Acessibilidade / Reportar erro

Neuromuscular disorders: genes, genetic counseling and therapeutic trials

Abstract

Neuromuscular disorders (NMD) are a heterogeneous group of genetic conditions, with autosomal dominant, recessive, or X-linked inheritance. They are characterized by progressive muscle degeneration and weakness. Here, we are presenting our major contributions to the field during the past 30 years. We have mapped and identified several novel genes responsible for NMD. Genotype-phenotype correlations studies enhanced our comprehension on the effect of gene mutations on related proteins and their impact on clinical findings. The search for modifier factors allowed the identification of a novel "protective"; variant which may have important implication on therapeutic developments. Molecular diagnosis was introduced in the 1980s and new technologies have been incorporated since then. Next generation sequencing greatly improved our capacity to identify disease-causing mutations with important benefits for research and prevention through genetic counseling of patients' families. Stem cells researches, from and for patients, have been used as tools to study human genetic diseases mechanisms and for therapies development. The clinical effect of preclinical trials in mice and canine models for muscular dystrophies are under investigation. Finally, the integration of our researches and genetic services with our post-graduation program resulted in a significant output of new geneticists, spreading out this expertise to our large country.

Keywords
genetic diseases; genetic counseling; neuromuscular disorders; stem cells; therapies

Introduction

Neuromuscular disorders (NMD) include a wide group of genetic conditions that affect about 1 in 1000 individuals worldwide. They are characterized by progressive muscle degeneration and weakness due to genetic mutations which primarily or secondarily impair skeletal muscle function. Most of these mutations display autosomal recessive, autosomal dominant or X-linked inheritance. The onset can occur in childhood and have a severe progression or later in life with a slower course. A complete list of diseases/genes/phenotypes is available at http://www.musclegenetable.fr/.

Our group, at the Institute of Biosciences, at the University of São Paulo, was pioneer in establishing a center for diagnosis, genetic counseling and research in neuromuscular disorders in the late 1970s. In the 1980s, we founded the Brazilian Muscular Dystrophy Association, which has recently joined with a larger entity for handicapped patients, the AACD. In 2000, the Human Genome Research Center was founded, and in 2005 stem cells research was introduced, to the now called Human Genome and Stem Cells Research Center (HUG-CELL). To date, we have attended about 26,000 patients with NMD and at-risk relatives at the HUG-CELL. Here, we will summarize our main contributions to the field.

Mapping and identification of new genes

Although the human genome project was declared completed in 2003, it has been recently estimated that only 50% of the 7.315 Mendelian phenotypes, usually represented by rare disorders, have been associated to given genes (Chong et al., 2015Chong JX, Buckingham KJ, Jhangiani SN, Boehm C, Sobreira N, Smith JD, Harrell TM, McMillin MJ, Wiszniewski W, Gambin T et al. (2015) The Genetic Basis of Mendelian Phenotypes: Discoveries, Challenges, and Opportunities. Am J Hum Genet 7:199-215.). This scenario is even more complex and challenging for disorders with multifactorial inheritance, in which multiple variants, genes, environmental effects and epigenetic phenomena may be involved and are usually highly heterogeneous. Therefore, the identification of putative causative variants of clinical phenotypes is still an important approach in human genetics, providing a direct link between a particular phenotype and a given gene.

We have mapped 8 loci responsible for neuromuscular disorders and identified for the first time five of their mutational gene mechanisms. We have also collaborated with the identification of another five genes.

Large families with many affected members, as well as isolates with high degree of consanguinity were very important to identify new disease genes (Figueiredo et al., 2015Figueiredo T, Melo US, Pessoa AL, Nobrega PR, Kitajima JP, Correa I, Zatz M, Kok F and Santos S (2015) Homozygous missense mutation in MED25 segregates with syndromic intellectual disability in a large consanguineous family. J Med Genet 52:123-127.).

Duchenne Muscular Dystrophy

In 1981, we described a case of a girl affected by Duchenne muscular dystrophy (DMD), carrying an X-autosomal translocation with a breakpoint at Xp21 (Zatz et al., 1981Zatz M, Vianna-Morgante AM, Campos P and Diament AJ (1981) Translocation (X;6) in a female with Duchenne muscular dystrophy: Implications for the localisation of the DMD locus. J Med Genet 18:442-447.). This case, together with similar cases reported in the literature, was key to confirm the mapping of the Duchenne locus at Xp21, leading to the identification of the DMD gene in 1988 by the group of Louis M. Kunkel (Koenig et al., 1998Koenig M, Monaco AP and Kunkel LM (1998) The complete sequence of dystrophin predicts a rod-shaped cytoskeletal protein. Cell 53:219-228.; Monaco et al., 1988Monaco AP, Bertelson CJ, Liechti-Gallati S, Moser H and Kunkel LM (1988) An explanation for the phenotypic differences between patients bearing partial deletions of the DMD locus. Genomics 2:90-95.).

Limb-girdle muscular dystrophies

Subsequently we focused on limb-girdle muscular dystrophies (LGMD), a group of disorders that affect primarily the pelvic and scapular limb-girdles. LGMD can be transmitted through autosomal recessive (AR) and, less frequently, autosomal dominant (AD) inheritance. Currently there are 19 AR (LGMD2A-2T) and eight AD genes (LGMD1A-1H) already identified. Affected patients can have a mild disease course remaining ambulant until late in life or have a severe phenotype, clinically very similar to X-linked DMD. Among the severe forms, LGMD2C, LGMD2D, LGMD2E and LGMD2F are sarcoglycanopathies, caused by mutations in the SGCG, SGCA, SGCB and SGCD genes, coding for γ-SG, α-SG, β-SG, and δ-SG, components of the sarcoglycan (SG) complex. These transmembrane glycoproteins, together with sarcospan, dystrophin, dystroglycans, syntrophins and α-dystrobrevin, constitute the Dystrophin-glycoprotein complex. This complex acts as a linker between the cytoskeleton of the muscle cell and the extracellular matrix, providing mechanical support to the plasma membrane during myofiber contraction (Yoshida and Ozawa, 1990Yoshida M and Ozawa E (1990) Glycoprotein complex anchoring dystrophin to sarcolemma. J Biochem 108:748-52.). Among patients diagnosed with AR LGMD (through DNA and/or muscle protein analysis) from 120 families in Brazil, the LGMD2A is the most frequent form of sarcoglycanophaty (Zatz et al., 2003Zatz M, de Paula F, Starling A and Vainzof M (2003) The 10 autosomal recessive limb-girdle muscular dystrophies. Neuromuscul Disord 13:532-544.).

The first LGMD gene identified by our group is responsible for LGMD2F, caused by mutations in δ-SG gene coding for one of the four sarcoglycan proteins. This gene causes a severe childhood form of muscular dystrophy (Nigro et al., 1996Nigro V, de Sa Moreira E, Piluso G, Vainzof M, Belsito A, Politano L, Puca AA, Passos-Bueno MR and Zatz M (1996) Autosomal recessive limb-girdle muscular dystrophy, LGMD2F, is caused by a mutation in the delta-sarcoglycan gene. Nat Genet 14:195-198.; Passos-Bueno et al., 1996Passos-Bueno MR, Moreira ES, Vainzof M, Marie SK and Zatz M (1996) Linkage analysis in autosomal recessive limb-girdle muscular dystrophy (AR LGMD) maps a sixth form to 5q33-34 (LGMD2F) and indicates that there is at least one more subtype of AR LGMD. Hum Mol Genet 5:815-820.). Another gene mapped and identified by our group is responsible for AR-LGMD2G, caused by a mutation in the TCAP gene, coding for the protein telethonin (Moreira et al., 1997Moreira ES, Vainzof M, Marie SK, Sertie AL, Zatz M and Passos-Bueno MR (1997) The seventh form of autosomal recessive limb-girdle muscular dystrophy is mapped to 17q11-12. Am J Hum Genet 61:151-159., 2000Moreira ES, Wiltshire TJ, Faulkner G, Nilforoushan A, Vainzof M, Suzuki OT, Valle G, Reeves R, Zatz M, Passos-Bueno MR et al. (2000) Limb-girdle muscular dystrophy type 2G is caused by mutations in the gene encoding the sarcomeric protein telethonin. Nat Genet 24:163-166.). Although identified in compound heterozygous patients belonging to a family of Italian origin, LGMD2G has been shown afterwards to be very rare, and few cases have been reported outside Brazil. LGMD2G is characterized by a variable phenotype, with onset in childhood or adulthood (Vainzof et al., 2002Vainzof M, Moreira ES, Suzuki OT, Faulkner G, Valle G, Beggs AH, Carpen O, Ribeiro AF, Zanoteli E, Gurgel-Gianneti J et al. (2002) Telethonin protein expression in neuromuscular disorders. Biochim Biophys Acta 1588:33-40.).

The protein telethonin is a 19 kDa component of the sarcomere Z-disk in striated and cardiac muscles (Valle et al., 1997Valle G, Faulkner G, De Antoni A, Pacchioni B, Pallavicini A, Pandolfo D, Tiso N, Toppo S, Trevisan S and Lanfranchi G (1997) Telethonin, a novel sarcomeric protein of heart and skeletal muscle. FEBS Lett 415:163-168.). Telethonin is the first example of a sarcomeric protein, of which deficiency was associated with a form of muscular dystrophy, without disruption of the sarcomere structure (Vainzof et al., 2002Vainzof M, Moreira ES, Suzuki OT, Faulkner G, Valle G, Beggs AH, Carpen O, Ribeiro AF, Zanoteli E, Gurgel-Gianneti J et al. (2002) Telethonin protein expression in neuromuscular disorders. Biochim Biophys Acta 1588:33-40.).

More recently we have identified the gene for the AD LGMD1G muscular dystrophy (Vieira et al., 2014Vieira NM, Naslavsky MS, Licinio L, Kok F, Schlesinger D, Vainzof M, Sanchez N, Kitajima JP, Gal L, Cavacana N et al. (2014) A defect in the RNA-processing protein HNRPDL causes limb-girdle muscular dystrophy 1G (LGMD1G). Hum Mol Genet 23:4103-4110.), which had been mapped by our group 10 years earlier (Starling et al., 2004Starling A, Kok F, Passos-Bueno MR, Vainzof M and Zatz M (2004) A new form of autosomal dominant limb-girdle muscular dystrophy (LGMD1G) with progressive fingers and toes flexion limitation maps to chromosome 4p21. Eur J Hum Genet 12:1033-1040.). Mutations in the RNA-processing protein HNRPDL, a heterogeneous ribonucleoprotein family member, which participates in mRNA biogenesis and metabolism, were identified in two large, unrelated families: one from Brazil and the other from Uruguay. The identification of the LGMD1G gene revealed a novel association between a muscular disorder and an RNA-related gene and reinforces the importance of RNA binding/processing proteins in muscle development and muscle disease.

We also collaborated in the identification of LGMD2A (Beckmann et al., 1991Beckmann JS, Richard I, Hillaire D, Broux O, Antignac C, Bois E, Cann H, Cottingham RW Jr., Feingold N, Feingold J et al. (1991) A gene for limb-girdle muscular dystrophy maps to chromosome 15 by linkage. C R Acad Sci III 312:141-148.; Richard et al., 1995Richard I, Broux O, Allamand V, Fougerousse F, Chiannilkulchai N, Bourg N, Brenguier L, Devaud C, Pasturaud P, Roudaut C et al. (1995) Mutations in the proteolytic enzyme calpain 3 cause limb-girdle muscular dystrophy type 2A. Cell 81:27-40.; Spencer et al., 1997Spencer MJ, Tidball JG, Anderson LV, Bushby KM, Harris JB, Passos-Bueno MR, Somer H, Vainzof M and Zatz M (1997) Absence of calpain 3 in a form of limb-girdle muscular dystrophy (LGMD2A). J Neurol Sci 146:173-178.), LGMD2B (Passos-Bueno et al., 1995Passos-Bueno MR, Bashir R, Moreira ES, Vainzof M, Marie SK, Vasquez L, Iughetti P, Bakker E, Keers S, Stephenson A et al. (1995) Confirmation of the 2p locus for the mild autosomal recessive limb-girdle muscular dystrophy gene (LGMD2B) in three families allows refinement of the candidate region. Genomics 27:192-195.; Bushby et al., 1996Bushby K, Bashir R, Keers S, Britton S, Zatz M, Passos-Bueno MR, Lovett M, Mahjneh I, Marconi G and Strachan T (1996) The molecular biology of LGMD2B—towards the identification of the LGMD gene on chromosome 2p13. Neuromuscul Disord 6:491-492.; Bashir et al., 1998Bashir R, Britton S, Strachan T, Keers S, Vafiadaki E, Lako M, Richard I, Marchand S, Bourg N, Argov Z et al. (1998) A gene related to Caenorhabditis elegans spermatogenesis factor fer-1 is mutated in limb-girdle muscular dystrophy type 2B. Nat Genet 20:37-42.;), LGMD2C (McNally et al., 1996bMcNally EM, Passos-Bueno MR, Bonnemann CG, Vainzof M, de Sa Moreira E, Lidov HG, Othmane KB, Denton PH, Vance JM, Zatz M et al. (1996b) Mild and severe muscular dystrophy caused by a single gamma-sarcoglycan mutation. Am J Hum Genet 59:1040-1047.), LGMD2D and LGMD2E genes (Bonnemann et al., 1996Bonnemann CG, Passos-Bueno MR, McNally EM, Vainzof M, de Sa Moreira E, Marie SK, Pavanello RC, Noguchi S, Ozawa E, Zatz M et al. (1996) Genomic screening for beta-sarcoglycan gene mutations: Missense mutations may cause severe limb-girdle muscular dystrophy type 2E (LGMD 2E). Hum Mol Genet 5:1953-1961.), responsible for several forms of AR-LGMDs (Zatz et al., 2003Zatz M, de Paula F, Starling A and Vainzof M (2003) The 10 autosomal recessive limb-girdle muscular dystrophies. Neuromuscul Disord 13:532-544.).

Spastic paraplegia (SPG)

We have mapped and/or identified several genes responsible for pure spastic paraplegia, namely SPG8 (Rocco et al., 2000Rocco P, Vainzof M, Froehner SC, Peters MF, Marie SK, Passos-Bueno MR and Zatz M (2000) Brazilian family with pure autosomal dominant spastic paraplegia maps to 8q: Analysis of muscle beta 1 syntrophin. Am J Med Genet 92:122-127.; Valdmanis et al., 2007Valdmanis PN, Meijer IA, Reynolds A, Lei A, MacLeod P, Schlesinger D, Zatz M, Reid E, Dion PA, Drapeau P et al. (2007) Mutations in the KIAA0196 gene at the SPG8 locus cause hereditary spastic paraplegia. Am J Hum Genet 80:152-161.), SPG4 (Starling et al., 2002bStarling A, Rocco P, Passos-Bueno MR, Hazan J, Marie SK and Zatz M (2002b) Autosomal dominant (AD) pure spastic paraplegia (HSP) linked to locus SPG4 affects almost exclusively males in a large pedigree. J Med Genet 39:e77.; Mitne-Neto et al., 2007aMitne-Neto M, Kok F, Beetz C, Pessoa A, Bueno C, Graciani Z, Martyn M, Monteiro CB, Mitne G, Hubert P et al. (2007a) A multi-exonic SPG4 duplication underlies sex-dependent penetrance of hereditary spastic paraplegia in a large Brazilian pedigree. Eur J Hum Genet 15:1276-1279.), or X-linked SPG (Starling et al., 2002aStarling A, Rocco P, Cambi F, Hobson GM, Passos Bueno MR and Zatz M (2002a) Further evidence for a fourth gene causing X-linked pure spastic paraplegia. Am J Med Genet 111:152-156.). Our group also described a new complicated autosomal recessive form of spastic paraplegia named SPOAN (spastic paraplegia, optic atrophy, neuropathy) (Macedo-Souza et al., 2005Macedo-Souza LI, Kok F, Santos S, Amorim SC, Starling A, Nishimura A, Lezirovitz K, Lino AM and Zatz M (2005) Spastic paraplegia, optic atrophy, and neuropathy is linked to chromosome 11q13. Ann Neurol 57:730-737., 2009Macedo-Souza LI, Kok F, Santos S, Licinio L, Lezirovitz K, Cavacana N, Bueno C, Amorim S, Pessoa A, Graciani Z et al. (2009) Spastic paraplegia, optic atrophy, and neuropathy: New observations, locus refinement, and exclusion of candidate genes. Ann Hum Genet 73:382-387.). SPOAN was identified in a geographically isolated region in the backlands of Northeastern Brazil, known for a high incidence of consanguineous marriages. The mutation, recently identified through whole genome sequencing, is a homozygous deletion in the noncoding region of the kinesin light chain-2 (KLC2) gene, a novel mechanism, which reinforces the importance of noncoded regions in human pathology (Melo et al., 2015Melo US, Macedo-Souza LI, Figueiredo T, Muotri AR, Gleeson JG, Coux G, Armas P, Calcaterra NB, Kitajima JP, Amorim S et al. (2015) Overexpression of KLC2 due to a homozygous deletion in the non-coding region causes SPOAN syndrome. Hum Mol Genet 24:6877-6885.).

Motor neuron diseases

Motor neuron diseases are a group of neurodegenerative disorders with involvement of upper and/or lower motor neurons, such as amyotrophic lateral sclerosis (ALS), spinal muscular atrophy, progressive bulbar palsy, and primary lateral sclerosis. A new locus for a recessive X-linked juvenile form of distal muscular atrophy has also been mapped in our lab (Takata et al., 2004Takata RI, Speck Martins CE, Passos-Bueno MR, Abe KT, Nishimura AL, Da Silva MD, Monteiro Jr A, Lima MI, Kok F and Zatz M (2004) A new locus for recessive distal spinal muscular atrophy at Xq13.1-q21. J Med Genet 41:224-229.).

However, a more important contribution from our group was the mapping and subsequent identification of the amyotrophic lateral sclerosis gene 8 or ALS8 in a very large Brazilian genealogy (Nishimura et al., 2004aNishimura AL, Mitne-Neto M, Silva HC, Oliveira JR, Vainzof M and Zatz M (2004a) A novel locus for late onset amyotrophic lateral sclerosis/motor neurone disease variant at 20q13. J Med Genet 41:315-320.,bNishimura AL, Mitne-Neto M, Silva HC, Richieri-Costa A, Middleton S, Cascio D, Kok F, Oliveira JR, Gillingwater T, Webb J et al. (2004b) A mutation in the vesicle-trafficking protein VAPB causes late-onset spinal muscular atrophy and amyotrophic lateral sclerosis. Am J Hum Genet 75:822-831.). This gene codes for the vesicle-associated membrane protein/synaptobrevin-associated membrane protein B (VAPB) gene. Members of the vesicle-associated proteins are intracellular membrane proteins that can associate with microtubules and that have been shown to have a function in membrane transport (Mitne-Neto et al., 2007bMitne-Neto M, Ramos CR, Pimenta DC, Luz JS, Nishimura AL, Gonzales FA, Oliveira CC and Zatz M (2007b) A mutation in human VAP-B—MSP domain, present in ALS patients, affects the interaction with other cellular proteins. Protein Expr Purif 55:139-146.). We have also shown that the VAPB mutation, which is now identified in hundreds of Brazilian patients, is due to a founder effect (Nishimura et al., 2005Nishimura AL, Al-Chalabi A and Zatz M (2005) A common founder for amyotrophic lateral sclerosis type 8 (ALS8) in the Brazilian population. Hum Genet 118:499-500.). VAPB mutations have been subsequently identified in patients with different origin – from Japan and Europe (Funke et al., 2010Funke AD, Esser M, Kruttgen A, Weis J, Mitne-Neto M, Lazar M, Nishimura AL, Sperfeld AD, Trillenberg P, Senderek J et al. (2010) The p.P56S mutation in the VAPB gene is not due to a single founder: The first European case. Clin Genet 77:302-303.). This finding has attracted a lot of attention from researchers in the field because VAPB seems to be involved in other forms of ALS (Coatti et al., 2015Coatti GC, Beccari MS, Olavio TR, Mitne-Neto M, Okamoto OK and Zatz M (2015) Stem cells for amyotrophic lateral sclerosis modeling and therapy: Myth or fact? Cytometry A 87:197-211.; Teuling et al., 2007Teuling E, Ahmed S, Haasdijk E, Demmers J, Steinmetz MO, Akhmanova A, Jaarsma D and Hoogenraad CC (2007) Motor neuron disease-associated mutant vesicle-associated membrane protein-associated protein (VAP) B recruits wild-type VAPs into endoplasmic reticulum-derived tubular aggregates. J Neurosci 27:9801-9815.).

Genotype-phenotype correlations

Neuromuscular disorders are characterized by marked phenotypic and genotypic heterogeneity, with a similar clinical course caused by mutations in different genes while several different phenotypes can be associated with mutations in one particular gene (Zatz et al., 2000Zatz M, Vainzof M and Passos-Bueno MR (2000) Limb-girdle muscular dystrophy: One gene with different phenotypes, one phenotype with different genes. Curr Opin Neurol 13:511-517.). Our group has contributed with this characterization for several forms of muscular dystrophies.

After the identification of the dystrophin gene as the protein involved in DMD, a correlation between dystrophin presence/quantity and the severity of the phenotype in dystrophinopathies was strongly suggested. Genotype, phenotype and protein analysis enabled us to pinpoint the important functional domains of the dystrophin protein (Vainzof et al., 1993Vainzof M, Takata RI, Passos-Bueno MR, Pavanello RC and Zatz M (1993) Is the maintainance of the C-terminus domain of dystrophin enough to ensure a milder Becker muscular dystrophy phenotype? Hum Mol Genet 2:39-42.; Passos-Bueno et al., 1994Passos-Bueno MR, Vainzof M, Marie SK and Zatz M (1994) Half the dystrophin gene is apparently enough for a mild clinical course: Confirmation of its potential use for gene therapy. Hum Mol Genet 3:919-922.), and question the previously suggested correlation (Vainzof et al., 1990Vainzof M, Pavanello RC, Pavanello Filho I, Passos-Bueno MR, Rapaport D, Hsi CT and Zatz M (1990) Dystrophin immunostaining in muscles from patients with different types of muscular dystrophy: A Brazilian study. J Neurol Sci 98:221-233., 1991bVainzof M, Pavanello RC, Pavanello I, Tsanaclis AM, Levy JA, Passos-Bueno MR, Rapaport D and Zatz M (1991b) Dystrophin immunofluorescence pattern in manifesting and asymptomatic carriers of Duchenne's and Becker muscular dystrophies of different ages. Neuromuscul Disord 1:177-183., cVainzof M, Zubrzycka-Gaarn EE, Rapaport D, Passos-Bueno MR, Pavanello RC, Pavanello-Filho I and Zatz M (1991c) Immunofluorescence dystrophin study in Duchenne dystrophy through the concomitant use of two antibodies directed against the carboxy-terminal and the amino-terminal region of the protein. J Neurol Sci 101:141-147., 1995bVainzof M, Passos-Bueno MR, Pavanello RC and Zatz M (1995b) Is dystrophin always altered in Becker muscular dystrophy patients? J Neurol Sci 131:99-104.).

For most forms of autosomal recessive LGMDs, missense mutations, on average, have been associated with a milder phenotype as compared to null mutations. (Richard et al., 1999Richard I, Roudaut C, Saenz A, Pogue R, Grimbergen JE, Anderson LV, Beley C, Cobo AM, de Diego C, Eymard B et al. (1999) Calpainopathy- A survey of mutations and polymorphisms. Am J Hum Genet 64:1524-40.; de Paula et al., 2002de Paula F, Vainzof M, Passos-Bueno MR, de Cassia MPR, Matioli SR, Anderson LVB, Nigro V and Zatz M (2002) Clinical variability in calpainopathy: What makes the difference? Eur J Hum Genet 10:825-832.; Starling et al., 2003Starling A, de Paula F, Silva H, Vainzof M and Zatz M (2003) Calpainopathy: How broad is the spectrum of clinical variability? J Mol Neurosci 21:233-236.). We also observed that mutations in the LGMD2I genes, first reported in severe congenital forms, could be also found in adult forms with slow progression or even asymptomatic cases (de Paula et al., 2003de Paula F, Vieira N, Starling A, Yamamoto LU, Lima B, de Cassia Pavanello R, Vainzof M, Nigro V and Zatz M (2003) Asymptomatic carriers for homozygous novel mutations in the FKRP gene: The other end of the spectrum. Eur J Hum Genet 11:923-930.), associated with specific protein alterations in the muscle (Yamamoto et al., 2008Yamamoto LU, Velloso FJ, Lima BL, Fogaca LL, de Paula F, Vieira NM, Zatz M and Vainzof M (2008) Muscle protein alterations in LGMD2I patients with different mutations in the Fukutin-related protein gene. J Histochem Cytochem 56:995-1001.). Muscle protein characterizations have contributed to elucidate the organization of the dystrophin-glycoprotein complex (Vainzof et al., 1991aVainzof M, Pavanello RC, Pavanello-Filho I, Rapaport D, Passos-Bueno MR, Zubrzycka-Gaarn EE, Bulman DE and Zatz M (1991a) Screening of male patients with autosomal recessive Duchenne dystrophy through dystrophin and DNA studies. Am J Med Genet 39:38-41., 1996Vainzof M, Passos-Bueno MR, Canovas M, Moreira ES, Pavanello RC, Marie SK, Anderson LV, Bonnemann CG, McNally EM, Nigro V et al. (1996) The sarcoglycan complex in the six autosomal recessive limb-girdle muscular dystrophies. Hum Mol Genet 5:1963-1969., 1999Vainzof M, Passos-Bueno MR, Pavanello RC, Marie SK, Oliveira AS and Zatz M (1999) Sarcoglycanopathies are responsible for 68% of severe autosomal recessive limb-girdle muscular dystrophy in the Brazilian population. J Neurol Sci 164:44-49.), for genotype-phenotype correlation studies and had an important role in NMD diagnosis. After the introduction of next generation sequencing (NGS), molecular diagnosis is achieved directly with DNA analysis and therefore muscle biopsies have been obtained only for research purposes.

The search for modifier (protective) variants/factors

Genotype-phenotype correlation studies revealed that, for several disorders such as LGMD, facioscapulohumeral muscular dystrophy, the same disease mutation usually associated with a severe phenotype could be also found in individuals only mildly affected or even asymptomatic (Bonnemann et al., 1996Bonnemann CG, Passos-Bueno MR, McNally EM, Vainzof M, de Sa Moreira E, Marie SK, Pavanello RC, Noguchi S, Ozawa E, Zatz M et al. (1996) Genomic screening for beta-sarcoglycan gene mutations: Missense mutations may cause severe limb-girdle muscular dystrophy type 2E (LGMD 2E). Hum Mol Genet 5:1953-1961.; de Paula et al., 2002de Paula F, Vainzof M, Passos-Bueno MR, de Cassia MPR, Matioli SR, Anderson LVB, Nigro V and Zatz M (2002) Clinical variability in calpainopathy: What makes the difference? Eur J Hum Genet 10:825-832., 2003de Paula F, Vieira N, Starling A, Yamamoto LU, Lima B, de Cassia Pavanello R, Vainzof M, Nigro V and Zatz M (2003) Asymptomatic carriers for homozygous novel mutations in the FKRP gene: The other end of the spectrum. Eur J Hum Genet 11:923-930.; McNally et al., 1996aMcNally EM, Duggan D, Gorospe JR, Bonnemann CG, Fanin M, Pegoraro E, Lidov HG, Noguchi S, Ozawa E, Finkel RS et al. (1996a) Mutations that disrupt the carboxyl-terminus of gamma-sarcoglycan cause muscular dystrophy. Hum Mol Genet 59:1841-1847.; Moreira et al., 2003Moreira ES, Vainzof M, Suzuki OT, Pavanello RC, Zatz M and Passos-Bueno MR (2003) Genotype-phenotype correlations in 35 Brazilian families with sarcoglycanopathies including the description of three novel mutations. J Med Genet 40:E12.; Starling et al., 2003Starling A, de Paula F, Silva H, Vainzof M and Zatz M (2003) Calpainopathy: How broad is the spectrum of clinical variability? J Mol Neurosci 21:233-236.; Tonini et al., 2004Tonini MM, Passos-Bueno MR, Cerqueira A, Matioli SR, Pavanello R and Zatz M (2004) Asymptomatic carriers and gender differences in facioscapulohumeral muscular dystrophy (FSHD). Neuromuscul Disord 14:33-38.; Ricci et al., 2014Ricci G, Zatz M and Tupler R (2014) Facioscapulohumeral muscular dystrophy: More complex than it appears. Curr Mol Med [Epub ahead of print].; Scionti et al., 2012Scionti I, Greco F, Ricci G, Govi M, Arashiro P, Vercelli L, Berardinelli A, Angelini C, Antonini G, Cao M et al. (2012) Large-scale population analysis challenges the current criteria for the molecular diagnosis of fascioscapulohumeral muscular dystrophy. Am J Hum Genet 90:628-635.). Although rare, this has also been observed for DMD (Zatz et al., 2014Zatz M, Pavanello RC, Lazar M, Yamamoto GL, Lourenco NC, Cerqueira A, Nogueira L and Vainzof M (2014) Milder course in Duchenne patients with nonsense mutations and no muscle dystrophin. Neuromuscul Disord 24:986-989.; Castro-Gago, 2015Castro-Gago M (2015) Milder course in Duchenne patients with nonsense mutations and no muscle dystrophin. Neuromuscul Disord 25:443.; Zatz, 2015Zatz M (2015) Response to: Milder course in Duchenne patients with nonsense mutations and no muscle dystrophin. Neuromuscul Disord 25:444.). Utrophin, an autosomal ubiquitously expressed protein with structural homology to dystrophin, has been suggested as a possible modulator of DMD severity and thus as a therapeutic target for treating DMD. However, we observed that utrophin expression does not differ between severely and mildly affected DMD patients (Vainzof et al., 1995aVainzof M, Passos-Bueno MR, Man N and Zatz M (1995a) Absence of correlation between utrophin localization and quantity and the clinical severity in Duchenne/Becker dystrophies. Am J Med Genet 58:305-309., Vainzof 2016Vainzof M, Feitosa L, Ayub-Guerrieri D, Canovas M, Pavanello RCM and Zatz M (2016) Mild course in atypical Duchenne muscular dystrophy patients is not caused by utrophin overexpression. Neuromuscul. Disord 26:197-200.).

Therefore, the search for protective variants or mechanisms continues to be a great challenge. Interestingly, the identification of two golden retriever muscular dystrophy (GRMD) dogs with a very mild phenotype and a normal lifespan (Zucconi et al., 2010Zucconi E, Valadares MC, Vieira NM, Bueno Jr CR, Secco M, Jazedje T, da Silva HC, Vainzof M and Zatz M (2010) Ringo: Discordance between the molecular and clinical manifestation in a golden retriever muscular dystrophy dog. Neuromuscul Disord 20:64-70.; Zatz et al., 2014Zatz M, Pavanello RC, Lazar M, Yamamoto GL, Lourenco NC, Cerqueira A, Nogueira L and Vainzof M (2014) Milder course in Duchenne patients with nonsense mutations and no muscle dystrophin. Neuromuscul Disord 24:986-989.) have been the subject of much investigation. We have recently shown that up-regulation of Jagged1, which is a known regulator of the Notch pathway, is responsible for the milder course in these two dogs (Vieira et al., 2015Vieira NM, Elvers I, Alexander MS, Moreira YB, Eran A, Gomes JP, Marshall JL, Karlsson EK, Verjovski-Almeida S, Lindblad-Toh K et al. (2015) Jagged 1 rescues the Duchenne Muscular Dystrophy phenotype. Cell 163:1204-1213.). In addition, in the mdx mouse model for DMD, we observed a milder course associated with less fibrosis and more regeneration when the mutation was transferred to a different background (unpublished observations). Identifying what protects some exceptional dogs, mice or individuals from the deleterious effect of a disease-causing mutation is a great challenge which could open new venues for treatment (Cohn and Dubowitz, 2016Cohn RD and Dubowitz V (2016) Duchenne muscular dystrophy: Ringo to the rescue? Neuromuscul Disord 26:5-6.).

Genetic testing

Research in human and medical genetics allied to genetic counseling services in Brazil initiated in the late 1960s. During those years, the model implemented by Oswaldo Frota-Pessoa in our Department consisted of research associated with services to patients and families, in a mutually beneficial scenario: patients contributed to new findings, while those new findings helped patients. This model has been maintained by the geneticists at the Human Genome and Stem Cells Research Center. Genetic testing and genetic counseling in our community, has been one of the main activities of HUG-CELL with emphasis to the group of disorders associated with research projects, including: neuromuscular, neurodegenerative, craniofacial, deafness, intellectual disability, and Autism Spectrum disorders.

From Southern blot to next generation sequencing

Molecular diagnosis for neuromuscular disorders was introduced in our center in the late 1980s based on Southern blot and PCR (polymerase chain reaction) analysis. The introduction of these methods have largely contributed for the development of research as well as for the prevention of new cases, based on diagnosis of NMD families, carrier detection in DMD/DMB families and genetic counseling. The detection of deletions in the dystrophin gene (which account for 60–70% of the mutations responsible for Duchenne dystrophy) were first performed by Southern blot, followed by multiplex analysis of the most common deleted exons by PCR. In the 1990's, the use of microsatellite markers throughout the genome led to the mapping of novel NMD genes. In addition, microsatellite markers within and flanking the dystrophin gene, allowed us to improve DMD/DMB carriers detection tests by segregation analysis, particularly in familial cases in which no deletion had been detected in the dystrophin gene. In the mid 1990's, genetic testing for diagnosis of Myotonic muscular dystrophy and Facioscapulohumeral disorder were introduced, both based on Southern blot methodology. In 2000, with the inauguration of the Human Genome Center, a non-profit DNA diagnostic lab was installed. The acquisition of a semi-automated Sanger sequencing equipment opened the possibility to again expand and improve genetic testing (by allowing the larger scale use of Sanger sequencing methodology) and include other NMD, particularly the limb-girdle muscular dystrophies. This expansion also enabled us to reach a much wider public. In addition to benefiting patients/families seen at our center it allowed the performance of updated molecular diagnosis in DNA samples from patients throughout Brazil. Our laboratory was also one of the first core sequencing facilities at University of Sao Paulo, a service that has been offered since then and which has been continuously updated. A recent new improvement in 2013-2014 was the acquisition of next generation sequencing (NGS) equipment (MiSeq and HiSeq, Illumina) and standardization of its methodology. NGS, based on a panel of 80 genes, increased significantly the efficacy of NMD diagnosis, with molecular alterations being identified in ~73% of the cases. In addition to decreasing the cost of NMD testing, the use of a panel of genes avoids the ethical issues associated to the identification of incidental findings through exome sequencing. NGS has also imposed a great challenge in bioinformatics training and expertise development on how to deal, store and analyze large data sets.

Whole exome sequencing (WES) which was standardized in our center in 2014 has brought important contributions to basic research, and to identification of new disease loci and pathogenic mutations. WES also enables a better characterization of the Brazilian population genetic variability, which is crucial for interpreting sequence analysis and for obtaining accurate diagnosis (Zatz et al., 2012Zatz M, Pavanello R de C, Lourenco NC, Cerqueira A, Lazar M and Vainzof M (2012) Assessing pathogenicity for novel mutation/sequence variants: The value of healthy older individuals. Neuromolecular Med 14:281-284.) (http://laboratorio.genoma.ib.usp.br; http://genoma.ib.usp.br).

Genetic counseling

Genetic counseling (GC) is of upmost importance for the prevention of genetic diseases, especially for the untreatable ones. The process of GC includes diagnosis confirmation, identification of at-risk members, prenatal and pre-implantation diagnosis, as well as family orientation on management of affected patients.

Several ethical issues regarding the use of genetic tests have been the subject of much debate since the pre-molecular era, particularly regarding asymptomatic at-risk relatives in late-onset disorders. In accordance with an international consensus, we do not test asymptomatic children at-risk for late-onset disorders for which there is no treatment, such as cerebellar ataxias or myotonic dystrophy, as this decision should be taken solely by the subjects themselves when they reach adulthood. On the other hand, the identification of asymptomatic carriers for recessive autosomal or X-linked diseases can be crucial for the reproductive decisions of the parents.

Before offering genetic testing, several issues are discussed with patients or family members, such as which individuals should be tested, implications of test results, and how to deal with unexpected findings, such as false paternity.

More recently, the introduction of exome sequencing has opened new ethical debates particularly related to incidental findings. What should be disclosed? What is the geneticist's responsibility? Do patients and family members understand all the possibilities when signing informed consents about their willingness to be informed or not? As there is still no Brazilian regulation on this matter, we have adopted the criteria of: ACMG (American College of Medical Genetics and Genomics) Board of Directors (2013)ACMG (American College of Medical Genetics) Board of Directors (2013) Points to consider for informed consent for genome/exome sequencing. Genet Med 15:748-749.. We provide information to those who are interested about the at-risk variants associated with disorders, to which preventive measures and/or treatment are available. Our experience has shown that there is no rigid rule and each case has to be discussed by the genetic counseling team before deciding how to better approach the patients' family.

Most services in Brazil offer prenatal diagnosis (PND) in spite of abortion not being allowed for genetic disorders reasons (the only exception for a legal abortion is the confirmation of anencephaly). In our service we decided to perform PND in the 1990s for at-risk women, who would otherwise interrupt their pregnancy, fearing that their fetus could be affected by a genetic disorder (for example mothers or sisters of Duchenne muscular dystrophy patients or parents at-risk for congenital muscular dystrophy 1A, (Yamamoto et al., 2004Yamamoto LU, Gollop TR, Naccache NF, Pavanello RC, Zanoteli E, Zatz M and Vainzof M (2004) Protein and DNA analysis for the prenatal diagnosis of alpha2-laminin-deficient congenital muscular dystrophy. Diagn Mol Pathol 13:167-171., Vainzof et al., 2005Vainzof M, Richard P, Herrmann R, Jimenez-Mallebrera C, Talim B, Yamamoto LU, Ledeuil C, Mein R, Abbs S, Brockington M et al. (2005) Prenatal diagnosis in laminin alpha2 chain (merosin)-deficient congenital muscular dystrophy: A collective experience of five international centers. Neuromuscul Disord 15:588-594.). In the majority of cases referred for PND, the test results are negative for the mutation present in the family, and as a result, more women are encouraged to continue their pregnancies and unaffected babies are saved. Moreover, since many of our patients come from a poor social background, orientation on management of the genetic disease has been an important part of our service. This approach was the subject of a paper celebrating 125 years of Science magazine in 2005 (Zatz, 2005Zatz M (2005) Global voices of science. When science is not enough: Fighting genetic disease in Brazil. Science 308:55-57.). More recently, psychoanalytical support and follow-up for patients and at-risk relatives has also been offered in our center, which has been very important particularly for patients with degenerative disorders. Finally, the integration of community services with our post-graduation program results in a significant contribution in the training of a number of geneticists, enabling them to start new Centers in other Brazilian cities and spread out this expertise to our large country.

Stem cells: from patients and for patients

Stem cells from patients: IPSc cells

The groundbreaking discovery of induced pluripotent stem cells (iPSCs) by Dr. Yamanaka's group in 2006 (Takahashi and Yamanaka, 2006Takahashi K and Yamanaka S (2006) Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126:663-676.), demonstrating the possibility to reprogram differentiated cells to an embryonic-like stem cell, opened a new venue for research. While applications of iPSCs in cell therapy are envisioned but still in a premature stage of development, the use of iPSCs as tools to study human genetic diseases mechanisms boomed in the last few years. With this in mind, we have established an IPS cells bank from patients with different neuromuscular disorders. One interesting result was obtained with patients affected by amyotrophic lateral sclerosis type 8 which had been identified by our group. We have successfully reprogrammed fibroblasts from ALS8 patients and generated motor neurons. Our results suggest that optimal levels of VAPB may play a central role in the pathogenesis of ALS8, which is in agreement with the observed reduction of VAPB in sporadic ALS and SOD1 murine model (Mitne-Neto et al., 2011Mitne-Neto M, Machado-Costa M, Marchetto MC, Bengtson MH, Joazeiro CA, Tsuda H, Bellen HJ, Silva HC, Oliveira AS, Lazar M et al. (2011) Downregulation of VAPB expression in motor neurons derived from induced pluripotent stem cells of ALS8 patients. Hum Mol Genet 20:3642-3652.).

The most recent gene editing CRISPR-cas9 technology (Doudna and Charpentier, 2014Doudna JA and Charpentier E (2014) Genome editing. The new frontier of genome engineering with CRISPR-Cas9. Science 346:1258096.) applied to different cells derived from IPSCs will certainly bring important contributions to functional studies, enhancing our comprehension on pathological mechanisms underlying neuromuscular disorders and providing new opportunities for treatment.

Stem cells for patients: cell therapy

Preclinical trials in neuromuscular and neurodegenerative disorders

The possibility to treat progressive muscular dystrophies, particularly DMD, with stem cell therapy has been of great interest and the focus of many investigations. Before starting therapeutic trials, several questions need to be addressed: What is the effect of stem-cell therapy for muscular dystrophies in animal models? What is the best source of adult stem cells? Is immunosuppression necessary? Are the experiments reproducible with different cell lines, or from different donors? Should injections be local or systemic? Most importantly, what is the safety level of non autologous stem cell transplantation? This last issue is very important for genetic conditions such as neuromuscular disorders where autologous stem cell transplantation is unlikely to be beneficial. In order to address these questions we have performed a series of pre-clinical experiments with human-derived stem cells transplanted into different murine models and GRMD dogs.

Comparison between different sources of stem cells revealed that cord tissue is a much richer source of stem cells than umbilical cord blood, and that mesenchymal stem cells from blood and tissue have a different expression profile (Secco et al., 2008aSecco M, Zucconi E, Vieira NM, Fogaca LL, Cerqueira A, Carvalho MD, Jazedje T, Okamoto OK, Muotri AR and Zatz M (2008a) Mesenchymal stem cells from umbilical cord: Do not discard the cord! Neuromuscul Disord 18:17-18.,bSecco M, Zucconi E, Vieira NM, Fogaca LL, Cerqueira A, Carvalho MD, Jazedje T, Okamoto OK, Muotri AR and Zatz M (2008b) Multipotent stem cells from umbilical cord: Cord is richer than blood! Stem Cells 26:146-150., 2009Secco M, Moreira YB, Zucconi E, Vieira NM, Jazedje T, Muotri AR, Okamoto OK, Verjovski-Almeida S and Zatz M (2009) Gene expression profile of mesenchymal stem cells from paired umbilical cord units: Cord is different from blood. Stem Cell Rev 5:387-401.). The relevance of this observation, which resulted in a highly cited paper, was based on the fact that umbilical cord blood banks (both public and private) had been storing blood and discarding tissue. Next, we also identified fallopian tubes as an important source of stem cells, with potential to enhance bone regeneration (Jazedje et al., 2009Jazedje T, Perin PM, Czeresnia CE, Maluf M, Halpern S, Secco M, Bueno DF, Vieira NM, Zucconi E and Zatz M (2009) Human fallopian tube: A new source of multipotent adult mesenchymal stem cells discarded in surgical procedures. J Transl Med 7:46., 2012Jazedje T, Bueno DF, Almada BV, Caetano H, Czeresnia CE, Perin PM, Halpern S, Maluf M, Evangelista LP, Nisenbaum MG et al. (2012) Human fallopian tube mesenchymal stromal cells enhance bone regeneration in a xenotransplanted model. Stem Cell Rev 8:355-362.).

Several reports on stem cell transplantation have been published by other groups in murine as well as canine models of muscular dystrophy using immunosuppressed animals (Di Rocco et al., 2006Di Rocco G, Iachininoto MG, Tritarelli A, Straino S, Zacheo A, Germani A, Crea F and Capogrossi MC (2006) Myogenic potential of adipose-tissue-derived cells. J Cell Sci 119:2945-2952.; Sampaolesi et al., 2006Sampaolesi M, Blot S, D'Antona G, Granger N, Tonlorenzi R, Innocenzi A, Mognol P, Thibaud JL, Galvez BG, Barthelemy I et al. (2006) Mesoangioblast stem cells ameliorate muscle function in dystrophic dogs. Nature 444:574-579.; Rouger et al., 2011Rouger K, Larcher T, Dubreil L, Deschamps JY, Le Guiner C, Jouvion G, Delorme B, Lieubeau B, Carlus M, Fornasari B et al. (2011) Systemic delivery of allogenic muscle stem cells induces long-term muscle repair and clinical efficacy in Duchenne muscular dystrophy dogs. Am J Pathol 179:2501-2518.; Nitahara-Kasahara et al., 2012Nitahara-Kasahara Y, Hayashita-Kinoh H, Ohshima-Hosoyama S, Okada H, Wada-Maeda M, Nakamura A, Okada T and Takeda S (2012) Long-term engraftment of multipotent mesenchymal stromal cells that differentiate to form myogenic cells in dogs with Duchenne muscular dystrophy. Mol Ther 20:168-177.). This approach may hinder the interpretation of clinical effects of stem cell injections since immunosuppressant drugs have a beneficial effect on muscular dystrophy (Davies and Grounds, 2006Davies KE and Grounds MD (2006) Treating muscular dystrophy with stem cells? Cell 127:1304-1306.). Therefore, we performed our xenotransplantation experiments without any immunosuppression therapy. We first analyzed the effect of human adipose-derived stem cells (hASCs), which were injected in SJL mice (the murine model for dysferlinopathy). Human cells were well tolerated and treated mice performed significantly better than untreated controls in functional tests (Vieira et al., 2008Vieira NM, Bueno Jr CR, Brandalise V, Moraes LV, Zucconi E, Secco M, Suzuki MF, Camargo MM, Bartolini P, Brum PC et al. (2008) SJL dystrophic mice express a significant amount of human muscle proteins following systemic delivery of human adipose-derived stromal cells without immunosuppression. Stem Cells 26:2391-2398.). We next compared the effect of human umbilical cord stem cells injected in SJL mice using the same protocol. Differently from the experiment with hASCs, injected mice did not show any functional improvement but untreated controls showed a decline in functional tests (Vieira et al., 2010Vieira NM, Zucconi E, Bueno Jr CR, Secco M, Suzuki MF, Bartolini P, Vainzof M and Zatz M (2010) Human multipotent mesenchymal stromal cells from distinct sources show different in vivo potential to differentiate into muscle cells when injected in dystrophic mice. Stem Cell Rev 6:560-566.; 2012Vieira NM, Valadares M, Zucconi E, Secco M, Bueno Jr. CR, Brandalise V, Assoni A, Gomes J, Landini V, Andrade T et al. (2012) Human adipose-derived mesenchymal stromal cells injected systemically into GRMD dogs without immunosuppression are able to reach the host muscle and express human dystrophin. Cell Transplant 21:1407-1417.).

The next question we wanted to answer was whether observed discrepancies were due to donors' different genetic background or different stem cell sources. Therefore, we compared the effect of pericytes (which are precursors of mesenchymal stem cells, Caplan and Sorrell, 2015Caplan AI and Sorrell JM (2015) The MSC curtain that stops the immune system. Immunol Lett. 168:136-139.) derived from different tissues (adipose tissue, endometrium, abdominal muscle and fallopian tube) from a female donor undergoing hysterectomy. Pericytes were injected intraperitoneally in double-knockout mdx/utrophin mice. We observed a beneficial effect only in pericyte-injected animals, which lived significantly longer (~ 25%) (Valadares et al., 2014Valadares MC, Gomes JP, Castello G, Assoni A, Pellati M, Bueno C, Corselli M, Silva H, Bartolini P, Vainzof M et al. (2014) Human adipose tissue derived pericytes increase life span in Utrn (tm1Ked) Dmd (mdx) /J mice. Stem Cell Rev 10:830-840.).

The danger of stem cell contamination using Parkinson's disease as a model

Preliminary trials to treat highly prevalent neurodegenerative diseases, such as Parkinson's disease (PD), with mesenchymal stem cells have generated controversial results. In a rat model of PD induced by the MPTP neurotoxin, we first observed a significant bilateral preservation of dopaminergic neurons in the substantia nigra and prevention of motor deficits typically observed in PD, following intracerebral administration of human umbilical cord-derived mesenchymal stem cells (UC-MSC) early after MPTP injury. However, surprisingly, administration of fibroblasts -mesenchymal cells without stem cell properties, as a xenotransplantation control was highly detrimental, causing significant neurodegeneration and motor dysfunction independently of MPTP administration. Our pre-clinical study suggests that fibroblasts may be common cell contaminants affecting the clinical outcome in stem cell therapy protocols, which might also explain the discrepant clinical results (Pereira et al., 2011Pereira MC, Secco M, Suzuki DE, Janjoppi L, Rodini CO, Torres LB, Araujo BH, Cavalheiro EA, Zatz M and Okamoto OK (2011) Contamination of mesenchymal stem-cells with fibroblasts accelerates neurodegeneration in an experimental model of Parkinson's disease. Stem Cell Rev 7:1006-1017.). These observations should be widely disseminated, since many private clinics claim to be injecting stem cells to treat many different diseases while, in reality, it is unknown how well characterized these injected cells are.

Safety of nonautologous stem cell transplantation: GRMD dogs

To investigate the safety of nonautologous stem cell transplantation, we injected hASCs in the best available animal model for DMD: the golden retriever muscular dystrophy (GRMD) model. Affected animals carry a frameshift point mutation resulting in the absence of the muscle protein dystrophin (Sharp et al., 1992Sharp NJ, Kornegay JN, Van Camp SD, Herbstreith MH, Secore SL, Kettle S, Hung WY, Constantinou CD, Dykstra MJ, Roses AD et al. (1992) An error in dystrophin mRNA processing in golden retriever muscular dystrophy, an animal homologue of Duchenne muscular dystrophy. Genomics 13:115-121.). These dogs have a severe disease course and most do not survive beyond two years, despite some may have a variable phenotype. Although no human dystrophin was found in muscles from recipient dogs, a functional improvement was observed shortly after a series of injections followed by an apparent stabilization afterwards, without noticeable side effects. A growing body of evidence indicates that although mesenchymal stem cells are partially defined by their ability to differentiate into various tissues in vitro, it is their trophic, paracrine and immunomodulatory functions that may have the greatest therapeutic impact in vivo, decreasing inflammation and fibrosis (Murphy et al., 2013Murphy MB, Moncivais K and Caplan AI (2013) Mesenchymal stem cells: Environmentally responsive therapeutics for regenerative medicine. Exp Mol Med 45:e54.; Caplan and Sorrell, 2015Caplan AI and Sorrell JM (2015) The MSC curtain that stops the immune system. Immunol Lett. 168:136-139.). This would explain the beneficial effect observed in the injected GRMD dogs. In short, we showed that repeated injections of hASCs, from different donors, are well tolerated in immunocompetent GRMD dogs. We also observed functional benefits in three dogs followed from four to six years post-transplantation, without tumor formation. This study has the longest follow-up of human cells transplanted animals ever reported. These observations, which should be replicated in larger samples, might have important applications for future therapy in patients with different forms of muscular dystrophies (Zatz et al., 2015Zatz M, Gomes J, Pelatti M, Secco M, Vieira N, Zucconi E, Vainzof M, Landini V et al. (2015) Long term follow-up of GRMD dogs transplanted with human adipose derived stem cells. Neuromuscul Disord 25(suppl 2):S290.).

Acknowledgments

This work would not have been possible without the financial support of FAPESP, CNPq, INCT, FINEP, ABDIM/AACD, Duchenne research fund and the University of Sao Paulo. We are extremely grateful to all students and the team at the Human Genome and stem-cell center, and the Genedog kennel, who participated in these studies. Special thanks go to thousands of patients and their families for their great support and encouragement, and for showing us what is really important in life.

References

  • ACMG (American College of Medical Genetics) Board of Directors (2013) Points to consider for informed consent for genome/exome sequencing. Genet Med 15:748-749.
  • Bashir R, Britton S, Strachan T, Keers S, Vafiadaki E, Lako M, Richard I, Marchand S, Bourg N, Argov Z et al. (1998) A gene related to Caenorhabditis elegans spermatogenesis factor fer-1 is mutated in limb-girdle muscular dystrophy type 2B. Nat Genet 20:37-42.
  • Beckmann JS, Richard I, Hillaire D, Broux O, Antignac C, Bois E, Cann H, Cottingham RW Jr., Feingold N, Feingold J et al. (1991) A gene for limb-girdle muscular dystrophy maps to chromosome 15 by linkage. C R Acad Sci III 312:141-148.
  • Bonnemann CG, Passos-Bueno MR, McNally EM, Vainzof M, de Sa Moreira E, Marie SK, Pavanello RC, Noguchi S, Ozawa E, Zatz M et al. (1996) Genomic screening for beta-sarcoglycan gene mutations: Missense mutations may cause severe limb-girdle muscular dystrophy type 2E (LGMD 2E). Hum Mol Genet 5:1953-1961.
  • Bushby K, Bashir R, Keers S, Britton S, Zatz M, Passos-Bueno MR, Lovett M, Mahjneh I, Marconi G and Strachan T (1996) The molecular biology of LGMD2B—towards the identification of the LGMD gene on chromosome 2p13. Neuromuscul Disord 6:491-492.
  • Caplan AI and Sorrell JM (2015) The MSC curtain that stops the immune system. Immunol Lett. 168:136-139.
  • Castro-Gago M (2015) Milder course in Duchenne patients with nonsense mutations and no muscle dystrophin. Neuromuscul Disord 25:443.
  • Chong JX, Buckingham KJ, Jhangiani SN, Boehm C, Sobreira N, Smith JD, Harrell TM, McMillin MJ, Wiszniewski W, Gambin T et al. (2015) The Genetic Basis of Mendelian Phenotypes: Discoveries, Challenges, and Opportunities. Am J Hum Genet 7:199-215.
  • Coatti GC, Beccari MS, Olavio TR, Mitne-Neto M, Okamoto OK and Zatz M (2015) Stem cells for amyotrophic lateral sclerosis modeling and therapy: Myth or fact? Cytometry A 87:197-211.
  • Cohn RD and Dubowitz V (2016) Duchenne muscular dystrophy: Ringo to the rescue? Neuromuscul Disord 26:5-6.
  • Davies KE and Grounds MD (2006) Treating muscular dystrophy with stem cells? Cell 127:1304-1306.
  • de Paula F, Vainzof M, Passos-Bueno MR, de Cassia MPR, Matioli SR, Anderson LVB, Nigro V and Zatz M (2002) Clinical variability in calpainopathy: What makes the difference? Eur J Hum Genet 10:825-832.
  • de Paula F, Vieira N, Starling A, Yamamoto LU, Lima B, de Cassia Pavanello R, Vainzof M, Nigro V and Zatz M (2003) Asymptomatic carriers for homozygous novel mutations in the FKRP gene: The other end of the spectrum. Eur J Hum Genet 11:923-930.
  • Di Rocco G, Iachininoto MG, Tritarelli A, Straino S, Zacheo A, Germani A, Crea F and Capogrossi MC (2006) Myogenic potential of adipose-tissue-derived cells. J Cell Sci 119:2945-2952.
  • Doudna JA and Charpentier E (2014) Genome editing. The new frontier of genome engineering with CRISPR-Cas9. Science 346:1258096.
  • Figueiredo T, Melo US, Pessoa AL, Nobrega PR, Kitajima JP, Correa I, Zatz M, Kok F and Santos S (2015) Homozygous missense mutation in MED25 segregates with syndromic intellectual disability in a large consanguineous family. J Med Genet 52:123-127.
  • Funke AD, Esser M, Kruttgen A, Weis J, Mitne-Neto M, Lazar M, Nishimura AL, Sperfeld AD, Trillenberg P, Senderek J et al. (2010) The p.P56S mutation in the VAPB gene is not due to a single founder: The first European case. Clin Genet 77:302-303.
  • Jazedje T, Perin PM, Czeresnia CE, Maluf M, Halpern S, Secco M, Bueno DF, Vieira NM, Zucconi E and Zatz M (2009) Human fallopian tube: A new source of multipotent adult mesenchymal stem cells discarded in surgical procedures. J Transl Med 7:46.
  • Jazedje T, Bueno DF, Almada BV, Caetano H, Czeresnia CE, Perin PM, Halpern S, Maluf M, Evangelista LP, Nisenbaum MG et al. (2012) Human fallopian tube mesenchymal stromal cells enhance bone regeneration in a xenotransplanted model. Stem Cell Rev 8:355-362.
  • Koenig M, Monaco AP and Kunkel LM (1998) The complete sequence of dystrophin predicts a rod-shaped cytoskeletal protein. Cell 53:219-228.
  • Macedo-Souza LI, Kok F, Santos S, Amorim SC, Starling A, Nishimura A, Lezirovitz K, Lino AM and Zatz M (2005) Spastic paraplegia, optic atrophy, and neuropathy is linked to chromosome 11q13. Ann Neurol 57:730-737.
  • Macedo-Souza LI, Kok F, Santos S, Licinio L, Lezirovitz K, Cavacana N, Bueno C, Amorim S, Pessoa A, Graciani Z et al. (2009) Spastic paraplegia, optic atrophy, and neuropathy: New observations, locus refinement, and exclusion of candidate genes. Ann Hum Genet 73:382-387.
  • McNally EM, Duggan D, Gorospe JR, Bonnemann CG, Fanin M, Pegoraro E, Lidov HG, Noguchi S, Ozawa E, Finkel RS et al. (1996a) Mutations that disrupt the carboxyl-terminus of gamma-sarcoglycan cause muscular dystrophy. Hum Mol Genet 59:1841-1847.
  • McNally EM, Passos-Bueno MR, Bonnemann CG, Vainzof M, de Sa Moreira E, Lidov HG, Othmane KB, Denton PH, Vance JM, Zatz M et al. (1996b) Mild and severe muscular dystrophy caused by a single gamma-sarcoglycan mutation. Am J Hum Genet 59:1040-1047.
  • Melo US, Macedo-Souza LI, Figueiredo T, Muotri AR, Gleeson JG, Coux G, Armas P, Calcaterra NB, Kitajima JP, Amorim S et al. (2015) Overexpression of KLC2 due to a homozygous deletion in the non-coding region causes SPOAN syndrome. Hum Mol Genet 24:6877-6885.
  • Mitne-Neto M, Kok F, Beetz C, Pessoa A, Bueno C, Graciani Z, Martyn M, Monteiro CB, Mitne G, Hubert P et al. (2007a) A multi-exonic SPG4 duplication underlies sex-dependent penetrance of hereditary spastic paraplegia in a large Brazilian pedigree. Eur J Hum Genet 15:1276-1279.
  • Mitne-Neto M, Ramos CR, Pimenta DC, Luz JS, Nishimura AL, Gonzales FA, Oliveira CC and Zatz M (2007b) A mutation in human VAP-B—MSP domain, present in ALS patients, affects the interaction with other cellular proteins. Protein Expr Purif 55:139-146.
  • Mitne-Neto M, Machado-Costa M, Marchetto MC, Bengtson MH, Joazeiro CA, Tsuda H, Bellen HJ, Silva HC, Oliveira AS, Lazar M et al. (2011) Downregulation of VAPB expression in motor neurons derived from induced pluripotent stem cells of ALS8 patients. Hum Mol Genet 20:3642-3652.
  • Monaco AP, Bertelson CJ, Liechti-Gallati S, Moser H and Kunkel LM (1988) An explanation for the phenotypic differences between patients bearing partial deletions of the DMD locus. Genomics 2:90-95.
  • Moreira ES, Vainzof M, Marie SK, Sertie AL, Zatz M and Passos-Bueno MR (1997) The seventh form of autosomal recessive limb-girdle muscular dystrophy is mapped to 17q11-12. Am J Hum Genet 61:151-159.
  • Moreira ES, Wiltshire TJ, Faulkner G, Nilforoushan A, Vainzof M, Suzuki OT, Valle G, Reeves R, Zatz M, Passos-Bueno MR et al. (2000) Limb-girdle muscular dystrophy type 2G is caused by mutations in the gene encoding the sarcomeric protein telethonin. Nat Genet 24:163-166.
  • Moreira ES, Vainzof M, Suzuki OT, Pavanello RC, Zatz M and Passos-Bueno MR (2003) Genotype-phenotype correlations in 35 Brazilian families with sarcoglycanopathies including the description of three novel mutations. J Med Genet 40:E12.
  • Murphy MB, Moncivais K and Caplan AI (2013) Mesenchymal stem cells: Environmentally responsive therapeutics for regenerative medicine. Exp Mol Med 45:e54.
  • Nigro V, de Sa Moreira E, Piluso G, Vainzof M, Belsito A, Politano L, Puca AA, Passos-Bueno MR and Zatz M (1996) Autosomal recessive limb-girdle muscular dystrophy, LGMD2F, is caused by a mutation in the delta-sarcoglycan gene. Nat Genet 14:195-198.
  • Nishimura AL, Mitne-Neto M, Silva HC, Oliveira JR, Vainzof M and Zatz M (2004a) A novel locus for late onset amyotrophic lateral sclerosis/motor neurone disease variant at 20q13. J Med Genet 41:315-320.
  • Nishimura AL, Mitne-Neto M, Silva HC, Richieri-Costa A, Middleton S, Cascio D, Kok F, Oliveira JR, Gillingwater T, Webb J et al. (2004b) A mutation in the vesicle-trafficking protein VAPB causes late-onset spinal muscular atrophy and amyotrophic lateral sclerosis. Am J Hum Genet 75:822-831.
  • Nishimura AL, Al-Chalabi A and Zatz M (2005) A common founder for amyotrophic lateral sclerosis type 8 (ALS8) in the Brazilian population. Hum Genet 118:499-500.
  • Nitahara-Kasahara Y, Hayashita-Kinoh H, Ohshima-Hosoyama S, Okada H, Wada-Maeda M, Nakamura A, Okada T and Takeda S (2012) Long-term engraftment of multipotent mesenchymal stromal cells that differentiate to form myogenic cells in dogs with Duchenne muscular dystrophy. Mol Ther 20:168-177.
  • Passos-Bueno MR, Vainzof M, Marie SK and Zatz M (1994) Half the dystrophin gene is apparently enough for a mild clinical course: Confirmation of its potential use for gene therapy. Hum Mol Genet 3:919-922.
  • Passos-Bueno MR, Bashir R, Moreira ES, Vainzof M, Marie SK, Vasquez L, Iughetti P, Bakker E, Keers S, Stephenson A et al. (1995) Confirmation of the 2p locus for the mild autosomal recessive limb-girdle muscular dystrophy gene (LGMD2B) in three families allows refinement of the candidate region. Genomics 27:192-195.
  • Passos-Bueno MR, Moreira ES, Vainzof M, Marie SK and Zatz M (1996) Linkage analysis in autosomal recessive limb-girdle muscular dystrophy (AR LGMD) maps a sixth form to 5q33-34 (LGMD2F) and indicates that there is at least one more subtype of AR LGMD. Hum Mol Genet 5:815-820.
  • Pereira MC, Secco M, Suzuki DE, Janjoppi L, Rodini CO, Torres LB, Araujo BH, Cavalheiro EA, Zatz M and Okamoto OK (2011) Contamination of mesenchymal stem-cells with fibroblasts accelerates neurodegeneration in an experimental model of Parkinson's disease. Stem Cell Rev 7:1006-1017.
  • Ricci G, Zatz M and Tupler R (2014) Facioscapulohumeral muscular dystrophy: More complex than it appears. Curr Mol Med [Epub ahead of print].
  • Richard I, Broux O, Allamand V, Fougerousse F, Chiannilkulchai N, Bourg N, Brenguier L, Devaud C, Pasturaud P, Roudaut C et al. (1995) Mutations in the proteolytic enzyme calpain 3 cause limb-girdle muscular dystrophy type 2A. Cell 81:27-40.
  • Richard I, Roudaut C, Saenz A, Pogue R, Grimbergen JE, Anderson LV, Beley C, Cobo AM, de Diego C, Eymard B et al. (1999) Calpainopathy- A survey of mutations and polymorphisms. Am J Hum Genet 64:1524-40.
  • Rocco P, Vainzof M, Froehner SC, Peters MF, Marie SK, Passos-Bueno MR and Zatz M (2000) Brazilian family with pure autosomal dominant spastic paraplegia maps to 8q: Analysis of muscle beta 1 syntrophin. Am J Med Genet 92:122-127.
  • Rouger K, Larcher T, Dubreil L, Deschamps JY, Le Guiner C, Jouvion G, Delorme B, Lieubeau B, Carlus M, Fornasari B et al. (2011) Systemic delivery of allogenic muscle stem cells induces long-term muscle repair and clinical efficacy in Duchenne muscular dystrophy dogs. Am J Pathol 179:2501-2518.
  • Sampaolesi M, Blot S, D'Antona G, Granger N, Tonlorenzi R, Innocenzi A, Mognol P, Thibaud JL, Galvez BG, Barthelemy I et al. (2006) Mesoangioblast stem cells ameliorate muscle function in dystrophic dogs. Nature 444:574-579.
  • Scionti I, Greco F, Ricci G, Govi M, Arashiro P, Vercelli L, Berardinelli A, Angelini C, Antonini G, Cao M et al. (2012) Large-scale population analysis challenges the current criteria for the molecular diagnosis of fascioscapulohumeral muscular dystrophy. Am J Hum Genet 90:628-635.
  • Secco M, Zucconi E, Vieira NM, Fogaca LL, Cerqueira A, Carvalho MD, Jazedje T, Okamoto OK, Muotri AR and Zatz M (2008a) Mesenchymal stem cells from umbilical cord: Do not discard the cord! Neuromuscul Disord 18:17-18.
  • Secco M, Zucconi E, Vieira NM, Fogaca LL, Cerqueira A, Carvalho MD, Jazedje T, Okamoto OK, Muotri AR and Zatz M (2008b) Multipotent stem cells from umbilical cord: Cord is richer than blood! Stem Cells 26:146-150.
  • Secco M, Moreira YB, Zucconi E, Vieira NM, Jazedje T, Muotri AR, Okamoto OK, Verjovski-Almeida S and Zatz M (2009) Gene expression profile of mesenchymal stem cells from paired umbilical cord units: Cord is different from blood. Stem Cell Rev 5:387-401.
  • Sharp NJ, Kornegay JN, Van Camp SD, Herbstreith MH, Secore SL, Kettle S, Hung WY, Constantinou CD, Dykstra MJ, Roses AD et al. (1992) An error in dystrophin mRNA processing in golden retriever muscular dystrophy, an animal homologue of Duchenne muscular dystrophy. Genomics 13:115-121.
  • Spencer MJ, Tidball JG, Anderson LV, Bushby KM, Harris JB, Passos-Bueno MR, Somer H, Vainzof M and Zatz M (1997) Absence of calpain 3 in a form of limb-girdle muscular dystrophy (LGMD2A). J Neurol Sci 146:173-178.
  • Starling A, Rocco P, Cambi F, Hobson GM, Passos Bueno MR and Zatz M (2002a) Further evidence for a fourth gene causing X-linked pure spastic paraplegia. Am J Med Genet 111:152-156.
  • Starling A, Rocco P, Passos-Bueno MR, Hazan J, Marie SK and Zatz M (2002b) Autosomal dominant (AD) pure spastic paraplegia (HSP) linked to locus SPG4 affects almost exclusively males in a large pedigree. J Med Genet 39:e77.
  • Starling A, de Paula F, Silva H, Vainzof M and Zatz M (2003) Calpainopathy: How broad is the spectrum of clinical variability? J Mol Neurosci 21:233-236.
  • Starling A, Kok F, Passos-Bueno MR, Vainzof M and Zatz M (2004) A new form of autosomal dominant limb-girdle muscular dystrophy (LGMD1G) with progressive fingers and toes flexion limitation maps to chromosome 4p21. Eur J Hum Genet 12:1033-1040.
  • Takata RI, Speck Martins CE, Passos-Bueno MR, Abe KT, Nishimura AL, Da Silva MD, Monteiro Jr A, Lima MI, Kok F and Zatz M (2004) A new locus for recessive distal spinal muscular atrophy at Xq13.1-q21. J Med Genet 41:224-229.
  • Takahashi K and Yamanaka S (2006) Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126:663-676.
  • Teuling E, Ahmed S, Haasdijk E, Demmers J, Steinmetz MO, Akhmanova A, Jaarsma D and Hoogenraad CC (2007) Motor neuron disease-associated mutant vesicle-associated membrane protein-associated protein (VAP) B recruits wild-type VAPs into endoplasmic reticulum-derived tubular aggregates. J Neurosci 27:9801-9815.
  • Tonini MM, Passos-Bueno MR, Cerqueira A, Matioli SR, Pavanello R and Zatz M (2004) Asymptomatic carriers and gender differences in facioscapulohumeral muscular dystrophy (FSHD). Neuromuscul Disord 14:33-38.
  • Vainzof M, Pavanello RC, Pavanello Filho I, Passos-Bueno MR, Rapaport D, Hsi CT and Zatz M (1990) Dystrophin immunostaining in muscles from patients with different types of muscular dystrophy: A Brazilian study. J Neurol Sci 98:221-233.
  • Vainzof M, Pavanello RC, Pavanello-Filho I, Rapaport D, Passos-Bueno MR, Zubrzycka-Gaarn EE, Bulman DE and Zatz M (1991a) Screening of male patients with autosomal recessive Duchenne dystrophy through dystrophin and DNA studies. Am J Med Genet 39:38-41.
  • Vainzof M, Pavanello RC, Pavanello I, Tsanaclis AM, Levy JA, Passos-Bueno MR, Rapaport D and Zatz M (1991b) Dystrophin immunofluorescence pattern in manifesting and asymptomatic carriers of Duchenne's and Becker muscular dystrophies of different ages. Neuromuscul Disord 1:177-183.
  • Vainzof M, Zubrzycka-Gaarn EE, Rapaport D, Passos-Bueno MR, Pavanello RC, Pavanello-Filho I and Zatz M (1991c) Immunofluorescence dystrophin study in Duchenne dystrophy through the concomitant use of two antibodies directed against the carboxy-terminal and the amino-terminal region of the protein. J Neurol Sci 101:141-147.
  • Vainzof M, Takata RI, Passos-Bueno MR, Pavanello RC and Zatz M (1993) Is the maintainance of the C-terminus domain of dystrophin enough to ensure a milder Becker muscular dystrophy phenotype? Hum Mol Genet 2:39-42.
  • Vainzof M, Passos-Bueno MR, Man N and Zatz M (1995a) Absence of correlation between utrophin localization and quantity and the clinical severity in Duchenne/Becker dystrophies. Am J Med Genet 58:305-309.
  • Vainzof M, Passos-Bueno MR, Pavanello RC and Zatz M (1995b) Is dystrophin always altered in Becker muscular dystrophy patients? J Neurol Sci 131:99-104.
  • Vainzof M, Passos-Bueno MR, Canovas M, Moreira ES, Pavanello RC, Marie SK, Anderson LV, Bonnemann CG, McNally EM, Nigro V et al. (1996) The sarcoglycan complex in the six autosomal recessive limb-girdle muscular dystrophies. Hum Mol Genet 5:1963-1969.
  • Vainzof M, Passos-Bueno MR, Pavanello RC, Marie SK, Oliveira AS and Zatz M (1999) Sarcoglycanopathies are responsible for 68% of severe autosomal recessive limb-girdle muscular dystrophy in the Brazilian population. J Neurol Sci 164:44-49.
  • Vainzof M, Moreira ES, Suzuki OT, Faulkner G, Valle G, Beggs AH, Carpen O, Ribeiro AF, Zanoteli E, Gurgel-Gianneti J et al. (2002) Telethonin protein expression in neuromuscular disorders. Biochim Biophys Acta 1588:33-40.
  • Vainzof M, Richard P, Herrmann R, Jimenez-Mallebrera C, Talim B, Yamamoto LU, Ledeuil C, Mein R, Abbs S, Brockington M et al. (2005) Prenatal diagnosis in laminin alpha2 chain (merosin)-deficient congenital muscular dystrophy: A collective experience of five international centers. Neuromuscul Disord 15:588-594.
  • Vainzof M, Feitosa L, Ayub-Guerrieri D, Canovas M, Pavanello RCM and Zatz M (2016) Mild course in atypical Duchenne muscular dystrophy patients is not caused by utrophin overexpression. Neuromuscul. Disord 26:197-200.
  • Valadares MC, Gomes JP, Castello G, Assoni A, Pellati M, Bueno C, Corselli M, Silva H, Bartolini P, Vainzof M et al. (2014) Human adipose tissue derived pericytes increase life span in Utrn (tm1Ked) Dmd (mdx) /J mice. Stem Cell Rev 10:830-840.
  • Valdmanis PN, Meijer IA, Reynolds A, Lei A, MacLeod P, Schlesinger D, Zatz M, Reid E, Dion PA, Drapeau P et al. (2007) Mutations in the KIAA0196 gene at the SPG8 locus cause hereditary spastic paraplegia. Am J Hum Genet 80:152-161.
  • Valle G, Faulkner G, De Antoni A, Pacchioni B, Pallavicini A, Pandolfo D, Tiso N, Toppo S, Trevisan S and Lanfranchi G (1997) Telethonin, a novel sarcomeric protein of heart and skeletal muscle. FEBS Lett 415:163-168.
  • Vieira NM, Bueno Jr CR, Brandalise V, Moraes LV, Zucconi E, Secco M, Suzuki MF, Camargo MM, Bartolini P, Brum PC et al. (2008) SJL dystrophic mice express a significant amount of human muscle proteins following systemic delivery of human adipose-derived stromal cells without immunosuppression. Stem Cells 26:2391-2398.
  • Vieira NM, Zucconi E, Bueno Jr CR, Secco M, Suzuki MF, Bartolini P, Vainzof M and Zatz M (2010) Human multipotent mesenchymal stromal cells from distinct sources show different in vivo potential to differentiate into muscle cells when injected in dystrophic mice. Stem Cell Rev 6:560-566.
  • Vieira NM, Valadares M, Zucconi E, Secco M, Bueno Jr. CR, Brandalise V, Assoni A, Gomes J, Landini V, Andrade T et al. (2012) Human adipose-derived mesenchymal stromal cells injected systemically into GRMD dogs without immunosuppression are able to reach the host muscle and express human dystrophin. Cell Transplant 21:1407-1417.
  • Vieira NM, Naslavsky MS, Licinio L, Kok F, Schlesinger D, Vainzof M, Sanchez N, Kitajima JP, Gal L, Cavacana N et al. (2014) A defect in the RNA-processing protein HNRPDL causes limb-girdle muscular dystrophy 1G (LGMD1G). Hum Mol Genet 23:4103-4110.
  • Vieira NM, Elvers I, Alexander MS, Moreira YB, Eran A, Gomes JP, Marshall JL, Karlsson EK, Verjovski-Almeida S, Lindblad-Toh K et al. (2015) Jagged 1 rescues the Duchenne Muscular Dystrophy phenotype. Cell 163:1204-1213.
  • Yamamoto LU, Gollop TR, Naccache NF, Pavanello RC, Zanoteli E, Zatz M and Vainzof M (2004) Protein and DNA analysis for the prenatal diagnosis of alpha2-laminin-deficient congenital muscular dystrophy. Diagn Mol Pathol 13:167-171.
  • Yamamoto LU, Velloso FJ, Lima BL, Fogaca LL, de Paula F, Vieira NM, Zatz M and Vainzof M (2008) Muscle protein alterations in LGMD2I patients with different mutations in the Fukutin-related protein gene. J Histochem Cytochem 56:995-1001.
  • Yoshida M and Ozawa E (1990) Glycoprotein complex anchoring dystrophin to sarcolemma. J Biochem 108:748-52.
  • Zatz M (2005) Global voices of science. When science is not enough: Fighting genetic disease in Brazil. Science 308:55-57.
  • Zatz M (2015) Response to: Milder course in Duchenne patients with nonsense mutations and no muscle dystrophin. Neuromuscul Disord 25:444.
  • Zatz M, Vianna-Morgante AM, Campos P and Diament AJ (1981) Translocation (X;6) in a female with Duchenne muscular dystrophy: Implications for the localisation of the DMD locus. J Med Genet 18:442-447.
  • Zatz M, Vainzof M and Passos-Bueno MR (2000) Limb-girdle muscular dystrophy: One gene with different phenotypes, one phenotype with different genes. Curr Opin Neurol 13:511-517.
  • Zatz M, de Paula F, Starling A and Vainzof M (2003) The 10 autosomal recessive limb-girdle muscular dystrophies. Neuromuscul Disord 13:532-544.
  • Zatz M, Pavanello R de C, Lourenco NC, Cerqueira A, Lazar M and Vainzof M (2012) Assessing pathogenicity for novel mutation/sequence variants: The value of healthy older individuals. Neuromolecular Med 14:281-284.
  • Zatz M, Pavanello RC, Lazar M, Yamamoto GL, Lourenco NC, Cerqueira A, Nogueira L and Vainzof M (2014) Milder course in Duchenne patients with nonsense mutations and no muscle dystrophin. Neuromuscul Disord 24:986-989.
  • Zatz M, Gomes J, Pelatti M, Secco M, Vieira N, Zucconi E, Vainzof M, Landini V et al. (2015) Long term follow-up of GRMD dogs transplanted with human adipose derived stem cells. Neuromuscul Disord 25(suppl 2):S290.
  • Zucconi E, Valadares MC, Vieira NM, Bueno Jr CR, Secco M, Jazedje T, da Silva HC, Vainzof M and Zatz M (2010) Ringo: Discordance between the molecular and clinical manifestation in a golden retriever muscular dystrophy dog. Neuromuscul Disord 20:64-70.
  • Associate Editor: Francisco Mauro Salzano

Publication Dates

  • Publication in this collection
    Jul-Sep 2016

History

  • Received
    28 Jan 2016
  • Accepted
    29 Mar 2016
Sociedade Brasileira de Genética Rua Cap. Adelmio Norberto da Silva, 736, 14025-670 Ribeirão Preto SP Brazil, Tel.: (55 16) 3911-4130 / Fax.: (55 16) 3621-3552 - Ribeirão Preto - SP - Brazil
E-mail: editor@gmb.org.br