anatomi penting abstrak

Embed Size (px)

Citation preview

  • 8/11/2019 anatomi penting abstrak

    1/14

    R E V I E W A R T I C L E

    Spinal muscular atrophy: a motor neuron disorder or a

    multi-organ disease?Monir Shababi,1,2, Christian L. Lorson1,2 and Sabine S. Rudnik-Schoneborn3

    1Department of Veterinary Pathobiology, Life Sciences Center, University of Missouri, Columbia, MO, USA2Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, USA3Institute of Human Genetics, Medical Faculty, University Hospital Aachen, Aachen, Germany

    Abstract

    Spinal muscular atrophy (SMA) is an autosomal recessive disorder that is the leading genetic cause of infantile

    death. SMA is characterized by loss of motor neurons in the ventral horn of the spinal cord, leading to

    weakness and muscle atrophy. SMA occurs as a result of homozygous deletion or mutations in Survival MotorNeuron-1 (SMN1). Loss of SMN1 leads to a dramatic reduction in SMN protein, which is essential for motor

    neuron survival. SMA disease severity ranges from extremely severe to a relatively mild adult onset form of

    proximal muscle atrophy. Severe SMA patients typically die mostly within months or a few years as a

    consequence of respiratory insufficiency and bulbar paralysis. SMA is widely known as a motor neuron disease;

    however, there are numerous clinical reports indicating the involvement of additional peripheral organs

    contributing to the complete picture of the disease in severe cases. In this review, we have compiled clinical

    and experimental reports that demonstrate the association between the loss of SMN and peripheral organ

    deficiency and malfunction. Whether defective peripheral organs are a consequence of neuronal damage/

    muscle atrophy or a direct result of SMN loss will be discussed.

    Key words: Autonomic Nervous System; Cardiac Defects; Peripheral Organs; SMA.

    Introduction

    Clinical classification of SMA

    Spinal muscular atrophy (SMA) is an autosomal recessive dis-

    order that is the most common inherited motor neuron dis-

    ease and the leading genetic cause of newborn mortality.

    SMA occurs in approximately 1 : 10 000 live births with a

    carrier frequency of 1 : 50 (Sugarman et al. 2012). SMA

    Type I was identified more than a century ago by Werdnig

    and Hoffmann as a disorder starting with progressive weak-

    ness at infancy and resulting in death at a very early age.

    They also characterized the pathology of the disease as loss

    of anterior horn cells. Following international consensus,

    four types (IIV) of SMA are classified based on the

    clinical criteria including physical milestones, the onset of

    symptoms, and life span (Munsat & Davies, 1992). The most

    common types are acute infantile (SMA Type I, or Werdnig

    Hoffmann disease), chronic infantile (SMA Type II), chronic

    juvenile (SMA Type III, or KugelbergWelander disease),

    and adult onset (SMA Type IV). Infants with the most severe

    form of SMA can have decreased movement at prenatal

    stage and rarely present with fetal akinesia sequence, con-

    genital contractures and postnatal respiratory insufficiency.

    The majority of SMA Type I patients have normal strength

    at birth but exhibit progressive weakness within a few

    weeks or months. Death usually ensues within 12 months

    (Burnett et al. 2009; Rudnik-Schoneborn et al. 2009),

    although this typical life span can be extended with

    improved nutritional and respiratory care (Oskoui et al.

    2007). Types II and III SMA present with weakness during

    childhood and usually exhibit variable degrees of physical

    impairment (Zerres et al. 1997; Bosboom et al. 2009). Type

    IV occurs at adulthood and is the mildest form of the dis-

    ease; approximately 70% of Type II patients reach adult-

    hood but the life expectancy of Types IIIIV is normal

    (Zerres et al. 1997; Lewelt et al. 2012).

    Genetic determinants of SMA

    The cause of SMA was identified as the loss of a gene called

    Survival Motor Neuron-1 (SMN1) located on chromosome

    Correspondence

    Monir Shababi, Department of Veterinary Pathobiology, Life Sciences

    Center, University of Missouri, Room 404, Columbia, MO 65211, USA.

    T: + 1 573 8842680; F: + 1 573 8849395; E: [email protected]

    Accepted for publication26 June 2013

    2013 Anatomical Society

    J. Anat. (2013) doi: 10.1111/joa.12083

    Journal of Anatomy

  • 8/11/2019 anatomi penting abstrak

    2/14

    5q (Brzustowicz et al. 1990; Lefebvre et al. 1995). A human-

    specific copy gene,SMN2, exists on the same region and dif-

    fers from SMN1 by only a few silent nucleotides (Lefebvre

    et al. 1995; Rochette et al. 2001). Therefore, SMN2encodes

    an identical protein to SMN1, although it cannot fullyrestore the function of SMN1. The critical difference

    between SMN1 and SMN2 is a C to T transition at the 6th

    position of exon 7, leading to an alternatively spliced mRNA

    isoform that lacks exon 7 (SMND7) (Lorson et al. 1999;

    Monani et al. 1999). While SMN1-derived transcripts

    produce full-length and functional SMN protein, nearly

    90% ofSMN2-derived transcripts generate a truncated and

    unstable protein (SMND7), which lacks the 16 amino acids

    encoded by exon 7 (Lorson et al. 1998, 1999; Monani et al.

    1999). Importantly, however,SMN2produces a low amount

    of full-length SMN, comparable to ~ 10% of SMN1 levels

    (Lorson & Androphy, 2000).SMN2is paradoxically a double-

    edged sword: it provides only a sufficient level of SMN pro-

    tein to allow survival and subsequent development of SMA;

    it is also the most potent disease modifier and a bona fide

    therapeutic target (Mailman et al. 2002; Wirth et al. 2006).

    The number of SMN2copies, and therefore the level of the

    full-length SMN created by SMN2 transcripts, directly

    impacts disease severity. Type I individuals typically have

    one to two copies of SMN2, whereas milder forms of SMA

    are mostly associated with three to four copies of SMN2

    (Feldkotter et al. 2002). SMA Type IV is only exceptionally

    caused by SMN deficiency. In addition, clinically unaffected

    individuals have been identified who lack SMN1, pointing

    towards further modifiers for the disease phenotype in rareinstances.

    SMN protein and function

    SMN protein is a 38-kD protein localized in the cytoplasm

    and nucleus. It is ubiquitously expressed and intimately

    involved in a well-defined biochemical pathway that relates

    to global gene expression: snRNP biogenesis (Fischer et al.

    1997; Liu et al. 1997), as well as a less defined role in RNP

    trafficking in neurons (Zhang et al. 2003). Currently, it is

    unclear which SMN-associated function underlies SMA

    development. snRNPs are the building blocks for the

    general splicing machinery and are essential in all tissues.

    Initially, it was difficult to envision how a general cellular

    defect such as snRNP assembly could account for the motor

    neuron pathology associated with SMA, but recent reports

    have demonstrated that not all snRNP complexes are

    equally affected by reduced SMN levels (Lotti et al. 2012).

    The recent identification of specific gene targets that are

    dysregulated in SMN-deficient contexts including Drosoph-

    ilaand mice has supported the hypothesis that SMA devel-

    ops due to splicing abnormalities (Imlach et al. 2012; Lotti

    et al. 2012). However, there is still considerable debate over

    the specific function(s) that leads to SMA development.

    Several theories have been suggested to explain the specific

    role of SMN in the survival of motor neurons including:

    (i) SMN functions in splicing of the transcripts essential for

    integrity and differentiation of motor neurons (Winkler

    et al. 2005); (ii) there is a requirement for higher levels of

    SMN in motor neurons than other tissues (Battaglia et al.1997); (iii) SMN functions specifically in axonal transport

    (Pagliardini et al. 2000) and neuronal development such as

    growth cones and neurites (Bechade et al. 1999; Jablonka

    et al. 2000; Zhang et al. 2003). Certainly, the snRNP assem-

    bly activity is the best characterized function of SMN, but a

    conclusive link between SMA and snRNP dysreglation has

    not been established.

    SMN expression

    SMN expression is developmentally regulated (Battaglia

    et al. 1997) and there is a high level of SMN expression in

    most tissues during embryogenesis (Novelli et al. 1997), fol-

    lowed by a significant reduction after birth in all tissues

    except in the brain and spinal cord, which remains relatively

    high until 2 weeks post birth (La Bella et al. 1998). Based

    on this pattern of expression, it is likely that most tissues

    require SMN for normal development and that extremely

    low SMN levels at prenatal stage are detrimental for nearly

    all tissues. The necessity of SMN during embryogenesis for

    development of axons and motoneuron dendrites in zebra-

    fish (Hao et al. 2013), neuromuscular junction in Drosophila

    (Chan et al. 2003), cranial nerves as well as lumbar spinal

    nerves and innervations in mice (McGovern et al. 2008; Liu

    et al. 2010) is established.Interestingly, prenatal immunoblot studies on SMA Type I

    fetuses showed that SMN protein was greatly reduced in all

    tissues examined, i.e. skeletal muscle, heart, brain, kidney,

    thymus, pancreas and lung (Burlet et al. 1998). However,

    when SMN1 and SMN2 expression was analyzed separately

    in SMA fetuses and controls at 15 weeks gestation, the

    contribution of the SMN2 expression to the amount of full-

    length SMN protein was greater in unaffected tissues such

    as muscle and kidney compared with the spinal cord (Soler-

    Botija et al. 2005). These results suggest the importance of

    SMN2 as a potential contributor to the disease mechanism

    in SMA and its possible role in compensating pathology in

    tissues clinically unaffected by the disease.

    Morphological studies of motor neurons in SMA Type I

    fetuses gave evidence that programmed motoneuron death

    is prolonged in comparison with controls (Fidzianska &

    Rafalowska, 2002; Soler-Botija et al. 2002). Nuclear abnor-

    malities were found as early as 16 weeks gestation in

    affected fetuses (Fidzianska & Rafalowska, 2002). More

    recent neuro-pathological analyses of affected fetuses and

    patients suggest a fetal developmental maturation error

    and a postnatal retrograde dying-back degeneration of

    lower motor neurons in SMA (Ito et al. 2011).

    This review will summarize the results of studies in SMA

    patients and SMA mouse models to highlight the possible

    2013 Anatomical Society

    SMA: a motor neuron or multi-organ disease?, M. Shababi et al.2

  • 8/11/2019 anatomi penting abstrak

    3/14

    role of SMN in development and function of the nervous

    system and internal organs. We will also examine the stud-

    ies focusing on restoration of SMN and the impact of their

    results on understanding the disease pathology.

    Clinical reports of cardiac/autonomic defectsin SMA patients

    Type I patients

    Cardiac defects

    Less than 10% of SMA Type I patients retain only one copy

    of SMN2 gene and generally have a congenitally lethal

    course (also sometimes denoted as SMA Type 0), whereas

    80% of patients have two copies of SMN2. Most of the car-

    diac defects have been reported in SMA Type I with one

    SMN2 copy number, occasionally identified at prenatal

    stages (Rijhsinghani et al. 1997; Sarnat & Trevenen, 2007;

    Parra et al. 2012). However, the SMN2 copy numbers of

    patients showing cardiac defects was not always reported

    or determined due to lack of genetic information. Mean-

    while, numerous cases of congenital heart defects were

    reported in genetically confirmed SMA patients. These

    reports indicated a series of heart malformation features

    including atrial septal defects, atrioventricular septal

    defects, valvular aortic stenosis, hypoplastic aortic arch,

    severe coarctation of the aorta, partial atrioventricular

    canal, tricuspid atresia, univentricular heart, and several

    characteristics of hypoplastic left heart syndrome (Moller

    et al. 1990; Burglen et al. 1995; Mulleners et al. 1996;El-Matary et al. 2004; Cook et al. 2006; Bach, 2007; Vaidla

    et al. 2007; Menke et al. 2008; Rudnik-Schoneborn et al.

    2008). Some of these cases were associated with bradycar-

    dia (Bach, 2007), digital necrosis and vascular thrombosis

    (Rudnik-Schoneborn et al. 2010). Autonomic nervous sys-

    tem (ANS) dysfunction was considered to be the major

    cause of distal necrosis in SMA patients.

    These clinical reports clearly indicate a connection

    between cardiac defects and the most severe forms of SMA,

    making the previous assumption that this association is

    coincidental truly doubtful. Consistent with this, one study

    estimated the ratio of SMN2copy numbers and occurrence

    of cardiac defects in 65 SMA Type I patients diagnosed

    genetically within the first 6 months of age. Three of four

    (75%) patients with one copy of SMN2 demonstrated he-

    modynamically relevant atrial or ventricular septal defects.

    The authors suggested that the probability of simultaneous

    occurrence of both conditions is < 1 out of 50 million

    patients, suggesting a role for SMN in cardiogenesis in

    severe SMA cases (Rudnik-Schoneborn et al. 2008).

    Despite these findings there is only little evidence for car-

    diac rhythm abnormalities in SMA Type I. Electrocardio-

    graphic abnormalities were found in 12 of 13 patients with

    an average age of ~37 months (Coletta et al. 1989). Echo-

    cardiography did not reveal cardiac structural defects;

    therefore, the abnormal ECG was considered to be caused

    by muscle fasciculations. Similar reports of ECG abnormali-

    ties such as right ventricle overload were documented in

    37% of the examined patients who were mostly SMA Type I

    (Distefano et al. 1994). The authors speculated that thecause could be pulmonary hypertension due to respiration

    anomalies (Distefano et al. 1994). An additional study docu-

    mented that 80% of 47 SMA cases (mean age ~40 months,

    including all three types) exhibited isoelectric line tremors

    in electrocardiogram (ECG) tracings (Huang et al. 1996)

    which did not have consequences for heart rhythm and

    function.

    Autonomic nervous system defects

    Patients with SMA Type I normally do not live long enough

    to exhibit other organ dysfunction despite progressive mus-

    cle weakness, but new clinical features were observed in

    patients who survived many years under assisted ventila-

    tion. A large retrospective study of long-term ventilated

    SMA Type I patients demonstrated that 15 of 63 patients

    experienced severe, symptomatic bradycardia (Bach, 2007).

    A series of autonomic tests on Type I SMA patients revealed

    a sympathetic-vagal imbalance, fluctuation of blood pres-

    sure, and irregular skin responses to temperature changes

    (Hachiya et al. 2005). Additionally, vascular abnormalities

    such as distal necrosis have been reported in SMA Type I

    patients, occasionally with ASD and asymmetric left ventric-

    ular hypertrophy (Araujo Ade et al. 2009; Rudnik-Schone-

    born et al. 2010). In some cases, the distal necrosis occurs

    with normal cardiac function (Araujo Ade et al. 2009), dem-onstrating that the autonomic nervous system dysfunction

    may lead to impaired regulation of vascular tone. However,

    the structural defects of the heart tissue at prenatal stages

    cannot be explained by the ANS deficiency. To make a dis-

    tinction between the two, it would be remarkable to iden-

    tify the intrinsic heart defects independent of ANS and

    analyze the ANS defects in relation to the function of other

    organs including the heart. Further experiments should

    address the question as to whether SMN interacts with

    other genes involved in cardio- and neurogenesis. A com-

    mon pathway might be provided by functional defects of

    neural crest cells which migrate into the developing heart

    and the large vessels and also differentiate into autonomic

    and sensory nerves (Crane & Trainor, 2006).

    SMA type II-III patients

    Although the literature supports the conclusion that SMA

    Type I patients are more likely to develop cardiac disease,

    cardiac function and rhythm remain remarkably stable even

    in severely handicapped SMA Type II and III patients in end

    stages of the disease. Previous case reports of cardiac dys-

    function, mostly AV conduction and other rhythm distur-

    bances and dilative cardiomyopathy, in SMA Type III

    patients have to be interpreted with caution as none of

    2013 Anatomical Society

    SMA: a motor neuron or multi-organ disease?, M. Shababi et al. 3

  • 8/11/2019 anatomi penting abstrak

    4/14

    these patients had been genetically confirmed for SMN

    deficiency. It has to be taken into account that patients

    with SMA have a normal population risk to develop cardiac

    insufficiency at an advanced age for unrelated reasons, e.g.

    on the basis of a coronary heart disease, as this was seen ina patient with SMA Type III who was diagnosed with

    enlargement and diffuse hypokinesia of cardiac ventricles

    at age 65 and died at age 67 from respiratory arrest (Kuru

    et al. 2009). The situation is also different in patients with

    long-standing respiratory dysfunction who are prone to

    right heart overloading (Distefano et al. 1994). Nonetheless,

    if early-onset cardiomyopathy is seen in patients with mild

    SMA, it is most likely that this patient has a different

    genetic condition. Phenocopies of SMA Type III include met-

    abolic diseases (e.g. acid maltase deficiency) or muscular

    dystrophies. In patients with cardiac arrhythmias and AV

    conduction disturbances, EmeryDreifuss muscular dystro-

    phy or myotonic dystrophy type 2 have to be considered.

    Lamin A/C gene mutations were seen in 10% of patients

    with the diagnosis of autosomal dominant proximal SMA

    (Rudnik-Schoneborn et al. 2007).

    One study performed a standard ECG and a routine echo-

    cardiography for 37 genetically confirmed Type II/III SMA

    patients, aged 665 years, to investigate the possibility of

    cardiac involvement (Palladino et al. 2011). Elevated heart

    rate and left ventricular dilation were detected in only two

    patients, aged 63 and 65, whose heart problems were con-

    sidered to be caused by hypertension and/or coronary

    artery disease (Palladino et al. 2011). These authors con-

    cluded that heart dysfunction is not associated with SMATypes II-III, an observation that is in line with longer natural

    history studies and clinical practice.

    Cardiac defects in SMA mouse models

    In contrast to humans, rodents and all other vertebrates

    have a single copy of theSMNgene. MurineSmnencodes a

    C at the 6th position of exon 7 and is therefore analogous

    to humanSMN1. Two widely used severe SMA mouse mod-

    els (SMN2 and SMND7) lack murineSmnand contain two

    copies of the human SMN2 transgene. The SMN2 model

    lives on average 45 days (Monani et al. 2000) and the

    SMND7 model which contains the cDNA of the D7 transcript

    lives slightly longer, achieving 1317 days (Le et al. 2005).

    Therefore, both models likely represent very severe SMA

    disease contexts. Recently, three individual labs identified

    functional and structural heart defects in murine SMA,

    demonstrating a link between extremely low levels of SMN

    and cardiac defects (Bevan et al. 2010; Heier et al. 2010;

    Shababi et al. 2010a).

    Pathological/structural defects

    Cardiac structural defects include inter-ventricular septum

    (IVS) remodeling, under-developed left ventricular wall,

    and dilated ventricles occurring at embryonic stage in

    SMN2 model and shortly after birth in SMND7 model pre-

    ceding motor neuron degeneration (Shababi et al. 2010a).

    Analysis of the vasculature in the SMA heart also revealed a

    significant reduction in the width of arterial wall and a pro-nounced decrease in the density of capillary bed (Shababi

    et al. 2010a, 2012), which can certainly hinder the ability of

    the heart to pump blood into other tissues/organs. A

    reduced number of capillaries was also shown in the skele-

    tal muscle of SMA mice (Somers et al. 2012). Additional

    structural defects are marked disorganization and degener-

    ation of myofibers with swollen mitochondria in SMND7

    mice at postnatal day 14 (P14) (Bevan et al. 2010).

    Cardiac functional defects and autonomic nervous

    system

    Cardiac functional analysis indicated significantly reduced

    heart rate, stroke volume (SV), cardiac output, fractional

    shortening (FS), and LV mass in the SMND7 heart at P7 and

    14 (Bevan et al. 2010). Interestingly, systemic injection of

    self-complementary (sc) AAV9 expressing the full-length

    SMN cDNA which rescues the SMA phenotype (Foust et al.

    2010) could not fully rescue the functional defects except

    the heart rate. Therefore, the possibility of SMN functioning

    in cardiogenesis and/or ANS abnormalities that contribute

    to defective heart function was suggested (Bevan et al.

    2010). Consistent with this, whole-mount heart immuno-

    staining indicated a lack of prominent sympathetic nerves

    and fewer, thinner branches in the SMA heart comparedwith age-matched heterozygous littermates (Heier et al.

    2010). To distinguish the intrinsic cardiac defects from ANS

    defects in SMA mice, we examined cardiac function using

    cine-MRI in parallel with the structural/biochemical proper-

    ties of the rescued SMND7 heart following systemic delivery

    of scAAV9-SMN. Rescued mice demonstrated a significant

    repair of motor function, a high level of SMN protein in the

    heart, and a great improvement in cardiac structural/bio-

    chemical defects (Shababi et al. 2012). However, the heart

    function including the heart rate was only partially restored

    compared with the age-matched wildtype, further confirm-

    ing that ANS deficiency leads to inadequate heart function.

    Similar to the features seen in few SMA Type I patients, dis-

    tal necrosis has also been reported in intermediate SMA

    mice models and therapeutically rescued severe SMA mice

    (Avila et al. 2007; Gavrilina et al. 2008; Passini et al. 2010).

    Extension of life span has led to increased recognition of

    necrosis in the tail and ear tips of these mice. It is likely that

    the necrosis is a result of impaired vascularization due to

    autonomic nervous system dysfunction, which is consistent

    with vascular defects detected in the SMND7 heart (Shababi

    et al. 2012).

    The reduced contractility in the SMA heart is attributed

    to decreased sympathetic stimulation and bradycardia.

    However, restoring the heart rate in the rescued SMA mice

    2013 Anatomical Society

    SMA: a motor neuron or multi-organ disease?, M. Shababi et al.4

  • 8/11/2019 anatomi penting abstrak

    5/14

    or increasing the heart rate in SMA mice by using drugs

    cannot restore the contractile dysfunction, suggesting a

    very low contractile reserve in the SMA heart (Bevan et al.

    2010). Abnormalities in ATP-generating pathways (e.g.

    mitochondrial function, glycolysis, glycogenolysis and phos-photransferase reactions) can reduce ATP supply in failing

    hearts (Ingwall, 2009). Consistent with this, abnormal car-

    diac mitochondria and hypoglycemia in SMA mice and

    patients have been reported (Bruce et al. 1995; Butchbach

    et al. 2009; Bevan et al. 2010). Measurement of contractility

    in isolated cardiomyocytes of the SMA heart will clarify

    whether the contractile dysfunction is intrinsic to the heart

    or depends on ANS function. To detect ANS defects related

    to cardiac function, tail-cuff and telemetric measurements

    will be suitable to reveal differences in sympatho-vagal bal-

    ances between the rescued SMA and wildtype mice. The

    activity of autonomic nervous system regarding cardiac

    function can be measured more directly during telemetry

    using pharmacological drugs and examining significant

    alterations in the heart rate under the inhibition of

    parasympathetic and/or sympathetic nervous activity in the

    rescued SMA mice.

    Additional organ deficiencies in SMA patientsand mice

    In contrast to numerous reports of cardiac defects in SMA

    patients, the number of instances showing defects in other

    peripheral organs is limited. This may be due to the fact

    that severe SMA patients have various complications anddie prematurely before the damage to other organs is fully

    recognized. In the following section, we examine additional

    organs with reported deficiencies in SMA patients and ani-

    mals models. The peripheral organ defects of SMA mice

    models are summarized in Table 1.

    Brain and sensory neurons

    Earlier studies of SMN expression in rat, monkey and human

    revealed a widespread but uneven SMN expression within

    several areas of the brain. Intense expression was observed

    in specific neuronal cell populations, such as in layer V pyra-

    midal neurons of the neocortex, the pallidal neurons in the

    basal ganglia, and the neurons of the deep cerebellar nuclei

    as well as the motor neurons of the brainstem and spinal

    cord (Battaglia et al. 1997). High SMN expression in spinal

    preganglionic sympathetic neurons and the neocortical

    pyramidal neurons, which was considered to have no effect

    in SMA development at the time, may possibly be responsi-

    ble for peripheral organ function and normal cognitive abil-

    ity. Cognitive function is well preserved in chronic SMA,

    and the overall experience in clinical practice is that infants

    with SMA Type II have an early speech development in com-

    parison to healthy (normally moving) toddlers. However,

    formal assessments of cognitive functions are hampered by

    the fact that these are standardized only for children

    beyond 5 years of age. Therefore, only few data from natu-

    ral history studies are available for SMA Type I patients.

    Along with the observation of ANS dysfunction arising in

    ventilated SMA Type I patients, it would be important tostudy cognitive development and function in long-term sur-

    vivors. One larger cognitive and intelligence study of SMA

    Types I-III, aged 6.018 years, was undertaken in Germany

    and demonstrated no difference between SMA patients

    and healthy controls (von Gontard et al. 2002).

    Defective hippocampus development due to abnormali-

    ties in cellular proliferation and neurogenesis is reported in

    severe SMN2 mice model (Wishart et al. 2010). Additionally,

    several cases of defective sensory neurons and thalamic

    lesions have been detected in genetically confirmed SMA

    Type I patients (Rudnik-Schoneborn et al. 2003; Ito et al.

    2004), although reports of sensory neuron disorder are

    more common in SMA mice (Jablonka et al. 2006; Ling

    et al. 2010; Mentis et al. 2011; Gogliotti et al. 2012; Marti-

    nez et al. 2012). The role of SMN in development and main-

    tenance of sensory neurons is supported by high SMN levels

    in dorsal root ganglia and posterior horn of the spinal cord

    in human fetus, albeit with less intensity than anterior horn

    (Tizzano et al. 1998). There is a considerable debate

    whether the loss of synapse in sensory neurons results in

    motor neuron cell death or is a consequence of SMN loss in

    motor neurons. Studies in zebrafish andDrosophilasuggest

    that abnormalities in sensory-motor circuit create the motor

    system defects and repair of motor neural network activity

    is essential to ameliorate the disease phenotype (Imlachet al. 2012; Lotti et al. 2012). On the other hand, restora-

    tion of SMN in motor neurons of SMA mice repairs the NMJ

    defects and also restores synapses in the sensory neurons

    (Gogliotti et al. 2012; Martinez et al. 2012), suggesting that

    the sensory motor circuit function is dependent on SMN lev-

    els in motor neurons.

    Muscle and neuromuscular junction (NMJ)

    A direct effect of the SMN loss on NMJ dysfunction and

    SMA progression is fully established and reviewed in great

    detail (Murray et al. 2010b; Bottai & Adami, 2013; Goulet

    et al. 2013); therefore, we briefly mention the reported

    defects in NMJ and muscle.

    In human muscle, SMN protein is localized at NMJ (Fan &

    Simard, 2002) and its role in NMJ development was verified

    by the failure of the cultured muscle cells derived from SMA

    patients to cluster acetylcholine receptors (AChRs) at the

    junction (Arnold et al. 2004). Delayed maturation of myotu-

    bes in SMA fetuses has been identified (Martinez-Hernandez

    et al. 2009). In addition, neurofilament accumulation along

    with poor terminal arborization in postnatal diaphragm

    samples of SMA Type I have been reported (Kariya et al.

    2008). Further evidence for the role of NMJ in human

    SMA pathology comes from a recent study that provided a

    2013 Anatomical Society

    SMA: a motor neuron or multi-organ disease?, M. Shababi et al. 5

  • 8/11/2019 anatomi penting abstrak

    6/14

  • 8/11/2019 anatomi penting abstrak

    7/14

    detailed structural characterization of NMJ defects in SMA

    fetuses (Martinez-Hernandez et al. 2013). The main prena-

    tal defects were abnormal modification of acetylcholine

    receptor clustering, irregular accumulation and positioning

    of synaptic vesicles, and atypical nerve terminals in motorendplates of SMA Type I samples, whereas SMA Type II

    fetuses were similar to controls.

    In SMA mice, disruption of skeletal muscle molecular

    composition with increased activity of cell death pathways

    (Mutsaers et al. 2011), abnormal differentiation in muscle

    satellite cells, deficient formation of myotubes, and

    decreased muscle fiber size are reported (Le et al. 2005; Lee

    et al. 2011; Hayhurst et al. 2012). Interestingly, loss of mur-

    ine Smn specifically in the skeletal muscle causes muscle

    necrosis, paralysis, and death (Cifuentes-Diaz et al. 2001).

    Yet, restoration of SMN in the muscle or increasing the mus-

    cle mass by using compounds has limited therapeutic bene-

    fits in treated mice (Gavrilina et al. 2008; Rose et al. 2009;

    Bosch-Marce et al. 2011), further confirming that NMJ mat-

    uration defects and abnormal synapses are the hallmark of

    SMA pathology (Kariya et al. 2008).

    Interestingly, there are no detectable pre-symptomatic

    neurodevelopmental defects in SMA mice models (Kariya

    et al. 2008; McGovern et al. 2008; Murray et al. 2010a). Yet,

    rapid degeneration of motor neurons at disease onset cause

    several post-natal NMJ defects in SMA mice which clearly

    resemble human defects. NMJ pathology in SMA mice

    includes neurofilament buildup and poor axonal sprouting,

    denervation, abnormal calcium homeostasis leading to

    defects in motor neuron excitability, reduced terminalarborization, disruption in synaptic vesicle release, aberrant

    expression of synaptic proteins, delayed post-synaptic matu-

    ration, loss of Schwann cells leading to defects in compensa-

    tory endplate remodeling and nerve-directed maturation of

    acetylcholine receptor (AChR) clusters (Cifuentes-Diaz et al.

    2002; Jablonka et al. 2007; Kariya et al. 2008; Murray

    et al. 2008, 2012; Kong et al. 2009; Ling et al. 2010; Ruiz

    et al. 2010; Torres-Benito et al. 2011).

    A recent report demonstrates that not all the muscles in

    SMA mice are uniformly affected (Ling et al. 2012). The sen-

    sitive muscles in SMND7 mice which have normal NMJ for-

    mation at P1, become highly denervated by P4 as the

    disease progresses (Ling et al. 2012). Severely denervated

    muscles in SMA mice are among axial and appendicular

    muscles which are clinically related to SMA patients. This

    study shows that the NMJ denervation in these muscles is

    likely due to the loss in preservation of the synapse which is

    consistent with other studies (Kariya et al. 2008; Murray

    et al. 2012).

    Liver and metabolic disorders

    Reye-like syndrome of fulminant liver failure is reported in

    only one case of SMA type II patient following a spinal sur-

    gery, which was suggested to be caused by a combination

    of different factors such as pre-operative fasting, prolonged

    anesthesia, post-operative stress, and reduced fatty acid oxi-

    dation (Zolkipli et al. 2012). Abnormal fatty acid metabo-

    lism is the most common metabolic defect reported in

    severe patients and some younger SMA Type II patients,which may return to normal with aging (Tein et al. 1995;

    Crawford et al. 1999). Mild to moderate dicarboxylic acidu-

    ria is consistently found in SMA Types I and II patients which

    is very similar to mitochondrial b oxidation abnormalities

    (Tein et al. 1995; Crawford et al. 1999). In addition, one

    study reported an increased level of esterified carnitine, a

    factor that performs a role in fatty acid transport from the

    cytosol to the mitochondria in plasma of severe SMA

    patients, concomitant with abnormalities in mitochondrial

    multifunctional enzyme complex (Tein et al. 1995). How-

    ever, it is not certain whether data of patients with other

    entities, e.g. mitochondrial disease, were included in this

    study, as it was published before SMN gene testing was

    available. The level of ketone bodies (secondary products of

    mitochondrial fatty acid b-oxidation in liver) in SMA

    patients with metabolic disorder was normal or not signifi-

    cantly reduced (Crawford et al. 1999), suggesting a normal

    fatty acid utilization by the liver and, therefore, a muscle

    specific defect in fatty acid metabolism. To investigate

    whether fatty acid metabolism defect is a consequence of

    denervation and muscle atrophy, fatty acid levels in the

    plasma of 33 infants with severe SMA were measured and

    compared with that of normal infants and six diseased con-

    trol infants affected with equally severe denervating non-

    SMA (Crawford et al. 1999). They found a significantlyincreased ratio of dodecanoic acid (C12) to tetradecanoic

    acid (C14) in the plasma of all SMA patients compared with

    control patients. The authors suggested that the fatty acid

    metabolism disorder in SMA is directly related to the loss of

    SMN1 or possibly other genes in the chromosome 5q and is

    not the consequence of muscle denervation and/or atrophy

    (Crawford et al. 1999). Further evidence of fatty acid oxida-

    tion disorder is based on the autopsy samples of some

    infants with severe SMA who showed fatty vacuolization of

    the liver (Crawford et al. 1999).

    The role of SMN in the development and function of liver

    in mice was demonstrated by a study in which a mutation

    in the exon 7 of murine Smn directed to liver led to liver

    failure and late embryonic lethality of transgenic mice (Vitte

    et al. 2004). Importantly, however, this animal model may

    produce SMN levels that are comparable to a null-state

    which is not SMA, but would be predicted to be lethal for

    any tissue. The importance of liver in SMA pathology was

    indicated by the experiments in which restoration of SMN

    and subsequent increase in insulin-like growth factor (IGF)1

    levels in the liver of SMA mice, through subcutaneous injec-

    tion of therapeutic oligonucleotide (ASO), was an impor-

    tant factor for high degree of rescue (Hua et al. 2011).

    Premature death of SMA severe patients in infancy is a

    significant hurdle for continuous analysis of the urine and

    2013 Anatomical Society

    SMA: a motor neuron or multi-organ disease?, M. Shababi et al. 7

  • 8/11/2019 anatomi penting abstrak

    8/14

    plasma to investigate the exact role that SMN may play in

    metabolic defects. While controversial in the community, a

    specific diet has not been scientifically evaluated. On the

    other hand, extreme muscle wasting in severe SMA patients

    could be explained by defects in fatty acid transport/oxida-tion, which plays a major role in energy production during

    prolonged fasting, resulting in a detrimental effect on

    working cardiac and skeletal muscle. A thorough investiga-

    tion of lipid content in the liver and possibly muscle of SMA

    mice models, before and after the disease onset, can pro-

    vide valuable information regarding the direct or indirect

    effect of SMN deficiency on metabolic abnormalities.

    Pancreas

    While single case reports do not provide sufficient evidence

    for an association of diabetes mellitus and SMA, consider-

    ing the frequency of both conditions, there are occasional

    cases reporting diabetes and abnormalities in the glucose

    metabolism in a few genetically confirmed SMA Type II and

    III patients (Bowerman et al. 2012c; Lamarca et al. 2012). In

    addition, three cases of acute pancreatitis among long-term

    survivors of ventilated SMA Type I patients (Bach, 2007) and

    pathological pancreatic defects in autopsied pancreas speci-

    mens from infants and an intermediate SMA mouse model

    (SMA2B/) have been documented (Bowerman et al. 2012c).

    The metabolic defects in SMA2B/ mice were characterized

    by fasting hyperglycemia, glucose intolerance, hypersensi-

    tivity to insulin, and hyperglucagonemia. Pathological

    defects were identified by loss of insulin-producing b cellsand a corresponding increase in the number of the gluca-

    gon-producing a cells in pancreatic islets of mice and

    human specimens. Based on the observation that the pan-

    creatic pathology and fasting hyperglycemia occurred

    before the onset of SMA symptoms, the authors suggested

    that the pancreatic phenotype is independent of the neuro-

    nal SMA phenotype and is rather a direct consequence of

    SMN deficiency within the pancreas (Bowerman et al.

    2012c).

    Careful observation of SMA patients regarding their glu-

    cose homeostasis is essential to shed light on the role of

    SMN in glucose metabolism and pancreas function. In the

    study of Bach (2007), one patient who had acute pancreati-

    tis was taking valproic acid in a research protocol. Valproic

    acid is well known to be associated with an increased pan-

    creatitis risk (Asconape et al. 1993). Therefore, considering

    a possible disposition to pancreatic dysfunction in severe

    SMA, clinicians have to be alerted about corresponding side

    effects of medications used in clinical trials or clinical care.

    Intestine

    Thus far, the existence of intestinal issues in SMA patients is

    considered to be the result of muscle weakness. Intestinal

    problems have been recorded in SMA mice (Le et al. 2011;

    Schreml et al. 2012) and two SMA patients (Type II and Type

    IV) represented by severe constipation (Ionasescu et al.

    1994; Khawaja et al. 2004). In addition, a micro-dissection

    study revealed significantly low values for fractional area of

    neural tissue in small intestine and colon of severe SMApatients (Galvis et al. 1992), suggesting that the inadequacy

    of the autonomic nervous system contributes to functional

    impairments.

    Intestinal problems in mice were recorded as impacted

    bowel and pockets of fluid and gas (pneumoperitoneum) in

    SMN-induced transgenic mice following extended survival

    (Le et al. 2011). Pathological defects in intestine of severe

    Taiwanese SMA mice are characterized by string-like intes-

    tine, reduced numbers of villi, intracytoplasmic vacuoles

    predominantly at the tips of the villi, severe intramural

    edema in the lamina propria, severe diarrhea, and opaque

    fluid in the abdomen of most late-stage SMA animals

    (Schreml et al. 2012). Similar to other organs, the pathologi-

    cal impact of SMN deficiency on the intestine may not be

    fully recognized due to the premature death of severe SMA

    patients and mice.

    Lung

    Pulmonary complications are the most reported condition

    and by far the most devastating aspect of the disease

    leading to the death of severe SMA patients at infancy.

    In contrast to infantile SMA with respiratory distress

    Type 1 (SMARD1), where diaphragmatic palsy is a hallmark

    (Rudnik-Schoneborn et al. 2004), SMA with SMN deficiencyresults in intercostal muscle weakness and thoracic cage

    deformation, thereby reducing pulmonary volume and ven-

    tilatory capacity. SMA Type I patients frequently show para-

    doxical breathing caused by preserved diaphragmatic

    movements. Complications include airway obstruction, air-

    way inflammation, increased mucus production, aspiration

    causing pneumonia, lung damage, diminished ability to

    clear secretions, weakened pulmonary defenses, restrictive

    lung disease, and respiratory arrest. Non-invasive mechani-

    cal ventilation and tracheostomy can both extend survival

    of SMA Type I patients, but tracheostomy results in perma-

    nent ventilator reliance and prevents speech development

    (Bach et al. 2007).

    Since respiratory complications mostly arise from hypo-

    ventilation and subsequent respiratory infections, there is

    little data focusing upon the structural lung damage in

    SMA autopsies. Several cases of aletectasis (one was associ-

    ated with bradycardia) are reported (Collado-Ortiz et al.

    2007; Modi et al. 2010; Henrichsen et al. 2012). Most

    recently, lung structural defects were indicated in the severe

    Taiwanese mice (Schreml et al. 2012). These defects were

    described as discolorations of the lungs compatible with

    atelectasis or pulmonary infarctions, ruptured alveolar sep-

    tum, and emphysema. Whether the pathological defects in

    the lungs of SMA mice are relevant to SMA patients is an

    2013 Anatomical Society

    SMA: a motor neuron or multi-organ disease?, M. Shababi et al.8

  • 8/11/2019 anatomi penting abstrak

    9/14

    open question. Extension of survival through mechanical

    means or any other therapeutic strategies may reveal more

    potential pathological defects in the lungs of SMA patients

    than presently known.

    Restoration of SMN only in motor neuronsdoes not fully rescue severe SMA mice

    Many researchers have focused on different methods to

    restore SMN protein in motor neurons of SMA mice. The

    outcome of these experiments demonstrate that increasing

    SMN levels exclusively in motor neurons has a surprisingly

    small impact upon survival and weight gain of severe SMA

    mice. As therapeutics move forward towards the clinic, it

    is essential to fully understand the pathology of SMA

    and determine the temporal requirements for SMN restora-

    tion within these tissues. Different approaches designed

    to increase the SMN levels in motor neurons and their

    outcomes are summarized.

    Transgenic mouse models

    Transgenic models have been created to examine the con-

    tribution of specific cell types, most notably, skeletal muscle

    and neurons (Gavrilina et al. 2008). When full-length SMN

    is expressed under the control of the HSA promoter (which

    should be highly expressed in skeletal muscle), the severe

    SMA phenotype was only marginally improved. In contrast,

    SMNexpression driven by the PrP promoter (which should

    be highly expressed in several populations within the cen-tral nervous system, including neuronal and glial popula-

    tions) significantly extended survival (Gavrilina et al. 2008).

    Interestingly, although average life span was extended

    from ~5 days to 150210 days, some mice died prematurely

    for unknown reasons (Gavrilina et al. 2008). This could be

    due to the nature of transgenic animals or may suggest that

    additional tissues are required for a complete rescue. In a

    recent study, doxycycline-dependent induction of SMN lev-

    els in a transgenic SMA model at P1 rescued the SMA phe-

    notype (Le et al. 2011). However, 70% of the animals died

    within a month once SMN induction was stopped, even

    though the physiology and morphology of the neuromus-

    cular junctions were largely normal at the time of death (Le

    et al. 2011). Lutz et al. (2011) have used an inducible SMN

    rescue allele that was induced at different time points to

    investigate the temporal requirement for SMN in order to

    achieve rescue. They observed a significant extension of life

    span in approximately 50% of the rescued mice when the

    SMN allele was induced at symptomatic stage of PND46.

    They concluded that the beneficial effect of SMN induction

    in their rescued mice was due to SMN restoration to all tis-

    sues, and not exclusively to motor neurons. Their justifica-

    tion for the lack of rescue in 50% of their SMA mice relies

    on the possibility of the heart abnormalities. Another

    report described using an inducible allele (Hb9-Cre) that

    restored SMN solely in motor neurons and repaired the

    motor neuron phenotype but resulted in a very modest

    increase in survival (Gogliotti et al. 2012). The authors spec-

    ulated that the minimal extension of life span may be due

    to lack of autonomic innervations of the heart. In a comple-mentary study, SMN was depleted specifically in motor neu-

    rons and resulted in only a mild cellular phenotype rather

    than the predicted severe SMA phenotype (Park et al.

    2010). These studies suggest that the composite SMA phe-

    notype and disease progression is the result of SMN short-

    age in additional tissues and the full phenotypic rescue

    requires SMN restoration in all tissues.

    Therapeutics and delivery methods

    The delivery method of the therapeutics to restore the SMN

    in the CNS as well as the peripheral organs may have a

    great impact on the rate of the rescue. One of the most

    effective RNA therapeutics is a short, antisense oligonucleo-

    tide (ASO) that blocks the inhibitory activity of ISS-N1

    located within intron 7 and increases the inclusion of exon 7

    (Singh et al. 2009, 2010; Hua et al. 2010, 2011; Passini et al.

    2011; Osman et al. 2012; Porensky et al. 2012). Extension in

    life span and preservation of neuromuscular junctions of

    SMA mice has been observed with 2-O-2-methoxyethyl-

    modified ASO, referred to as ASO-10-27, when delivered

    via a single intracerebroventricular (ICV) injection into

    SMND7 mice (Passini et al. 2011). Surprisingly, a recent

    report described the positive effect of the ASO-10-27 on

    the survival of severe Taiwanese mice by subcutaneous(SC) injection (Hua et al. 2011). Their results indicated that

    SC injection of ASO extended the survival much more

    effectively than ICV injection, even though the ASO dose

    used in ICV injection was lower than that in SC injection.

    Interestingly, combination of both injection methods

    increased the life span even longer than each individual

    injection. They concluded that SMN restoration in periph-

    eral tissues in combination with partial restoration in the

    CNS can achieve efficient rescue of severe SMA mice. On

    the other hand, a recent study reported a life span of

    more than 100 days for SMND7 mice following a single

    ICV injection of morpholino (MO) oligomer against ISS-N1

    (Porensky et al. 2012). This study concluded that there was

    no difference in the survival of the ICV-injected animals vs.

    those treated with combined ICV and systemic injection.

    According to their results, SMN increase was minimal in

    the peripheral organs of ICV-injected mice, suggesting that

    the rescue was due to early salvage of the motor pheno-

    type. Additional therapeutic intervention is gene replace-

    ment therapy using scAAV9 expressing full-length SMN

    cDNA that has provided the most substantial improvement

    in severe SMA mice models (Foust et al., 2010; Valori et al.

    2010; Dominguez et al. 2011; Glascock et al. 2012a,b;

    Shababi et al. 2012; Benkhelifa-Ziyyat et al. 2013). How-

    ever, sudden deaths, cardiac/respiratory complications, and

    2013 Anatomical Society

    SMA: a motor neuron or multi-organ disease?, M. Shababi et al. 9

  • 8/11/2019 anatomi penting abstrak

    10/14

    average life spans of 6570 days are common following

    intravenous delivery of scAAV9-SMN. We have recently

    reported a significantly increased level of strength and

    weight gain in scAAV9-SMN injected mice through ICV

    delivery than IV delivery using a low dose of scAAV9-SMN(Glascock et al. 2012b). Additionally, our survival analysis

    demonstrated that ICV-treated mice displayed fewer early

    deaths than IV-treated animals. ICV injection at P2 can also

    result in dissemination of the virus into the peripheral

    organs (due to immature state of the blood brain barrier)

    and, therefore, the possibility remains that direct delivery

    of SMN into the CNS along with partial restoration in the

    peripheral organs leads to an efficient rescue. An alterna-

    tive possibility is that ICV delivery of the therapeutic virus

    is more suitable than IV delivery for efficient transduction

    of the ANS and subsequently leads to more improved

    function of the peripheral organs.

    Chemical compounds

    A variety of chemical compounds have been examined

    within the context of SMA models as well as SMA patients.

    Recent reviews have focused upon these compounds and

    their modes-of-action (Lorson et al., 2010; Shababi et al.

    2010b; Cherry & Androphy, 2012; Lewelt et al. 2012; Lorson

    & Lorson, 2012). Several therapeutic compounds used in

    SMA treatment such as Rock inhibitors and follistatin had a

    positive effect on NMJ maturation (Rock inhibitors) and

    increased the numbers of motor neurons in the lumbar

    spinal cord (Follistatin) (Rose et al. 2009; Bowerman et al.2010, 2012b). However, their effect is SMN-independent,

    resulting in a very modest increase in the survival of the

    SMA mice.

    Additional compounds such as HDACi and prolactin (PLR)

    are capable of SMN induction, but their effect on survival

    is either insignificant or modest, respectively. The HDACi-

    treated severe Taiwanese mice demonstrated normal motor

    endplate and NMJ morphology but significant structural

    damages to the heart, intestine, and lung at P5 (Schreml

    et al. 2012). PLR-treated SMND7 mice had a modest exten-

    sion in the survival (~70%), even though SMN protein

    induction in the CNS of these mice was higher than that in

    wildtype or heterozygous mice (Farooq et al. 2011). One of

    the factors considered to contribute to the moderate rescue

    was the lack of SMN induction in the peripheral organs,

    including cardiac tissue.

    Conclusion

    Although it is clear that the primary pathology in SMA is

    neurodegeneration, there is increasing evidence from clini-

    cal reports and animal studies that other tissues are

    involved in the overall phenotype, especially in the most

    severe forms of the disease. Additional complications in

    patients include autonomic nervous system involvement,

    congenital heart defects, liver, pancreas and intestinal dys-

    function, and metabolic deficiencies. In SMA mouse models,

    further features are observed, such as cardiac structural and

    functional defects along with rhythm disturbances, defec-

    tive development of specific brain areas, and moreextended necrosis of tail and ears. However, mice are not

    men, and there remain significant differences in the pheno-

    typic features of patients and mouse models which cannot

    be explained so far. Taking the impact of respiratory, limbic,

    cardiac, and autonomic nervous system dysfunction in

    severe SMA patients into account, it will be beneficial to

    reveal the potential pathological defects in each organ

    before and after the disease onset to distinguish the specific

    defects occurring as a direct result of SMN shortage from

    those which are the byproduct of disease progression. Since

    the total loss of SMN results in embryonic lethality, it is not

    surprising that the extremely low levels of SMN in severely

    affected patients lead to detrimental damages in every tis-

    sue. Therefore, to develop the most efficient therapeutic

    approach and also prevent further complications that may

    arise with extended survival following therapeutic interven-

    tions, it is necessary to investigate the specific damages to

    every system independently in detail. The comparison of

    the defects in SMA mice models will provide valuable

    insights if they are accompanied by similar studies in autop-

    sied specimens of SMA patients. One of the most important

    issues in regard to treating SMA patients by any therapeutic

    means is to maintain a broad and extensive outlook to

    ensure the ability of clinicians to predict and contain atypi-

    cal complications which may arise due to the strain onperipheral organs as a result of increased survival.

    References

    Araujo Ade Q, Araujo M, Swoboda KJ (2009) Vascular perfusion

    abnormalities in infants with spinal muscular atrophy. J Pedi-

    atr155, 292294.

    Arnold AS, Gueye M, Guettier-Sigrist S, et al. (2004) Reduced

    expression of nicotinic AChRs in myotubes from spinal muscu-

    lar atrophy I patients. Lab Invest84, 12711278.

    Asconape JJ, Penry JK, Dreifuss FE, et al. (1993) Valproate-asso-

    ciated pancreatitis. Epilepsia 34, 177183.

    Avila AM, Burnett BG, Taye AA, et al. (2007) Trichostatin A

    increases SMN expression and survival in a mouse model of

    spinal muscular atrophy. J Clin Invest117 , 659671.

    Bach JR (2007) Medical considerations of long-term survival

    of Werdnig-Hoffmann disease. Am J Phys Med Rehabil 86,

    349355.

    Bach JR, Saltstein K, Sinquee D, et al. (2007) Long-term survival

    in Werdnig-Hoffmann disease. Am J Phys Med Rehabil 86,

    339345 quiz 346-8, 379.

    Battaglia G, Princivalle A, Forti F, et al. (1997) Expression of the

    SMN gene, the spinal muscular atrophy determining gene, in

    the mammalian central nervous system. Hum Mol Genet 6,

    19611971.

    Bechade C, Rostaing P, Cisterni C, et al. (1999) Subcellular distri-

    bution of survival motor neuron (SMN) protein: possible

    2013 Anatomical Society

    SMA: a motor neuron or multi-organ disease?, M. Shababi et al.10

  • 8/11/2019 anatomi penting abstrak

    11/14

  • 8/11/2019 anatomi penting abstrak

    12/14

  • 8/11/2019 anatomi penting abstrak

    13/14

    Lorson CL, Strasswimmer J, Yao JM, et al. (1998) SMN oligomer-

    ization defect correlates with spinal muscular atrophy severity.

    Nat Genet19, 6366.

    Lorson CL, Hahnen E, Androphy EJ, et al. (1999) A single nucleo-

    tide in the SMN gene regulates splicing and is responsible for

    spinal muscular atrophy.Proc Natl Acad Sci U S A96, 63076311.Lorson CL, Rindt H, Shababi M (2010) Spinal muscular atrophy:

    mechanisms and therapeutic strategies. Hum Mol Genet 19,

    R111R118.

    Lotti F, Imlach WL, Saieva L, et al. (2012) An SMN-dependent

    U12 splicing event essential for motor circuit function. Cell

    151, 440454.

    Lutz CM, Kariya S, Patruni S, et al. (2011) Postsymptomatic res-

    toration of SMN rescues the disease phenotype in a mouse

    model of severe spinal muscular atrophy. J Clin Invest 121,

    30293041.

    Mailman MD, Heinz JW, Papp AC, et al. (2002) Molecular analy-

    sis of spinal muscular atrophy and modification of the pheno-

    type by SMN2. Genet Med4, 2026.

    Martinez TL, Kong L, Wang X, et al. (2012) Survival motor neu-ron protein in motor neurons determines synaptic integrity in

    spinal muscular atrophy. J Neurosci32, 87038715.

    Martinez-Hernandez R, Soler-Botija C, Also E, et al. (2009) The

    developmental pattern of myotubes in spinal muscular atro-

    phy indicates prenatal delay of muscle maturation. J Neuropa-

    thol Exp Neurol68, 474481.

    Martinez-Hernandez R, Bernal S, Also-Rallo E, et al. (2013) Syn-

    aptic defects in type I spinal muscular atrophy in human

    development. Journal of Pathology229, 4961.

    McGovern VL, Gavrilina TO, Beattie CE, et al. (2008) Embryonic

    motor axon development in the severe SMA mouse. Hum Mol

    Genet17, 29002909.

    Menke LA, Poll-The BT, Clur SA, et al. (2008) Congenital heart

    defects in spinal muscular atrophy type I: a clinical report oftwo siblings and a review of the literature. Am J Med Genet

    A 146A, 740744.

    Mentis GZ, Blivis D, Liu W, et al. (2011) Early functional impair-

    ment of sensory-motor connectivity in a mouse model of

    spinal muscular atrophy. Neuron 69, 453467.

    Modi HN, Suh SW, Hong JY, et al. (2010) Treatment and compli-

    cations in flaccid neuromuscular scoliosis (Duchenne muscular

    dystrophy and spinal muscular atrophy) with posterior-only

    pedicle screw instrumentation. Eur Spine J19, 384393.

    Moller P, Moe N, Saugstad OD, et al. (1990) Spinal muscular

    atrophy type I combined with atrial septal defect in three sibs.

    Clin Genet38, 8183.

    Monani UR, Lorson CL, Parsons DW, et al. (1999) A single nucle-

    otide difference that alters splicing patterns distinguishes the

    SMA gene SMN1 from the copy gene SMN2. Hum Mol Genet

    8, 11771183.

    Monani UR, Sendtner M, Coovert DD, et al. (2000) The human

    centromeric survival motor neuron gene (SMN2) rescues

    embryonic lethality inSmn/ mice and results in a mouse with

    spinal muscular atrophy. Hum Mol Genet9, 333339.

    Mulleners WM, van Ravenswaay CM, Gabreels FJ, et al. (1996)

    Spinal muscular atrophy combined with congenital heart dis-

    ease: a report of two cases. Neuropediatrics 27, 333334.

    Munsat TL, Davies KE (1992) International SMA consortium

    meeting, 2628 June 1992, Bonn, Germany. Neuromuscul Dis-

    ord2, 423428.

    Murray LM, Comley LH, Thomson D, et al. (2008) Selective

    vulnerability of motor neurons and dissociation of pre- and

    post-synaptic pathology at the neuromuscular junction in

    mouse models of spinal muscular atrophy. Hum Mol Genet17,

    949962.

    Murray LM, Lee S, Baumer D, et al. (2010a) Pre-symptomatic

    development of lower motor neuron connectivity in a mouse

    model of severe spinal muscular atrophy. Hum Mol Genet 19,420433.

    Murray LM, Talbot K, Gillingwater TH (2010b) Review: neuro-

    muscular synaptic vulnerability in motor neurone disease:

    amyotrophic lateral sclerosis and spinal muscular atrophy.

    Neuropathol Appl Neurobiol36, 133156.

    Murray LM, Beauvais A, Bhanot K, et al. (2012) Defects in neu-

    romuscular junction remodelling in the Smn(2B/) mouse

    model of spinal muscular atrophy. Neurobiol Dis 49C , 5767.

    Mutsaers CA, Wishart TM, Lamont DJ, et al. (2011) Reversible

    molecular pathology of skeletal muscle in spinal muscular

    atrophy.Hum Mol Genet20, 43344344.

    Novelli G, Calza L, Amicucci P, et al. (1997) Expression study of

    survival motor neuron gene in human fetal tissues. Biochem

    Mol Med61, 102106.Oskoui M, Levy G, Garland CJ, et al. (2007) The changing natu-

    ral history of spinal muscular atrophy type 1. Neurology 69,

    19311936.

    Osman EY, Yen PF, Lorson CL (2012) Bifunctional RNAs target-

    ing the intronic splicing silencer N1 increase SMN levels and

    reduce disease severity in an animal model of spinal muscular

    atrophy.Mol Ther20, 119126.

    Pagliardini S, Giavazzi A, Setola V, et al. (2000) Subcellular local-

    ization and axonal transport of the survival motor neuron

    (SMN) protein in the developing rat spinal cord. Hum Mol

    Genet9, 4756.

    Palladino A, Passamano L, Taglia A, et al. (2011) Cardiac involve-

    ment in patients with spinal muscular atrophies. Acta Myol

    30, 175

    178.Park GH, Maeno-Hikichi Y, Awano T, et al. (2010) Reduced sur-

    vival of motor neuron (SMN) protein in motor neuronal pro-

    genitors functions cell autonomously to cause spinal muscular

    atrophy in model mice expressing the human centromeric

    (SMN2) gene.J Neurosci30, 1200512019.

    Parra J, Alias L, Also-Rallo E, et al. (2012) Evaluation of fetal

    nuchal translucency in 98 pregnancies at risk for severe spinal

    muscular atrophy: possible relevance of the SMN2 copy num-

    ber.J Matern Fetal Neonatal Med25, 12461249.

    Passini MA, Bu J, Roskelley EM, et al. (2010) CNS-targeted

    gene therapy improves survival and motor function in a

    mouse model of spinal muscular atrophy. J Clin Invest 120,

    12531264.

    Passini MA, Bu J, Richards AM, et al. (2011) Antisense oligonu-

    cleotides delivered to the mouse CNS ameliorate symptoms of

    severe spinal muscular atrophy. Sci Transl Med3, 72ra18.

    Porensky PN, Mitrpant C, McGovern VL, et al. (2012) A single

    administration of morpholino antisense oligomer rescues spinal

    muscular atrophy in mouse. Hum Mol Genet21, 16251638.

    Riessland M, Ackermann B, Forster A, et al.(2010) SAHA amelio-

    rates the SMA phenotype in two mouse models for spinal

    muscular atrophy. Hum Mol Genet19, 14921506.

    Rijhsinghani A, Yankowitz J, Howser D, et al. (1997) Sonograph-

    ic and maternal serum screening abnormalities in fetuses

    affected by spinal muscular atrophy. Prenat Diagn 17,

    166169.

    Rochette CF, Gilbert N, Simard LR (2001) SMN gene duplication

    and the emergence of the SMN2 gene occurred in distinct

    2013 Anatomical Society

    SMA: a motor neuron or multi-organ disease?, M. Shababi et al. 13

  • 8/11/2019 anatomi penting abstrak

    14/14

    hominids: SMN2 is unique to Homo sapiens. Hum Genet 108,

    255266.

    Rose FF Jr, Mattis VB, Rindt H, et al. (2009) Delivery of recombi-

    nant follistatin lessens disease severity in a mouse model of

    spinal muscular atrophy. Hum Mol Genet18, 9971005.

    Rudnik-Schoneborn S, Goebel HH, Schlote W, et al. (2003) Clas-sical infantile spinal muscular atrophy with SMN deficiency

    causes sensory neuronopathy. Neurology60, 983987.

    Rudnik-Schoneborn S, Stolz P, Varon R, et al. (2004) Long-term

    observations of patients with infantile spinal muscular atrophy

    with respiratory distress type 1 (SMARD1). Neuropediatrics 3 5,

    174182.

    Rudnik-Schoneborn S, Botzenhart E, Eggermann T, et al.

    (2007) Mutations of the LMNA gene can mimic autosomal

    dominant proximal spinal muscular atrophy. Neurogenetics

    8, 137142.

    Rudnik-Schoneborn S, Heller R, Berg C, et al. (2008) Congenital

    heart disease is a feature of severe infantile spinal muscular

    atrophy.J Med Genet45, 635638.

    Rudnik-Schoneborn S, Berg C, Zerres K, et al. (2009) Genotype-phenotype studies in infantile spinal muscular atrophy (SMA)

    type I in Germany: implications for clinical trials and genetic

    counselling.Clin Genet76, 168178.

    Rudnik-Schoneborn S, Vogelgesang S, Armbrust S, et al. (2010)

    Digital necroses and vascular thrombosis in severe spinal mus-

    cular atrophy. Muscle Nerve 42, 144147.

    Ruiz R, Casanas JJ, Torres-Benito L, et al. (2010) Altered intracel-

    lular Ca2+ homeostasis in nerve terminals of severe spinal mus-

    cular atrophy mice. J Neurosci30, 849857.

    Sarnat HB, Trevenen CL (2007) Motor neuron degeneration in a

    20-week male fetus: spinal muscular atrophy type 0. Can J

    Neurol Sci34, 215220.

    Schreml J, Riessland M, Paterno M, et al. (2012) Severe SMA

    mice show organ impairment that cannot be rescued bytherapy with the HDACi JNJ-26481585. Eur J Hum Genet 21,

    643652.

    Shababi M, Habibi J, Yang HT, et al. (2010a) Cardiac defects

    contribute to the pathology of Spinal Muscular Atrophy mod-

    els. Hum Mol Genet19, 40594071.

    Shababi M, Mattis VB, Lorson CL (2010b) Therapeutics that

    directly increase SMN expression to treat spinal muscular atro-

    phy. Drug News Perspect23, 475482.

    Shababi M, Habibi J, Ma L, et al. (2012) Partial restoration of

    cardio-vascular defects in a rescued severe model of spinal

    muscular atrophy.J Mol Cell Cardiol52, 10741082.

    Singh NN, Shishimorova M, Cao LC, et al. (2009) A short anti-

    sense oligonucleotide masking a unique intronic motif pre-

    vents skipping of a critical exon in spinal muscular atrophy.

    RNA Biol6, 341350.

    Singh NN, Hollinger K, Bhattacharya D, et al. (2010) An anti-

    sense microwalk reveals critical role of an intronic position

    linked to a unique long-distance interaction in pre-mRNA

    splicing.RNA 16, 11671181.

    Soler-Botija C, Ferrer I, Gich I, et al. (2002) Neuronal death is

    enhanced and begins during foetal development in type I

    spinal muscular atrophy spinal cord. Brain 125, 16241634.

    Soler-Botija C, Cusco I, Caselles L, et al. (2005) Implication of

    fetal SMN2 expression in type I SMA pathogenesis: protection

    or pathological gain of function? J Neuropathol Exp Neurol64, 215223.

    Somers E, Stencel Z, Wishart TM, et al. (2012) Density, calibre

    and ramification of muscle capillaries are altered in a mouse

    model of severe spinal muscular atrophy. Neuromuscul Disord

    22, 435442.

    Sugarman EA, Nagan N, Zhu H, et al. (2012) Pan-ethnic carrier

    screening and prenatal diagnosis for spinal muscular atrophy:

    clinical laboratory analysis of >72,400 specimens. Eur J Hum

    Genet20, 2732.

    Tein I, Sloane AE, Donner EJ, et al. (1995) Fatty acid oxidation

    abnormalities in childhood-onset spinal muscular atrophy: pri-

    mary or secondary defect(s)? Pediatr Neurol12, 2130.

    Tizzano EF, Cabot C, Baiget M (1998) Cell-specific survival motor

    neuron gene expression during human development of thecentral nervous system: implications for the pathogenesis of

    spinal muscular atrophy. Am J Pathol153, 355361.

    Torres-Benito L, Neher MF, Cano R, et al. (2011) SMN require-

    ment for synaptic vesicle, active zone and microtubule postna-

    tal organization in motor nerve terminals. PLoS One 6,

    e26164.

    Vaidla E, Talvik I, Kulla A, et al. (2007) Neonatal spinal muscular

    atrophy type 1 with bone fractures and heart defect. J Child

    Neurol22, 6770.

    Valori CF, Ning K, Wyles M, et al. (2010) Systemic delivery of

    scAAV9 expressing SMN prolongs survival in a model of spinal

    muscular atrophy. Sci Transl Med2, 35ra42.

    Vitte JM, Davoult B, Roblot N, et al. (2004) Deletion of murine

    Smn exon 7 directed to liver leads to severe defect of liverdevelopment associated with iron overload. Am J Pathol 165,

    17311741.

    Winkler C, Eggert C, Gradl D, et al. (2005) Reduced U snRNP

    assembly causes motor axon degeneration in an animal model

    for spinal muscular atrophy. Genes Dev19, 23202330.

    Wirth B, Brichta L, Hahnen E (2006) Spinal muscular atrophy:

    from gene to therapy. Semin Pediatr Neurol13, 121131.

    Wishart TM, Huang JP, Murray LM, et al. (2010) SMN deficiency

    disrupts brain development in a mouse model of severe spinal

    muscular atrophy. Hum Mol Genet19, 42164228.

    Zerres K, Wirth B, Rudnik-Schoneborn S (1997) Spinal muscular

    atrophy clinical and genetic correlations. Neuromuscul Dis-

    ord7, 202207.

    Zhang HL, Pan F, Hong D, et al. (2003) Active transport of the

    survival motor neuron protein and the role of exon-7 in cyto-

    plasmic localization. J Neurosci23, 66276637.

    Zolkipli Z, Sherlock M, Biggar WD, et al. (2012) Abnormal fatty

    acid metabolism in spinal muscular atrophy may predispose to

    perioperative risks. Eur J Paediatr Neurol16, 549553.

    2013 Anatomical Society

    SMA: a motor neuron or multi-organ disease?, M. Shababi et al.14