Skip to main content
Top
Published in: Orphanet Journal of Rare Diseases 1/2019

Open Access 01-12-2019 | Research

Tracking sex-dependent differences in a mouse model of CLN6-Batten disease

Authors: McKayla J. Poppens, Jacob T. Cain, Tyler B. Johnson, Katherine A. White, Samantha S. Davis, Rachel Laufmann, Alexander D. Kloth, Jill M. Weimer

Published in: Orphanet Journal of Rare Diseases | Issue 1/2019

Login to get access

Abstract

Background

CLN6-Batten disease is a rare neurodevelopmental disorder characterized pathologically by the accumulation of lysosomal storage material, glial activation and neurodegeneration, and phenotypically by loss of vision, motor coordination, and cognitive ability, with premature death occurring in the second decade of life. In this study, we investigate whether sex differences in a mouse model of CLN6-Batten disease impact disease onset and progression.

Results

A number of noteworthy differences were observed including elevated accumulation of mitochondrial ATP synthase subunit C in the thalamus and cortex of female Cln6 mutant mice at 2 months of age. Moreover, female mutant mice showed more severe behavioral deficits. Beginning at 9 months of age, female mice demonstrated learning and memory deficits and suffered a more severe decline in motor coordination. Further, compared to their male counterparts, female animals succumbed to the disease at a slightly younger age, indicating an accelerated disease progression. Conversely, males showed a marked increase in microglial activation at 6 months of age in the cortex relative to females.

Conclusions

Thus, as female Cln6 mutant mice exhibit cellular and behavioral deficits that precede similar pathologies in male mutant mice, our findings suggest the need for consideration of sex-based differences in CLN6 disease progression during development of preclinical and clinical studies.
Appendix
Available only for authorised users
Literature
1.
go back to reference Cooper JD. Progress towards understanding the neurobiology of batten disease or neuronal ceroid lipofuscinosis. Curr Opin Neurol. 2003;16(2):121–8.PubMedCrossRef Cooper JD. Progress towards understanding the neurobiology of batten disease or neuronal ceroid lipofuscinosis. Curr Opin Neurol. 2003;16(2):121–8.PubMedCrossRef
2.
go back to reference Mole SE, Williams RE, Goebel HH. Correlations between genotype, ultrastructural morphology and clinical phenotype in the neuronal ceroid lipofuscinoses. Neurogenetics. 2005;6(3):107–26.PubMedCrossRef Mole SE, Williams RE, Goebel HH. Correlations between genotype, ultrastructural morphology and clinical phenotype in the neuronal ceroid lipofuscinoses. Neurogenetics. 2005;6(3):107–26.PubMedCrossRef
3.
go back to reference Goebel HH, Wisniewski KE. Current state of clinical and morphological features in human NCL. Brain Pathol. 2004;14(1):61–9.PubMedCrossRef Goebel HH, Wisniewski KE. Current state of clinical and morphological features in human NCL. Brain Pathol. 2004;14(1):61–9.PubMedCrossRef
4.
go back to reference Palmer DN, Barry LA, Tyynela J, Cooper JD. NCL disease mechanisms. Biochim Biophys Acta. 2013;1832(11):1882–93.PubMedCrossRef Palmer DN, Barry LA, Tyynela J, Cooper JD. NCL disease mechanisms. Biochim Biophys Acta. 2013;1832(11):1882–93.PubMedCrossRef
5.
6.
go back to reference Jalanko A, Braulke T. Neuronal ceroid lipofuscinoses. Biochim Biophys Acta. 2009;1793(4):697–709.PubMedCrossRef Jalanko A, Braulke T. Neuronal ceroid lipofuscinoses. Biochim Biophys Acta. 2009;1793(4):697–709.PubMedCrossRef
7.
go back to reference Warrier V, Vieira M, Mole SE. Genetic basis and phenotypic correlations of the neuronal ceroid lipofusinoses. Biochim Biophys Acta (BBA) - Mol Basis Dis. 2013;1832(11):1827–30.CrossRef Warrier V, Vieira M, Mole SE. Genetic basis and phenotypic correlations of the neuronal ceroid lipofusinoses. Biochim Biophys Acta (BBA) - Mol Basis Dis. 2013;1832(11):1827–30.CrossRef
8.
go back to reference Gao H, Boustany RM, Espinola JA, Cotman SL, Srinidhi L, Antonellis KA, et al. Mutations in a novel CLN6-encoded transmembrane protein cause variant neuronal ceroid lipofuscinosis in man and mouse. Am J Hum Genet. 2002;70(2):324–35.PubMedCrossRef Gao H, Boustany RM, Espinola JA, Cotman SL, Srinidhi L, Antonellis KA, et al. Mutations in a novel CLN6-encoded transmembrane protein cause variant neuronal ceroid lipofuscinosis in man and mouse. Am J Hum Genet. 2002;70(2):324–35.PubMedCrossRef
9.
go back to reference Sharp JD, Wheeler RB, Parker KA, Gardiner RM, Williams RE, Mole SE. Spectrum of CLN6 mutations in variant late infantile neuronal ceroid lipofuscinosis. Hum Mutat. 2003;22(1):35–42.PubMedCrossRef Sharp JD, Wheeler RB, Parker KA, Gardiner RM, Williams RE, Mole SE. Spectrum of CLN6 mutations in variant late infantile neuronal ceroid lipofuscinosis. Hum Mutat. 2003;22(1):35–42.PubMedCrossRef
10.
go back to reference Teixeira CA, Espinola J, Huo L, Kohlschutter J, Persaud Sawin DA, Minassian B, et al. Novel mutations in the CLN6 gene causing a variant late infantile neuronal ceroid lipofuscinosis. Hum Mutat. 2003;21(5):502–8.PubMedCrossRef Teixeira CA, Espinola J, Huo L, Kohlschutter J, Persaud Sawin DA, Minassian B, et al. Novel mutations in the CLN6 gene causing a variant late infantile neuronal ceroid lipofuscinosis. Hum Mutat. 2003;21(5):502–8.PubMedCrossRef
11.
go back to reference Bronson RT, Donahue LR, Johnson KR, Tanner A, Lane PW, Faust JR. Neuronal ceroid lipofuscinosis (nclf), a new disorder of the mouse linked to chromosome 9. Am J Med Genet. 1998;77(4):289–97.PubMedCrossRef Bronson RT, Donahue LR, Johnson KR, Tanner A, Lane PW, Faust JR. Neuronal ceroid lipofuscinosis (nclf), a new disorder of the mouse linked to chromosome 9. Am J Med Genet. 1998;77(4):289–97.PubMedCrossRef
12.
go back to reference Jolly RD, Palmer DN. The neuronal ceroid-lipofuscinoses (batten disease): comparative aspects. Neuropathol Appl Neurobiol. 1995;21(1):50–60.PubMedCrossRef Jolly RD, Palmer DN. The neuronal ceroid-lipofuscinoses (batten disease): comparative aspects. Neuropathol Appl Neurobiol. 1995;21(1):50–60.PubMedCrossRef
13.
go back to reference Jolly RD, West DM. Blindness in South Hampshire sheep: a neuronal ceroidlipofuscinosis. N Z Vet J. 1976;24(6):123.PubMedCrossRef Jolly RD, West DM. Blindness in South Hampshire sheep: a neuronal ceroidlipofuscinosis. N Z Vet J. 1976;24(6):123.PubMedCrossRef
14.
go back to reference Golden LC, Voskuhl R. The importance of studying sex differences in disease: the example of multiple sclerosis. J Neurosci Res. 2017;95(1–2):633–43.PubMedPubMedCentralCrossRef Golden LC, Voskuhl R. The importance of studying sex differences in disease: the example of multiple sclerosis. J Neurosci Res. 2017;95(1–2):633–43.PubMedPubMedCentralCrossRef
15.
go back to reference Beery AK, Zucker I. Sex bias in neuroscience and biomedical research. Neurosci Biobehav Rev. 2011;35(3):565–72.PubMedCrossRef Beery AK, Zucker I. Sex bias in neuroscience and biomedical research. Neurosci Biobehav Rev. 2011;35(3):565–72.PubMedCrossRef
18.
go back to reference Davies W. Sex differences in attention deficit hyperactivity disorder: candidate genetic and endocrine mechanisms. Front Neuroendocrinol. 2014;35(3):331–46.PubMedCrossRef Davies W. Sex differences in attention deficit hyperactivity disorder: candidate genetic and endocrine mechanisms. Front Neuroendocrinol. 2014;35(3):331–46.PubMedCrossRef
19.
go back to reference Schaafsma SM, Pfaff DW. Etiologies underlying sex differences in autism Spectrum disorders. Front Neuroendocrinol. 2014;35(3):255–71.PubMedCrossRef Schaafsma SM, Pfaff DW. Etiologies underlying sex differences in autism Spectrum disorders. Front Neuroendocrinol. 2014;35(3):255–71.PubMedCrossRef
20.
21.
go back to reference Cialone J, Adams H, Augustine EF, Marshall FJ, Kwon JM, Newhouse N, et al. Females experience a more severe disease course in batten disease. J Inherit Metab Dis. 2012;35(3):549–55.PubMedCrossRef Cialone J, Adams H, Augustine EF, Marshall FJ, Kwon JM, Newhouse N, et al. Females experience a more severe disease course in batten disease. J Inherit Metab Dis. 2012;35(3):549–55.PubMedCrossRef
22.
go back to reference Isolation of a novel gene underlying Batten disease, CLN3. The international batten disease consortium. Cell. 1995;82(6):949–57.CrossRef Isolation of a novel gene underlying Batten disease, CLN3. The international batten disease consortium. Cell. 1995;82(6):949–57.CrossRef
23.
go back to reference Cotman SL, Vrbanac V, Lebel LA, Lee RL, Johnson KA, Donahue LR, et al. Cln3(Deltaex7/8) knock-in mice with the common JNCL mutation exhibit progressive neurologic disease that begins before birth. Hum Mol Genet. 2002;11(22):2709–21.PubMedCrossRef Cotman SL, Vrbanac V, Lebel LA, Lee RL, Johnson KA, Donahue LR, et al. Cln3(Deltaex7/8) knock-in mice with the common JNCL mutation exhibit progressive neurologic disease that begins before birth. Hum Mol Genet. 2002;11(22):2709–21.PubMedCrossRef
24.
go back to reference Kovacs AD, Pearce DA. Finding the most appropriate mouse model of juvenile CLN3 (batten) disease for therapeutic studies: the importance of genetic background and gender. Dis Model Mech. 2015;8(4):351–61.PubMedPubMedCentralCrossRef Kovacs AD, Pearce DA. Finding the most appropriate mouse model of juvenile CLN3 (batten) disease for therapeutic studies: the importance of genetic background and gender. Dis Model Mech. 2015;8(4):351–61.PubMedPubMedCentralCrossRef
25.
go back to reference Guarneri R, Russo D, Cascio C, D'Agostino S, Galizzi G, Bigini P, et al. Retinal oxidation, apoptosis and age- and sex-differences in the mnd mutant mouse, a model of neuronal ceroid lipofuscinosis. Brain Res. 2004;1014(1–2):209–20.PubMedCrossRef Guarneri R, Russo D, Cascio C, D'Agostino S, Galizzi G, Bigini P, et al. Retinal oxidation, apoptosis and age- and sex-differences in the mnd mutant mouse, a model of neuronal ceroid lipofuscinosis. Brain Res. 2004;1014(1–2):209–20.PubMedCrossRef
26.
go back to reference Cannelli N, Garavaglia B, Simonati A, Aiello C, Barzaghi C, Pezzini F, et al. Variant late infantile ceroid lipofuscinoses associated with novel mutations in CLN6. Biochem Biophys Res Commun. 2009;379(4):892–7.PubMedCrossRef Cannelli N, Garavaglia B, Simonati A, Aiello C, Barzaghi C, Pezzini F, et al. Variant late infantile ceroid lipofuscinoses associated with novel mutations in CLN6. Biochem Biophys Res Commun. 2009;379(4):892–7.PubMedCrossRef
27.
go back to reference Canafoglia L, Gilioli I, Invernizzi F, Sofia V, Fugnanesi V, Morbin M, et al. Electroclinical spectrum of the neuronal ceroid lipofuscinoses associated with CLN6 mutations. Neurology. 2015;85(4):316–24.PubMedPubMedCentralCrossRef Canafoglia L, Gilioli I, Invernizzi F, Sofia V, Fugnanesi V, Morbin M, et al. Electroclinical spectrum of the neuronal ceroid lipofuscinoses associated with CLN6 mutations. Neurology. 2015;85(4):316–24.PubMedPubMedCentralCrossRef
28.
go back to reference Munroe PB, Mitchison HM, O'Rawe AM, Anderson JW, Boustany RM, Lerner TJ, et al. Spectrum of mutations in the batten disease gene, CLN3. Am J Hum Genet. 1997;61(2):310–6.PubMedPubMedCentralCrossRef Munroe PB, Mitchison HM, O'Rawe AM, Anderson JW, Boustany RM, Lerner TJ, et al. Spectrum of mutations in the batten disease gene, CLN3. Am J Hum Genet. 1997;61(2):310–6.PubMedPubMedCentralCrossRef
29.
go back to reference Brann DW, Dhandapani K, Wakade C, Mahesh VB, Khan MM. Neurotrophic and neuroprotective actions of estrogen: basic mechanisms and clinical implications. Steroids. 2007;72(5):381–405.PubMedPubMedCentralCrossRef Brann DW, Dhandapani K, Wakade C, Mahesh VB, Khan MM. Neurotrophic and neuroprotective actions of estrogen: basic mechanisms and clinical implications. Steroids. 2007;72(5):381–405.PubMedPubMedCentralCrossRef
30.
go back to reference McEwen BS, Alves SE. Estrogen actions in the central nervous system. Endocr Rev. 1999;20(3):279–307.PubMed McEwen BS, Alves SE. Estrogen actions in the central nervous system. Endocr Rev. 1999;20(3):279–307.PubMed
31.
go back to reference Garcia-Estrada J, Del Rio JA, Luquin S, Soriano E, Garcia-Segura LM. Gonadal hormones down-regulate reactive gliosis and astrocyte proliferation after a penetrating brain injury. Brain Res. 1993;628(1–2):271–8.PubMedCrossRef Garcia-Estrada J, Del Rio JA, Luquin S, Soriano E, Garcia-Segura LM. Gonadal hormones down-regulate reactive gliosis and astrocyte proliferation after a penetrating brain injury. Brain Res. 1993;628(1–2):271–8.PubMedCrossRef
32.
go back to reference Barreto G, Veiga S, Azcoitia I, Garcia-Segura LM, Garcia-Ovejero D. Testosterone decreases reactive astroglia and reactive microglia after brain injury in male rats: role of its metabolites, oestradiol and dihydrotestosterone. Eur J Neurosci. 2007;25(10):3039–46.PubMedCrossRef Barreto G, Veiga S, Azcoitia I, Garcia-Segura LM, Garcia-Ovejero D. Testosterone decreases reactive astroglia and reactive microglia after brain injury in male rats: role of its metabolites, oestradiol and dihydrotestosterone. Eur J Neurosci. 2007;25(10):3039–46.PubMedCrossRef
33.
go back to reference Arevalo MA, Santos-Galindo M, Acaz-Fonseca E, Azcoitia I, Garcia-Segura LM. Gonadal hormones and the control of reactive gliosis. Horm Behav. 2013;63(2):216–21.PubMedCrossRef Arevalo MA, Santos-Galindo M, Acaz-Fonseca E, Azcoitia I, Garcia-Segura LM. Gonadal hormones and the control of reactive gliosis. Horm Behav. 2013;63(2):216–21.PubMedCrossRef
34.
go back to reference Barreto G, Santos-Galindo M, Diz-Chaves Y, Pernia O, Carrero P, Azcoitia I, et al. Selective estrogen receptor modulators decrease reactive astrogliosis in the injured brain: effects of aging and prolonged depletion of ovarian hormones. Endocrinology. 2009;150(11):5010–5.PubMedCrossRef Barreto G, Santos-Galindo M, Diz-Chaves Y, Pernia O, Carrero P, Azcoitia I, et al. Selective estrogen receptor modulators decrease reactive astrogliosis in the injured brain: effects of aging and prolonged depletion of ovarian hormones. Endocrinology. 2009;150(11):5010–5.PubMedCrossRef
36.
go back to reference Yanguas-Casas N, Crespo-Castrillo A, de Ceballos ML, Chowen JA, Azcoitia I, Arevalo MA, et al. Sex differences in the phagocytic and migratory activity of microglia and their impairment by palmitic acid. Glia. 2018;66(3):522–37.PubMedCrossRef Yanguas-Casas N, Crespo-Castrillo A, de Ceballos ML, Chowen JA, Azcoitia I, Arevalo MA, et al. Sex differences in the phagocytic and migratory activity of microglia and their impairment by palmitic acid. Glia. 2018;66(3):522–37.PubMedCrossRef
37.
go back to reference Sochocka M, Diniz BS, Leszek J. Inflammatory response in the CNS: friend or foe? Mol Neurobiol. 2017;54(10):8071–89.PubMedCrossRef Sochocka M, Diniz BS, Leszek J. Inflammatory response in the CNS: friend or foe? Mol Neurobiol. 2017;54(10):8071–89.PubMedCrossRef
38.
go back to reference Schwarz JM, Sholar PW, Bilbo SD. Sex differences in microglial colonization of the developing rat brain. J Neurochem. 2012;120(6):948–63.PubMedPubMedCentral Schwarz JM, Sholar PW, Bilbo SD. Sex differences in microglial colonization of the developing rat brain. J Neurochem. 2012;120(6):948–63.PubMedPubMedCentral
39.
go back to reference Kim S, Kim MJ, Kim S, Kang HS, Lim SW, Myung W, et al. Gender differences in risk factors for transition from mild cognitive impairment to Alzheimer's disease: a CREDOS study. Compr Psychiatry. 2015;62:114–22.PubMedCrossRef Kim S, Kim MJ, Kim S, Kang HS, Lim SW, Myung W, et al. Gender differences in risk factors for transition from mild cognitive impairment to Alzheimer's disease: a CREDOS study. Compr Psychiatry. 2015;62:114–22.PubMedCrossRef
40.
go back to reference Kang S, Kim JB, Heo TH, Kim SJ. Cell cycle arrest in batten disease lymphoblast cells. Gene. 2013;519(2):245–50.PubMedCrossRef Kang S, Kim JB, Heo TH, Kim SJ. Cell cycle arrest in batten disease lymphoblast cells. Gene. 2013;519(2):245–50.PubMedCrossRef
41.
go back to reference Grimaldi CM. Sex and systemic lupus erythematosus: the role of the sex hormones estrogen and prolactin on the regulation of autoreactive B cells. Curr Opin Rheumatol. 2006;18(5):456–61.PubMedCrossRef Grimaldi CM. Sex and systemic lupus erythematosus: the role of the sex hormones estrogen and prolactin on the regulation of autoreactive B cells. Curr Opin Rheumatol. 2006;18(5):456–61.PubMedCrossRef
42.
go back to reference Seehafer SS, Ramirez-Montealegre D, Wong AM, Chan CH, Castaneda J, Horak M, et al. Immunosuppression alters disease severity in juvenile batten disease mice. J Neuroimmunol. 2011;230(1–2):169–72.PubMedPubMedCentralCrossRef Seehafer SS, Ramirez-Montealegre D, Wong AM, Chan CH, Castaneda J, Horak M, et al. Immunosuppression alters disease severity in juvenile batten disease mice. J Neuroimmunol. 2011;230(1–2):169–72.PubMedPubMedCentralCrossRef
43.
go back to reference Augustine EF, Beck CA, Adams HR, Defendorf S, Vierhile A, Timm D, et al. Short-term Administration of Mycophenolate is Well-Tolerated in CLN3 disease (juvenile neuronal ceroid Lipofuscinosis). JIMD Rep. 2018. Augustine EF, Beck CA, Adams HR, Defendorf S, Vierhile A, Timm D, et al. Short-term Administration of Mycophenolate is Well-Tolerated in CLN3 disease (juvenile neuronal ceroid Lipofuscinosis). JIMD Rep. 2018.
44.
go back to reference Morgan JP, Magee H, Wong A, Nelson T, Koch B, Cooper JD, et al. A murine model of variant late infantile ceroid lipofuscinosis recapitulates behavioral and pathological phenotypes of human disease. PLoS One. 2013;8(11):e78694.PubMedPubMedCentralCrossRef Morgan JP, Magee H, Wong A, Nelson T, Koch B, Cooper JD, et al. A murine model of variant late infantile ceroid lipofuscinosis recapitulates behavioral and pathological phenotypes of human disease. PLoS One. 2013;8(11):e78694.PubMedPubMedCentralCrossRef
Metadata
Title
Tracking sex-dependent differences in a mouse model of CLN6-Batten disease
Authors
McKayla J. Poppens
Jacob T. Cain
Tyler B. Johnson
Katherine A. White
Samantha S. Davis
Rachel Laufmann
Alexander D. Kloth
Jill M. Weimer
Publication date
01-12-2019
Publisher
BioMed Central
Published in
Orphanet Journal of Rare Diseases / Issue 1/2019
Electronic ISSN: 1750-1172
DOI
https://doi.org/10.1186/s13023-019-0994-8

Other articles of this Issue 1/2019

Orphanet Journal of Rare Diseases 1/2019 Go to the issue