touchNEUROLOGY touchNEUROLOGY
Movement Disorders
Read Time: 6 mins

Understanding Multiple System Atrophy— Could Genetics Lead the Way?

Copy Link
Published Online: Oct 20th 2016 US Neurology, 2016;12(2):110–2 DOI: https://doi.org/10.17925/USN.2016.12.02.110
Authors: Miriam Sklerov, Cheryl Waters
Quick Links:
Abstract
Article
Article Information
Abstract:
Overview

Multiple system atrophy (MSA) is a progressive, adult-onset, neurodegenerative disorder characterized by parkinsonism, cerebellar ataxia, autonomic failure, and corticospinal tract dysfunction. It is considered a rare disease, similar in frequency to the more well-known neurodegenerative disease amyotrophic lateral sclerosis or Lou Gehrig’s disease. Though MSA has not traditionally been considered a genetic disease, new evidence is emerging supporting some genetic predisposition in many patients. In this short review, we will discuss advances in the knowledge of genetics in MSA and how this has furthered our understanding of the pathophysiology of the disease. There is still much unknown about this disease, but some of the recent advances made in MSA genetics may give important clues to understanding the pathogenesis and treatment of this disease.

Keywords

Multiple System Atrophy, Genetics, Parkinsonism

Article:

Multiple system atrophy (MSA) is a is an adult-onset, sporadic, progressive neurodegenerative disease characterized by a varying combination of parkinsonism, cerebellar ataxia, autonomic failure, and corticospinal dysfunction. Patients either have a predominance of parkinsonian symptoms (MSA-P) such that they are often misdiagnosed with Parkinson’s disease, or a predominance of cerebellar ataxia (MSA-C). Onset is usually in the 6th and 7th decades of life. The prevalence of MSA is roughly 3–4 per 100,000 in the US.1 The disease progression tends to be faster than that of Parkinson’s disease, with an average disease duration of 8–10 years from onset of symptoms to demise. MSA is neurologically devastating and quickly leaves patients unable walk, speak, and take care of themselves. Patients suffering from MSA often suffer from orthostatic hypotension that can be debilitating, urinary retention requiring catheterization, and various breathing problems. The cause of death in MSA is usually related to dysphagia, respiratory problems, or cardiovascular complications of dysautonomia. There is a very limited arsenal of medications that can be used to treat this disease. Currently available treatments are symptomatic, and include medications to treat parkinsonian symptoms, orthostatic hypotension, urinary symptoms, and sexual dysfunction. Patients typically get brief and modest responses to these medications. There are no treatments available to slow down or stop the progression of MSA.

There are no objective, quantitative tests to reliably make or confirm this diagnosis in living patients. The diagnosis in this disease, as in many movement disorders, is clinical – i.e., it is made based on history, examination, sometimes using supplementary information such as the results of a magnetic resonance image (MRI) of the brain, iodine-123-metaiodobenzylguanidine (MIBG) nuclear medicine scan,2,3 serum norepinephrine levels, or testing of the autonomic nervous system.4 The only certain diagnosis is made with brain autopsy, which shows accumulation of the protein α-synuclein within glial cells in the brain and degeneration in the striatum and substantia nigra, and/or degeneration in the olivoponto- cerebellar regions of the brain. The lack of highly sensitive and specific confirmatory tests for the diagnosis of MSA, in addition to the rarity of the disease, have been major factors that have made research in MSA difficult.

More recently there have been some advances in laboratory and translational research in MSA. In particular, genetics of MSA has been a recent topic of interest, and is opening new doors in the understanding of the pathogenesis of MSA. In this short review, we will discuss several past and recent advances in the knowledge of genetics in MSA and how this has furthered our understanding of the pathophysiology of the disease and paved the way for novel therapeutic options.

Genetics of multiple system atrophy
Traditionally, most adult-onset neurodegenerative diseases have been considered sporadic rather than genetic in origin. However, more evidence is emerging that many neurodegenerative diseases may have not only a Mendelian pattern of inheritance, but also genes that strongly predispose to the condition. Examples of this include the ApoE gene for Alzheimer’s disease and GBA in Parkinson disease. There have been several multiplex families with MSA in the scientific literature,5–7 though a candidate gene has only been identified in two of these families. A large genome-wide association study is currently underway; however, few studies have attempted such modern approaches to analyzing genetic predispositions in MSA. Genetics research in MSA is clearly lagging behind that of other α-synucleinopathies (Parkinson’s disease and dementia with Lewy bodies [DLB]), though recently a few candidate genes have given researchers clues as to the etiology of MSA.

Alpha-synuclein gene
Mutations and duplications in the α-synuclein (SNCA) gene have been clearly linked with α-synucleinopathies. The clinical syndromes and pathologic findings in several patients with known mutations or duplications have shown significant overlap with MSA,8 including the finding of frequent glial cytoplasmic inclusions on pathologic examination. Some of the families described with SNCA mutations had members with a clinical picture consistent with a diagnosis of MSA. A few studies found significant associations between variants in the SNCA gene and sporadic cases of clinical MSA9,10 though there is some controversy about the generalizability of this finding in Asian populations.11,12 These findings were supported by the discovery of α-synuclein mRNA overexpression in the brains of patients with MSA.13 This supports a hypothesis that the toxicity of α-synuclein plays an important role in the pathogenesis of this disease, and may help to pave the way for therapeutic options.

Glucocerebrosidase gene
Glucocerebrosidase (GCase), encoded by the Glucocerebrosidase gene (GBA), is an enzyme involved in the lysosomal degradation of sphingolipids.14 Homozygous mutations of this gene result in Gaucher’s disease (GD), a rare lysosomal storage disease that is particularly prevalent in certain populations, such as the Ashkenazi Jewish population. Parkinson’s disease and DLB patients are roughly 5–10-fold15 and 8 fold16 (respectively) more likely to carry GBA mutations than the general population. These genetic findings have been followed up with findings of reduced GCase activity in the brains17 and peripheral blood18 of patients with Parkinson‘s disease. The prevalence of GBA mutations among patients with MSA has been studied by a few groups in recent years, and has yielded varied results.19–25

Six studies have found no association between GBA mutations and MSA. The most recent and largest study included 969 clinically diagnosed MSA patients and 1,509 controls from Japan, several sites in Europe, and several sites in North America.21 They found a significantly increased odds ratio for GBA variants in MSA patients compared to controls. In fact, two patients with MSA had homozygous mutations in the GBA gene without a known clinical diagnosis of GD, while none of the controls had homozygous or compound heterozygous mutations. This study certainly indicates that further study of the relationship between GBA and MSA is required. It implies that genetic testing for GD may be indicated in certain populations with MSA, and introduces another area for research into therapeutic options for MSA.

Coenzyme Q2 gene
Recently, four multiplex MSA families were studied with whole exome and subsequent linkage analysis in the attempt at identifying a candidate genetic locus for MSA.26 In two of four Japanese families, the researchers detected mutations in the coenzyme Q2 (CoQ2) gene, which plays a role in the synthetic pathway for CoQ10. They followed this with genetic analysis of 726 Japanese patients with sporadic MSA and 1040 Japanese controls, which revealed 35 CoQ2 mutations among MSA patients and 17 among controls (OR=3.05, p=0.00015). An association between sporadic MSA and CoQ2 variants was found in Japanese, Taiwanese and Chinese populations, but these results were not replicated in other cohorts.26–31 Further studies have revealed reduction in CoQ10 levels in the cerebellum and serum of patients with MSA.32,33 These findings may be particularly important in implicating CoQ10 supplementation as a therapeutic option in MSA, though this has not yet been systematically studied.

Other implicated genes
A hexanucleotide expansion in the C9orf72 gene located on chromosome 9 has been found to be present in patients with familial forms of frontotemporal dementia and amyotrophic lateral sclerosis (ALS). There is also a report of clinically diagnosed MSA with C9orf72 expansion in a family with ALS.34 Though C9orf72 expansions are associated with parkinsonian symptoms and syndromes, this is the only report of suspected MSA with C9orf72. Subsequent studies, including a series of 100 pathologically confirmed MSA cases,35 have failed to produce a link between this expansion and MSA.

Microtubule-associated protein tau (MAPT) gene has been associated with frontotemporal dementia and ALS phenotypes as well as parkinsonism. There does not appear to be an association between mutations in this gene and MSA, however this has not been extensively studied.36

Mutations in the Leucine-rich repeat kinase 2 (LRRK2) have been investigated for a causal link with MSA. Most studies have not found an association between LRRK2 mutations and MSA.37,38 However one large series from the US and UK found a protective effect of LRRK2 variants on MSA.39

Genetic mutations in the spinocerebellar ataxia family (SCA) are associated with varying degrees of ataxia, parkinsonism, dystonia, and even autonomic dysfunction. Given the clinical overlap with symptoms seen in MSA, it can be difficult to distinguish SCA from MSA. Patients with SCA syndromes typically present earlier in life than MSA, are usually slower in progression, and will have a family history. In clinical practice, it is often advisable to test for SCA mutations when evaluating patients presenting with adult-onset cerebellar ataxia, with or without a known family history.40 Most studies looking at the association between clinically diagnosed MSA and mutations in the SCA genes have focused on the utility of SCA genetic testing in patients presenting clinically with MSA. There are no large pathologic series, to our knowledge, of MSA that have been tested for mutations in SCA genes. However, there is one case report of a patient with adult-onset cerebellar ataxia with dysautonomia and parkinsonism who was clinically diagnosed with MSA, who subsequently underwent testing for SCA mutations and was found to have an SCA-3 mutation. On autopsy, this patient had findings typical of MSA. This finding indicates that further studies are required to fully analyze this association.

Conclusion
MSA is not generally considered a genetic disease, and in fact only rarely has been described in families. More recent efforts in the field of MSA genetics have revealed several candidate genes that may be involved in the pathogenesis of the disease. The pathogenesis of MSA remains an enigma, though it is becoming clear that genetic associations exist. Many mechanisms for the development and propagation of MSA have been postulated, including impaired elimination of α-synuclein within the cell,41–44 mitochondrial dysfunction,32,33,45,46 direct toxicity of α-synuclein,47 oxidative stress,48 and neuroinflammation.49,50 More recently, there are also data that implicate a prion-like propagation of MSA.51 We are hopeful that genetic associations may give us further clues into the pathogenesis of MSA, as well as targets for therapeutic interventions. For example, if GBA mutations confer a higher risk of developing MSA, it follows that treatment with enzyme replacement therapy may offer some benefit to patients who have MSA, with or without GBA mutations. It is clear that none of the genetic associations discussed above confer a high penetrance for development of MSA; rather they likely create a pre-disposition for this rare and devastating disease. With advances in the knowledge of genetic contributions to MSA we may find clues to the understanding and treatment of this disease.

Article Information:
Disclosure

Miriam Sklerov was supported by NIH/NINDS T32 NS07153, under funding. Cheryl Waters has nothing to disclose in relation to this article. No funding was received for the publication of this article. This article involves a review of the literature and did not involve any studies with human or animal subjects performed by any of the authors.

Correspondence

Cheryl Waters, 710 West 168th street, 3rd floor, New York, NY 10032. E: cw345@cumc.columbia.edu

Access

This article is published under the Creative Commons Attribution Noncommercial License, which permits any noncommercial use, distribution, adaptation, and reproduction provided the original author(s) and source are given appropriate credit.

Received

2016-06-21T00:00:00

References

1. Bower JH, Maraganore DM, McDonnell SK, Rocca WA, Incidence of progressive supranuclear palsy and multiple system atrophy in Olmsted County, Minnesota, 1976 to 1990, Neurology, 1997;49:1284–8.
2. Braune S, Reinhardt M, Schnitzer R, et al., Cardiac uptake of [123I]MIBG separates Parkinson's disease from multiple system atrophy, Neurology, 1999;53:1020–5.
3. Courbon F, Brefel-Courbon C, Thalamas C, et al., Cardiac MIBG scintigraphy is a sensitive tool for detecting cardiac sympathetic denervation in Parkinson's disease, Mov Disord, 2003;18:890–7.
4. Gilman S, Wenning GK, Low PA, et al., Second consensus statement on the diagnosis of multiple system atrophy, Neurology, 2008;71:670–6.
5. Hara K, Momose Y, Tokiguchi S, et al., Multiplex families with multiple system atrophy, Arch Neurol, 2007;64:545–51.
6. Wullner U, Schmitt I, Kammal M, et al., Definite multiple system atrophy in a German family, Journal of Neurology, Neurosurgery, and Psychiatry, 2009;80:449–50.
7. Hohler AD, Singh VJ, Probable hereditary multiple system atrophy-autonomic (MSA-A) in a family in the United States, J Clin Neurosci, 2012;19:479–80.
8. Ahmed Z, Asi YT, Sailer A, et al., The neuropathology, pathophysiology and genetics of multiple system atrophy, Neuropathol Appl Neurobiol, 2012;38:4–24.
9. Al-Chalabi A, Durr A, Wood NW, et al., Genetic variants of the alpha-synuclein gene SNCA are associated with multiple system atrophy, PloS One, 2009;4:e7114.
10. Scholz SW, Houlden H, Schulte C, et al., SNCA variants are associated with increased risk for multiple system atrophy, Ann Neurol, 2009;65:610–4.
11. Ozawa T, Takano H, O Onodera, et al., No mutation in the entire coding region of the alpha-synuclein gene in pathologically confirmed cases of multiple system atrophy, Neurosci Lett, 1999:110–2.
12. Yun JY, Lee WW, Lee JY, et al., SNCA variants and multiple system atrophy, Ann Neurol, 2010;67:554–5.
13. Asi YT, Simpson JE, Heath PR, et al., Alpha-synuclein mRNA expression in oligodendrocytes in MSA, Glia, 2014;62:964–70.
14. Grabowski GA, Gaucher disease. Enzymology, genetics, and treatment, Adv Hum Genet, 1993;21:377–441.
15. Zhao F, Bi L, Wang W, et al., Mutations of glucocerebrosidase gene and susceptibility to Parkinson's disease: An updated meta-analysis in a European population, Neuroscience, 2016;320:239–46.
16. Nalls MA, Duran R, Lopez G, et al., A multicenter study of glucocerebrosidase mutations in dementia with Lewy bodies, JAMA Neurol, 2013;70:727–35.
17. Gegg ME, Burke D, Heales SJ, et al., Glucocerebrosidase deficiency in substantia nigra of parkinson disease brains, Ann Neurol, 2012;72:455–63.
18. Alcalay RN, Levy OA, Waters CC, et al., Glucocerebrosidase activity in Parkinson's disease with and without GBA mutations, Brain, 2015;138:2648–58.
19. Eblan MJ, Nguyen J, Ziegler SG, et al., Glucocerebrosidase mutations are also found in subjects with early-onset parkinsonism from Venezuela, Mov Disord, 2006;21:282–3.
20. Jamrozik Z, Lugowska A, Slawek J, Kwiecinski H, Glucocerebrosidase mutations p.L444P and p.N370S are not associated with multisystem atrophy, progressive supranuclear palsy and corticobasal degeneration in Polish patients, J Neurol, 2010;257:459–60.
21. Mitsui J, Matsukawa T, Sasaki H, et al., Variants associated with Gaucher disease in multiple system atrophy, Ann Clin Transl Neur, 2015;2:417–26.
22. Nishioka K, Ross OA, Vilarino-Guell C, et al., Glucocerebrosidase mutations in diffuse Lewy body disease, Parkinsonism Relat Disord, 2011;17:55–7.
23. Segarane B, Li A, Paudel R, et al., Glucocerebrosidase Mutations in 108 Neuropathologically Confirmed Cases of Multiple System Atrophy, Neurology, 2009;72:1185–6.
24. Srulijes K, Hauser AK, Guella I, et al., No association of GBA mutations and multiple system atrophy, Eur J Neurol, 2013;20:e61–e62.
25. Sun QY, Guo JF, Han WW, et al., Genetic association study of glucocerebrosidase gene L444P mutation in essential tremor and multiple system atrophy in mainland China, J Clin Neurosci, 2013;20:217–9.
26. Multiple-System Atrophy Research Collaboration, Mutations in COQ2 in familial and sporadic multiple-system atrophy, N Engl J Med, 2013;369:233–244.
27. Chen YP, Zhao B, Cao B, et al., Mutation scanning of the COQ2 gene in ethnic Chinese patients with multiple-system atrophy, Neurobiol Aging, 2015;36:1222 e7–11.
28. Lin CH, Tan EK, Yang CC, et al., COQ2 gene variants associate with cerebellar subtype of multiple system atrophy in Chinese, Mov Disord, 2015;30:436–7.
29. Sun Z, Ohta Y, Yamashita T, et al., New susceptible variant of COQ2 gene in Japanese patients with sporadic multiple system atrophy, Neurol Genet, 2016;2:e54.
30. Wen XD, Li HF, Wang HX, et al., Mutation Analysis of COQ2 in Chinese Patients with Cerebellar Subtype of Multiple System Atrophy, CNS Neurosci Ther, 2015;21:626–30.
31. Zhao Q, Yang X, Tian S, et al., Association of the COQ2 V393A variant with risk of multiple system atrophy in East Asians: a case-control study and meta-analysis of the literature, Neurol Sci, 2016;37:423–30.
32. Kasai T, Tokuda T, Ohmichi T, et al., Serum Levels of Coenzyme Q10 in Patients with Multiple System Atrophy, PloS One, 2016;11: e0147574.
33. Schottlaender LV, Bettencourt C, Kiely AP, et al., Coenzyme Q10 Levels Are Decreased in the Cerebellum of Multiple-System Atrophy Patients, PloS One, 2016;11:e0149557.
34. Goldman JS, Quinzii C, Dunning-Broadbent J, et al., Multiple system atrophy and amyotrophic lateral sclerosis in a family with hexanucleotide repeat expansions in C9orf72, JAMA Neurol, 2014;71:771–4.
35. Scholz SW, Majounie E, Revesz T, et al., Multiple system atrophy is not caused by C9orf72 hexanucleotide repeat expansions, Neurobiol Aging, 2015;36:1223 e1–2.
36. Chen Y, Cao B, Ou R, et al., Association analysis of the GRN rs5848 and MAPT rs242557 polymorphisms in Parkinson's disease and multiple system atrophy: a large-scale population-based study and meta-analysis, Int J Neurosci, 2015;126: 947–54.
37. Fujioka S, Ogaki K, Tacik PM, et al., Update on novel familial forms of Parkinson's disease and multiple system atrophy, Parkinsonism Relat Disord, 2014;20 Suppl 1:S29–34.
38. Lu CS, Chang HC, Weng YH, et al., Analysis of the LRRK2 Gly2385Arg variant in primary dystonia and multiple system atrophy in Taiwan, Parkinsonism Relat Disord, 2008;14:393–6.
39. Heckman MG, Schottlaender L, Soto-Ortolaza AI, et al., LRRK2 exonic variants and risk of multiple system atrophy, Neurology, 2014;83:2256–61.
40. Doherty KM, Warner TT, Lees AJ, Late onset ataxia: MSA-C or SCA 17? A gene penetrance dilemma, Mov Disord, 2014;29:36–8.
41. Makioka K, Yamazaki T, Takatama M, et al., Activation and alteration of lysosomes in multiple system atrophy, Neuroreport, 2012;23:270–6.
42. Pukass K, Richter-Landsberg C, Inhibition of UCH-L1 in oligodendroglial cells results in microtubule stabilization and prevents alpha-synuclein aggregate formation by activating the autophagic pathway: implications for multiple system atrophy, Front Cell Neurosci, 2015;9:163.
43. Stefanova N, Kaufmann WA, Humpel C, et al., Systemic proteasome inhibition triggers neurodegeneration in a transgenic mouse model expressing human alpha-synuclein under oligodendrocyte promoter: implications for multiple system atrophy, Acta Neuropathol, 2012;124:51–65.
44. Tanji K, Odagiri S, Maruyama A, et al., Alteration of autophagosomal proteins in the brain of multiple system atrophy, Neurobiol Dis, 2013;49:190–8.
45. Mitsui J, Matsukawa T, Ishiura H, et al., Mutations in COQ2 in Familial and Sporadic Multiple-System Atrophy, New Engl J Med, 2013;369:233–44.
46. Ubhi K, Rockenstein E, Kragh C, et al., Widespread microRNA dysregulation in multiple system atrophy - disease-related alteration in miR-96, Eur J Neurosci, 2014;39:1026–41.
47. Scholz SW, Houlden H, Schulte C, et al., SNCA Variants Are Associated with Increased Risk for Multiple System Atrophy, Ann Neurol, 2009;65:610–4.
48. Stefanova N, Reindl M, Neumann M, et al., Oxidative stress in transgenic mice with oligodendroglial alpha-synuclein overexpression replicates the neuropathology of multiple system atrophy, Am J Pathol, 2005;166:869–76.
49. Radford R, Rcom-H'cheo-Gauthier A, Wong MB, et al., The degree of astrocyte activation in multiple system atrophy is inversely proportional to the distance to alpha-synuclein inclusions, Mol Cell Neurosci, 2015;65:68–81.
50. Valera E, Masliah E, Immunotherapy for neurodegenerative diseases: Focus on alpha-synucleinopathies, Pharmacol Ther, 2013;138:311–22.
51. Prusiner SB, Woerman AL, Mordes DA, et al., Evidence for alphasynuclein prions causing multiple system atrophy in humans with parkinsonism, P Natl Acad Sci USA, 2015;112:E5308–E17.

Further Resources

Share this Article
Related Content In Movement Disorders
  • Copied to clipboard!
    accredited arrow-down-editablearrow-downarrow_leftarrow-right-bluearrow-right-dark-bluearrow-right-greenarrow-right-greyarrow-right-orangearrow-right-whitearrow-right-bluearrow-up-orangeavatarcalendarchevron-down consultant-pathologist-nurseconsultant-pathologistcrosscrossdownloademailexclaimationfeedbackfiltergraph-arrowinterviewslinkmdt_iconmenumore_dots nurse-consultantpadlock patient-advocate-pathologistpatient-consultantpatientperson pharmacist-nurseplay_buttonplay-colour-tmcplay-colourAsset 1podcastprinter scenerysearch share single-doctor social_facebooksocial_googleplussocial_instagramsocial_linkedin_altsocial_linkedin_altsocial_pinterestlogo-twitter-glyph-32social_youtubeshape-star (1)tick-bluetick-orangetick-red tick-whiteticktimetranscriptup-arrowwebinar Sponsored Department Location NEW TMM Corporate Services Icons-07NEW TMM Corporate Services Icons-08NEW TMM Corporate Services Icons-09NEW TMM Corporate Services Icons-10NEW TMM Corporate Services Icons-11NEW TMM Corporate Services Icons-12Salary £ TMM-Corp-Site-Icons-01TMM-Corp-Site-Icons-02TMM-Corp-Site-Icons-03TMM-Corp-Site-Icons-04TMM-Corp-Site-Icons-05TMM-Corp-Site-Icons-06TMM-Corp-Site-Icons-07TMM-Corp-Site-Icons-08TMM-Corp-Site-Icons-09TMM-Corp-Site-Icons-10TMM-Corp-Site-Icons-11TMM-Corp-Site-Icons-12TMM-Corp-Site-Icons-13TMM-Corp-Site-Icons-14TMM-Corp-Site-Icons-15TMM-Corp-Site-Icons-16TMM-Corp-Site-Icons-17TMM-Corp-Site-Icons-18TMM-Corp-Site-Icons-19TMM-Corp-Site-Icons-20TMM-Corp-Site-Icons-21TMM-Corp-Site-Icons-22TMM-Corp-Site-Icons-23TMM-Corp-Site-Icons-24TMM-Corp-Site-Icons-25TMM-Corp-Site-Icons-26TMM-Corp-Site-Icons-27TMM-Corp-Site-Icons-28TMM-Corp-Site-Icons-29TMM-Corp-Site-Icons-30TMM-Corp-Site-Icons-31TMM-Corp-Site-Icons-32TMM-Corp-Site-Icons-33TMM-Corp-Site-Icons-34TMM-Corp-Site-Icons-35TMM-Corp-Site-Icons-36TMM-Corp-Site-Icons-37TMM-Corp-Site-Icons-38TMM-Corp-Site-Icons-39TMM-Corp-Site-Icons-40TMM-Corp-Site-Icons-41TMM-Corp-Site-Icons-42TMM-Corp-Site-Icons-43TMM-Corp-Site-Icons-44TMM-Corp-Site-Icons-45TMM-Corp-Site-Icons-46TMM-Corp-Site-Icons-47TMM-Corp-Site-Icons-48TMM-Corp-Site-Icons-49TMM-Corp-Site-Icons-50TMM-Corp-Site-Icons-51TMM-Corp-Site-Icons-52TMM-Corp-Site-Icons-53TMM-Corp-Site-Icons-54TMM-Corp-Site-Icons-55TMM-Corp-Site-Icons-56TMM-Corp-Site-Icons-57TMM-Corp-Site-Icons-58TMM-Corp-Site-Icons-59TMM-Corp-Site-Icons-60TMM-Corp-Site-Icons-61TMM-Corp-Site-Icons-62TMM-Corp-Site-Icons-63TMM-Corp-Site-Icons-64TMM-Corp-Site-Icons-65TMM-Corp-Site-Icons-66TMM-Corp-Site-Icons-67TMM-Corp-Site-Icons-68TMM-Corp-Site-Icons-69TMM-Corp-Site-Icons-70TMM-Corp-Site-Icons-71TMM-Corp-Site-Icons-72