Elsevier

Neuroscience

Volume 429, 1 March 2020, Pages 143-155
Neuroscience

Research Article
Lysosomal Dysregulation in the Murine AppNL-G-F/NL-G-F Model of Alzheimer’s Disease

https://doi.org/10.1016/j.neuroscience.2019.12.042Get rights and content

Highlights

  • LAMP1 accumulates at amyloid beta plaques in AppNL-G-F/NL-G-F (knock-in) mouse brain.

  • Lysosomal hydrolases are enriched at amyloid beta plaques in AppNL-G-F/NL-G-F mice.

  • Some lysosomal network proteins are elevated in AppNL-G-F/NL-G-F cortex.

  • Lysosomal network dysfunction in AppNL-G-F/NL-G-F mice resembles human AD.

Abstract

Lysosomal network dysfunction is a prominent feature of Alzheimer’s disease (AD). Although transgenic mouse models of AD are known to model some aspects of lysosomal network dysfunction, the lysosomal network has not yet been examined in the knock-in AppNL-G-F/NL-G-F mouse. We aimed to determine whether AppNL-G-F/NL-G-F mice exhibit disruptions to the lysosomal network in the brain. Lysosome-associated membrane protein 1 (LAMP1) and cathepsins B, L and D accumulated at amyloid beta plaques in the AppNL-G-F/NL-G-F mice, as occurs in human Alzheimer’s patients. The accumulation of these lysosomal proteins occurred early in the development of neuropathology, presenting at the earliest and smallest amyloid beta plaques observed. AppNL-G-F/NL-G-F mice also exhibited elevated activity of β-hexosaminidase and cathepsins D/E and elevated levels of selected lysosomal network proteins, namely LAMP1, cathepsin D and microtubule-associated protein light chain 3 (LC3-II) in the cerebral cortex, as determined by western blot. Elevation of cathepsin D did not change the extent of co-localisation between cathepsin D and LAMP1 in the AppNL-G-F/NL-G-F mice. These findings demonstrate that perturbations of the lysosomal network occur in the AppNL-G-F/NL-G-F mouse model, further validating its use an animal model of pre-symptomatic AD.

Introduction

It is estimated that there are 50 million people worldwide living with dementia, which is associated with a cost of US$1 trillion (Patterson, 2018). Alzheimer’s disease (AD), the most common cause of dementia, has no effective disease-altering treatments. Despite hundreds of clinical trials to evaluate treatments in humans, only four drugs, which do not alter the course of the disease, have been approved (Cummings et al., 2014, Patterson, 2018). Pathogenic mechanisms, as well as most hereditary contributions to the sporadic form of AD remain to be elucidated. It is therefore crucial that authentic animal models are generated and characterised for biomedical research, to determine the causative disease pathways.

Transgenic mice that overexpress amyloid precursor protein (APP) and/or presenilin-1 have been successful in modelling amyloid plaque deposition and accompanying cognitive deficits. These models, such as 5XFAD (Oakley et al., 2006) and TgCRND8 (Chishti et al., 2001) have been instrumental to AD research. More recently, mouse models of AD have been generated through the knock-in of humanised amyloid beta and familial AD mutations, rather than transgenic overexpression of APP and/or presenilin-1. One such model is the AppNL-G-F/NL-G-F mouse, which has humanised amyloid beta sequence with three additional familial mutations: Swedish (NL), Arctic (G) and Iberian (F) incorporated into the App locus. This mouse has severe amyloidosis, with onset of plaque deposition in the cerebral cortex commencing by two-months of age and spreading to subcortical regions by four-months (Saito et al., 2014), preceding behavioural changes (Masuda et al., 2016, Sakakibara et al., 2018, Whyte et al., 2018, Latif-Hernandez et al., 2019, Mehla et al., 2019). AppNL-G-F/NL-G-F mice also develop microgliosis and astrogliosis, (Saito et al., 2014, Mehla et al., 2019). However, a number of findings from transgenic, APP-overexpressing mouse models of AD are not replicated in knock-in models, leading Sasaguri et al. (2017) to suggest that re-examining findings obtained in transgenic mouse models in knock-in models is good practice. The AppNL-G-F/NL-G-F model is still being characterised, and it is important to establish which features of human AD are recapitulated in this mouse.

Progressive perturbation of the lysosomal network (comprising endo-lysosomal and autophagic pathways) is a prominent neuropathological feature of AD (Cataldo et al., 2000, Nixon and Cataldo, 2006, Peric and Annaert, 2015, Nixon, 2017, Whyte et al., 2017), while genetic variation in lysosomal network genes is associated with AD (Lambert et al., 2013, Gao et al., 2018). Examples of the manifestation of lysosomal network dysfunction in AD include enlargement of RAB5-positive endosomes, (Cataldo et al., 2000, Nixon, 2017) and altered expression and activity of lysosomal proteins (reviewed in Whyte, et al., 2017). For example, in human AD patients, cathepsin D and β-hexosaminidase A protein levels are elevated in a substantial portion of neurons from vulnerable brain regions such as pyramidal neurons in layers III and V in the neocortex and the CA2 and CA3 fields of the hippocampus, as well as a smaller portion of neurons from less severely affected regions such as Purkinje cells, striatal neurons, and neurons from the thalamus and medulla (Cataldo et al., 1994, Cataldo et al., 1995, Cataldo et al., 1996). Furthermore, extracellular amyloid plaques are associated with dystrophic neurites that are filled with lysosomal network cargo vesicles (Terry et al., 1964, Nixon et al., 2005). In humans, lysosome-associated membrane protein 1 (LAMP1) can be observed histologically at most amyloid plaques (Barrachina et al., 2006, Hassiotis et al., 2018). Lysosomal hydrolases, including cathepsins, are also observed extracellularly, in association with amyloid plaques (Bernstein et al., 1989, Cataldo and Nixon, 1990, Cataldo et al., 1991, Cataldo et al., 1996). Abundant LAMP1 staining around amyloid plaques is modelled well in transgenic mouse models of AD (Condello et al., 2011, Gowrishankar et al., 2015). However, there are some inconsistencies in the existing literature in relation to the presence of lysosomal hydrolases at amyloid plaques in AD mouse models: cathepsin D has been observed around amyloid plaques in APPSWE/PS1-dE9 mice (Ta et al., 2013); however, Gowrishankar and colleagues (2015) reported that cathepsins B, D and L were only very weakly detected around plaques in 5xFAD mice compared with prominent staining in surrounding neurons.

The lysosomal network has not been examined in an App knock-in line of mice, such as the AppNL-G-F/NL-G-F model. It is important to determine whether this mouse model exhibits disruptions to the lysosomal network similar to those observed in human AD and transgenic mouse models to determine the scope of relevance for this knock-in model. Lysosomal abnormalities have been proposed as a useful criterion for evaluating how well animal models of AD recapitulate the human condition (Cataldo et al., 1994). Here, we describe evidence of lysosomal network dysfunction in AppNL-G-F/NL-G-F mice. The AppNL-G-F/NL-G-F mouse model therefore provides an additional tool to probe the lysosomal network in AD and will permit spatio-temporal investigations of lysosomal network pathology at various disease stages.

Section snippets

Animals

AppNL-G-F/NL-G-F male founder mice were obtained from RIKEN (Saito et al., 2014) and a breeding colony established at SAHMRI following re-derivation. F3-5 AppNL-G-F/NL-G-F and wild-type controls on the same C57BL/6J background were aged until 3-, 6-, 12-, 16- (female mice) or 29-weeks (male mice) and humanely killed via carbon dioxide asphyxiation. All animal experimentation was approved by the SAHMRI (SAM129) and University of Adelaide (M-2015-082) Animal Ethics Committees and conducted

LAMP1 accumulates at the smallest and earliest-appearing amyloid beta plaques in AppNL-G-F/NL-G-F mice

Amyloid beta plaques, detectable by 6E10 staining, were not observed in wild-type mice at any of the ages examined (3-, 6-, 12-, 16- and 29-weeks; Fig. 1E). Plaques were also not evident in three- or six-week-old AppNL-G-F/NL-G-F mice, but by 12-weeks of age, extracellular amyloid beta plaques began to develop in these mice (Fig. 1E). The plaques resembled diffuse, primitive and classic plaques that are observed in human AD (Thal et al., 2000, Peiffer et al., 2002). At 12-weeks, plaques were

Discussion

We have shown that some aspects of the lysosomal network, previously unassessed in AppNL-G-F/NL-G-F mice, are disrupted in this knock-in model of AD. Consistent with human AD (Barrachina et al., 2006, Hassiotis et al., 2018), AppNL-G-F/NL-G-F mice have stark accumulation of the lysosomal marker LAMP1 associated with amyloid plaques. This may be an important component of Alzheimer’s pathogenesis, as lysosomal network vesicles around amyloid plaques have been hypothesised to be a source of local

Funding

This work was supported by an Australian Rotary Health/Rotary Club of Adelaide Funding Partner Scholarship and a Research Training Program Scholarship, awarded to LSW. Funding was also provided by the Hopwood Centre for Neurobiology, Lifelong Health Theme, SAHMRI.

Author contributions

LSW contributed to experimental design and concept, performed immunofluorescence studies, performed the enzyme activity assays and western blots, analysed the data, prepared figures and wrote the manuscript. SH performed immunofluorescence studies, contributed to experimental design and concept, analysed the data and reviewed the manuscript. KJH performed western blots, analysed the immunofluorescence and western blotting data, and reviewed the manuscript. AAL, KMH, JJH and contributed to

Declaration Of Interests

None

Acknowledgments

We thank Drs Takashi Saito and Takaomi Saido from the RIKEN Center for Brain Science Laboratory for Proteolytic Neuroscience for provision of the AppNL-G-F/NL-G-F mice. We also thank Leanne Hein for assistance with genotyping, Meghan Douglass for assistance with tissue collection, Yi Ng for slide scanner operation, and the Bioresources team at SAHMRI, especially Amanda Wilson, for caring for the mice.

Glossary

Lysosomal network
network comprising the endosomal and autophagic pathways, as well as lysosomes.

References (57)

  • M. Barrachina et al.

    Lysosome-associated membrane protein 1 (LAMP-1) in Alzheimer's disease

    Neuropathol Appl Neurobiol

    (2006)
  • H.G. Bernstein et al.

    Immunodetection of cathepsin D in neuritic plaques found in brains of patients with dementia of Alzheimer type

    J Hirnforsch

    (1989)
  • S. Bolte et al.

    A guided tour into subcellular colocalization analysis in light microscopy

    J Microsc

    (2006)
  • M. Bordi et al.

    Autophagy flux in CA1 neurons of Alzheimer hippocampus: increased induction overburdens failing lysosomes to propel neuritic dystrophy

    Autophagy

    (2016)
  • A.M. Cataldo et al.

    Gene expression and cellular content of cathepsin D in Alzheimer's disease brain: evidence for early up-regulation of the endosomal-lysosomal system

    Neuron

    (1995)
  • A.M. Cataldo et al.

    Colocalization of lysosomal hydrolase and beta-amyloid in diffuse plaques of the cerebellum and striatum in Alzheimer's disease and Down's syndrome

    J Neuropathol Exp Neurol

    (1996)
  • A.M. Cataldo et al.

    Increased neuronal endocytosis and protease delivery to early endosomes in sporadic Alzheimer's disease: neuropathologic evidence for a mechanism of increased beta-amyloidogenesis

    J Neurosci

    (1997)
  • A.M. Cataldo et al.

    Lysosomal abnormalities in degenerating neurons link neuronal compromise to senile plaque development in Alzheimer disease

    Brain Res

    (1994)
  • A.M. Cataldo et al.

    Enzymatically active lysosomal proteases are associated with amyloid deposits in Alzheimer brain

    Proc Natl Acad Sci U S A

    (1990)
  • A.M. Cataldo et al.

    Lysosomal hydrolases of different classes are abnormally distributed in brains of patients with Alzheimer disease

    Proc Natl Acad Sci U S A

    (1991)
  • A.M. Cataldo et al.

    Endocytic pathway abnormalities precede amyloid beta deposition in sporadic Alzheimer's disease and Down syndrome: differential effects of APOE genotype and presenilin mutations

    Am J Pathol

    (2000)
  • M.A. Chishti et al.

    Early-onset amyloid deposition and cognitive deficits in transgenic mice expressing a double mutant form of amyloid precursor protein 695

    J. Biol. Chem.

    (2001)
  • K. Coen et al.

    Lysosomal calcium homeostasis defects, not proton pump defects, cause endo-lysosomal dysfunction in PSEN-deficient cells

    J Cell Biol

    (2012)
  • C. Condello et al.

    Multicolor time-stamp reveals the dynamics and toxicity of amyloid deposition

    Sci Rep

    (2011)
  • J.L. Cummings et al.

    Alzheimer's disease drug-development pipeline: few candidates, frequent failures

    Alzheimers Res Ther

    (2014)
  • C. Esselens et al.

    Presenilin 1 mediates the turnover of telencephalin in hippocampal neurons via an autophagic degradative pathway

    J Cell Biol

    (2004)
  • S. Gao et al.

    Genetic variation within endolysosomal system is associated with late-onset Alzheimer's disease

    Brain

    (2018)
  • E.J. Goetzl et al.

    Altered lysosomal proteins in neural-derived plasma exosomes in preclinical Alzheimer disease

    Neurology

    (2015)
  • S. Gowrishankar et al.

    Massive accumulation of luminal protease-deficient axonal lysosomes at Alzheimer's disease amyloid plaques

    Proc Natl Acad Sci U S A

    (2015)
  • S. Hassiotis et al.

    Lysosomal LAMP1 immunoreactivity exists in both diffuse and neuritic amyloid plaques in the human hippocampus

    Eur J Neurosci

    (2018)
  • M.W. Ho et al.

    Differential effect of chloride ions on -galactosidase isoenzymes: a method for separate assay

    Clin Chim Acta

    (1971)
  • K. Ii et al.

    Abnormal distribution of cathepsin proteinases and endogenous inhibitors (cystatins) in the hippocampus of patients with Alzheimer's disease, parkinsonism-dementia complex on Guam, and senile dementia and in the aged

    Virchows Arch A Pathol Anat Histopathol

    (1993)
  • Y.M. Kuo et al.

    Comparative analysis of amyloid-beta chemical structure and amyloid plaque morphology of transgenic mouse and Alzheimer's disease brains

    J Biol Chem

    (2001)
  • J.C. Lambert et al.

    Meta-analysis of 74,046 individuals identifies 11 new susceptibility loci for Alzheimer's disease

    Nat Genet

    (2013)
  • A. Latif-Hernandez et al.

    Subtle behavioral changes and increased prefrontal-hippocampal network synchronicity in APP(NL-G-F) mice before prominent plaque deposition

    Behav Brain Res

    (2019)
  • D.H. Leaback et al.

    Studies on glucosaminidase. 4. The fluorimetric assay of N-acetyl-beta-glucosaminidase

    Biochem J

    (1961)
  • J.H. Lee et al.

    Lysosomal proteolysis and autophagy require presenilin 1 and are disrupted by Alzheimer-related PS1 mutations

    Cell

    (2010)
  • A. Magini et al.

    Abnormal cortical lysosomal beta-hexosaminidase and beta-galactosidase activity at post-synaptic sites during Alzheimer's disease progression

    Int J Biochem Cell Biol

    (2015)
  • Cited by (11)

    • Lysosomal gene Hexb displays haploinsufficiency in a knock-in mouse model of Alzheimer's disease

      2022, IBRO Neuroscience Reports
      Citation Excerpt :

      Brains were post-fixed in 4% (w/v) paraformaldehyde in PBS for seven days and stored in PBS at 4 °C, then embedded in paraffin wax. Cortical tissue was homogenised for assay of cathepsins D/E, β-galactosidase, cathepsins B/L, and β-hexosaminidase activities, as described in Whyte et al., 2020. Six micrometre sagittal sections were cut on a microtome (RM2235 Leica, Wetzlar, Germany) 0.36–0.96 mm lateral from the midline (based on stereotaxic coordinates in a mouse brain atlas (Paxinos and Franklin, 2001)).

    • PICALM regulates cathepsin D processing and lysosomal function

      2021, Biochemical and Biophysical Research Communications
      Citation Excerpt :

      Further, CTSD is known to accumulate in early endosomes [10], is over-expressed in neurons, and is not fully proteolytically processed in the lysosomal system [21,22]. Accumulation of immature CTSD and autophagic proteins such as LC3-II is a robust phenomenon in AD and has also been observed in AD animal models [23,24]. These key disease features were recapitulated by impairing key interactions of PICALM with PIP2 and AP2.

    • The composition of the gut microbiota following early-life antibiotic exposure affects host health and longevity in later life

      2021, Cell Reports
      Citation Excerpt :

      Portal, lobular and necrosis inflammation: 0, no inflammatory infiltrate; 1, low level of inflammatory cell infiltration; 2, moderate level of inflammatory cell infiltration; 3, severe inflammation. Half brains were post-fixed with 10% neutral buffered formalin (ThermoFisher) for routine paraffin embedment as previously described (Whyte et al., 2020). Six-micron paraffin sections of mouse brain were cut using a Leica microtome (RM2235) and subjected to immunohistochemical staining for Ionized calcium binding adaptor molecule 1, IBA-1 (FUJIFILM Wako Pure Chemical Corporation) and Doublecortin DCX (Abcam) (Hassiotis et al., 2018).

    View all citing articles on Scopus

    Present address: Childhood Dementia Research Group, College of Medicine and Public Health, Flinders University, Bedford Park, South Australia 5042, Australia.

    Present address: Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY, US.

    View full text