Skip to main content
Top
Published in: Orphanet Journal of Rare Diseases 1/2018

Open Access 01-12-2018 | Research

Hypermanganesemia due to mutations in SLC39A14: further insights into Mn deposition in the central nervous system

Authors: L. Marti-Sanchez, J. D. Ortigoza-Escobar, A. Darling, M. Villaronga, H. Baide, M. Molero-Luis, M. Batllori, M. I. Vanegas, J. Muchart, L. Aquino, R. Artuch, A. Macaya, M. A. Kurian, Pérez Dueñas

Published in: Orphanet Journal of Rare Diseases | Issue 1/2018

Login to get access

Abstract

Background

The SLC39A14, SLC30A10 and SLC39A8 are considered to be key genes involved in manganese (Mn) homeostasis in humans. Mn levels in plasma and urine are useful tools for early recognition of these disorders. We aimed to explore further biomarkers of Mn deposition in the central nervous system in two siblings presenting with acute dystonia and hypermanganesemia due to mutations in SLC39A14. These biomarkers may help clinicians to establish faster and accurate diagnosis and to monitor disease progression after chelation therapy is administered.

Results

A customized gene panel for movement disorders revealed a novel missense variant (c.311G > T; p.Ser104Ile) in SLC39A14 gene in two siblings presenting at the age of 10 months with acute dystonia and motor regression. Mn concentrations were analyzed using inductively coupled mass spectrometry in plasma and cerebrospinal fluid, disclosing elevated Mn levels in the index case compared to control patients. Surprisingly, Mn values were 3-fold higher in CSF than in plasma. We quantified the pallidal index, defined as the ratio between the signal intensity in the globus pallidus and the subcortical frontal white matter in axial T1-weighted MRI, and found significantly higher values in the SLC39A14 patient than in controls. These values increased over a period of 10 years, suggesting the relentless pallidal accumulation of Mn. Following genetic confirmation, a trial with the Mn chelator Na2CaEDTA led to a reduction in plasma Mn, zinc and selenium levels. However, parents reported worsening of cervical dystonia, irritability and sleep difficulties and chelation therapy was discontinued.

Conclusions

Our study expands the very few descriptions of patients with SLC39A14 mutations. We report for the first time the elevation of Mn in CSF of SLC39A14 mutated patients, supporting the hypothesis that brain is an important organ of Mn deposition in SLC39A14-related disease. The pallidal index is an indirect and non-invasive method that can be used to rate disease progression on follow-up MRIs. Finally, we propose that patients with inherited defects of manganese transport should be initially treated with low doses of Na2CaEDTA followed by gradual dose escalation, together with a close monitoring of blood trace elements in order to avoid side effects.
Literature
1.
go back to reference Santos D, Batoreu MC, Almeida I, Ramos R, Sidoryk-Wegrzynowicz M, Aschner M, et al. Manganese alters rat brain amino acids levels. Biol Trace Elem Res. 2012;150(1–3):337–41.CrossRefPubMedPubMedCentral Santos D, Batoreu MC, Almeida I, Ramos R, Sidoryk-Wegrzynowicz M, Aschner M, et al. Manganese alters rat brain amino acids levels. Biol Trace Elem Res. 2012;150(1–3):337–41.CrossRefPubMedPubMedCentral
2.
go back to reference Fujishiro H, Yoshida M, Nakano Y, Himeno S. Interleukin-6 enhances manganese accumulation in SH-SY5Y cells: implications of the up-regulation of ZIP14 and the down-regulation of ZnT10. Metallomics. 2014;6(4):944–9.CrossRefPubMed Fujishiro H, Yoshida M, Nakano Y, Himeno S. Interleukin-6 enhances manganese accumulation in SH-SY5Y cells: implications of the up-regulation of ZIP14 and the down-regulation of ZnT10. Metallomics. 2014;6(4):944–9.CrossRefPubMed
4.
go back to reference Aydemir TB, Kim MH, Kim J, Colon-Perez LM, Banan G, Mareci TH, et al. Metal transporter Zip14 (Slc39a14) deletion in mice increases manganese deposition and produces Neurotoxic signatures and diminished motor activity. J Neurosci. 2017;37(25):5996–6006.CrossRefPubMedPubMedCentral Aydemir TB, Kim MH, Kim J, Colon-Perez LM, Banan G, Mareci TH, et al. Metal transporter Zip14 (Slc39a14) deletion in mice increases manganese deposition and produces Neurotoxic signatures and diminished motor activity. J Neurosci. 2017;37(25):5996–6006.CrossRefPubMedPubMedCentral
5.
go back to reference Girijashanker K, He L, Soleimani M, Reed JM, Li H, Liu Z, et al. Slc39a14 gene encodes ZIP14, a metal/bicarbonate symporter: similarities to the ZIP8 transporter. Mol Pharmacol. 2008;73(5):1413–23.CrossRefPubMedPubMedCentral Girijashanker K, He L, Soleimani M, Reed JM, Li H, Liu Z, et al. Slc39a14 gene encodes ZIP14, a metal/bicarbonate symporter: similarities to the ZIP8 transporter. Mol Pharmacol. 2008;73(5):1413–23.CrossRefPubMedPubMedCentral
6.
go back to reference Hutchens S, Liu C, Jursa T, Shawlot W, Chaffee BK, Yin W, et al. Deficiency in the manganese efflux transporter SLC30A10 induces severe hypothyroidism in mice. J Biol Chem. 2017;292(23):9760–73.CrossRefPubMed Hutchens S, Liu C, Jursa T, Shawlot W, Chaffee BK, Yin W, et al. Deficiency in the manganese efflux transporter SLC30A10 induces severe hypothyroidism in mice. J Biol Chem. 2017;292(23):9760–73.CrossRefPubMed
7.
go back to reference Lin W, Vann DR, Doulias PT, Wang T, Landesberg G, Li X, et al. Hepatic metal ion transporter ZIP8 regulates manganese homeostasis and manganese-dependent enzyme activity. J Clin Invest. 2017;127(6):2407–17.CrossRefPubMedPubMedCentral Lin W, Vann DR, Doulias PT, Wang T, Landesberg G, Li X, et al. Hepatic metal ion transporter ZIP8 regulates manganese homeostasis and manganese-dependent enzyme activity. J Clin Invest. 2017;127(6):2407–17.CrossRefPubMedPubMedCentral
9.
go back to reference Pinilla-Tenas JJ, Sparkman BK, Shawki A, Illing AC, Mitchell CJ, Zhao N, et al. Zip14 is a complex broad-scope metal-ion transporter whose functional properties support roles in the cellular uptake of zinc and nontransferrin-bound iron. Am J Physiol Cell Physiol. 2011;301(4):C862–71.CrossRefPubMedPubMedCentral Pinilla-Tenas JJ, Sparkman BK, Shawki A, Illing AC, Mitchell CJ, Zhao N, et al. Zip14 is a complex broad-scope metal-ion transporter whose functional properties support roles in the cellular uptake of zinc and nontransferrin-bound iron. Am J Physiol Cell Physiol. 2011;301(4):C862–71.CrossRefPubMedPubMedCentral
10.
go back to reference Xin Y, Gao H, Wang J, Qiang Y, Imam MU, Li Y, et al. Manganese transporter Slc39a14 deficiency revealed its key role in maintaining manganese homeostasis in mice. Cell Discov. 2017;3:17025.CrossRefPubMedPubMedCentral Xin Y, Gao H, Wang J, Qiang Y, Imam MU, Li Y, et al. Manganese transporter Slc39a14 deficiency revealed its key role in maintaining manganese homeostasis in mice. Cell Discov. 2017;3:17025.CrossRefPubMedPubMedCentral
11.
go back to reference Riley LG, Cowley MJ, Gayevskiy V, Roscioli T, Thorburn DR, Prelog K, et al. A SLC39A8 variant causes manganese deficiency, and glycosylation and mitochondrial disorders. J Inherit Metab Dis. 2017;40(2):261–9.CrossRefPubMed Riley LG, Cowley MJ, Gayevskiy V, Roscioli T, Thorburn DR, Prelog K, et al. A SLC39A8 variant causes manganese deficiency, and glycosylation and mitochondrial disorders. J Inherit Metab Dis. 2017;40(2):261–9.CrossRefPubMed
12.
go back to reference Park JH, Hogrebe M, Grüneberg M, DuChesne I, von der Heiden AL, Reunert J, et al. SLC39A8 deficiency: a disorder of manganese transport and Glycosylation. Am J Hum Genet. 2015;97(6):894–903.CrossRefPubMedPubMedCentral Park JH, Hogrebe M, Grüneberg M, DuChesne I, von der Heiden AL, Reunert J, et al. SLC39A8 deficiency: a disorder of manganese transport and Glycosylation. Am J Hum Genet. 2015;97(6):894–903.CrossRefPubMedPubMedCentral
13.
go back to reference Aschner M, Erikson KM, Herrero Hernández E, Tjalkens R. Manganese and its role in Parkinson's disease: from transport to neuropathology. NeuroMolecular Med. 2009;11(4):252–66.CrossRefPubMedPubMedCentral Aschner M, Erikson KM, Herrero Hernández E, Tjalkens R. Manganese and its role in Parkinson's disease: from transport to neuropathology. NeuroMolecular Med. 2009;11(4):252–66.CrossRefPubMedPubMedCentral
14.
go back to reference Tuschl K, Mills PB, Parsons H, Malone M, Fowler D, Bitner-Glindzicz M, et al. Hepatic cirrhosis, dystonia, polycythaemia and hypermanganesaemia - a new metabolic disorder. J Inherit Metab Dis. 2008;31(2):151–63.CrossRefPubMed Tuschl K, Mills PB, Parsons H, Malone M, Fowler D, Bitner-Glindzicz M, et al. Hepatic cirrhosis, dystonia, polycythaemia and hypermanganesaemia - a new metabolic disorder. J Inherit Metab Dis. 2008;31(2):151–63.CrossRefPubMed
15.
go back to reference Tuschl K, Meyer E, Valdivia LE, Zhao N, Dadswell C, Abdul-Sada A, et al. Mutations in SLC39A14 disrupt manganese homeostasis and cause childhood-onset parkinsonism-dystonia. Nat Commun. 2016;7:11601.CrossRefPubMedPubMedCentral Tuschl K, Meyer E, Valdivia LE, Zhao N, Dadswell C, Abdul-Sada A, et al. Mutations in SLC39A14 disrupt manganese homeostasis and cause childhood-onset parkinsonism-dystonia. Nat Commun. 2016;7:11601.CrossRefPubMedPubMedCentral
16.
go back to reference Tuschl K, Clayton PT, Gospe SM Jr, Gulab S, Ibrahim S, Singhi P, et al. Syndrome of hepatic cirrhosis, dystonia, polycythemia, and hypermanganesemia caused by mutations in SLC30A10, a manganese transporter in man. Am J Hum Genet. 2012;90(3):457–66.CrossRefPubMedPubMedCentral Tuschl K, Clayton PT, Gospe SM Jr, Gulab S, Ibrahim S, Singhi P, et al. Syndrome of hepatic cirrhosis, dystonia, polycythemia, and hypermanganesemia caused by mutations in SLC30A10, a manganese transporter in man. Am J Hum Genet. 2012;90(3):457–66.CrossRefPubMedPubMedCentral
17.
go back to reference Zaki MS, Issa MY, Elbendary HM, El-Karaksy H, Hosny H, Ghobrial C, et al. Hypermanganesemia with dystonia, polycythemia and cirrhosis in 10 patients: six novel SLC30A10 mutations and further phenotype delineation. Clin Genet. 2017; https://doi.org/10.1111/cge.13184. Zaki MS, Issa MY, Elbendary HM, El-Karaksy H, Hosny H, Ghobrial C, et al. Hypermanganesemia with dystonia, polycythemia and cirrhosis in 10 patients: six novel SLC30A10 mutations and further phenotype delineation. Clin Genet. 2017; https://​doi.​org/​10.​1111/​cge.​13184.
18.
go back to reference Quadri M, Federico A, Zhao T, Breedveld GJ, Battisti C, Delnooz C, et al. Mutations in SLC30A10 cause parkinsonism and dystonia with hypermanganesemia, polycythemia, and chronic liver disease. Am J Hum Genet. 2012;90(3):467–77.CrossRefPubMedPubMedCentral Quadri M, Federico A, Zhao T, Breedveld GJ, Battisti C, Delnooz C, et al. Mutations in SLC30A10 cause parkinsonism and dystonia with hypermanganesemia, polycythemia, and chronic liver disease. Am J Hum Genet. 2012;90(3):467–77.CrossRefPubMedPubMedCentral
19.
go back to reference Quadri M, Kamate M, Sharma S, Olgiati S, Graafland J, Breedveld GJ, et al. Manganese transport disorder: novel SLC30A10 mutations and early phenotypes. Mov Disord. 2015;30(7):996–1001.CrossRefPubMed Quadri M, Kamate M, Sharma S, Olgiati S, Graafland J, Breedveld GJ, et al. Manganese transport disorder: novel SLC30A10 mutations and early phenotypes. Mov Disord. 2015;30(7):996–1001.CrossRefPubMed
20.
go back to reference Gospe SM Jr, Caruso RD, Clegg MS, Keen CL, Pimstone NR, Ducore JM, et al. Paraparesis, hypermanganesaemia, and polycythaemia: a novel presentation of cirrhosis. Arch Dis Child. 2000;83(5):439–42.CrossRefPubMedPubMedCentral Gospe SM Jr, Caruso RD, Clegg MS, Keen CL, Pimstone NR, Ducore JM, et al. Paraparesis, hypermanganesaemia, and polycythaemia: a novel presentation of cirrhosis. Arch Dis Child. 2000;83(5):439–42.CrossRefPubMedPubMedCentral
21.
go back to reference Brna P, Gordon K, Dooley JM, Price V. Manganese toxicity in a child with iron deficiency and polycythemia. J Child Neurol. 2011;26(7):891–4.CrossRefPubMed Brna P, Gordon K, Dooley JM, Price V. Manganese toxicity in a child with iron deficiency and polycythemia. J Child Neurol. 2011;26(7):891–4.CrossRefPubMed
22.
go back to reference Stamelou M, Tuschl K, Chong WK, Burroughs AK, Mills PB, Bhatia KP, et al. Dystonia with brain manganese accumulation resulting from SLC30A10 mutations: a new treatable disorder. Mov Disord. 2012;27(10):1317–22.CrossRefPubMedPubMedCentral Stamelou M, Tuschl K, Chong WK, Burroughs AK, Mills PB, Bhatia KP, et al. Dystonia with brain manganese accumulation resulting from SLC30A10 mutations: a new treatable disorder. Mov Disord. 2012;27(10):1317–22.CrossRefPubMedPubMedCentral
23.
go back to reference Lechpammer M, Clegg MS, Muzar Z, Huebner PA, Jin LW, Gospe SM Jr. Pathology of inherited manganese transporter deficiency. Ann Neurol. 2014;75(4):608–12.CrossRefPubMed Lechpammer M, Clegg MS, Muzar Z, Huebner PA, Jin LW, Gospe SM Jr. Pathology of inherited manganese transporter deficiency. Ann Neurol. 2014;75(4):608–12.CrossRefPubMed
24.
go back to reference Mukhtiar K, Ibrahim S, Tuschl K, Mills P. Hypermanganesemia with Dystonia, Polycythemia and cirrhosis (HMDPC) due to mutation in the SLC30A10 gene. Brain and Development. 2016;38(9):862–5.CrossRefPubMed Mukhtiar K, Ibrahim S, Tuschl K, Mills P. Hypermanganesemia with Dystonia, Polycythemia and cirrhosis (HMDPC) due to mutation in the SLC30A10 gene. Brain and Development. 2016;38(9):862–5.CrossRefPubMed
25.
go back to reference Boycott KM, Beaulieu CL, Kernohan KD, Gebril OH, Mhanni A, Chudley AE, et al. Autosomal-recessive intellectual disability with Cerebellar atrophy syndrome caused by mutation of the manganese and zinc transporter gene SLC39A8. Am J Hum Genet. 2015;97(6):886–93.CrossRefPubMedPubMedCentral Boycott KM, Beaulieu CL, Kernohan KD, Gebril OH, Mhanni A, Chudley AE, et al. Autosomal-recessive intellectual disability with Cerebellar atrophy syndrome caused by mutation of the manganese and zinc transporter gene SLC39A8. Am J Hum Genet. 2015;97(6):886–93.CrossRefPubMedPubMedCentral
26.
go back to reference Heitland P, Köster HD. Biomonitoring of 37 trace elements in blood samples from inhabitants of northern Germany by ICP-MS. J Trace Elem Med Biol. 2006;20(4):253–62.CrossRefPubMed Heitland P, Köster HD. Biomonitoring of 37 trace elements in blood samples from inhabitants of northern Germany by ICP-MS. J Trace Elem Med Biol. 2006;20(4):253–62.CrossRefPubMed
27.
go back to reference Wahlen R, Evans L, Turner J, Hearn R. The use of collision/reaction cell ICP-MS for the simultaneous determination of 18 elements in blood and serum samples. Santa Clara: Agilent Technologies. 2005. http://www.agilent.com/chem. Accesed 28 Ago 2009. Wahlen R, Evans L, Turner J, Hearn R. The use of collision/reaction cell ICP-MS for the simultaneous determination of 18 elements in blood and serum samples. Santa Clara: Agilent Technologies. 2005. http://​www.​agilent.​com/​chem. Accesed 28 Ago 2009.
28.
go back to reference Krieger D, Krieger S, Jansen O, Gass P, Theilmann L, Lichtnecker H. Manganese and chronic hepatic encephalopathy. Lancet. 1995;346(8970):270–4.CrossRefPubMed Krieger D, Krieger S, Jansen O, Gass P, Theilmann L, Lichtnecker H. Manganese and chronic hepatic encephalopathy. Lancet. 1995;346(8970):270–4.CrossRefPubMed
29.
go back to reference Harrington JM, Young DJ, Essader AS, Sumner SJ, Levine KE. Analysis of human serum and whole blood for mineral content by ICP-MS and ICP-OES: development of a mineralomics method. Biol Trace Elem Res. 2014;160(1):132–42.CrossRefPubMedPubMedCentral Harrington JM, Young DJ, Essader AS, Sumner SJ, Levine KE. Analysis of human serum and whole blood for mineral content by ICP-MS and ICP-OES: development of a mineralomics method. Biol Trace Elem Res. 2014;160(1):132–42.CrossRefPubMedPubMedCentral
30.
go back to reference Chang Y, Kim Y, Woo ST, Song HJ, Kim SH, Lee H, et al. High signal intensity on magnetic resonance imaging is a better predictor of neurobehavioral performances than blood manganese in asymptomatic welders. Neurotoxicology. 2009;30(4):555–63.CrossRefPubMed Chang Y, Kim Y, Woo ST, Song HJ, Kim SH, Lee H, et al. High signal intensity on magnetic resonance imaging is a better predictor of neurobehavioral performances than blood manganese in asymptomatic welders. Neurotoxicology. 2009;30(4):555–63.CrossRefPubMed
31.
go back to reference Li SJ, Jiang L, Fu X, Huang S, Huang YN, Li XR, et al. Pallidal index as biomarker of manganese brain accumulation and associated with manganese levels in blood: a meta-analysis. PLoS One. 2014;9(4):1–7. Li SJ, Jiang L, Fu X, Huang S, Huang YN, Li XR, et al. Pallidal index as biomarker of manganese brain accumulation and associated with manganese levels in blood: a meta-analysis. PLoS One. 2014;9(4):1–7.
32.
go back to reference Di Toro Mammarella L, Mignarri A, Battisti C, Monti L, Bonifati V, Rasi F, et al. Two-year follow-up after chelating therapy in a patient with adult-onset parkinsonism and hypermanganesaemia due to SLC30A10 mutations. Neurol. 2014;261(1):227–8.CrossRef Di Toro Mammarella L, Mignarri A, Battisti C, Monti L, Bonifati V, Rasi F, et al. Two-year follow-up after chelating therapy in a patient with adult-onset parkinsonism and hypermanganesaemia due to SLC30A10 mutations. Neurol. 2014;261(1):227–8.CrossRef
33.
go back to reference Elmer M. Cranton MD. A Textbook on EDTA Chelation Therapy. 2nd ed. Hampton Roads Publishing Company, Inc. 2001. Elmer M. Cranton MD. A Textbook on EDTA Chelation Therapy. 2nd ed. Hampton Roads Publishing Company, Inc. 2001.
Metadata
Title
Hypermanganesemia due to mutations in SLC39A14: further insights into Mn deposition in the central nervous system
Authors
L. Marti-Sanchez
J. D. Ortigoza-Escobar
A. Darling
M. Villaronga
H. Baide
M. Molero-Luis
M. Batllori
M. I. Vanegas
J. Muchart
L. Aquino
R. Artuch
A. Macaya
M. A. Kurian
Pérez Dueñas
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Orphanet Journal of Rare Diseases / Issue 1/2018
Electronic ISSN: 1750-1172
DOI
https://doi.org/10.1186/s13023-018-0758-x

Other articles of this Issue 1/2018

Orphanet Journal of Rare Diseases 1/2018 Go to the issue