial connection? Human reproduction (Oxford, England) 15 Suppl 2, 160–172.

A

Schroeder, E.K., Kelsey, N.A., Doyle, J., Breed, E., Bouchard, R.J., Loucks, F.A., Harbison, R.A., Linseman, D.A., 2009. Green tea epigallocatechin 3-gallate accumulates in mitochondria and displays a selective antiapoptotic effect against inducers of mitochondrial oxidative stress in neurons. Antioxidants & redox signaling 11, 469–480. https://doi.org/10.1089/ars.2008.2215 Scott-McKean, J.J., Costa, A.C.S., 2011. Exaggerated NMDA mediated LTD in a mouse model of Down syndrome and pharmacological rescuing by memantine. Learning & Memory 18, 774–778. https://doi.org/10.1101/lm.024182.111 Scudellari, M., 2016. How iPS cells changed the world. Nature 534, 310–312. https://doi.org/10.1038/534310a 74

Serrano-Pérez, M.C., Fernández, M., Neria, F., Berjón-Otero, M., Doncel-Pérez, E., Cano, E., Tranque, P., 2015. NFAT transcription factors regulate survival, proliferation, migration, and differentiation of neural precursor cells: NFAT Effects on Neural Precursor Cells. Glia 63, 987– 1004. https://doi.org/10.1002/glia.22797 Sherman, S.L., Allen, E.G., Bean, L.H., Freeman, S.B., 2007. Epidemiology of Down syndrome. Mental retardation and developmental disabilities research reviews 13, 221–227. https://doi.org/10.1002/mrdd.20157

IP T

Shi, W.-L., Liu, Z.-Z., Wang, H.-D., Wu, D., Zhang, H., Xiao, H., Chu, Y., Hou, Q.-F., Liao, S.-X., 2016. Integrated miRNA and mRNA expression profiling in fetal hippocampus with Down syndrome. Journal of biomedical science 23, 48. https://doi.org/10.1186/s12929-016-0265-0

SC R

Shichiri, M., Yoshida, Y., Ishida, N., Hagihara, Y., Iwahashi, H., Tamai, H., Niki, E., 2011. αTocopherol suppresses lipid peroxidation and behavioral and cognitive impairments in the Ts65Dn mouse model of Down syndrome. Free Radical Biology & Medicine 50, 1801–1811. https://doi.org/10.1016/j.freeradbiomed.2011.03.023

U

Sies, H., 2010. Polyphenols and health: Update and perspectives. Archives of Biochemistry and Biophysics 501, 2–5. https://doi.org/10.1016/j.abb.2010.04.006

A

N

Singh, M., Jadhav, H.R., 2014. Melatonin: functions and ligands. Drug Discovery Today 19, 1410– 1418. https://doi.org/10.1016/j.drudis.2014.04.014

TE D

M

Škovierová, H., Vidomanová, E., Mahmood, S., Sopková, J., Drgová, A., Červeňová, T., Halašová, E., Lehotský, J., 2016. The Molecular and Cellular Effect of Homocysteine Metabolism Imbalance on Human Health. International Journal of Molecular Sciences 17, 1733. https://doi.org/10.3390/ijms17101733 Śmigielska-Kuzia, J., Boćkowski, L., Sobaniec, W., Kułak, W., Sendrowski, K., 2010. Amino acid metabolic processes in the temporal lobes assessed by proton magnetic resonance spectroscopy (1H MRS) in children with Down syndrome. Pharmacol Rep 62, 1070–1077.

CC

EP

Smith, I., Calegari, F., 2015. Cyclin D1 Again Caught in the Act: Dyrk1a Links G1 and Neurogenesis in Down Syndrome. EBioMedicine 2, 96–97. https://doi.org/10.1016/j.ebiom.2015.02.003

A

Song, C., He, J., Chen, J., Liu, Y., Xiong, F., Wang, Y., Li, T., 2015. Effect of the onecarbon unit cycle on overall DNA methylation in children with Down’s syndrome. Molecular medicine reports 12, 8209–8214. https://doi.org/10.3892/mmr.2015.4439 Souchet, B., Guedj, F., Penke-Verdier, Z., Daubigney, F., Duchon, A., Herault, Y., Bizot, J.-C., Janel, N., Creau, N., Delatour, B., Delabar, J.M., 2015. Pharmacological correction of excitation/inhibition imbalance in Down syndrome mouse models. Frontiers in behavioral neuroscience 9, 267. https://doi.org/10.3389/fnbeh.2015.00267 Souchet, B., Guedj, F., Sahun, I., Duchon, A., Daubigney, F., Badel, A., Yanagawa, Y., Barallobre, M.J., Dierssen, M., Yu, E., Herault, Y., Arbones, M., Janel, N., Creau, N., Delabar, J.M., 2014. 75

Excitation/inhibition balance and learning are modified by Dyrk1a gene dosage. Neurobiology of disease 69, 65–75. https://doi.org/10.1016/j.nbd.2014.04.016 Spalding, K.L., Bergmann, O., Alkass, K., Bernard, S., Salehpour, M., Huttner, H.B., Boström, E., Westerlund, I., Vial, C., Buchholz, B.A., Possnert, G., Mash, D.C., Druid, H., Frisén, J., 2013. Dynamics of Hippocampal Neurogenesis in Adult Humans. Cell 153, 1219–1227. https://doi.org/10.1016/j.cell.2013.05.002

IP T

Stagi, S., Lapi, E., Romano, S., Bargiacchi, S., Brambilla, A., Giglio, S., Seminara, S., de Martino, M., 2015. Determinants of Vitamin D Levels in Children and Adolescents with Down Syndrome. International Journal of Endocrinology 2015, 1–11. https://doi.org/10.1155/2015/896758

SC R

Stagni, F., Giacomini, A., Emili, M., Guidi, S., Bartesaghi, R., 2018. Neurogenesis impairment: An early developmental defect in Down syndrome. Free Radical Biology and Medicine 114, 15–32. https://doi.org/10.1016/j.freeradbiomed.2017.07.026

U

Stagni, F., Giacomini, A., Emili, M., Trazzi, S., Guidi, S., Sassi, M., Ciani, E., Rimondini, R., Bartesaghi, R., 2016. Short- and long-term effects of neonatal pharmacotherapy with epigallocatechin-3-gallate on hippocampal development in the Ts65Dn mouse model of Down syndrome. Neuroscience. https://doi.org/10.1016/j.neuroscience.2016.07.031

M

A

N

Stagni, F., Giacomini, A., Guidi, S., Ciani, E., Bartesaghi, R., 2015. Timing of therapies for Down syndrome: the sooner, the better. Frontiers in behavioral neuroscience 9, 265. https://doi.org/10.3389/fnbeh.2015.00265

TE D

Stagni, F., Magistretti, J., Guidi, S., Ciani, E., Mangano, C., Calzà, L., Bartesaghi, R., 2013. Pharmacotherapy with Fluoxetine Restores Functional Connectivity from the Dentate Gyrus to Field CA3 in the Ts65Dn Mouse Model of Down Syndrome. PLoS ONE 8, e61689. https://doi.org/10.1371/journal.pone.0061689

EP

Stagni, F., Raspanti, A., Giacomini, A., Guidi, S., Emili, M., Ciani, E., Giuliani, A., Bighinati, A., Calzà, L., Magistretti, J., Bartesaghi, R., 2017. Long-term effect of neonatal inhibition of APP gamma-secretase on hippocampal development in the Ts65Dn mouse model of Down syndrome. Neurobiology of Disease 103, 11–23. https://doi.org/10.1016/j.nbd.2017.03.012

CC

Stefanidis, K., Belitsos, P., Fotinos, A., Makris, N., Loutradis, D., Antsaklis, A., 2011. Causes of infertility in men with Down syndrome: Down Syndrome associated male infertility. Andrologia 43, 353–357. https://doi.org/10.1111/j.1439-0272.2010.01043.x

A

Stringer, M., Abeysekera, I., Dria, K.J., Roper, R.J., Goodlett, C.R., 2015. Low dose EGCG treatment beginning in adolescence does not improve cognitive impairment in a Down syndrome mouse model. Pharmacology Biochemistry and Behavior. https://doi.org/10.1016/j.pbb.2015.09.002 Stringer, M., Abeysekera, I., Thomas, J., LaCombe, J., Stancombe, K., Stewart, R.J., Dria, K.J., Wallace, J.M., Goodlett, C.R., Roper, R.J., 2017. Epigallocatechin-3-gallate (EGCG) consumption in the Ts65Dn model of Down syndrome fails to improve behavioral deficits and is detrimental to skeletal phenotypes. Physiology & Behavior 177, 230–241. https://doi.org/10.1016/j.physbeh.2017.05.003 76

Suetsugu, M., Mehraein, P., 1980. Spine distribution along the apical dendrites of the pyramidal neurons in Down’s syndrome. Acta neuropathologica 50, 207–210. Sukla, K.K., Jaiswal, S.K., Rai, A.K., Mishra, O.P., Gupta, V., Kumar, A., Raman, R., 2015. Role of folate-homocysteine pathway gene polymorphisms and nutritional cofactors in Down syndrome: A triad study. Human reproduction (Oxford, England) 30, 1982–1993. https://doi.org/10.1093/humrep/dev126

IP T

Sullivan, K.D., Evans, D., Pandey, A., Hraha, T.H., Smith, K.P., Markham, N., Rachubinski, A.L., Wolter-Warmerdam, K., Hickey, F., Espinosa, J.M., Blumenthal, T., 2017. Trisomy 21 causes changes in the circulating proteome indicative of chronic autoinflammation. Scientific Reports 7. https://doi.org/10.1038/s41598-017-13858-3

SC R

Sun, X., He, G., Song, W., 2006. BACE2, as a novel APP θ-secretase, is not responsible for the pathogenesis of Alzheimer’s disease in Down syndrome. The FASEB Journal 20, 1369–1376. https://doi.org/10.1096/fj.05-5632com

U

Sutherland, B.A., Shaw, O.M., Clarkson, A.N., Jackson, D.N., Sammut, I.A., Appleton, I., 2005. Neuroprotective effects of (–)-epigallocatechin gallate following hypoxia-ischemia-induced brain damage: novel mechanisms of action. The FASEB journal 19, 258–260.

M

A

N

Szemes, M., Davies, R.L., Garden, C.L., Usowicz, M.M., 2013. Weaker control of the electrical properties of cerebellar granule cells by tonically active GABAA receptors in the Ts65Dn mouse model of Down’s syndrome. Molecular Brain 6, 33. https://doi.org/10.1186/1756-6606-6-33

TE D

Takashima, S., Iida, K., Mito, T., Arima, M., 1994. Dendritic and histochemical development and ageing in patients with Down’s syndrome. J Intellect Disabil Res 38 ( Pt 3), 265–273. Tamaoka A., 1998. Characterization of Amyloid .BETA. Protein Species in the Plasma, Cerebrospinal fluid and Brains of Patients with Alzheimer’s Disease. Nippon Ronen Igakkai Zasshi. Japanese Journal of Geriatrics 35, 273–277. https://doi.org/10.3143/geriatrics.35.273

CC

EP

Tamasaki, A., Saito, Y., Ueda, R., Ohno, K., Yokoyama, K., Satake, T., Sakuma, H., Takahashi, Y., Kondoh, T., Maegaki, Y., 2016. Effects of donepezil and serotonin reuptake inhibitor on acute regression during adolescence in Down syndrome. Brain and Development 38, 113–117. https://doi.org/10.1016/j.braindev.2015.06.006

A

Tan, D.-X., Hardeland, R., Manchester, L.C., Paredes, S.D., Korkmaz, A., Sainz, R.M., Mayo, J.C., Fuentes-Broto, L., Reiter, R.J., 2010. The changing biological roles of melatonin during evolution: from an antioxidant to signals of darkness, sexual selection and fitness. Biological Reviews of the Cambridge Philosophical Society 85, 607–623. https://doi.org/10.1111/j.1469-185X.2009.00118.x Tapiola, T., Soininen, H., Pirttila, T., 2001. CSF tau and A 42 levels in patients with Down’s syndrome. Neurology 56, 979–980. https://doi.org/10.1212/WNL.56.7.979 Teipel, S.J., Hampel, H., 2006. Neuroanatomy of Down syndrome in vivo: a model of preclinical Alzheimer’s disease. Behav. Genet. 36, 405–415. https://doi.org/10.1007/s10519-006-9047-x Thomazeau, A., Lassalle, O., Iafrati, J., Souchet, B., Guedj, F., Janel, N., Chavis, P., Delabar, J., 77

Manzoni, O.J., 2014. Prefrontal deficits in a murine model overexpressing the down syndrome candidate gene dyrk1a. The Journal of neuroscience : the official journal of the Society for Neuroscience 34, 1138–1147. https://doi.org/10.1523/JNEUROSCI.2852-13.2014 Tiano, L., Padella, L., Santoro, L., Carnevali, P., Principi, F., Bruge, F., Gabrielli, O., Littarru, G.P., 2012. Prolonged coenzyme Q10 treatment in Down syndrome patients: effect on DNA oxidation. Neurobiology of aging 33, 626.e1–8. https://doi.org/10.1016/j.neurobiolaging.2011.03.025

IP T

Torres, M.D., Garcia, O., Tang, C., Busciglio, J., 2018. Dendritic spine pathology and thrombospondin-1 deficits in Down syndrome. Free Radic. Biol. Med. 114, 10–14. https://doi.org/10.1016/j.freeradbiomed.2017.09.025

SC R

Troca-Marín, J.A., Casañas, J.J., Benito, I., Montesinos, M.L., 2014. The Akt-mTOR pathway in Down’s syndrome: the potential use of rapamycin/rapalogs for treating cognitive deficits. CNS & neurological disorders drug targets 13, 34–40.

U

Tyler, W.A., Haydar, T.F., 2013. Multiplex Genetic Fate Mapping Reveals a Novel Route of Neocortical Neurogenesis, Which Is Altered in the Ts65Dn Mouse Model of Down Syndrome. Journal of Neuroscience 33, 5106–5119. https://doi.org/10.1523/JNEUROSCI.5380-12.2013

A

N

Uberos, J., Romero, J., Molina-Carballo, A., Muñoz-Hoyos, A., 2010. Melatonin and elimination of kynurenines in children with Down’s syndrome. Journal of pediatric endocrinology & metabolism: JPEM 23, 277–282.

TE D

M

Usowicz, M.M., Garden, C.L.P., 2012. Increased excitability and altered action potential waveform in cerebellar granule neurons of the Ts65Dn mouse model of Down syndrome. Brain Research 1465, 10–17. https://doi.org/10.1016/j.brainres.2012.05.027 Vacca, R.A., Valenti, D., 2015. Green tea EGCG plus fish oil omega-3 dietary supplements rescue mitochondrial dysfunctions and are safe in a Down’s syndrome child. Clin Nutr 34, 783–784. https://doi.org/10.1016/j.clnu.2015.04.012

CC

EP

Vacca, R.A., Valenti, D., Caccamese, S., Daglia, M., Braidy, N., Nabavi, S.M., 2016. Plant polyphenols as natural drugs for the management of Down syndrome and related disorders. Neuroscience and biobehavioral reviews 71, 865–877. https://doi.org/10.1016/j.neubiorev.2016.10.023

A

Valenti, D., Braidy, N., De Rasmo, D., Signorile, A., Rossi, L., Atanasov, A.G., Volpicella, M., Henrion-Caude, A., Nabavi, S.M., Vacca, R.A., 2018. Mitochondria as pharmacological targets in Down syndrome. Free Radical Biology and Medicine 114, 69–83. https://doi.org/10.1016/j.freeradbiomed.2017.08.014 Valenti, D., de Bari, L., De Filippis, B., Henrion-Caude, A., Vacca, R.A., 2014. Mitochondrial dysfunction as a central actor in intellectual disability-related diseases: an overview of Down syndrome, autism, Fragile X and Rett syndrome. Neurosci Biobehav Rev 46 Pt 2, 202–217. https://doi.org/10.1016/j.neubiorev.2014.01.012 Valenti, D., de Bari, L., de Rasmo, D., Signorile, A., Henrion-Caude, A., Contestabile, A., Vacca, 78

R.A., 2016. The polyphenols resveratrol and epigallocatechin-3-gallate restore the severe impairment of mitochondria in hippocampal progenitor cells from a Down syndrome mouse model. Biochimica et biophysica acta 1862, 1093–1104. https://doi.org/10.1016/j.bbadis.2016.03.003 Valenti, D., De Rasmo, D., Signorile, A., Rossi, L., de Bari, L., Scala, I., Granese, B., Papa, S., Vacca, R.A., 2013. Epigallocatechin-3-gallate prevents oxidative phosphorylation deficit and promotes mitochondrial biogenesis in human cells from subjects with Down’s syndrome. Biochimica et Biophysica Acta (BBA)-Molecular Basis of Disease 1832, 542–552.

SC R

IP T

Valenti, D., Manente, G.A., Moro, L., Marra, E., Vacca, R.A., 2011. Deficit of complex I activity in human skin fibroblasts with chromosome 21 trisomy and overproduction of reactive oxygen species by mitochondria: involvement of the cAMP/PKA signalling pathway. The Biochemical journal 435, 679–688. https://doi.org/10.1042/BJ20101908

U

Valenti, D., Rossi, L., Marzulli, D., Bellomo, F., De Rasmo, D., Signorile, A., Vacca, R.A., 2017. Inhibition of Drp1-mediated mitochondrial fission improves mitochondrial dynamics and bioenergetics stimulating neurogenesis in hippocampal progenitor cells from a Down syndrome mouse model. Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease 1863, 3117– 3127. https://doi.org/10.1016/j.bbadis.2017.09.014

M

A

N

Valenti, D., Tullo, A., Caratozzolo, M.F., Merafina, R.S., Scartezzini, P., Marra, E., Vacca, R.A., 2010. Impairment of F1F0-ATPase, adenine nucleotide translocator and adenylate kinase causes mitochondrial energy deficit in human skin fibroblasts with chromosome 21 trisomy. The Biochemical Journal 431, 299–310. https://doi.org/10.1042/BJ20100581

TE D

Van Oekelen, D., Megens, A., Meert, T., Luyten, W.H.M.L., Leysen, J.E., 2003. Functional study of rat 5-HT2A receptors using antisense oligonucleotides. J. Neurochem. 85, 1087–1100. Varghese, J., 2010. BACE: lead target for orchestrated therapy of Alzheimer’s disease. Hoboken: Wiley.

CC

EP

Vassar, R., Bennett, B.D., Babu-Khan, S., Kahn, S., Mendiaz, E.A., Denis, P., Teplow, D.B., Ross, S., Amarante, P., Loeloff, R., Luo, Y., Fisher, S., Fuller, J., Edenson, S., Lile, J., Jarosinski, M.A., Biere, A.L., Curran, E., Burgess, T., Louis, J.C., Collins, F., Treanor, J., Rogers, G., Citron, M., 1999. Beta-secretase cleavage of Alzheimer’s amyloid precursor protein by the transmembrane aspartic protease BACE. Science (New York, N.Y.) 286, 735–741. Verma, I.C., 2000. Burden of genetic disorders in India. Indian journal of pediatrics 67, 893–898.

A

Verma, I.C., Bijarnia, S., 2002. The burden of genetic disorders in India and a framework for community control. Community genetics 5, 192–196. https://doi.org/66335 Victorino, D.B., Bederman, I.R., Costa, A.C.S., 2017. Pharmacokinetic Properties of Memantine after a Single Intraperitoneal Administration and Multiple Oral Doses in Euploid Mice and in the Ts65Dn Mouse Model of Down’s Syndrome. Basic & Clinical Pharmacology & Toxicology 121, 382–389. https://doi.org/10.1111/bcpt.12816 Wang, H., Cheng, E., Brooke, S., Chang, P., Sapolsky, R., 2003. Over‐expression of antioxidant 79

enzymes protects cultured hippocampal and cortical neurons from necrotic insults. Journal of neurochemistry 87, 1527–1534. Wang, J., Gallagher, D., DeVito, L.M., Cancino, G.I., Tsui, D., He, L., Keller, G.M., Frankland, P.W., Kaplan, D.R., Miller, F.D., 2012. Metformin activates an atypical PKC-CBP pathway to promote neurogenesis and enhance spatial memory formation. Cell Stem Cell 11, 23–35. https://doi.org/10.1016/j.stem.2012.03.016

SC R

IP T

Wang, X., Zhao, Y., Zhang, X., Badie, H., Zhou, Y., Mu, Y., Loo, L.S., Cai, L., Thompson, R.C., Yang, B., Chen, Y., Johnson, P.F., Wu, C., Bu, G., Mobley, W.C., Zhang, D., Gage, F.H., Ranscht, B., Zhang, Y., Lipton, S.A., Hong, W., Xu, H., 2013. Loss of sorting nexin 27 contributes to excitatory synaptic dysfunction by modulating glutamate receptor recycling in Down’s syndrome. Nature Medicine 19, 473–480. https://doi.org/10.1038/nm.3117

U

Weick, J.P., Held, D.L., Bonadurer, G.F., Doers, M.E., Liu, Y., Maguire, C., Clark, A., Knackert, J.A., Molinarolo, K., Musser, M., Yao, L., Yin, Y., Lu, J., Zhang, X., Zhang, S.-C., Bhattacharyya, A., 2013. Deficits in human trisomy 21 iPSCs and neurons. Proceedings of the National Academy of Sciences 110, 9962–9967. https://doi.org/10.1073/pnas.1216575110

A

N

Weitzdoerfer, R., Dierssen, M., Fountoulakis, M., Lubec, G., 2001. Fetal life in Down syndrome starts with normal neuronal density but impaired dendritic spines and synaptosomal structure. Journal of neural transmission. Supplementum. https://doi.org/10.1007/978-3-7091-6262-0_5

M

Whitaker-Azmitia, P.M., 2001. Serotonin and brain development: role in human developmental diseases. Brain Res. Bull. 56, 479–485.

TE D

Whittle, N., Sartori, S.B., Dierssen, M., Lubec, G., Singewald, N., 2007. Fetal Down Syndrome Brains Exhibit Aberrant Levels of Neurotransmitters Critical for Normal Brain Development. PEDIATRICS 120, e1465–e1471. https://doi.org/10.1542/peds.2006-3448

EP

Wong, H., Levenga, J., Cain, P., Rothermel, B., Klann, E., Hoeffer, C., 2015. RCAN1 overexpression promotes age-dependent mitochondrial dysregulation related to neurodegeneration in Alzheimer’s disease. Acta Neuropathologica 130, 829–843. https://doi.org/10.1007/s00401-0151499-8

CC

Wuang, Y.-P., Chiang, C.-S., Su, C.-Y., Wang, C.-C., 2011. Effectiveness of virtual reality using Wii gaming technology in children with Down syndrome. Research in Developmental Disabilities 32, 312–321. https://doi.org/10.1016/j.ridd.2010.10.002

A

Xie, W., Ramakrishna, N., Wieraszko, A., Hwang, Y.-W., 2008. Promotion of Neuronal Plasticity by (−)-Epigallocatechin-3-Gallate. Neurochemical Research 33, 776–783. https://doi.org/10.1007/s11064-007-9494-7 Yang, X., Zhang, Y., Xu, H., Luo, X., Yu, J., Liu, J., Chang, R.C.-C., 2016. Neuroprotection of Coenzyme Q10 in Neurodegenerative Diseases. Current Topics in Medicinal Chemistry 16, 858– 866. Yin, X., Jin, N., Shi, J., Zhang, Y., Wu, Y., Gong, C.-X., Iqbal, K., Liu, F., 2017. Dyrk1A 80

overexpression leads to increase of 3R-tau expression and cognitive deficits in Ts65Dn Down syndrome mice. Scientific reports 7, 619. https://doi.org/10.1038/s41598-017-00682-y Yu, J., Jia, Y., Guo, Y., Chang, G., Duan, W., Sun, M., Li, B., Li, C., 2010. Epigallocatechin-3gallate protects motor neurons and regulates glutamate level. FEBS letters 584, 2921–2925. https://doi.org/10.1016/j.febslet.2010.05.011

IP T

Yu, T., Clapcote, S.J., Li, Z., Liu, C., Pao, A., Bechard, A.R., Carattini-Rivera, S., Matsui, S.-I., Roder, J.C., Baldini, A., Mobley, W.C., Bradley, A., Yu, Y.E., 2010. Deficiencies in the region syntenic to human 21q22.3 cause cognitive deficits in mice. Mammalian Genome 21, 258–267. https://doi.org/10.1007/s00335-010-9262-x

SC R

Zaki, M.E., El-Bassyouni, H.T., Tosson, A.M.S., Youness, E., Hussein, J., 2017. Coenzyme Q10 and pro-inflammatory markers in children with Down syndrome: clinical and biochemical aspects. Jornal de pediatria 93, 100–104. https://doi.org/10.1016/j.jped.2016.04.012

U

Zamponi, E., Zamponi, N., Coskun, P., Quassollo, G., Lorenzo, A., Cannas, S.A., Pigino, G., Chialvo, D.R., Gardiner, K., Busciglio, J., Helguera, P., 2018. Nrf2 stabilization prevents critical oxidative damage in Down syndrome cells. Aging cell 17, e12812. https://doi.org/10.1111/acel.12812

A

N

Zana, M., Janka, Z., Kálmán, J., 2007. Oxidative stress: A bridge between Down’s syndrome and Alzheimer’s disease. Neurobiology of Aging 28, 648–676. https://doi.org/10.1016/j.neurobiolaging.2006.03.008

TE D

M

Zhang, Z.-X., Li, Y.-B., Zhao, R.-P., 2017. Epigallocatechin Gallate Attenuates β-Amyloid Generation and Oxidative Stress Involvement of PPARγ in N2a/APP695 Cells. Neurochemical Research 42, 468–480. https://doi.org/10.1007/s11064-016-2093-8

EP

Zhao, Z., Yu, W., Shi, C., Liang, R., Chen, X., Feng, X., Zhang, X., Mu, Q., Shen, H., Guo, J., 2017. Correlation between receptor-interacting protein 140 expression and directed differentiation of human embryonic stem cells into neural stem cells. Neural Regeneration Research 12, 118. https://doi.org/10.4103/1673-5374.198997

CC

Zis, P., Strydom, A., 2018. Clinical aspects and biomarkers of Alzheimer’s disease in Down syndrome. Free Radical Biology and Medicine 114, 3–9. https://doi.org/10.1016/j.freeradbiomed.2017.08.024

A

Zorrilla de San Martin, J., Delabar, J.-M., Bacci, A., Potier, M.-C., 2018. GABAergic overinhibition, a promising hypothesis for cognitive deficits in Down syndrome. Free Radical Biology and Medicine 114, 33–39. https://doi.org/10.1016/j.freeradbiomed.2017.10.002

Legends to the figures

81

Figure 1. Diagram of Down syndrome neuropathology. Hsa21 trisomy in the brain leads to alterations in neurotransmission, neurogenesis and synaptic plasticity with consequent impairments in neurodevelopment (see the neurobiological alterations in the sectors of upper part of the scheme). Amyloid pathology together with an increase in oxidative stress and neural cell death leads to neurodegeneration associated with AD-like dementia and early aging in DS (see the sectors of the

A

CC

EP

TE D

M

A

N

U

SC R

IP T

lower part of the scheme).

Figure 2. Therapeutic action of selected drugs to correct some neurobiological alterations in Down syndrome. Presynaptic GABAergic, glutamatergic and serotonergic and cholinergic transmissions in DS are depicted. In DS post synaptic neurons, overexpression of Hsa21 genes (in red) determines impairment of target genes or pathways (red dotted line ---I) possibly removed by 82

the therapeutic actions of the selected drugs which restore the normal functions (back line continue arrows); the red arrows indicate up () or down () regulation of Hsa21-downstream proteins/receptors in DS. Impairment in signalling pathways controlling mitochondrial functions (involving miR155, DYRK1A, RCAN1, RIP140) induces a reduction in mitochondrial ATP production, an increase in ROS formation, and impairment in many ATP-dependent neurological

IP T

processes including synaptic activity and neurogenesis. Molecular targets of bumetanide, memantine, epigallocatechine-3-gallate (EGCG), fluoxetine, donepezil, β- and γ-secretase

SC R

modulators melatonin, metformin, resveratrol, coenzyme Q and folinic acid are indicated with continued line with the same colours of the respective drug; specifically, () indicates activation effects and (___I) indicate inhibition of overexpression/hyperactivity. NMDAR, NMDA receptor;

U

SerotoninR, serotonin receptor; AchE, acetylcholine esterase; nAchR, nicotine acetylcholine

N

receptor; P, phosphorylation; Ac, acetylation; NM, nuclear membrane; MOM, mitochondrial outer

A

membrane; IMS, mitochondrial intermembrane space; MIM, mitochondrial inner membrane.

M

Respiratory chain complexes: II, complex II; Q, coenzyme Q; III, complex III; c, cytochrome c; IV,

A

CC

EP

TE D

complex IV.

83

84

EP

CC

A TE D

IP T

SC R

U

N

A

M

Table 1. Mouse and cellular models for Down syndrome studies. The mouse models, e.g. transgenic mice carrying triplication of orthologous Hsa21 regions, as well as cellular systems, cited in this review are reported. Model name

Origin

Endpoint gene region

References

Numbers of orthologous

Transgenic mouse

Dp(16)1Yey

Transgenic mouse

miR155-Zbtb21

Reeves et al., 1995;

100 Hsa21 genes

Xing et al., 2016

Lipi-Zbtb21

Li et al., 2007;

113 Hsa21 genes

Transgenic mouse

Xing et al., 2016 Sago et al., 1998;

70 Hsa21 genes

Xing et al., 2016

Cb1r1-Fam3b

Olson et al., 2004

A

Ts1Rhr

Sod1-Zbtb21

U

Transgenic mouse

N

Ts1Cje

SC R

Ts65Dn

IP T

genes in 3 copies

Transgenic mouse

TE D

Ts1Yah

M

29 Hsa21 genes

Transgenic mouse

CC

EP

Ts2Yah

A

TgDYRK1A

BAC-Tg1

Transgenic mouse

Abcg1-U2af1

Pereira et al., 2009

12 Hsa21 genes Hspa13-App

Brault et al., 2015;

19 Hsa21 genes

Xing et al., 2016

Dyrk1A

Altafaj et al., 2001

1 Hsa21 gene Transgenic mouse

Rcan1

Xing et al., 2013

1 Hsa21 gene Neural progenitor

From Ts65Dn transgenic

cells (NPCs)

mouse

miR155-Zbtb21

Contestabile et al., 2007; Valenti et al., 2016; 2017

100 Hsa21 genes 85

DS-iPSC-derived

Human fibroblasts

Hsa21 trisomy

Hibaoui et al., 2014

Human: foetal and

Hsa21 trisomy

Valenti et al., 2010; 2011

NPCs Skin fibroblasts

children Heart fibroblasts

Human: foetal

Hsa21 trisomy

Izzo et al., 2004

Lymphoblastoids

Human immortalized

Hsa21 trisomy

Valenti et al., 2013; Granese et al., 2013

A

CC

EP

TE D

M

A

N

U

SC R

IP T

lymphocytes: children

86

I N U SC R

Table 2. Main pharmacological interventions in Down syndrome

The main for human use-approved drugs tested in vitro and in vivo in DS, their targets, therapeutic actions and results relevant for DS are reported. In bracket the treatment duration. Abbreviations: o.d., oral dosage; I.P., intraperitoneal injection; I.C., subcutaneous injection, NPCs, neural progenitor cells. ID, ClinicalTrials.gov identification number. Target

Model system/

A

Drug

Dose

Ts65Dn mice

0.2 mg/kg/day

transmission

10-16 weeks old

I.P. (10-16 weeks) aberrant

GABAergic

Ts65Dn mice

(RG1662)

transmission

3-4 months old

A

CC E

PT

Basmisanil

Fluoxetine

Results

References

Improvement of

Deidda et al.,

hippocampal synaptic

2015

action

GABAergic

ED

Bumetanide

M

Trial (ClinicalTrials.gov)

Therapeutic

20 mg/kg/day

Reduction of

excitatory

plasticity and enhanced

GABAA signaling

memory

Negative

Rescue of deficient

Martínez-Cué et al.,

neurogenesis and

2013

o.d. (3-4 months) allosteric modulation of α5-

stabilization of

GABAA receptor

GABAergic synapse density

Phase II trial in DS subjects

20-240 mg

No significant

La Roche

12-30 years old

o.d. twice daily

improvement of

statement

(ID: NCT02024789)

(26 weeks)

cognitive deficits in DS

https://ds-

subjects

int.org/node/3547

Serotonergic

Ts65Dn mice

10 mg/kg/day

Inhibition of

Recovery of the

Stagni et al.,

transmission

(45 days old)

I.P., I.C.

serotonin reuptake

synaptic network

2013; Guidi et al.,

Ts65Dn mice (60 days

10 mg/kg/day

Rescue of hippocampal

2013

old)

o.d. daily (8

synaptic plasticity and

Begenesis et al., 87

I N U SC R weeks)

Continued

spatial memory

2014

Table 2. Main pharmacological interventions in Down syndrome (Continued)

A

Model system/Trial

Therapeutic

Results

References

action

Ts65Dn mice

30 mg/kg/day

Normalization of

Improvement of

Rueda et

transmission

9 months old

o.d. (10-16 weeks)

LTP and

memory, and cognitive

al., 2010;

improvement of

functions

Victorino et

PT CC E A Donepezil

Dose

Glutamatergic

ED

Memantine

Target

M

Drug

Cholinergic transmission

cognition

al., 2017

Pilot randomized double

5-10 mg/day

No differences between

Boada et

blinded trial in DS

o.d. (16 weeks)

drug treated and

al., 2012

subjects

placebo group in

18-32 years old

cognitive and adaptive

(ID N°: NCT01112683)

10 mg/day

functions.

Hanney et

Phase II trial in adults DS

o.d. (52 weeks)

Failed efficacy in

al., 2012

subjects

improving living skills

Above 40 years old

or counteracting

(ISRCTN registry ID N°:

memory and cognitive

ISRCTN47562898)

decline

Randomized double-blinded phase II trial in adult with DS Randomized double-blinded

3 mg/day o.d. (24 weeks)

Improvement of AD-like disease symptoms

Improvement overall functioning of DS patients

Improvement of

No differences between

Kondoh et al., 2011

88

I N U SC R

2.5-5.0 mg/day o.d. (10 weeks)

cognition

drug treated and placebo group in cognitive and adaptive functions.

Kishnani et al., 2010

A

phase III trial in children with DS (ID NCT00754052)

Continued

ED

Target

Model system/Trial

γ-Secretase

Amyloid

Ts65Dn mice

inhibitor

pathway

8 weeks old

(DAPT)

A

Metformin

Therapeutic

Results

References

100 mg/kg/day

Reduction of β-

Rescue of spatial

Netzer et al.,

amyloid levels

learning and

2010

memory deficits

Mitochondria

Randomized open trial

4 mg/kg/day

Rescue of reduced

Protective effect in

Tiano et al.,

and redox

in children and young

o.d. (20 months)

CoQ10 plasma

mitigating DNA

2012

homeostasis

DS subjects

levels.

oxidative damage

Activation of

Improvement of

Izzo et al.,

PGC1α signaling

mitochondrial

2017b

pathway.

dysfunction and

CC E

Coenzyme Q

Dose

action

PT

Drug

M

Table 2. Main pharmacological interventions in Down syndrome (Continued)

5-17 years old

Mitochondrial

Cultured fibroblasts

network and

with Has21 trisomy

0.05-0.5 mM

biogenesis

correction of altered mitochondrial network Melatonin

Mitochondria

Ts65Dn mice

0.5 mg/day

Antioxidant action

Attenuation of

Parisotto et 89

I N U SC R

and redox

6-12 months old

and modulation of

oxidative damage

al., 2016;

mitochondrial

and senescence;

Corrales et

integrity and

no improvement of

al., 2017

function

cognitive impairment

A

homeostasis

o.d.

ED

M

Continued

Table 2. Main pharmacological interventions in Down syndrome (Continued) Model system/Trial

Dose

Therapeutic

Results

References

Mitochondrial

Increase in cell

Valenti et

action 20 μM

DYRK1A,

Cultured fibroblasts with

mitochondria,

Has21 trisomy

ROS

energy and

al., 2013;

oxidative stress

Cultured Ts65Dn NPCs

scavenging.

metabolism;

2016; 2017

neurogenesis,

Activation of

decrease in

neurotrasmission

mitochondrial

oxidative stress;

bioenergetics

activation of

A

CC E

EGCG

Target

PT

Drug

neurogenesis.

Randomized double-

9 mg/kg per day

DYRK1A

Partial improving

de la Torre

blinded phase II trial in

o.d. (12 months)

inhibition

visual recognition

et al., 2016

DS young adults

memory, inhibitory 90

I N U SC R

(aged 16-34 years)

control, and adaptive

(ID N°: NCT01394796).

behavior;

Case study DS child 10

omega-3

year old

cognitive training.

10mg/kg EGCG plus

Activation of

Vacca and

8mg/kg omega-3

mitochondrial

Valenti,

(EPA+DHA)

respiratory

2015

chain

ED

M

A

EGCG and

potentiating effect in

PT

Continued

Table 2. Main pharmacological interventions in Down syndrome (Continued) Target

CC E

Drug

Resveratrol

Mitochondria,

Model system/Trial

Dose

Therapeutic

Results

References

Activation of

Rescue of

Valenti et

PGC1α/AMPK/

mitochondrial

al., 2016

Sirt1 axis

bioenergetics and

action Cultured Ts65Dn NPCs

10 μM

A

miR-155

biogenesis; Improvement of neurogenesis

Folinic acid

Folate

Randomized double-blinded

1 mg /kg per day

Antioxidant;

Improves the

Blehaut et

metabolism,

phase II/II trial in DS

o.d. (12 months)

improve folate

psychomotor

al., 2010 91

I N U SC R

homocysteine

children

pathway

(aged age 3 to 30 months)

in a subgroup of

(ID N°: NCT00294593)

children on thyroxin treatment.

Antioxidant;

Failed efficacy in

Ellis et al.,

blinded trial in DS

mg, Vitamin A 0.9 mg,

improved

improving

2008

children

Vitamin E 100 mg,

folate

cognitive tasks

(aged up to 7 Months)

Vitamin C 50 mg, folinic

metabolism

(ID N°: NCT00378456)

acid 0.1 mg

Randomized double-

vitamins and

metabolism,

minerals

homocysteine

M

A

Folate

o.d. (18 months)

A

CC E

PT

ED

development only

Selenium 10 mg, Zinc 5

Folinic acid

pathway

metabolism

92