Skip to main content
Top
Published in: Acta Neuropathologica Communications 1/2014

Open Access 01-12-2014 | Research

Possible involvement of lysosomal dysfunction in pathological changes of the brain in aged progranulin-deficient mice

Authors: Yoshinori Tanaka, James K Chambers, Takashi Matsuwaki, Keitaro Yamanouchi, Masugi Nishihara

Published in: Acta Neuropathologica Communications | Issue 1/2014

Login to get access

Abstract

Introduction

It has been shown that progranulin (PGRN) deficiency causes age-related neurodegenerative diseases such as frontotemporal lobar degeneration (FTLD) and neuronal ceroid lipofuscinosis (NCL), a lysosomal storage disease. Previous studies also suggested that PGRN is involved in modulating lysosomal function. To elucidate the pathophysiological role of PGRN in the aged brain, in the present study, lysosomal function and pathological changes of the brain were investigated using 10- and 90-week-old wild-type and PGRN-deficient mice.

Results

We showed that PGRN deficiency caused enhanced CD68 expression in activated microglia and astrogliosis in the cortex and thalamus, especially in the ventral posteromedial nucleus/ventral posterolateral nucleus (VPM/VPL), in the aged brain. Immunoreactivity for Lamp1 (lysosome marker) in the VPM/VPL and expression of lysosome-related genes, i.e. cathepsin D, V-type proton ATPase subunit d2, and transcription factor EB genes, were also increased by PGRN deficiency. Aggregates of p62, which is selectively degraded by the autophagy-lysosomal system, were observed in neuronal and glial cells in the VPM/VPL of aged PGRN-deficient mice. TAR DNA binding protein 43 (TDP-43) aggregates in the cytoplasm of neurons were also observed in aged PGRN-deficient mice. PGRN deficiency caused enhanced expression of glial cell-derived cytotoxic factors such as macrophage expressed gene 1, cytochrome b-245 light chain, cytochrome b-245 heavy chain, complement C4, tumor necrosis factor-α and lipocalin 2. In addition, neuronal loss and lipofuscinosis in the VPM/VPL and disrupted myelination in the cerebral cortex were observed in aged PGRN-deficient mice.

Conclusions

The present study shows that aged PGRN-deficient mice present with NCL-like pathology as well as TDP-43 aggregates in the VPM/VPL, where a particular vulnerability has been reported in NCL model mice. The present results also suggest that these pathological changes in the VPM/VPL are likely a result of lysosomal dysfunction. How PGRN prevents lysosomal dysfunction with aging remains to be elucidated.
Appendix
Available only for authorised users
Literature
1.
go back to reference Cenik B, Sephton CF, Kutluk Cenik B, Herz J, Yu G: Progranulin: a proteolytically processed protein at the crossroads of inflammation and neurodegeneration. J Biol Chem 2012, 287: 32298–32306. 10.1074/jbc.R112.399170CrossRefPubMedPubMedCentral Cenik B, Sephton CF, Kutluk Cenik B, Herz J, Yu G: Progranulin: a proteolytically processed protein at the crossroads of inflammation and neurodegeneration. J Biol Chem 2012, 287: 32298–32306. 10.1074/jbc.R112.399170CrossRefPubMedPubMedCentral
2.
go back to reference Bateman A, Bennett HP: The granulin gene family: from cancer to dementia. Bioessays 2009, 31: 1245–1254. 10.1002/bies.200900086CrossRefPubMed Bateman A, Bennett HP: The granulin gene family: from cancer to dementia. Bioessays 2009, 31: 1245–1254. 10.1002/bies.200900086CrossRefPubMed
3.
go back to reference Thurner L, Preuss KD, Fadle N, Regitz E, Klemm P, Zaks M, Kemele M, Hasenfus A, Csernok E, Gross WL, Pasquali JL, Martin T, Bohle RM, Pfreundschuh M: Progranulin antibodies in autoimmune diseases. J Autoimmun 2013, 42: 29–38. 10.1016/j.jaut.2012.10.003CrossRefPubMed Thurner L, Preuss KD, Fadle N, Regitz E, Klemm P, Zaks M, Kemele M, Hasenfus A, Csernok E, Gross WL, Pasquali JL, Martin T, Bohle RM, Pfreundschuh M: Progranulin antibodies in autoimmune diseases. J Autoimmun 2013, 42: 29–38. 10.1016/j.jaut.2012.10.003CrossRefPubMed
4.
go back to reference Matsubara T, Mita A, Minami K, Hosooka T, Kitazawa S, Takahashi K, Tamori Y, Yokoi N, Watanabe M, Matsuo E, Nishimura O, Seino S: PGRN is a key adipokine mediating high fat diet-induced insulin resistanceand obesity through IL-6 in adipose tissue. Cell Metab 2012, 15: 38–50. 10.1016/j.cmet.2011.12.002CrossRefPubMed Matsubara T, Mita A, Minami K, Hosooka T, Kitazawa S, Takahashi K, Tamori Y, Yokoi N, Watanabe M, Matsuo E, Nishimura O, Seino S: PGRN is a key adipokine mediating high fat diet-induced insulin resistanceand obesity through IL-6 in adipose tissue. Cell Metab 2012, 15: 38–50. 10.1016/j.cmet.2011.12.002CrossRefPubMed
5.
go back to reference Baker M, Mackenzie IR, Pickering-Brown SM, Gass J, Rademakers R, Lindholm C, Snowden J, Adamson J, Sadovnick AD, Rollinson S, Cannon A, Dwosh E, Neary D, Melquist S, Richardson A, Dickson D, Berger Z, Eriksen J, Robinson T, Zehr C, Dickey CA, Crook R, McGowan E, Mann D, Boeve B, Feldman H, Hutton M: Mutations in progranulin cause tau-negative frontotemporal dementia linked to chromosome 17. Nature 2006, 442: 916–919. 10.1038/nature05016CrossRefPubMed Baker M, Mackenzie IR, Pickering-Brown SM, Gass J, Rademakers R, Lindholm C, Snowden J, Adamson J, Sadovnick AD, Rollinson S, Cannon A, Dwosh E, Neary D, Melquist S, Richardson A, Dickson D, Berger Z, Eriksen J, Robinson T, Zehr C, Dickey CA, Crook R, McGowan E, Mann D, Boeve B, Feldman H, Hutton M: Mutations in progranulin cause tau-negative frontotemporal dementia linked to chromosome 17. Nature 2006, 442: 916–919. 10.1038/nature05016CrossRefPubMed
6.
go back to reference Cruts M, Gijselinck I, van der Zee J, Engelborghs S, Wils H, Pirici D, Rademakers R, Vandenberghe R, Dermaut B, Martin JJ, van Duijn C, Peeters K, Sciot R, Santens P, De Pooter T, Mattheijssens M, Van den Broeck M, Cuijt I, Vennekens K, De Deyn PP, Kumar-Singh S, Van Broeckhoven C: Null mutations in progranulin cause ubiquitin-positive frontotemporal dementia linked to chromosome 17q21. Nature 2006, 442: 920–924. 10.1038/nature05017CrossRefPubMed Cruts M, Gijselinck I, van der Zee J, Engelborghs S, Wils H, Pirici D, Rademakers R, Vandenberghe R, Dermaut B, Martin JJ, van Duijn C, Peeters K, Sciot R, Santens P, De Pooter T, Mattheijssens M, Van den Broeck M, Cuijt I, Vennekens K, De Deyn PP, Kumar-Singh S, Van Broeckhoven C: Null mutations in progranulin cause ubiquitin-positive frontotemporal dementia linked to chromosome 17q21. Nature 2006, 442: 920–924. 10.1038/nature05017CrossRefPubMed
7.
go back to reference Arai T, Hasegawa M, Akiyama H, Ikeda K, Nonaka T, Mori H, Mann D, Tsuchiya K, Yoshida M, Hashizume Y, Oda T: TDP-43 is a component of ubiquitin-positive tau-negative inclusions in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Biochem Biophys Res Commun 2006, 351: 602–611. 10.1016/j.bbrc.2006.10.093CrossRefPubMed Arai T, Hasegawa M, Akiyama H, Ikeda K, Nonaka T, Mori H, Mann D, Tsuchiya K, Yoshida M, Hashizume Y, Oda T: TDP-43 is a component of ubiquitin-positive tau-negative inclusions in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Biochem Biophys Res Commun 2006, 351: 602–611. 10.1016/j.bbrc.2006.10.093CrossRefPubMed
8.
go back to reference Tanaka Y, Matsuwaki T, Yamanouchi K, Nishihara M: Exacerbated inflammatory responses related to activated microglia after traumatic brain injury in progranulin-deficient mice. Neuroscience 2013, 231: 49–60. 10.1016/j.neuroscience.2012.11.032CrossRefPubMed Tanaka Y, Matsuwaki T, Yamanouchi K, Nishihara M: Exacerbated inflammatory responses related to activated microglia after traumatic brain injury in progranulin-deficient mice. Neuroscience 2013, 231: 49–60. 10.1016/j.neuroscience.2012.11.032CrossRefPubMed
9.
go back to reference Tanaka Y, Matsuwaki T, Yamanouchi K, Nishihara M: Increased lysosomal biogenesis in activated microglia and exacerbated neuronal damage after traumatic brain injury in progranulin-deficient mice. Neuroscience 2013, 250: 8–19. 10.1016/j.neuroscience.2013.06.049CrossRefPubMed Tanaka Y, Matsuwaki T, Yamanouchi K, Nishihara M: Increased lysosomal biogenesis in activated microglia and exacerbated neuronal damage after traumatic brain injury in progranulin-deficient mice. Neuroscience 2013, 250: 8–19. 10.1016/j.neuroscience.2013.06.049CrossRefPubMed
10.
go back to reference Conde JR, Streit WJ: Microglia in the aging brain. J Neuropathol Exp Neurol 2006, 65: 199–203. 10.1097/01.jnen.0000202887.22082.63CrossRefPubMed Conde JR, Streit WJ: Microglia in the aging brain. J Neuropathol Exp Neurol 2006, 65: 199–203. 10.1097/01.jnen.0000202887.22082.63CrossRefPubMed
11.
go back to reference Wils H, Kleinberger G, Pereson S, Janssens J, Capell A, Van Dam D, Cuijt I, Joris G, De Deyn PP, Haass C, Van Broeckhoven C, Kumar-Singh S: Cellular ageing, increased mortality and FTLD-TDP-associated neuropathology in progranulin knockout mice. J Pathol 2012, 228: 67–76.PubMed Wils H, Kleinberger G, Pereson S, Janssens J, Capell A, Van Dam D, Cuijt I, Joris G, De Deyn PP, Haass C, Van Broeckhoven C, Kumar-Singh S: Cellular ageing, increased mortality and FTLD-TDP-associated neuropathology in progranulin knockout mice. J Pathol 2012, 228: 67–76.PubMed
12.
go back to reference Smith KR, Damiano J, Franceschetti S, Carpenter S, Canafoglia L, Morbin M, Rossi G, Pareyson D, Mole SE, Staropoli JF, Sims KB, Lewis J, Lin WL, Dickson DW, Dahl HH, Bahlo M, Berkovic SF: Strikingly different clinicopathological phenotypes determined by progranulin-mutation dosage. Am J Hum Genet 2012, 90: 1102–1107. 10.1016/j.ajhg.2012.04.021CrossRefPubMedPubMedCentral Smith KR, Damiano J, Franceschetti S, Carpenter S, Canafoglia L, Morbin M, Rossi G, Pareyson D, Mole SE, Staropoli JF, Sims KB, Lewis J, Lin WL, Dickson DW, Dahl HH, Bahlo M, Berkovic SF: Strikingly different clinicopathological phenotypes determined by progranulin-mutation dosage. Am J Hum Genet 2012, 90: 1102–1107. 10.1016/j.ajhg.2012.04.021CrossRefPubMedPubMedCentral
13.
go back to reference Götzl JK, Damme M, Fellerer K, Tahirovic S, Kleinberger G, Janssens J, Van Der Zee J, Lang CM, Kremmer E, Martin JJ, Engelborghs S, Kretzschmar HA, Arzberger T, Van Broeckhoven C, Haass C, Capell A: Common pathobiochemical hallmarks of progranulin-associated frontotemporal lobar degeneration and neuronal ceroid lipofuscinosis. Acta Neuropathol 2014. in press. Götzl JK, Damme M, Fellerer K, Tahirovic S, Kleinberger G, Janssens J, Van Der Zee J, Lang CM, Kremmer E, Martin JJ, Engelborghs S, Kretzschmar HA, Arzberger T, Van Broeckhoven C, Haass C, Capell A: Common pathobiochemical hallmarks of progranulin-associated frontotemporal lobar degeneration and neuronal ceroid lipofuscinosis. Acta Neuropathol 2014. in press.
14.
go back to reference Jalanko A, Braulke T: Neuronal ceroid lipofuscinoses. Biochim Biophys Acta 2009, 1793: 697–709. 10.1016/j.bbamcr.2008.11.004CrossRefPubMed Jalanko A, Braulke T: Neuronal ceroid lipofuscinoses. Biochim Biophys Acta 2009, 1793: 697–709. 10.1016/j.bbamcr.2008.11.004CrossRefPubMed
15.
go back to reference Cooper JD: The neuronal ceroid lipofuscinoses: the same, but different? Biochem Soc Trans 2010, 38: 1448–1452. 10.1042/BST0381448CrossRefPubMed Cooper JD: The neuronal ceroid lipofuscinoses: the same, but different? Biochem Soc Trans 2010, 38: 1448–1452. 10.1042/BST0381448CrossRefPubMed
16.
go back to reference Kielar C, Maddox L, Bible E, Pontikis CC, Macauley SL, Griffey MA, Wong M, Sands MS, Cooper JD: Successive neuron loss in the thalamus and cortex in a mouse model of infantile neuronal ceroid lipofuscinosis. Neurobiol Dis 2007, 25: 150–162. 10.1016/j.nbd.2006.09.001CrossRefPubMed Kielar C, Maddox L, Bible E, Pontikis CC, Macauley SL, Griffey MA, Wong M, Sands MS, Cooper JD: Successive neuron loss in the thalamus and cortex in a mouse model of infantile neuronal ceroid lipofuscinosis. Neurobiol Dis 2007, 25: 150–162. 10.1016/j.nbd.2006.09.001CrossRefPubMed
17.
go back to reference Partanen S, Haapanen A, Kielar C, Pontikis C, Alexander N, Inkinen T, Saftig P, Gillingwater TH, Cooper JD, Tyynelä J: Synaptic changes in the thalamocortical system of cathepsin D-deficient mice: a model of human congenital neuronal ceroid-lipofuscinosis. J Neuropathol Exp Neurol 2008, 67: 16–29. 10.1097/nen.0b013e31815f3899CrossRefPubMed Partanen S, Haapanen A, Kielar C, Pontikis C, Alexander N, Inkinen T, Saftig P, Gillingwater TH, Cooper JD, Tyynelä J: Synaptic changes in the thalamocortical system of cathepsin D-deficient mice: a model of human congenital neuronal ceroid-lipofuscinosis. J Neuropathol Exp Neurol 2008, 67: 16–29. 10.1097/nen.0b013e31815f3899CrossRefPubMed
18.
go back to reference von Schantz C, Kielar C, Hansen SN, Pontikis CC, Alexander NA, Kopra O, Jalanko A, Cooper JD: Progressive thalamocortical neuron loss in Cln5 deficient mice: Distinct effects in Finnish variant late infantile NCL. Neurobiol Dis 2009, 34: 308–319. 10.1016/j.nbd.2009.02.001CrossRefPubMedPubMedCentral von Schantz C, Kielar C, Hansen SN, Pontikis CC, Alexander NA, Kopra O, Jalanko A, Cooper JD: Progressive thalamocortical neuron loss in Cln5 deficient mice: Distinct effects in Finnish variant late infantile NCL. Neurobiol Dis 2009, 34: 308–319. 10.1016/j.nbd.2009.02.001CrossRefPubMedPubMedCentral
19.
go back to reference Blom T, Schmiedt ML, Wong AM, Kyttälä A, Soronen J, Jauhiainen M, Tyynelä J, Cooper JD, Jalanko A: Exacerbated neuronal ceroid lipofuscinosis phenotype in Cln1/5 double-knockout mice. Dis Model Mech 2013, 6: 342–357. 10.1242/dmm.010140CrossRefPubMed Blom T, Schmiedt ML, Wong AM, Kyttälä A, Soronen J, Jauhiainen M, Tyynelä J, Cooper JD, Jalanko A: Exacerbated neuronal ceroid lipofuscinosis phenotype in Cln1/5 double-knockout mice. Dis Model Mech 2013, 6: 342–357. 10.1242/dmm.010140CrossRefPubMed
20.
go back to reference Kuronen M, Hermansson M, Manninen O, Zech I, Talvitie M, Laitinen T, Gröhn O, Somerharju P, Eckhardt M, Cooper JD, Lehesjoki AE, Lahtinen U, Kopra O: Galactolipid deficiency in the early pathogenesis of neuronal ceroid lipofuscinosis model Cln8 mnd : implications to delayed myelination and oligodendrocyte maturation. Neuropathol Appl Neurobiol 2012, 38: 471–486. 10.1111/j.1365-2990.2011.01233.xCrossRefPubMed Kuronen M, Hermansson M, Manninen O, Zech I, Talvitie M, Laitinen T, Gröhn O, Somerharju P, Eckhardt M, Cooper JD, Lehesjoki AE, Lahtinen U, Kopra O: Galactolipid deficiency in the early pathogenesis of neuronal ceroid lipofuscinosis model Cln8 mnd : implications to delayed myelination and oligodendrocyte maturation. Neuropathol Appl Neurobiol 2012, 38: 471–486. 10.1111/j.1365-2990.2011.01233.xCrossRefPubMed
21.
go back to reference Kayasuga Y, Chiba S, Suzuki M, Kikusui T, Matsuwaki T, Yamanouchi K, Kotaki H, Horai R, Iwakura Y, Nishihara M: Alteration of behavioural phenotype in mice by targeted disruption of the progranulin gene. Behav Brain Res 2007, 185: 110–118. 10.1016/j.bbr.2007.07.020CrossRefPubMed Kayasuga Y, Chiba S, Suzuki M, Kikusui T, Matsuwaki T, Yamanouchi K, Kotaki H, Horai R, Iwakura Y, Nishihara M: Alteration of behavioural phenotype in mice by targeted disruption of the progranulin gene. Behav Brain Res 2007, 185: 110–118. 10.1016/j.bbr.2007.07.020CrossRefPubMed
22.
go back to reference Oliveira VC, Carrara RC, Simoes DL, Saggioro FP, Carlotti CG Jr, Covas DT, Neder L: Sudan Black B treatment reduces autofluorescence and improves resolution of in situ hybridization specific fluorescent signals of brain sections. Histol Histopathol 2010, 25: 1017–1024.PubMed Oliveira VC, Carrara RC, Simoes DL, Saggioro FP, Carlotti CG Jr, Covas DT, Neder L: Sudan Black B treatment reduces autofluorescence and improves resolution of in situ hybridization specific fluorescent signals of brain sections. Histol Histopathol 2010, 25: 1017–1024.PubMed
23.
go back to reference Sofroniew MV, Vinters HV: Astrocytes: biology and pathology. Acta Neuropathol 2010, 119: 7–35. 10.1007/s00401-009-0619-8CrossRefPubMed Sofroniew MV, Vinters HV: Astrocytes: biology and pathology. Acta Neuropathol 2010, 119: 7–35. 10.1007/s00401-009-0619-8CrossRefPubMed
24.
go back to reference Boellaard JW, Schlote W, Hofer W: Species-specific ultrastructure of neuronal lipofuscin in hippocampus and neocortex of subhuman mammals and humans. Ultrastruct Pathol 2004, 28: 341–351. 10.1080/019131290882330CrossRefPubMed Boellaard JW, Schlote W, Hofer W: Species-specific ultrastructure of neuronal lipofuscin in hippocampus and neocortex of subhuman mammals and humans. Ultrastruct Pathol 2004, 28: 341–351. 10.1080/019131290882330CrossRefPubMed
25.
go back to reference Boggs JM: Myelin basic protein: a multifunctional protein. Cell Mol Life Sci 2006, 63: 1945–1961. 10.1007/s00018-006-6094-7CrossRefPubMed Boggs JM: Myelin basic protein: a multifunctional protein. Cell Mol Life Sci 2006, 63: 1945–1961. 10.1007/s00018-006-6094-7CrossRefPubMed
26.
go back to reference Ferguson CJ, Lenk GM, Meisler MH: Defective autophagy in neurons and astrocytes from mice deficient in PI(3,5)P2. Hum Mol Genet 2009, 18: 4868–4878. 10.1093/hmg/ddp460CrossRefPubMedPubMedCentral Ferguson CJ, Lenk GM, Meisler MH: Defective autophagy in neurons and astrocytes from mice deficient in PI(3,5)P2. Hum Mol Genet 2009, 18: 4868–4878. 10.1093/hmg/ddp460CrossRefPubMedPubMedCentral
27.
go back to reference Ju JS, Weihl CC: Inclusion body myopathy, Paget’s disease of the bone and fronto-temporal dementia: a disorder of autophagy. Hum Mol Genet 2010, 19: R38-R45. 10.1093/hmg/ddq157CrossRefPubMedPubMedCentral Ju JS, Weihl CC: Inclusion body myopathy, Paget’s disease of the bone and fronto-temporal dementia: a disorder of autophagy. Hum Mol Genet 2010, 19: R38-R45. 10.1093/hmg/ddq157CrossRefPubMedPubMedCentral
28.
go back to reference Yin F, Dumont M, Banerjee R, Ma Y, Li H, Lin MT, Beal MF, Nathan C, Thomas B, Ding A: Behavioral deficits and progressive neuropathology in progranulin-deficient mice: a mouse model of frontotemporal dementia. FASEB J 2010, 24: 4639–4647. 10.1096/fj.10-161471CrossRefPubMedPubMedCentral Yin F, Dumont M, Banerjee R, Ma Y, Li H, Lin MT, Beal MF, Nathan C, Thomas B, Ding A: Behavioral deficits and progressive neuropathology in progranulin-deficient mice: a mouse model of frontotemporal dementia. FASEB J 2010, 24: 4639–4647. 10.1096/fj.10-161471CrossRefPubMedPubMedCentral
29.
go back to reference Filimonenko M, Stuffers S, Raiborg C, Yamamoto A, Malerød L, Fisher EM, Isaacs A, Brech A, Stenmark H, Simonsen A: Functional multivesicular bodies are required for autophagic clearance of protein aggregates associated with neurodegenerative disease. J Cell Biol 2007, 179: 485–500. 10.1083/jcb.200702115CrossRefPubMedPubMedCentral Filimonenko M, Stuffers S, Raiborg C, Yamamoto A, Malerød L, Fisher EM, Isaacs A, Brech A, Stenmark H, Simonsen A: Functional multivesicular bodies are required for autophagic clearance of protein aggregates associated with neurodegenerative disease. J Cell Biol 2007, 179: 485–500. 10.1083/jcb.200702115CrossRefPubMedPubMedCentral
30.
go back to reference Wang X, Fan H, Ying Z, Li B, Wang H, Wang G: Degradation of TDP-43 and its pathogenic form by autophagy and the ubiquitin-proteasome system. Neurosci Lett 2010, 469: 112–116. 10.1016/j.neulet.2009.11.055CrossRefPubMed Wang X, Fan H, Ying Z, Li B, Wang H, Wang G: Degradation of TDP-43 and its pathogenic form by autophagy and the ubiquitin-proteasome system. Neurosci Lett 2010, 469: 112–116. 10.1016/j.neulet.2009.11.055CrossRefPubMed
31.
go back to reference Caccamo A, Majumder S, Deng JJ, Bai Y, Thornton FB, Oddo S: Rapamycin rescues TDP-43 mislocalization and the associated low molecular mass neurofilament instability. J Biol Chem 2009, 284: 27416–27424. 10.1074/jbc.M109.031278CrossRefPubMedPubMedCentral Caccamo A, Majumder S, Deng JJ, Bai Y, Thornton FB, Oddo S: Rapamycin rescues TDP-43 mislocalization and the associated low molecular mass neurofilament instability. J Biol Chem 2009, 284: 27416–27424. 10.1074/jbc.M109.031278CrossRefPubMedPubMedCentral
32.
go back to reference Gomes C, Escrevente C, Costa J: Mutant superoxide dismutase 1 overexpression in NSC-34 cells: effect of trehalose on aggregation, TDP-43 localization and levels of co-expressed glycoproteins. Neurosci Lett 2010, 475: 145–149. 10.1016/j.neulet.2010.03.065CrossRefPubMed Gomes C, Escrevente C, Costa J: Mutant superoxide dismutase 1 overexpression in NSC-34 cells: effect of trehalose on aggregation, TDP-43 localization and levels of co-expressed glycoproteins. Neurosci Lett 2010, 475: 145–149. 10.1016/j.neulet.2010.03.065CrossRefPubMed
33.
go back to reference Ahmed Z, Sheng H, Xu YF, Lin WL, Innes AE, Gass J, Yu X, Wuertzer CA, Hou H, Chiba S, Yamanouchi K, Leissring M, Petrucelli L, Nishihara M, Hutton ML, McGowan E, Dickson DW, Lewis J: Accelerated lipofuscinosis and ubiquitination in granulin knockout mice suggest a role for progranulin in successful aging. Am J Pathol 2010, 177: 311–324. 10.2353/ajpath.2010.090915CrossRefPubMedPubMedCentral Ahmed Z, Sheng H, Xu YF, Lin WL, Innes AE, Gass J, Yu X, Wuertzer CA, Hou H, Chiba S, Yamanouchi K, Leissring M, Petrucelli L, Nishihara M, Hutton ML, McGowan E, Dickson DW, Lewis J: Accelerated lipofuscinosis and ubiquitination in granulin knockout mice suggest a role for progranulin in successful aging. Am J Pathol 2010, 177: 311–324. 10.2353/ajpath.2010.090915CrossRefPubMedPubMedCentral
34.
go back to reference Kuronen M, Lehesjoki AE, Jalanko A, Cooper JD, Kopra O: Selective spatiotemporal patterns of glial activation and neuron loss in the sensory thalamocortical pathways of neuronal ceroid lipofuscinosis 8 mice. Neurobiol Dis 2012, 47: 444–457. 10.1016/j.nbd.2012.04.018CrossRefPubMed Kuronen M, Lehesjoki AE, Jalanko A, Cooper JD, Kopra O: Selective spatiotemporal patterns of glial activation and neuron loss in the sensory thalamocortical pathways of neuronal ceroid lipofuscinosis 8 mice. Neurobiol Dis 2012, 47: 444–457. 10.1016/j.nbd.2012.04.018CrossRefPubMed
35.
go back to reference Kao AW, Eisenhut RJ, Martens LH, Nakamura A, Huang A, Bagley JA, Zhou P, de Luis A, Neukomm LJ, Cabello J, Farese RV Jr, Kenyon C: A neurodegenerative disease mutation that accelerates the clearance of apoptotic cells. Proc Natl Acad Sci U S A 2011, 108: 4441–4446. 10.1073/pnas.1100650108CrossRefPubMedPubMedCentral Kao AW, Eisenhut RJ, Martens LH, Nakamura A, Huang A, Bagley JA, Zhou P, de Luis A, Neukomm LJ, Cabello J, Farese RV Jr, Kenyon C: A neurodegenerative disease mutation that accelerates the clearance of apoptotic cells. Proc Natl Acad Sci U S A 2011, 108: 4441–4446. 10.1073/pnas.1100650108CrossRefPubMedPubMedCentral
36.
go back to reference Schulz K, Kroner A, David S: Iron efflux from astrocytes plays a role in remyelination. J Neurosci 2012, 32: 4841–4847. 10.1523/JNEUROSCI.5328-11.2012CrossRefPubMed Schulz K, Kroner A, David S: Iron efflux from astrocytes plays a role in remyelination. J Neurosci 2012, 32: 4841–4847. 10.1523/JNEUROSCI.5328-11.2012CrossRefPubMed
37.
go back to reference Barateiro A, Domingues HS, Fernandes A, Relvas JB, Brites D: Rat cerebellar slice cultures exposed to bilirubin evidence reactive gliosis, excitotoxicity and impaired myelinogenesis that is prevented by AMPA and TNF-α inhibitors. Mol Neurobiol 2014, 49: 424–439. 10.1007/s12035-013-8530-7CrossRefPubMed Barateiro A, Domingues HS, Fernandes A, Relvas JB, Brites D: Rat cerebellar slice cultures exposed to bilirubin evidence reactive gliosis, excitotoxicity and impaired myelinogenesis that is prevented by AMPA and TNF-α inhibitors. Mol Neurobiol 2014, 49: 424–439. 10.1007/s12035-013-8530-7CrossRefPubMed
38.
go back to reference Appelqvist H, Wäster P, Kågedal K, Ollinger K: The lysosome: from waste bag to potential therapeutic target. J Mol Cell Biol 2013, 5: 214–226. 10.1093/jmcb/mjt022CrossRefPubMed Appelqvist H, Wäster P, Kågedal K, Ollinger K: The lysosome: from waste bag to potential therapeutic target. J Mol Cell Biol 2013, 5: 214–226. 10.1093/jmcb/mjt022CrossRefPubMed
39.
go back to reference Brady OA, Zheng Y, Murphy K, Huang M, Hu F: The frontotemporal lobar degeneration risk factor, TMEM106B, regulates lysosomal morphology and function. Hum Mol Genet 2013, 22: 685–695. 10.1093/hmg/dds475CrossRefPubMed Brady OA, Zheng Y, Murphy K, Huang M, Hu F: The frontotemporal lobar degeneration risk factor, TMEM106B, regulates lysosomal morphology and function. Hum Mol Genet 2013, 22: 685–695. 10.1093/hmg/dds475CrossRefPubMed
Metadata
Title
Possible involvement of lysosomal dysfunction in pathological changes of the brain in aged progranulin-deficient mice
Authors
Yoshinori Tanaka
James K Chambers
Takashi Matsuwaki
Keitaro Yamanouchi
Masugi Nishihara
Publication date
01-12-2014
Publisher
BioMed Central
Published in
Acta Neuropathologica Communications / Issue 1/2014
Electronic ISSN: 2051-5960
DOI
https://doi.org/10.1186/s40478-014-0078-x

Other articles of this Issue 1/2014

Acta Neuropathologica Communications 1/2014 Go to the issue