• Non ci sono risultati.

Alterations of brain circuits in Down syndrome murine models

N/A
N/A
Protected

Academic year: 2021

Condividi "Alterations of brain circuits in Down syndrome murine models"

Copied!
38
0
0

Testo completo

(1)

24 July 2021

Alterations of brain circuits in Down syndrome murine models / Gotti S.; Caricati E.; Panzica G.. - In: JOURNAL OF CHEMICAL NEUROANATOMY. - ISSN 0891-0618. - STAMPA. - 42(2011), pp. 317-326.

Original Citation:

Alterations of brain circuits in Down syndrome murine models

Published version:

DOI:10.1016/j.jchemneu.2011.09.002

Terms of use:

Open Access

(Article begins on next page)

Anyone can freely access the full text of works made available as "Open Access". Works made available under a Creative Commons license can be used according to the terms and conditions of said license. Use of all other works requires consent of the right holder (author or publisher) if not exempted from copyright protection by the applicable law.

Availability:

This is the author's manuscript

(2)

This Accepted Author Manuscript (AAM) is copyrighted and published by Elsevier. It is posted here by agreement between Elsevier and the University of Turin. Changes resulting from the publishing process - such as editing, corrections, structural formatting, and other quality control mechanisms - may not be reflected in this version of the text. The definitive version of the text was subsequently published in [Journal of Chemical Neuroanatomy, 42 (2011) 317–326,

DOI: 10.1016/j.jchemneu.2011.09.002].

You may download, copy and otherwise use the AAM for non-commercial purposes provided that your license is limited by the following restrictions:

(1) You may use this AAM for non-commercial purposes only under the terms of the CC-BY-NC-ND license.

(2) The integrity of the work and identification of the author, copyright owner, and publisher must be preserved in any copy.

(3) You must attribute this AAM in the following format: Creative Commons BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/deed.en), [+ Digital Object Identifier link to the published journal article on Elsevier’s ScienceDirect® platform]

(3)

Gotti et al. - Prepared for J. Chemical Neuroanatomy 04/09/2011

Alterations of brain circuits in Down syndrome murine models

Stefano Gotti * 1,2, Egidio Caricati1,2, GianCarlo Panzica1,2

1 - Laboratory of Neuroendocrinology, Neuroscience Institute of Torino (NIT), National Institute of Neuroscience (INN, Torino), Dept. Anatomy, Pharmacology and Forensic Medicine, University of Torino, corso M. D’Azeglio 52, 10126 – Torino, Italy

2 - Neuroscience Institute of Cavalieri-Ottolenghi foundation (NICO), Torino, Italy

* Corresponding author: Stefano Gotti Laboratory of Neuroendocrinology,

Neuroscience Institute Cavalieri-Ottolenghi (NICO), Regione Gonzole, 10 – 10043 Orbassano (TO), Italy Tel.: +39 011 6707054; fax: +39 011 6705931. e-mail: stefano.gotti@unito.it

(4)

Abstract

Trisomy 21, also referred to Down syndrome (DS), is the most common genetic cause of mental retardation, affecting 1 each 800-1000 newborn children all over the world. DS is a complex disease, determined by an extra copy of human chromosome 21 that causes an imbalanced gene dose effect. The syntenies that exist between mouse chromosomes 10, 16, and 17 and human chromosome 21 offer the opportunity for a genotype–phenotype correlation and several mouse models of DS have been developed to improve our knowledge about cognitive disabilities and brain alterations.

We present here the different murine models available up to now and we discuss the neural alterations that have been described in these strains. The largest amount of studies involved the so called Ts65Dn mouse showing early alterations of nitrergic, noradrenergic and cholinergic systems at the level of the basal forebrain. Neurogenesis and spine formations are decreased in the hippocampus, as well as the whole size of the cerebellum and the number of granule cells.

Keywords

(5)

1. Introduction

In 1959, Lejeune and coworkers discovered the association between the syndrome described in 1867 by John Langdon Down and the triplication of the human chromosome 21 (HSA21) (Lejeune et al., 1959). Today, the Down Syndrome (DS) is considered the most common genetic cause of mental retardation (Hassold and Jacobs, 1984) affecting 1 each 800-1000 newborn children all over the world (Roizen and Patterson, 2003).

DS is determined by a chromosomal aberration that involves the total or partial trisomy of HSA21, resulting from meiotic non-disjunction of the chromosome 21 pair (95%). About 4% of persons with DS have 46 chromosomes, one of which is carrying a Robertsonian translocation between chromosome 21q and the long arm of one of the other acrocentric chromosomes (usually chromosome 14 or 22). About 1% of persons with DS have a mosaicism, with cell populations showing either a normal or a trisomic 21 karyotype; in this case, the phenotype may be milder than that of typical 21 trisomy (Hook, 1981; Patterson, 1987; Nadal et al., 1996; Galdzicki et al., 2001).

A precise gene catalogue for HSA21 has not yet been determined, in fact its sequencing (Hattori et al., 2000) showed 225 genes and 59 pseudo genes, but the prediction based on estimation from the sequencing is of 364 genes (Gardiner et al., 2003; Gardiner and Costa, 2006). Most of the functions of these genes are still unknown. An additional, unexpected finding is that the actual fraction of chromosome 21 transcribed into RNA might be one order of magnitude higher than the fraction occupied by gene coding sequences (Kapranov et al., 2002).

Different hypotheses have been presented to account for the relationships between trisomy of HSA21 and the occurrence of DS phenotypes.

“Gene dosage effect hypothesis”: According to this hypotesis, DS phenotype is a direct consequence of the cumulative effects of the dosage imbalance of genes located on the triplicate chromosome (Epstein, 1986, 1990; Korenberg et al., 1990). A subset of genes on HSA21 is therefore directly responsible for the pathological traits associated with trisomy 21.

“Down Syndrome Critical Region (DSCR) hypothesis”: DS associated with partial 21 trisomy is very rarely observed (no more than 1% of living trisomic persons), but these persons are of particular interest because they allowed the

(6)

identification of a restricted region of HSA21, called Down syndrome critical region (DSCR), located on the distal third of the long arm of the chromosome (region 21q22). The DSCR hypothesis states that genes in this region, when present in three copies, are responsible for the DS phenotypes and may cause the disease (Hardy et al., 1989). DSCR was in fact associated with the expression of many features of DS like the dysmorphic phenotypes and the mental retardation (Delabar et al., 1993; Korenberg et al., 1994); this hypothesis is in agreement with the gene dosage effect hypothesis.

Recent studies performed in mice models of DS (see below) have shown that three copies of DSCR genes alone are not sufficient to generate the craniofacial dysmorphic phenotypes (Olson et al., 2004). On the contrary, other studies show that DSCR genes are necessary but not sufficient to determine the impairment of the hippocampal function (Olson et al., 2007).

“Amplified developmental instability hypothesis”: the dosage imbalance of

hundreds of genes on HSA21 determines a non-specific perturbation of genomic regulation and expression; in this case, it is not the presence of three copies of a particular gene but rather the presence of a threshold number of genes (no matter the genes) that leads to DS phenotypes (Patterson and Costa 2005).

In summary, these hypotheses led to the conclusion that DS depends on a genetic unbalance determined either by the expression of trisomic genes (Mao et al., 2005) or by a destabilization in the expression of many disomic genes induced by the increased expression of trisomic genes (Saran et al., 2003). This last hypothesis is supported for example by the reduction of the level of the alpha fetoprotein (Newby et al., 1997) or by the increase in the human chorionic gonadotrophin levels (Macri et al., 1993) in trisomy 21, while the corresponding genes are located not on HSA21, but on 4 and 19, respectively.

2. Phenotype of Down Syndrome

Adult individuals with DS present a wide range of phenotypic and physiological abnormalities, including craniofacial anomalies, and diminished male reproductive

(7)

capacities (Bovicelli et al., 1982).

More interesting for the aims of this paper, the most salient feature of DS is mental retardation. In fact, DS is characterized by abnormalities in learning, memory, and language that lead to mild-profound impairment of intellectual functions. Individuals with DS have more behavioral and psychiatric problems than other persons: 17% of individuals with DS less than 20 year old have a psychiatric disorder, most frequently a disruptive behavior disorder such as attention deficit hyperactivity disorder (6%), conduct/oppositional disorder (5%), or aggressive behavior (6%); 25% of adults with DS have a psychiatric disorder, most frequently a major depressive disorder (6%) or aggressive behavior (6%) (Roizen and Patterson, 2003).

Several differences of DS brains compared to non-DS ones have been described, with both post-mortem and magnetic resonance analyses. In particular, several macroscopical abnormalities were found: i.e., small cerebellum, reduced frontal, temporal, and occipital lobes, reduced number and depth of the cerebral sulci, reduced cerebral grey and white matter, reduced hippocampal volume, and an age-related decrease of corpus callosum (for a complete references see Roubertoux and Kerdhule, 2006; Teipel and Hampel, 2006).

Several neuronal abnormalities, including alteration of cortical lamination, reduced dendritic branching, and diminished number of synapses, have been also described (see for references Roizen and Patterson, 2003). Neuronal alterations are accompanied with changes of glial elements: astrocyte, oligodendrocite, microglia, and endothelial cells. Changes in the neuroendocrine activities are well documented. In particular modifications and alterations in the hypothalamo-pituitary axis correlates to thyroid, somatotropic, lactotropic, and gonadotropic functions (see Roubertoux and Kerdhule, 2006).

Adult persons with DS may develop by the fifth decade of life neuropathological changes typicals of Alzheimer’s disease (i.e., cortical atrophy, ventricular dilatation, presence of neurofibrillary tangles and senile plaques, reduction of basal forebrain cholinergic neurons). Clinical signs and symptoms of Alzheimer’s disease are noted in 75% of such individuals over 60 years of age, (Nadel, 1994; Roizen and Patterson, 2003).

(8)

3. Murine models of Down Syndrome

Based on previous description, it appears that DS is characterized by both genotypic and phenotypic variability, therefore, it is difficult to control for differences among individuals and improbable that all DS phenotypes may be explained by an unique genetic background. It is more reasonable to hypothesize that different genes could differentiallly participate to the DS phenotype determination, including neurological alterations and mental retardation. Moreover, only a few data are available on the anatomical and functional correlates in humans. Therefore, considering the necessity to determine the origin of the abnormalities and to follow their evolution, DS awaits the use of appropriate animal models with the possibility to manipulate their genome, and to study all tissues at different embryonic or postnatal stages.

Comparative genomics has drawn up syntenic maps across species and these maps are well documented for mouse and human genomes. HSA21 is syntenic with three mouse chromosomes: MMU10, MMU16, and MMU17 (Fig.1). About 80% of HSA21 and MMU16 were syntenic and the remaining 20% are syntenic with MMU10 and MMU17 (Hattori et al., 2000).

The first murine model for DS was developed about thirty years ago; other models were subsequently developed and periodically their utility and use were reviewed and discussed from different point of views (i.e. genomics, behavior, pharmacological treatment; see for instance the reviews of Dierssen et al 2001; Davisson et al., 2005; Contestabile et al., 2010).

Overall, there are at least three types of murine models for DS:  Single gene model

Several models have been generated by the insertion of extra copies of HSA21 single human genes into the mouse genome: SIM2 (Ema et al., 1999), S100beta (Gerlai et al., 1993), SOD1 (Gahtan et al., 1998), APP (Harris-Cerruti et al., 2004). In this case the working hypothesis is that even one gene may have an effect. However, transgenics for individual genes produced scarce effects and the longer is the triplicate fragment, the greater is the effect (Chabert et al., 2004).

(9)

 HSA21 inserted genes murine models

These models resulted from the insertion of all or part of HSA21 in the mouse genome (Smith et al., 1995; Shinohara et al., 2001; Branchi et al., 2004; Chabert et al., 2004; O’Doherthy et al., 2005).

 MMU Extra copy genes murine models

Numerous models have been developed inserting short or long sequences of murine chromosomes. These models are detailed below.

3.1 Ts16 Mice. Since 1980 and for the larger part of two decades, trisomy 16 mice (Ts16) were the only available model for the study of DS. Ts16 mice were developed using mice with Robertsonian chromosome and a breeding scheme that produce high frequencies of mouse embryos with specific whole chromosome trisomies (Fig.1). They were extensively studied and utilized because they demonstrated many similarities to the DS, at morphological, biochemical, immunological and haematological levels (Epstein et al., 1985). However, an important difference exists between DS and Ts16 mice: a significant proportion of human trisomy 21 fetuses (10%) survive to term, whereas Ts16 mice die shortly after birth (Kola and Hertzog, 1998). It is unclear whether this difference is based on a species difference (i.e. mouse versus human) or whether it is attributable to a gene dosage perturbation of genes not found on human chromosome 21 but found on mouse chromosome 16, that shows sequences synthenic to other human chromosomes (3, 8, 16 and 22). The inability of Ts16 mice to survive is a major limitation of the model, in fact many of the features of the human DS phenotype occur postnatal and sometimes even much later in life.

3.2 Ts65Dn mice. This mutant mouse was produced in order to generate mice trisomic for the syntenic segment of the MMU16 that carries genes of the HSA21 DSCR (Davisson et al., 1990; Davisson et al., 1993). In particular, Ts65Dn mouse carry a triplication of a genomic sequence from Mrpl39 to Znf295 genes (Kahlem et al., 2004) and contains several genes like App, Sod1, Sim2, Mx1 and so on).

(10)

Contrary to Ts16, the Ts65Dn mice have a longer life, therefore, they might be studied both during development and in adult age. Young Ts65Dn mice were found to be smaller in size compared to control mice but become obese in adult age (Davisson et al., 1993; Kola and Hertzog, 1998). Ts65Dn females have fewer litters than controls (3-7 instead of 10-12) and they exhibit poor care of offspring. Trisomic male mice are generally sterile, and fertile ones are very rare, thus preventing the development of stable colonies and reducing the impact of the model. However, in a recent study Moore et al. (2010) reported the identification and the selective breeding of rare fertile males from two working colonies of Ts65Dn mice, in this way trisomic offspring can be propagated to produce large cohorts of closely related siblings.

The Ts65Dn mouse shows several behavioral alterations resembling those observed in persons with DS. In fact, these mice have deficits in spatial learning and memory (Reeves et al., 1995; Demas et al., 1996; 1998) and altered sexual and aggressive behavior (Klein et al., 1996). The differences in mating behavior and aggression are indicative of deficits in adaptive behavior and persons with DS have been reported to have a variety of adaptive behavioral deficits (Collacott, 1993). As these impairments in the mating and aggressive behavior of Ts65Dn mice do not appear to be due to sensorimotor deficits (Klein et al., 1996), it has been suggested that these mice may serve as useful animal model for the study of other behavioral abnormalities associated with DS, such as learning and memory impairments (Escorihuela et al., 1995; Reeves et al., 1995; Demas et al., 1996).

3.3 Ts1Cje and Ts2Cje Mice. Ts1Cje mouse is a segmental trisomy mouse in which the non-centromeric end of mouse chromosome 16 was translocated to the mouse chromosome 12 (Sago et al., 1998). Ts1Cje mice contain 85 genes including Sod1, Ets2 and Mx1 genes, but not App gene (Fig.1); this segment corresponds to human 21q22.1 to 22.3; the Sod1 gene was disrupted by the chromosomal breakpoint. Both male and female Ts1Cje mice are fertile and produce 50% of Ts1Cje progeny when crossed to wild-type animals. Ts1Cje mice have survived more than 1 year and are indistinguishable from controls in gross appearance. Despite this, reduction in the dimension of the skulls and a pattern of craniofacial anomalies similar to Ts65Dn mice were demonstrated

(11)

(Richtsmeier et al., 2002). Moreover, Ts1Cje mice have distinct learning and behavioral abnormalities. Ts2Cje mouse was generated in 2005 (Villar et al., 2005) after a fortuitous translocation of the Ts65Dn marker chromosome to chromosome 12 and contain App gene.

3.4 Ms1Ts65 Mice. In 2000 Sago and co-worker (Sago et al., 2000) generated a new mutant mouse segmentally trisomic for the region by which Ts1Cje and Ts65Dn differ: the region from App to SOD1 genes in which there are 46 genes, to assess the contribution of this region to the Ts65Dn phenotype. These mice were generated crossing Ts65Dn females to Ts1Cje males and shown a much less severe phenotypic impact than other models.

3.5 Recent models

Starting from 2004 a number of new murine models for DS has been published (see Fig.1), in particular the Ts1Rhr Mice (Olson et al., 2004) trisomic for the DSCR; the MTs1Rhr Mice (Olson et al., 2004) disomic for the DSCR; the Tc1 (O’Doherthy et al., 2005) aneuploid for HSA21; the Dp(10)1Yey/+; Dp(16)1Yey/+; Dp(17)1Yey/+ Mice (Yu et al., 2010) with a full triplication of syntenic fragments of MMU10, MMU16 and MMU17; and the Ts1Yah Mice (Pereira et al.; 2009) that carries genes of the MMU17. Virtually no data on brain circuits are however available for these models, with the exception of Tc1 mice (Galante et al., 2009; Morice et al., 2008; O'Doherty et al., 2005; Sheppard et al., 2011).

4. Alterations of brain circuits

Due to the evident cognitive disabilities found in person with DS, most of murine models listed above have been studied almost exclusively from a behavioral point of view (cognitive and learning tasks; see for recent literature Seregaza et al., 2006; Roubertoux and Carlier, 2010). Very little is known about possible neurochemical impairments in the brain of these strains and investigations on the alterations of neuronal circuits in murine models converged principally to MMU16 extra copy genes models. From this point of

(12)

view Ts65Dn and Ts1Cje mice are the most studied models (see tables 1-2). Generally, these studies were directed to brain structural alterations or neurotransmitter/neuropeptide systems’ modifications.

4.1 Cholinergic system

The degeneration of basal forebrain cholinergic neurons is a typical hallmark observed in persons with DS and Alzheimer disease (Yates et al., 1983; Casanova et al., 1985), therefore, several investigations involved the cholinergic system of Ts65Dn mice (see table 1). In postnatal developmental studies a loss of cholinergic neurons in the medial septal nucleus and in the diagonal band of Broca, was observed (comparing Ts65Dn male mice at 4, 6, 8 and 10 months of age) starting from 6 month-old mice, the same age in which began the cognitive decline (Granholm et al., 2000). Other studies reported a much more delayed degeneration: 12 months (comparing Ts65Dn male mice of 4 and 12 months, Seo and Isacson, 2005;), or 18 months in Ts65Dn female mice (Hunter et al., 2004). At the age of 6 months several studies observed a reduction in the number of p75-NGFR- (a low affinity NGF receptor, localized in cholinergic neurons) and trkA- (a high-affinity NGF receptor) immunoreactive neurons of medial septal nucleus (Holtzman et al., 1996; Granholm et al., 2000; Cooper et al., 2001; Salehi et al., 2006) . The evaluation of choline acetyltransferase (ChAT) activity in other brain regions like hippocampus, olfactory bulb, olfactory cortex, pre-frontal cortex, and cerebellum, but not in other neocortical regions, demonstrated an up-regulation of the enzyme activity starting from the age of 10 months in Ts65Dn mice (Seo and Isacson, 2005; Contestabile et al., 2006). Remarkably, Ts1Cje mouse, although shows severe cognitive impairments, does not have degeneration of basal cholinergic forebrain neurons seen in Ts65Dn mouse (Sago et al., 1998).

4.2 Monoaminergic systems

Serotonergic system is altered in adult individuals with DS; this finding may be relevant to the pathogenesis and treatment of cognitive and non-cognitive (behavioral) features in DS (Seidl et al., 1999), therefore this system was investigated in Ts65Dn mice. However, a study performed in 3 month-old Ts65Dn mice found no alterations in the serotonergic

(13)

as well as in the catecholaminergic systems [identified by tyrosine hydroxylase (TH) immunoreactivity, Megias et al., 1997]. Interestingly, in this investigation no differences were found also in the cholinergic system, confirming that this system is altered later in life (Granholm et al., 2000). No subsequent investigations of TH or serotonin systems were performed in adults Ts65Dn or in other DS models.

Some studies were dedicated to the noradrenergic system that is involved in attentional and cognitive processes. In 5 month-old Ts65Dn mice, the central noradrenergic transmission, in particular the adenylate cyclase activity, was altered in the hippocampus and cerebral cortex, but not in other brain regions (Dierssen et al., 1997). A recent study reported that 8 month-old Ts65Dn mice exhibit progressive loss of norepinephrine phenotype in neurons of the Locus coeruleus, thus mimicking the early loss of locus coeruleus neurons in AD and in DS, that may contribute to these neuropathologies (Lockrow et al., 2011).

4.3 Nitrergic system

Nitric oxide (NO) is a gaseous neurotransmitters produced by the nitric oxide synthase (NOS) enzyme. NO is implicated in the control of several behaviors like aggressive and sexual behavior: for example, elevated aggressive behavior was reported in mice lacking the gene for neural isoform of NOS (Nelson et al., 1995). Several of the NO-related behaviors are altered in both persons with DS and in Ts65Dn mice, suggesting a possible involvement of nNOS-derived NO in DS. In a first study we demonstrated that Ts65Dn mice show both an increase of aggressive behavior and a precocious reduction of nitrergic neurons in the basal forebrain nuclei and in the hypothalamic paraventricular nucleus (PVN) at 3 months of age (Gotti et al., 2004; Fig.2) indicating a clear correlation of the behavioral aggressiveness with deficient NO production. The alteration of nNOS system is selective and don’t involve the whole nitrergic system; in fact other regions, as the pontine tegmental nuclei don’t show alterations of both nitrergic and cholinergic systems (Gotti et al., 2005). Tegmental nuclei are connected to the vertical part of the diagonal band of Broca (VDB) and medial septum (MS) and are important to start and stop locomotion; they represent a major mesopontine cholinergic system. Nitrergic and cholinergic systems are functionally related (de Vente, 2004), however the degree of

(14)

co-localization is variable in different brain regions. In our study (Gotti et al., 2005) we demonstrated a high degree of co-localization in pontine tegmental nuclei, whereas, in VDB-MS there is a low degree of co-localization (Fig.2). Noteworthy, at the age of 3 months the nitrergic system is altered only at the level of basal forebrain, while the cholinergic system is not altered (both in the basal forebrain, and in the mesopontine system). As previously reported, the cholinergic system starts its degeneration at later age (6 months, see Granholm et al., 2000) in VDB-MS, and no sign of degeneration have been reported for the mesopontine system. Therefore, it seems that the high degree of co-localization may represent a protective factor for the degeneration of neural circuits (mesopontine region), while in the basal telencephalon the precociuos alterations of nitrergic system could be causally related to the subsequent degeneration of the cholinergic system detected in basal forebrain of older animals.

4.4 Vasopressinergic system

Data collected in patients with DS demonstrated alterations in osmoregulation and autonomic functions, and indicated the vasopressinergic system of PVN as the major locus responsible of these changes (Mann et al., 1985; Labudova et al., 1998). Therefore, we investigated the distribution of arginine-vasopressin (AVP) immunoreactivity in the PVN of aged Ts65Dn female mice. The number of AVP immunoreactive neurons was significantly reduced in mutants as compared to controls (Panzica et al., 2006); therefore it seems that the AVP producing hypothalamic magnocellular system is one of major targets of the DS. On the other hand, AVP changes could reflect derangement of the adrenergic neurotransmitter system as the interaction between AVP and adrenergic neurons was proposed in literature (Labudova et al., 1998).

5. Structural alterations of the brain

As reported in the introduction, individuals with DS show several brain structural alterations in different regions. Similar studies have been performed in both Ts65Dn and Ts1Cje mice illustrating macro- and microscopical changes in the hippocampus and cortex (table 2).

(15)

5.1 Hippocampus

Starting from post-natal day 2, in Ts65Dn mice, it was observed a reduction of both hippocampal volume and cell proliferation (Contestabile et al., 2007). Moreover, at post-natal day 21 Ts65Dn mice show a reduction of hippocampal spine density (Belichenko et al., 2004) and, at 2 months of age, an alteration of neurogenesis (Clark et al., 2006). Also the electrical properties of hippocampus are altered, in fact, it was observed a reduction of long-term potentation (LTP) and an increase in long-term depression (LTD) in CA1 and DG regions (Siarey et al., 1999; Belichenko et al., 2007). In Ts1Cje and Ts1Rhr mice the decrease of hippocampal spine density was observed at 6 months of age (Belichenko et al., 2007, 2009). A reduction of neurogenesis within the subventricular zone of the lateral ventricle and of the hippocampal DG was found also in Ts1Cje mice at 3 months of age (Ishihara et al., 2010).

5.2 Cortex, cerebellum, and lateral ventricles

Development of the neocortex of the Ts16 mouse is characterized by a transient delay in the radial expansion of the cortical wall and a persistent reduction in cortical volume (Haydar et al., 1996). This is due to altered proliferative characteristics during Ts16 neuronogenesis and it results in a delay in the generation of neocortical neurons, leading to significant neocortical abnormalities such as those found in humans with DS (Haydar et al., 2000).

Pyramidal cells in environmentally enriched control animals become significantly more branched and more spinous than non-enriched controls. However, environmental enrichment had little effect on pyramidal cell structure in Ts65Dn mice. Differences in the number of spines found in the dendritic arbors of pyramidal cells in the two groups reflect differences in the number of excitatory inputs they receive and, consequently, complexity in cortical circuitry. These results suggest that behavioral deficits demonstrated in the Ts65Dn model could be attributed to abnormal circuit development (Dierssen et al., 2003). In addition, Ts65Dn mice show an increase of synaptophysin levels and calcium binding protein immunoreactive neurons from 4 months of age (Pérez-Cremades et al., 2010), and, starting from the age of 12 months, there is an increase of

(16)

VIP-immunoreactive neurons and of APP protein levels (Hill et al., 2003; Hunter et al., 2003). Other studies were performed in Ts1Cje mice demonstrating oxidative stress, mitochondrial dysfunction and hyperphosphorylation of tau, all of which may play critical roles in the pathogenesis of mental retardation in DS (Shukkur et al., 2006). Altered regulation of tau phosphorylation was also observed in Tc1 mice (Sheppard et al., 2011).

The whole size of the cerebellum and the number of granule cells was reduced very early in Ts65Dn mice (Roper et al., 2006; Aldridge et al., 2007; Baxter et al., 2000), as well as in Ts1Cje (Olson et al., 2004) and in Tc1 (O'Doherty et al., 2005). In particular, Tc1 mice revealed motor behavior alterations that may be related to cerebellar circuits alteration (Galante et al., 2009).

In recent studies it was observed a general reduction of the volume of ventricles in Ts65Dn mice (Bianchi et al., 2010a) and an enlargement of ventricles’ size in Ts1Cje mice (Ishihara et al., 2010).

6. Concluding remarks

DS is a complex disease and the mechanisms by which it produces phenotypic abnormalities are not fully understood (Dierssen et al., 2001; Antonarakis and Epstein, 2006). As explained in the introduction, in spite of many biochemical and physiological studies of the DS, it is still not fully known how the individual genes on HSA21, either singly or in association produce the phenotypical and behavioral alterations associated with the DS.

The number of murine models of DS is rapidly increasing. They have been generated, using different strategies (i.e., single gene, HSA21 inserted genes, or MMU extra copy genes models), but they are in some way under-used and scarcely known. These murine models provided a lot of new insights about cognitive impairments and disabilities and the possible correlation with a triplication of some genes. Nevertheless, detailed descriptions of neural circuits alterations and the comparison with those observed in the brain of humans with DS is still sporadic and not systematic, with the exception of the cholinergic system.

(17)

Changes in the size of the whole brain, of the cerebellum, and of the ventricles have been well documented in humans with DS and in Ts65Dn mice and other models (see table 2). Losses of brain functions, decreased levels of specific proteins, and alterations of brain circuits have also been discovered in trisomic mice, but there are serious gaps in the possible comparisons. For example: in DS brains (as well as in Alzheimer patients), gliosis and astrocytosis are frequently observed (Griffin et al., 1989). This is probably due to an overexpression of S100 beta gene (located in the DSCR of HSA21) that codes for S100, a calcium binding protein highly expressed in astrocytes, but also in cerebral neurons (Rickmann and Wolff, 1995; Castagna et al., 2003). In Ts65Dn mice there is no gliosis or astrocytes alteration, probably because the S100beta gene is located in mouse chromosome 10 and is not triplicated in this strain. The recent models in which there is a triplication of all syntenic genes of chromosome 10, 16 and 17 (Yu et al., 2010) could be very important for this point of view; unfortunately, this model was studied only for behavioral profiles. Another example is the Ts1Yah mice: the triplication of the genes of chromosome 17 in this case is not inducing cognitive impairment (Pereira et al., 2009) but nothing is known about the general brain morphology of this model.

At the moment, the Ts65Dn mouse is the only strain that have been investigated in details for behavioral deficits, neural circuits and brain structural alterations (see table 1-2). The earliest signal of brain circuits’ alteration in these mutant mice is represented by a decrease in the cell proliferation during the first week of life in the hippocampus (Contestabile et al., 2007), followed by the decrease of neurogenesis in the hippocampus at 2 months of life (Clark et al., 2006). In the basal forebrain, the nitrergic system shows a significant decrease of cell number at the age of 3 months (Gotti et al., 2004), followed by alteration of the noradrenergic transmission at the age of 5 months (Dierssen et al., 1997), and by the descrease of the cholinergic system at the age of 6 months (Granholm et al., 2000) or later (Seo and Isacson, 2005). These data, together with those collected at molecular level (Antonarakis and Epstein, 2006) could be very useful to understand the genesis of DS and provide hypothetical possibilities for exogenous correction of altered gene expression in DS.

However, present knowledge of the molecular pathogenesis of DS is poor, and the murine models have been largely used for descriptive and not for experimental purposes.

(18)

An overall analysis and comparative study of the existing models should be considered, but mice should be on the same genetic background and protocols should cover every aspect of mouse neurological, behavioral, biochemical, and histopathological features. Present descriptions are lacking of several details, for example studies comparing directly male and female expression of different markers are missing, many models have been studied only for their behavioral alterations and very few post-mortem neurochemical analyses in DS brain has been performed in order to validate and confirms data observed in murine models. The hope for the future is that the large amount of knowledge accumulated for some of the murine models (e.g. Ts65Dn mice) could be soon utilized to have a better understanding of the DS. In a complex disease like DS even each little “tessera” would be very important for the comprehension of the entire story.

Acknowledgements

This study was supported by University of Torino, Regione Piemonte and Cavalieri-Ottolenghi Foundation, Italy

(19)

References

Aldridge, K., Reeves, R.H., Olson, L.E., Richtsmeier, J.T., 2007. Differential effects of trisomy on brain shape and volume in related aneuploid mouse models. Am J Med Genet A, 143A, 1060-1070.

Alves-Sampaio, A., Troca-Marin, J.A., Montesinos, M.L., 2010. NMDA-mediated regulation of DSCAM dendritic local translation is lost in a mouse model of Down's syndrome. J Neurosci, 30, 13537-13548.

Antonarakis, S.E., Epstein, C.J., 2006. The challenge of Down syndrome. Trends Mol Med. 12, 473-9.

Baxter, L.L., Moran, T.H., Richtsmeier, J.T., Troncoso, J., Reeves, R.H., 2000. Discovery and genetic localization of Down syndrome cerebellar phenotypes using the Ts65Dn mouse. Hum Mol Genet, 9, 195-202.

Belichenko, N.P., Belichenko, P.V., Kleschevnikov, A.M., Salehi, A., Reeves, R.H., Mobley, W.C., 2009. The "Down syndrome critical region" is sufficient in the mouse model to confer behavioral, neurophysiological, and synaptic phenotypes characteristic of Down syndrome. J Neurosci, 29, 5938-5948.

Belichenko, P.V., Kleschevnikov, A.M., Salehi, A., Epstein, C.J., Mobley, W.C., 2007. Synaptic and cognitive abnormalities in mouse models of Down syndrome: exploring genotype-phenotype relationships. J Comp Neurol, 504, 329-345. Belichenko, P.V., Masliah, E., Kleschevnikov, A.M., Villar, A.J., Epstein, C.J., Salehi,

A., Mobley, W.C., 2004. Synaptic structural abnormalities in the Ts65Dn mouse model of Down Syndrome. J Comp Neurol, 480, 281-298.

Bianchi, P., Ciani, E., Contestabile, A., Guidi, S., Bartesaghi, R., 2010a. Lithium restores neurogenesis in the subventricular zone of the Ts65Dn mouse, a model for Down syndrome. Brain Pathol, 20, 106-118.

Bianchi, P., Ciani, E., Guidi, S., Trazzi, S., Felice, D., Grossi, G., Fernandez, M., Giuliani, A., Calza, L., Bartesaghi, R., 2010b. Early pharmacotherapy restores neurogenesis and cognitive performance in the Ts65Dn mouse model for Down syndrome. J Neurosci, 30, 8769-8779.

(20)

Bimonte-Nelson, H.A., Hunter, C.L., Nelson, M.E., Granholm, A.C., 2003. Frontal cortex BDNF levels correlate with working memory in an animal model of Down syndrome. Behav Brain Res, 139, 47-57.

Bovicelli, L., Orsini, L.F., Rizzo, N., Montacuti, V., Bacchetta, M., 1982. Reproduction in Down syndrome. Obstet Gynecol, 59, 13S-17S.

Branchi, I., Bichler, Z., Minghetti, L., Delabar, J.M., Malchiodi-Albedi, F., Gonzalez, M.C., Chettouh, Z., Nicolini, A., Chabert, C., Smith, D.J., Rubin, E.M., Migliore-Samour, D., Alleva, E., 2004. Transgenic mouse in vivo library of human Down syndrome critical region 1: association between DYRK1A overexpression, brain development abnormalities, and cell cycle protein alteration. J Neuropathol Exp Neurol, 63, 429-440.

Casanova, M.F., Walker, L.C., Whitehouse, P.J., Price, D.L., 1985. Abnormalities of the nucleus basalis in Down's syndrome. Ann Neurol, 18, 310-313.

Castagna, C., Viglietti-Panzica, C., Panzica, G.C., 2003. Protein S100 immunoreactivity in glial cells and neurons of the Japanese quail brain. Journal of Chemical Neuroanatomy. 25, 195-212.

Cataldo, A.M., Petanceska, S., Peterhoff, C.M., Terio, N.B., Epstein, C.J., Villar, A., Carlson, E.J., Staufenbiel, M., Nixon, R.A., 2003. App gene dosage modulates endosomal abnormalities of Alzheimer's disease in a segmental trisomy 16 mouse model of down syndrome. J Neurosci, 23, 6788-6792.

Chabert, C., Jamon, M., Cherfouh, A., Duquenne, V., Smith, D.J., Rubin, E., Roubertoux, P.L., 2004. Functional analysis of genes implicated in Down syndrome: 1. Cognitive abilities in mice transpolygenic for Down Syndrome Chromosomal Region-1 (DCR-1). Behav Genet, 34, 559-569.

Clark, S., Schwalbe, J., Stasko, M.R., Yarowsky, P.J., Costa, A.C., 2006. Fluoxetine rescues deficient neurogenesis in hippocampus of the Ts65Dn mouse model for Down syndrome. Exp Neurol, 200, 256-261.

Collacott, R.A., 1993. Epilepsy, dementia and adaptive behaviour in Down's syndrome. Intellect. Disabil. Res., 37, 153-160.

Contestabile, A., Fila, T., Bartesaghi, R., Contestabile, A., Ciani, E., 2006. Choline acetyltransferase activity at different ages in brain of Ts65Dn mice, an animal

(21)

model for Down's syndrome and related neurodegenerative diseases. J Neurochem, 97, 515-526.

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.

Contestabile, A., Benfenati, F., Gasparini, L., 2010. Communication breaks-Down: from neurodevelopment defects to cognitive disabilities in Down syndrome. Prog Neurobiol, 91, 1-22.

Cooper, J.D., Salehi, A., Delcroix, J.D., Howe, C.L., Belichenko, P.V., Chua-Couzens, J., Kilbridge, J.F., Carlson, E.J., Epstein, C.J., Mobley, W.C., 2001. Failed retrograde transport of NGF in a mouse model of Down's syndrome: reversal of cholinergic neurodegenerative phenotypes following NGF infusion. Proc Natl Acad Sci U S A, 98, 10439-10444.

Davisson, M.T., 2005. Mouse models of Down syndrome. Drug Discovery Today: Disease Models, 2, 103-109.

Davisson, M.T., Schmidt, C., Akeson, E.C., 1990. Segmental trisomy of murine chromosome 16: A new model system for studying Down syndrome. Progress in Clinical and Biological Research, 360, 263-280.

Davisson, M.T., Schmidt, C., Reeves, R.H., Irving, N.G., Akeson, E.C., Harris, B.S., Bronson, R.T., 1993. Segmental trisomy as a mouse model for Down syndrome. Prog Clin Biol Res, 384, 117-133.

de Vente, J., 2004. cGMP: a second messenger for acetylcholine in the brain? Neurochem Int, 45, 799-812.

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. Eur J Hum Genet, 1, 114-124.

Demas, G.E., Nelson, R.J., Krueger, B.K., Yarowsky, P.J., 1996. Spatial memory deficits in segmental trisomic Ts65Dn mice. Behav Brain Res, 82, 85-92.

(22)

Demas, G.E., Nelson, R.J., Krueger, B.K., Yarowsky, P.J., 1998. Impaired spatial working and reference memory in segmental trisomy (Ts65Dn) mice. Behav Brain Res, 90, 199-201.

Dierssen, M., Benavides-Piccione, R., Martinez-Cue, C., Estivill, X., Florez, 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. Cereb Cortex, 13, 758-764.

Dierssen, M., Fillat, C., Crnic, L., Arbones, M., Florez, J., Estivill, X., 2001. Murine models for Down syndrome. Physiol Behav, 73, 859-871.

Dierssen, M., Vallina, I.F., Baamonde, C., Garcia-Calatayud, S., Lumbreras, M.A., Florez, J., 1997. Alterations of central noradrenergic transmission in Ts65Dn mouse, a model for Down syndrome. Brain Res, 749, 238-244.

Down, J.L.H., 1867. Observations on an ethnic classification of idiots. J Mental Sci, 13, 121-123.

Ema, M., Ikegami, S., Hosoya, T., Mimura, J., Ohtani, H., Nakao, K., Inokuchi, K., Katsuki, M., Fujii-Kuriyama, Y., 1999. Mild impairment of learning and memory in mice overexpressing the mSim2 gene located on chromosome 16: an animal model of Down's syndrome. Hum Mol Genet, 8, 1409-1415.

Epstein, C.J., 1986. Developmental genetics. Experientia, 42, 1117-1128.

Epstein, C.J., Berger, C.N., Carlson, E.J., Chan, P.H., Huang, T.T. (1990) Models for Down syndrome: chromosome 21-specific genes in mice. In Patterson, D., Epstein, C.J. (eds.) Molecular genetics of chromosome 21 and Down syndrome. Wiley-Liss, New York, pp. 215-232.

Epstein, C.J., Cox, D.R., Epstein, L.B., 1985. Mouse trisomy 16: an animal model of human trisomy 21 (Down syndrome). Ann N Y Acad Sci, 450, 157-168.

Escorihuela, R.M., Fernandez-Teruel, A., Vallina, I.F., Baamonde, C., Lumbreras, M.A., Dierssen, M., Tobena, A., Florez, J., 1995. A behavioral assessment of Ts65Dn mice: a putative Down syndrome model. Neuroscience Letters, 199, 143-146. Gahtan, E., Auerbach, J.M., Groner, Y., Segal, M., 1998. Reversible impairment of

(23)

Galante, M., Jani, H., Vanes, L., Daniel, H., Fisher, E.M., Tybulewicz, V.L., Bliss, T.V., Morice, E., 2009. Impairments in motor coordination without major changes in cerebellar plasticity in the Tc1 mouse model of Down syndrome. Hum Mol Genet. 18, 1449-63.

Galdzicki, Z., Siarey, R., Pearce, R., Stoll, J., Rapoport, S.I., 2001. On the cause of mental retardation in Down syndrome: extrapolation from full and segmental trisomy 16 mouse models. Brain Res Rev, 35, 115-145.

Gardiner, K., Costa, A.C., 2006. The proteins of human chromosome 21. Am J Med Genet C Semin Med Genet, 142C, 196-205.

Gardiner, K., Fortna, A., Bechtel, L., Davisson, M.T., 2003. Mouse models of Down syndrome: how useful can they be? Comparison of the gene content of human chromosome 21 with orthologous mouse genomic regions. Gene, 318, 137-147. Gerlai, R., Friend, W., Becker, L., O'Hanlon, D., Marks, A., Roder, J., 1993. Female

transgenic mice carrying multiple copies of the human gene for S100 beta are hyperactive. Behavioral Brain Research, 55, 51-59.

Gotti, S., Caricati, E., Viglietti-Panzica, C., Panzica, G., 2005. Nitrinergic and cholinergic system in the tegmental nuclei of a murine model (Ts65Dn mice) for Down syndrome. Program No. 584.8. Neuroscience Meeting Planner. Washington, DC: Society for Neuroscience, 2005. Online.

Gotti, S., Chiavegatto, S., Sica, M., Viglietti-Panzica, C., Nelson, R.J., Panzica, G., 2004. Alteration of NO-producing system in the basal forebrain and hypothalamus of Ts65Dn mice: an immunohistochemical and histochemical study of a murine model for Down syndrome. Neurobiol Dis, 16, 563-571.

Granholm, A.-C.E., 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. Experimental Neurology, 161, 647-663.

Granholm, A.C., Sanders, L., Seo, H., Lin, L., Ford, K., Isacson, O., 2003. Estrogen alters amyloid precursor protein as well as dendritic and cholinergic markers in a mouse model of Down syndrome. Hippocampus, 13, 905-914.

Griffin, W.S., Stanley, L.C., Ling, C., White, L., MacLeod, V., Perrot, L.J., White, C.L., Araoz, C., 1989. Brain interleukin 1 and S-100 immunoreactivity are elevated in

(24)

Down syndrome and Alzheimer disease. Proceedings of the National Academy of Sciences of the United States of America, 86, 7611-7615.

Haydar, T.F., Blue, M.E., Molliver, M.E., Krueger, B.K., Yarowsky, P.J., 1996. Consequences of trisomy 16 for mouse brain development: corticogenesis in a model of Down syndrome. J Neurosci. 16, 6175-82.

Haydar, T.F., Nowakowski, R.S., Yarowsky, P.J., Krueger, B.K., 2000. Role of founder cell deficit and delayed neuronogenesis in microencephaly of the trisomy 16 mouse. J Neurosci. 20, 4156-4164.

Hardy, J., Irving, N., Kessling, A., 1989. Down on chromosome 21? Trends Neurosci, 12, 209-210.

Harris-Cerruti, C., Kamsler, A., Kaplan, B., Lamb, B., Segal, M., Groner, Y., 2004. Functional and morphological alterations in compound transgenic mice overexpressing Cu/Zn superoxide dismutase and amyloid precursor protein [correction]. Eur J Neurosci, 19, 1174-1190.

Hassold, T.J., Jacobs, P.A., 1984. Trisomy in man. Annu Rev Genet, 18, 69-97.

Hattori, M., Fujiyama, A., Taylor, T.D., Watanabe, H., Yada, T., Park, H.S., Toyoda, A., Ishii, K., Totoki, Y., Choi, D.K., Groner, Y., 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., Schibuya, K., Kawasaki, K., Asakawa, S., Shintani, A., Sasaki, T., Nagamine, K., Mitsuyama, S., Antonarakis, S.E., Minoshima, S., Shimizu, N., Nordsiek, G., Hornischer, K., Brant, P., Scharfe, M., Schon, O., Desario, A., Reichelt, J., Kauer, G., Blocker, 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.

Hewitt, C.A., Ling, K.H., Merson, T.D., Simpson, K.M., Ritchie, M.E., King, S.L., Pritchard, M.A., Smyth, G.K., Thomas, T., Scott, H.S., Voss, A.K., 2010. Gene network disruptions and neurogenesis defects in the adult Ts1Cje mouse model of Down syndrome. PLoS One, 5, e11561.

(25)

Hill, J.M., Ades, A.M., McCune, S.K., Sahir, N., Moody, E.M., Abebe, D.T., Crnic, L.S., Brenneman, D.E., 2003. Vasoactive intestinal peptide in the brain of a mouse model for Down syndrome. Exp Neurol, 183, 56-65.

Holtzman, D.M., Santucci, D., Kilbridge, J., Chua-Couzens, J., Fontana, D.J., Daniels, S.E., Johnson, R.M., Chen, K., Sun, Y., Carlson, E., Alleva, E., Epstein, C.J., Mobley, W.C., 1996. Developmental abnormalities and age-related neurodegeneration in a mouse model of Down syndrome. Proc Natl Acad Sci U S A, 93, 13333-13338.

Hook, E.B., 1981. Unbalanced Robertsonian translocations associated with Down's syndrome or Patau's syndrome: chromosome subtype, proportion inherited, mutation rates, and sex ratio. Hum Genet, 59, 235-239.

Hunter, C.L., Bachman, D., Granholm, A.C., 2004. Minocycline prevents cholinergic loss in a mouse model of Down's syndrome. Ann Neurol, 56, 675-688.

Hunter, C.L., Bimonte-Nelson, H.A., Nelson, M., Eckman, C.B., Granholm, A.C., 2004b. Behavioral and neurobiological markers of Alzheimer's disease in Ts65Dn mice: effects of estrogen. Neurobiol Aging, 25, 873-884.

Hunter, C.L., Isacson, O., Nelson, M., Bimonte-Nelson, H., Seo, H., Lin, L., Ford, K., Kindy, M.S., Granholm, A.C., 2003. Regional alterations in amyloid precursor protein and nerve growth factor across age in a mouse model of Down's syndrome. Neurosci Res, 45, 437-445.

Insausti, A.M., Megias, M., Crespo, D., Cruz-Orive, L.M., Dierssen, M., Vallina, I.F., Insausti, R., Florez, J., 1998. Hippocampal volume and neuronal number in Ts65Dn mice: a murine model of Down syndrome. Neurosci Lett, 253, 175-178. Ishihara, K., Amano, K., Takaki, E., Ebrahim, A.S., Shimohata, A., Shibazaki, N., Inoue,

I., Takaki, M., Ueda, Y., Sago, H., Epstein, C.J., Yamakawa, K., 2009. Increased lipid peroxidation in Down's syndrome mouse models. J Neurochem, 110, 1965-1976.

Ishihara, K., Amano, K., Takaki, E., Shimohata, A., Sago, H., Epstein, C.J., Yamakawa, K., 2010. Enlarged brain ventricles and impaired neurogenesis in the Ts1Cje and Ts2Cje mouse models of Down syndrome. Cereb Cortex, 20, 1131-1143.

(26)

Kahlem, P., Sultan, M., Herwig, R., Steinfath, M., Balzereit, D., Eppens, B., Saran, N.G., Pletcher, M.T., South, S.T., Stetten, G., Lehrach, H., Reeves, R.H., Yaspo, M.L., 2004. Transcript level alterations reflect gene dosage effects across multiple tissues in a mouse model of down syndrome. Genome Res, 14, 1258-1267.

Kapranov, P., Cawley, S.E., Drenkow, J., Bekiranov, S., Strausberg, R.L., Fodor, S.P., Gingeras, T.R., 2002. Large-scale transcriptional activity in chromosomes 21 and 22. Science, 296, 916-919.

Klein, S.L., Kriegsfeld, L.J., Hairston, J.E., Rau, V., Nelson, R.J., Yarowsky, P.J., 1996. Characterization of sensorimotor performance, reproductive and aggressive behaviors in segmental trisomic 16 (Ts65Dn) mice. Physiol Behav, 60, 1159-1164.

Kola, I., Hertzog, P.J., 1998. Down syndrome and mouse models. Curr Opin Genet Dev, 8, 316-321.

Korenberg, J.R., Chen, X.N., Schipper, R., Sun, Z., Gonsky, R., Gerwehr, S., Carpenter, N., Daumer, C., Dignan, P., Disteche, C., et al., 1994. Down syndrome phenotypes: the consequences of chromosomal imbalance. Proc Natl Acad Sci U S A, 91, 4997-5001.

Korenberg, J.R., Kawashima, H., Pulst, S.M., Ikeuchi, T., Ogasawara, N., Yamamoto, K., Schonberg, S.A., West, R., Allen, L., Magenis, E., et al., 1990. Molecular definition of a region of chromosome 21 that causes features of the Down syndrome phenotype. Am J Hum Genet, 47, 236-246.

Labudova, O., Fang-Kircher, S., Cairns, N., Moenkemann, H., Yeghiazaryan, K., Lubec, G., 1998. Brain vasopressin levels in Down syndrome and Alzheimer's disease. Brain Research. 806, 55-59.

Lejeune, J., Gautier, M., and Turpin, R., 1959. [Study of somatic chromosomes from 9 mongoloid children.]. C. R. Hebd Seances Acad. Sci, 248, 1721-1722.

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. J Alzheimers Dis, 23, 471-489.

(27)

Lockrow, J., Prakasam, A., Huang, P., Bimonte-Nelson, H., Sambamurti, K., Granholm, A.C., 2009. Cholinergic degeneration and memory loss delayed by vitamin E in a Down syndrome mouse model. Exp Neurol, 216, 278-289.

Lomoio, S., Scherini, E., Necchi, D., 2009. Beta-amyloid overload does not directly correlate with SAPK/JNK activation and tau protein phosphorylation in the cerebellar cortex of Ts65Dn mice. Brain Res, 1297, 198-206.

Lorenzi, H.A., Reeves, R.H., 2006. Hippocampal hypocellularity in the Ts65Dn mouse originates early in development. Brain Res, 1104, 153-159.

Macri, J.N., Spencer, K., Aitken, D., Garver, K., Buchanan, P.D., Muller, F., Boue, A., 1993. First-trimester free beta (hCG) screening for Down syndrome. Prenat Diagn, 13, 557-562.

Mann, D.M., Yates, P.O., Marcyniuk, B., 1985. Changes in Alzheimer's disease in the magnocellular neurones of the supraoptic and paraventricular nuclei of the hypothalamus and their relationship to the noradrenergic deficit. Clin Neuropathol. 4, 127-134.

Mao, R., Wang, X., Spitznagel, E.L., Jr., Frelin, L.P., Ting, J.C., Ding, H., Kim, J.W., Ruczinski, I., Downey, T.J., Pevsner, J., 2005. Primary and secondary transcriptional effects in the developing human Down syndrome brain and heart. Genome Biol, 6, R107.

Megias, M., Verduga, R., Dierssen, M., Florez, J., Insausti, R., Crespo, D., 1997. Cholinergic, serotonergic and catecholaminergic neurons are not affected in Ts65Dn mice. Neuroreport, 8, 3475-3478.

Moore, C.S., Hawkins, C., Franca, A., Lawler, A., Devenney, B., Das, I., Reeves, R.H., 2010. Increased male reproductive success in Ts65Dn "Down syndrome" mice. Mamm Genome, 21, 543-549.

Morice, E., Andreae, L.C., Cooke, S.F., Vanes, L., Fisher, E.M., Tybulewicz, V.L., Bliss, T.V., 2008. Preservation of long-term memory and synaptic plasticity despite short-term impairments in the Tc1 mouse model of Down syndrome. Learn Mem. 15, 492-500.

Nadal, M., Mila, M., Pritchard, M., Mur, A., Pujals, J., Blouin, J.L., Antonarakis, S.E., Ballesta, F., Estivill, X., 1996. YAC and cosmid FISH mapping of an unbalanced

(28)

chromosomal translocation causing partial trisomy 21 and Down syndrome. Hum Genet, 98, 460-466.

Nadel, L. (1994) Neural and cognitive development in Down syndrome. In Nadel, L., Rosenthal, D. (eds.) Down Syndrome: living and learning in the community. Wiley-Liss, New York, pp. 107-114.

Necchi, D., Lomoio, S., Scherini, E., 2008. Axonal abnormalities in cerebellar Purkinje cells of the Ts65Dn mouse. Brain Res, 1238, 181-188.

Nelson, R.J., Demas, G.E., Huang, P.L., Fishman, M.C., Dawson, V.L., Dawson, T.M., Snyder, S.H., 1995. Behavioural abnormalities in male mice lacking neuronal nitric oxide synthase. Nature, 378, 383-386.

Newby, D., Aitken, D.A., Crossley, J.A., Howatson, A.G., Macri, J.N., Connor, J.M., 1997. Biochemical markers of trisomy 21 and the pathophysiology of Down's syndrome pregnancies. Prenat Diagn, 17, 941-951.

O'Doherty, A., Ruf, S., Mulligan, C., Hildreth, V., Errington, M.L., Cooke, S., Sesay, A., Modino, S., Vanes, L., Hernandez, D., Linehan, J.M., Sharpe, P.T., Brandner, S., Bliss, T.V., Henderson, D.J., Nizetic, D., Tybulewicz, V.L., Fisher, E.M., 2005. An aneuploid mouse strain carrying human chromosome 21 with Down syndrome phenotypes. Science. 309, 2033-7.

Olson, L.E., Roper, R.J., Baxter, L.L., Carlson, E.J., Epstein, C.J., Reeves, R.H., 2004. Down syndrome mouse models Ts65Dn, Ts1Cje, and Ms1Cje/Ts65Dn exhibit variable severity of cerebellar phenotypes. Dev Dyn, 230, 581-589.

Olson, L.E., Roper, R.J., Sengstaken, C.L., Peterson, E.A., Aquino, V., Galdzicki, Z., Siarey, R., Pletnikov, M., Moran, T.H., Reeves, R.H., 2007. Trisomy for the Down syndrome 'critical region' is necessary but not sufficient for brain phenotypes of trisomic mice. Hum Mol Genet, 16, 774-782.

Panzica, G.C., Bassi, L., Gotti, S., 2006. Vasopressinergic system and aging in the female Ts65Dn mouse, a murine model for Down syndrome. Frontiers in Neuroendocrinology, 27, 2.

Patterson, D., 1987. The causes of Down syndrome. Sci Am, 257, 52-57, 60.

Patterson, D., Costa, A.C., 2005. Down syndrome and genetics - a case of linked histories. Nat Rev Genet, 6, 137-147.

(29)

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. Hum Mol Genet, 18, 4756-4769. Perez-Cremades, D., Hernandez, S., Blasco-Ibanez, J.M., Crespo, C., Nacher, J., Varea,

E., 2010. Alteration of inhibitory circuits in the somatosensory cortex of Ts65Dn mice, a model for Down's syndrome. J Neural Transm, 117, 445-455.

Pollonini, G., Gao, V., Rabe, A., Palminiello, S., Albertini, G., Alberini, C.M., 2008. Abnormal expression of synaptic proteins and neurotrophin-3 in the Down syndrome mouse model Ts65Dn. Neuroscience, 156, 99-106.

Ramakrishna, N., Meeker, H.C., Li, S., Brown, W.T., Rao, R., El Idrissi, A., 2009. Upregulation of beta-catenin expression in down syndrome model Ts65Dn mouse brain. Neuroscience, 161, 451-458.

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. Nat Genet, 11, 177-184.

Richtsmeier, J.T., Zumwalt, A., Carlson, E.J., Epstein, C.J., Reeves, R.H., 2002. Craniofacial phenotypes in segmentally trisomic mouse models for Down syndrome. Am J Med Genet, 107, 317-324.

Rickmann, M., Wolff, J.R., 1995. S100 protein expression in subpopulations of neurons of rat brain. Neuroscience. 67, 977-991.

Roizen, N.J., Patterson, D., 2003. Down's syndrome. Lancet, 361, 1281-1289.

Roper, R.J., Baxter, L.L., Saran, N.G., Klinedinst, D.K., Beachy, P.A., Reeves, R.H., 2006. Defective cerebellar response to mitogenic Hedgehog signaling in Down [corrected] syndrome mice. Proc Natl Acad Sci U S A, 103, 1452-1456.

Roubertoux, P.L., Carlier, M., 2010. Mouse models of cognitive disabilities in trisomy 21 (Down syndrome). Am J Med Genet C Semin Med Genet, 154C, 400-416.

Roubertoux, P.L., Kerdelhue, B., 2006. Trisomy 21: from chromosomes to mental retardation. Behav Genet, 36, 346-354.

(30)

Rueda, N., Mostany, R., Pazos, A., Florez, J., Martinez-Cue, C., 2005. Cell proliferation is reduced in the dentate gyrus of aged but not young Ts65Dn mice, a model of Down syndrome. Neurosci Lett, 380, 197-201.

Sago, H., Carlson, E.J., Smith, D.J., Kilbridge, J., Rubin, E.M., Mobley, W.C., Epstein, C.J., Huang, T.T., 1998. Ts1Cje, a partial trisomy 16 mouse model for Down syndrome, exhibits learning and behavioral abnormalities. Proc Natl Acad Sci U S A, 95, 6256-6261.

Sago, H., Carlson, E.J., Smith, D.J., Rubin, E.M., Crnic, L.S., Huang, T.T., Epstein, C.J., 2000. Genetic dissection of region associated with behavioral abnormalities in mouse models for Down syndrome. Pediatr Res, 48, 606-613.

Salehi, A., Delcroix, J.D., Belichenko, P.V., Zhan, K., Wu, C., Valletta, J.S., Takimoto-Kimura, R., Kleschevnikov, A.M., Sambamurti, K., Chung, P.P., Xia, W., Villar, A., Campbell, W.A., Kulnane, L.S., Nixon, R.A., Lamb, B.T., Epstein, C.J., Stokin, G.B., Goldstein, L.S., Mobley, W.C., 2006. Increased App expression in a mouse model of Down's syndrome disrupts NGF transport and causes cholinergic neuron degeneration. Neuron, 51, 29-42.

Saran, N.G., Pletcher, M.T., Natale, J.E., Cheng, Y., Reeves, R.H., 2003. Global disruption of the cerebellar transcriptome in a Down syndrome mouse model. Hum Mol Genet, 12, 2013-2019.

Seidl, R., Kaehler, S.T., Prast, H., Singewald, N., Cairns, N., Gratzer, M., Lubec, G., 1999. Serotonin (5-HT) in brains of adult patients with Down syndrome. Journal of Neural Transmission. Supplementum, 57, 221-232.

Seo, H., Isacson, O., 2005. Abnormal APP, cholinergic and cognitive function in Ts65Dn Down's model mice. Exp Neurol, 193, 469-480.

Seregaza, Z., Roubertoux, P.L., Jamon, M., Soumireu-Mourat, B., 2006. Mouse models of cognitive disorders in trisomy 21: a review. Behav Genet, 36, 387-404.

Sheppard, O., Plattner, F., Rubin, A., Slender, A., Linehan, J.M., Brandner, S., Tybulewicz, V.L., Fisher, E.M., Wiseman, F.K., 2011. Altered regulation of tau phosphorylation in a mouse model of down syndrome aging. Neurobiol Aging. Shinohara, T., Tomizuka, K., Miyabara, S., Takehara, S., Kazuki, Y., Inoue, J., Katoh,

(31)

Reeves, R.H., Oshimura, M., 2001. Mice containing a human chromosome 21 model behavioral impairment and cardiac anomalies of Down's syndrome. Hum Mol Genet, 10, 1163-1175.

Shukkur, E.A., Shimohata, A., Akagi, T., Yu, W., Yamaguchi, M., Murayama, M., Chui, D., Takeuchi, T., Amano, K., Subramhanya, K.H., Hashikawa, T., Sago, H., Epstein, C.J., Takashima, A., Yamakawa, K., 2006. Mitochondrial dysfunction and tau hyperphosphorylation in Ts1Cje, a mouse model for Down syndrome. Hum Mol Genet, 15, 2752-2762.

Siarey, R.J., Carlson, E.J., Epstein, C.J., Balbo, A., Rapoport, S.I., Galdzicki, Z., 1999. Increased synaptic depression in the Ts65Dn mouse, a model for mental retardation in Down syndrome. Neuropharmacology, 38, 1917-1920.

Smith, D.J., Zhu, Y., Zhang, J., Cheng, J.F., Rubin, E.M., 1995. Construction of a panel of transgenic mice containing a contiguous 2-Mb set of YAC/P1 clones from human chromosome 21q22.2. Genomics, 27, 425-434.

Teipel, S.J., Hampel, H., 2006. Neuroanatomy of Down syndrome in vivo: a model of preclinical Alzheimer's disease. Behav Genet, 36, 405-415.

Villar, A.J., Belichenko, P.V., Gillespie, A.M., Kozy, H.M., Mobley, W.C., Epstein, C.J., 2005. Identification and characterization of a new Down syndrome model, Ts[Rb(12.1716)]2Cje, resulting from a spontaneous Robertsonian fusion between T(171)65Dn and mouse chromosome 12. Mamm Genome, 16, 79-90.

Yates, C.M., Simpson, J., Gordon, A., Maloney, A.F., Allison, Y., Ritchie, I.M., Urquhart, A., 1983. Catecholamines and cholinergic enzymes in pre-senile and senile Alzheimer-type dementia and Down's syndrome. Brain Res, 280, 119-126. Yu, T., Li, Z., Jia, Z., Clapcote, S.J., Liu, C., Li, S., Asrar, S., Pao, A., Chen, R., Fan, N.,

Carattini-Rivera, S., Bechard, A.R., Spring, S., Henkelman, R.M., Stoica, G., Matsui, S., Nowak, N.J., Roder, J.C., Chen, C., Bradley, A., Yu, Y.E., 2010. A mouse model of Down syndrome trisomic for all human chromosome 21 syntenic regions. Hum Mol Genet, 19, 2780-2791.

(32)

Figure legends

Figure 1. Mouse models of trisomy 21. Human chromosome 21 (HSA21) is presented

with syntenic regions on mouse chromosomes (MMU16, MMU17, MMU10). The length of the bar is proportional to the number of genes carried by the fragment. (Partially modified from Seregaza et al., 2006; Roubertoux and Carlier, 2010).

Figure 2. Nitrergic system in basal forebrain, hypothalamus and mesopontine regions of

Ts65Dn mice: comparison with wild type ad co-localization with the cholinergic system.

Top panels: photomicrographs of sections at the level of the vertical part of the diagonal

band of Broca (VDB); middle panels: photomicrographs of sections at the level of the paraventricular hypothalamic nucleus (PVN). Nitrergic neurons are detected by means of immunohistochemistry for the enzyme n-NOS (NOS) or by histochemistry for NADPH-Diaphorase (ND). With both methods it is possible to note a dramatic reduction of the number of positive neurons in the trisomic (TS) mice compared to control (WT) mice.

Bottom panels: ChAT-ND double staining illustrating low co-localization in medial

septum (MS) and VDB, no co-localization in caudate-putamen complex (CPu) and high degree of co-localization in laterodorsal tegmental nucleus (LDT). Brown-orange cells: ChAT postive elements; Blu-violet cells: NADPH-Diaphorase positive elements; black cells: ChAT-ND double stained cells. Magnification bars = 150 µm in top and middle panels; 300 µm in double staining for MS-VDB and CPu; 150 µm in double staining for LDT. For methods see Gotti et al., 2004; Panzica and Garzino, 1997.

Table 1. Neurotransmitters’ systems altered in Ts65Dn mouse. Data are collected

according to different systems and different regions.

Table 2. Structural alterations and changes in neurogenesis in different trisomic models.

Left column reports the trisomic model. The changes are collected according to the different regions.

(33)

brain area and type of investigations

Comparison

TS Vs. Eu Age Sex References

CHOLINERGIC SYSTEM

MEDIAL SEPTAL NUCLEUS

6 months ns Holtzman et al. (1996)

number of p75-NGFR-ir neurons TS<Eu 12 months M Cooper et al. (2001)

19 months M Salehi et al. (2006)

ChAT-ir cell size TS<Eu 8 months M Granholm et al. (2000)

6 months M Granholm et al. (2000)*

number of ChAT-ir neurons TS<Eu 12 months ns Seo and Isacson (2005)

18 months F Hunter et al. (2004)

cell size and density of trkA-ir neurons TS<Eu 6 months M Granholm et al. (2000)

cell size and number of TrkA-ir neurons 19 months M Salehi et al. (2006)

BASAL FOREBRAIN

NGF protein levels TS<Eu 9 months F Hunter et al. (2003)

HIPPOCAMPUS

ChAT enzyme activity TS>Eu 10 monthsM and F Contestabile et al. (2006)

12 months ns Seo and Isacson (2005)

CEREBELLUM

ChAT enzyme activity TS>Eu 10 monthsM and F Contestabile et al. (2006)

NORADRENERGIC SYSTEM

CORTEX

basal level of cAMP TS<Eu 5 months ns Dierssen et al. (1997)

HIPPOCAMPUS

basal level of cAMP TS<Eu 5 months ns Dierssen et al. (1997)

LOCUS COERULEUS

loss of norepinephrine phenotype in neurons TS>Eu 8 months ns Lockrow et al. (2011)

NITRERGIC SYSTEM

number of NOS-ir neurons TS<Eu 3 months M Gotti et al. (2004)

PARAVENTRICULAR NUCLEUS

number of NOS-ir neurons TS<Eu 3 months M Gotti et al. (2004)

PEPTIDERGIC SYSTEMS

PARAVENTRICULAR NUCLEUS

number of vasopressin-ir neurons TS<Eu 18 months F Panzica et al. (2006)

HIPPOCAMPUS

VIP-ir neurons TS>Eu 12 months M Hill et al. (2003)

* also in Ventral part of the Diagonal band of Broca

(34)

Mouse model brain area and type of investigations

Comparison

TS Vs. Eu Age Sex References

STRUCTURAL ALTERATIONS

MEDIAL SEPTAL NUCLEUS

Ts65Dn enlarged endosome TS>Eu 6 months M and F Cataldo et al. (2003)

microglial activation (CD45-ir) TS>Eu 18 months F Hunter et al. (2004)

LATERAL VENTRICLES

Ts65Dn volume and lenght TS<Eu 12 months F Bianchi et al. (2010b)

Ts1Cje size TS>Eu PD12 M Ishihara et al. (2010)

CORTEX

Ts65Dn GAD-67, CB, CR, PV immunoreactivity in somatosensory synaptophysin levels in somatosensory cortex TS>Eu 4 months M Pérez-Cremades et al. (2010) cortex

BDNF protein in frontal cortex 6 months M Bimonte-Nelson et al. (2003)

dendritic arbors size of pyramidal cells TS<Eu 12 months F Dierssen et al. (2003)

branching pattern and dendritic lenght of pyramidal cells

VIP-ir neurons TS>Eu 12 months M Hill et al. (2003)

APP protein levels 13 months F Hunter et al. (2003)

CEREBELLUM

Ts65Dn size and number of granule cell (GC) PD6 ns Roper et al. (2006)

whole size TS<Eu 3 months ns Aldridge et al. (2007)

internal granule layer (IGL) and moleculare layer (ML) 4 months M and F Baxter et al. (2000)

Purkinje cells axonal alterations TS>Eu 10 months Necchi et al. (2008)

ML and IGL thickness and cell density TS<Eu ns

death of Punkinje cells TS>Eu 12 months ns Lomoio et al. (2009)

Ts1Cje volume and granule cells density TS<Eu 2,5 monthsM and F Olson et al. (2004)

Tc1 granule cells density TS<EU ns M O'Doherthy et al. (2005)

HIPPOCAMPUS

Ts65Dn volume of GCL PD2 ns Contestabile et al. (2007)

number and density of granule cells in DG TS<Eu PD6 M Lorenzi and Reeves (2006)

spine density PD21 M Belichenko et al. (2004)

size of Presynaptic terminals and dendritic spines TS>Eu

presence of EEs that contain NGF and App TS>Eu 3 months M Salehi et al. (2006)

synaptophysin levels TS<Eu 4 months ns Pollonini et al. (2008)

NT3 and CDK5 levels TS>Eu

CB-ir neurons in CA1/CA2 regions TS<Eu 4 months M Lockrow et al. (2009)

10 months M Hunter et al. (2004)

mean neuron number in granule cell layer (GCL) of DG TS<Eu 5 months ns Insausti et al. (1998)

mean neuron number in granule cell layer (GCL) of CA3

TS>Eu 5 months ns

NGF protein levels TS>Eu 6 months M Cooper et al. (2001)

TS<Eu 13 months F Hunter et al. (2003)

beta amyloid levels TS>Eu 6 months M Hunter et al. (2004b)

immunolabeling for Map2 in CA1 and DG TS<Eu 9 months F Granholm et al. (2003)

APP protein levels TS>Eu 13 months F Hunter et al. (2003)

microglial activation (CD45-ir) TS>Eu 18 months F Hunter et al. (2004)

Ts1Cje DSCAM levels in neurons TS>Eu adults (ns) ns Alves-Sampaio et al. (2010)

LPO-derived products TS>Eu 3 months M Ishihara et al. (2009)

spine density TS<Eu 6 months M Belichenko et al. (2007)

Ts1Rhr spine density TS<Eu 6 months M Belichenko et al. (2009)

STRIATUM

Ts65Dn APP protein levels TS>Eu 6 months F Hunter et al. (2003)

WHOLE BRAIN

Ts65Dn levels of MAP2 and NT3 TS>Eu PD1 ns Pollonini et al. (2008)

Riferimenti

Documenti correlati

The MCS paradigm alone does not involve the use of intermediate nodes to share data collected by mobile devices, and it considers only the device-to-device communication and the

d’acteurs, y compris lorsque ceux-ci revendiquent un comportement participatif : il peut aussi bien être adopté par des militants associatifs soucieux de s’engager dans les

Se alcuni dei nuclei narrativi e spettacolari più importanti, inoltre, sono già progettati nella prima scaletta (il terremoto di Catana, Anniba- le sulle Alpi, la battaglia

Although data on the expression of HSPB1 protein are more conflicting with some studies reporting enhanced [ 38 , 39 ] and others reduced [ 40 , 41 ] HSPB1 expression,

Un metodo utilizzato nella tecnica per attenuare questi fenomeni è la creazione di una finestra di comunicazione fra dorso e ventre della pala, in modo tale da creare

The provenance study of stone materials, which have been used in the construction of Nora monuments, has been tackled both petrographically and chemically, comparing the

Because the apical loop is an important site for regulation of miRNA biogene- sis ( 10 ), we hypothesized that Rbfox proteins would specif- ically bind to the sequence motif