You are on page 1of 22

Neurobiology of Disease 148 (2021) 105219

Contents lists available at ScienceDirect

Neurobiology of Disease
journal homepage: www.elsevier.com/locate/ynbdi

Leptin enhances adult neurogenesis and reduces pathological features in a


transgenic mouse model of Alzheimer’s disease
Michele Longoni Calió a, b, *, Amanda Cristina Mosini b, Darci Souza Marinho c,
Geisa Nogueira Salles d, Fernando Henrique Massinhani a, Gui Mi Ko e,
Marimélia Aparecida Porcionatto a, *
a
Department of Biochemistry, Laboratory of Neurobiology, Universidade Federal de São Paulo, São Paulo, SP 04039-032, Brazil
b
Department of Physiology, Laboratory of Neurobiology, Universidade Federal de São Paulo, São Paulo, SP 04039-032, Brazil
c
Department of Ginecology, Laboratory of Genetic Control, Universidade Federal de São Paulo, São Paulo, SP 04023-062, Brazil
d
Institute of Research and Development, Laboratory of Biomedical Nanotechnology, Universidade do Vale do Paraíba, São José dos Campos, SP, 12244-000, Brazil
e
Institute of Pharmacology and Molecular Biology INFAR, Universidade Federal de São Paulo, São Paulo, SP, 04044-020, Brazil

A R T I C L E I N F O A B S T R A C T

Keywords: Alzheimer’s disease (AD) is the most common dementia worldwide and is characterized by the presence of senile
Alzheimer’s disease plaques by amyloid-beta (Aβ) and neurofibrillary tangles of hyperphosphorylated Tau protein. These changes
amyloid-beta lead to progressive neuronal degeneration and dysfunction, resulting in severe brain atrophy and cognitive
leptin
deficits. With the discovery that neurogenesis persists in the adult mammalian brain, including brain regions
neurogenesis
neural stem cells
affected by AD, studies of the use of neural stem cells (NSCs) for the treatment of neurodegenerative diseases to
hippocampus repair or prevent neuronal cell loss have increased. Here we demonstrate that leptin administration increases the
neurogenic process in the dentate gyrus of the hippocampus as well as in the subventricular zone of lateral
ventricles of adult and aged mice. Chronic treatment with leptin increased NSCs proliferation with significant
effects on proliferation and differentiation of newborn cells. The expression of the long form of the leptin re­
ceptor, LepRb, was detected in the neurogenic niches by reverse qPCR and immunohistochemistry. Moreover,
leptin modulated astrogliosis, microglial cell number and the formation of senile plaques. Additionally, leptin led
to attenuation of Aβ-induced neurodegeneration and superoxide anion production as revealed by Fluoro-Jade B
and dihydroethidium staining. Our study contributes to the understanding of the effects of leptin in the brain that
may lead to the development of new therapies to treat Alzheimer’s disease.

1. Introduction microglial activation and oxidative stress, that are linked to abnormal
neuronal function, progressive degeneration and neuronal injury which
Described over a century ago, Alzheimer’s disease (AD) is the most cause memory loss and cognitive dysfunction (Armstrong, 2011; Laur­
common progressive dementia associated with aging (Gallaway et al., itzen et al., 2012; Zhang et al., 2011; Reitz and Mayeux, 2014; Heneka
2017; Dai et al., 2018). However, the precise mechanisms of AD still are et al., 2014; Jeong, 2017; Sanabria-Castro et al., 2017; Feitosa and da
not fully understood. The pathology of AD is mainly characterized by Silva Oliveira, 2018).
multiple etiological and biochemical aberrations including the forma­ Neurogenesis is the process of generating new neurons by neural
tion of senile plaques by aggregates of amyloid-beta (Aβ) peptides, stem cells (NSCs) that occurs mainly in two brain regions, named
neurofibrillary tangles (NFTs) of hyperphosphorylated Tau protein, neurogenic niches – the subventricular zone (SVZ) of the lateral

Abbreviation: 2xTgAD, double transgenic AD model; AD, Alzheimer’s disease; Aβ, amyloid-beta; BDNF, brain-derived neurotrophic factor; BrdU, 5-Bromo-
2’deoxyuridine; DCX, doublecortin; DG, dentate gyrus; DPX, Dibutylphthalate Polystyrene Xylene; DHE, dihydroethidium; FJB, fluorojade B; GFAP, glia fibrillary
acid protein; GPx1, glutathione peroxidase 1; LepRb, Leptin receptor b; Map2, microtubule-associated protein 2; NeuN, neuronal specific nuclear protein; NFT,
neurofibrillary tangles; NSC, neural stem cells; PBS, phosphate-buffered solution; PCNA, proliferating cell nuclear antigen; ROS, reactive oxygen species; SGZ,
subgranular zone; Sod2, superoxide dismutase 2; SVZ, subventricular zone; WT, wild type.
* Corresponding author at.: Rua Pedro de Toledo, 669 – 3◦ andar, CEP 04049-032, Vila Clementino, São Paulo, SP, Brazil
E-mail addresses: michele.longoni@unifesp.br (M.L. Calió), marimelia.pocionatto@unifesp.br (M.A. Porcionatto).

https://doi.org/10.1016/j.nbd.2020.105219
Received 22 April 2020; Received in revised form 18 November 2020; Accepted 3 December 2020
Available online 8 December 2020
0969-9961/© 2020 Published by Elsevier Inc. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
M.L. Calió et al. Neurobiology of Disease 148 (2021) 105219

ventricles (LVs) and the subgranular zone (SGZ) in the dentate gyrus treatment of AD and probably other neurodegenerative diseases.
(DG) of the hippocampus (Alvarez-Buylla and Lim, 2004; Fuentealba
et al., 2012) but also disseminated throughout the central nervous sys­ 2. Material and methods
tem (CNS) (Ming and Song, 2011; Lin and Iacovitti, 2015).
Although the adult brain of mammals, including humans, generates Animals - Three-month-old (Young) and twelve-month-old (Aged)
new neurons throughout life (Altman and Das, 1965; Eriksson et al., male heterozygous double transgenic APP/PS1 mice (2xTgAD) - over­
1998; Spalding et al., 2013; Ernst et al., 2014), the proliferative activity expressing a chimeric mouse/human mutant APP (amyloid precursor
of NSCs and the generation of new neurons markedly declines during protein) (Mo/HuAPP695swe) and a mutant human presenilin 1 (PS1-
aging and is deregulated in neurodegenerative diseases such as AD, dE9), - colony B6.Cg-Tg(APPswe,PSEN1dE9)85Dbo/J (from Jackson
which may contribute to a decline in memory function and some degree Laboratory, Bar Harbor, ME) and nontransgenic littermates C57BL/6J
of cognitive impairment (Clelland et al., 2009; Lazarov et al., 2010). mice (wild type – WT) were used. The animals, weighing 25–30g, were
Moreover, the differences observed in the neurogenic process of purchased from the Center of Development of Experimental Models for
neurodegenerative diseases may reflect different stages of the pathology Biology (CEDEME–UNIFESP), at the Universidade Federal de São Paulo,
(Jin et al., 2004a; Rodriguez et al., 2008; Rodriguez et al., 2011). The São Paulo, Brazil. Mice were genotyped to confirm the presence or
discovery that quiescent precursor cells can be activated even in the absence of the transgenes. Animals were housed and maintained on a
aged or damaged brain has contributed to the emergence of the hy­ 12-h light/dark cycle (lights on at 06:00 a.m.) in a humidity and
pothesis that neurodegenerative diseases could be treated by stimulating temperature-controlled environment with ad libitum access to food and
the adult neurogenesis. In that case, NSCs may be an endogenous source water. All procedures were carried out in accordance with the internal
of healthy cells where the derived progeny might be re-directed to areas Ethical Committee on Animal Research of the Universidade Federal de
of degeneration, where they would be involved in cell replacement, São Paulo UNIFESP/EPM (CEP 6882070814). All efforts were made to
providing a promising therapeutic approach (Gage, 2000; Lie et al., minimize animal suffering and to reduce the number of animals used.
2004; Hormoz, 2013; Zhang et al., 2017; Matarredona et al., 2018). Drugs - Recombinant mouse leptin (R&D Systems) was resuspended
Consequently, several studies have been conducted to investigate how in sterile saline solution (0.9 % NaCl).
the promotion of the neurogenic potential of these cells in AD improves Leptin treatment – To study the effect of leptin on cell proliferation in
the symptoms of the disease like spatial memory in experimental animal the neurogenic niches, a dose of 1mg/kg was used. This dose was chosen
models (Becker et al., 2007; Taniuchi et al., 2007; Naumann et al., 2009; based on its efficacy in inducing neurogenesis in mice (Garza et al.,
Markiewicz et al., 2011; Sahay et al., 2011; Kanemoto et al., 2013; 2008). Animals of both ages and lineages (n = 10 per group) received
Hamilton et al., 2015; Winner and Winkler, 2015; Zhang et al., 2017; Tai intraperitoneal (i.p.) injection of saline or leptin (1mg/kg), once a day at
et al., 2017). the end of the light cycle for 7 consecutive days. Bodyweight was
Clinical evidence indicates that diet and lifestyle are major risk assessed daily during the treatment. In the last two days of treatment,
factors for developing AD and disturbances in metabolism are also half of the mice (n = 5 per group) were injected with 5-Bromo-2-deox­
linked to the development of the pathology (Dosunmu et al., 2007; Yin yuridine (BrdU; Sigma, St. Louis, MO), an analog of the nitrogen base
et al., 2016). The hormone leptin is known to regulate food intake and thymidine that is incorporated into the DNA of the proliferative cells.
energy balance, being also involved in many other processes such as Animals received four intraperitoneal injections of BrdU (50mg/kg, in
immune response, stress, and neurodevelopment (Folch et al., 2012; 0.1M sodium phosphate buffer [PBS], pH7.4) within 48 h, at 8-h in­
Meyer et al., 2014; Bellefontaine et al., 2014). Indeed, lower leptin levels tervals between each dose, corresponding with the S-phase of cell di­
are associated with an elevated risk of AD, as well as aberrant leptin vision in mammals. These mice were transcardially perfused 8h after the
function (Lieb et al., 2009; Baranowska-Bik et al., 2015). Within the past last BrdU injection. The brains of these animals were used for immu­
decade, researchers showed a role for leptin in neuroprotection and nohistochemical evaluation of BrdU-labeled cells.
hippocampal neurogenesis. In animal models, leptin administration fa­ Biochemical measurements of leptin in serum - Serum samples were
cilitates learning and alters synaptic plasticity (Moult et al., 2010; Luo obtained from blood collected from all groups of aged animals and
et al., 2015; Ghasemi et al., 2016). Additionally, this adipokine atten­ stored at -80◦ C for correlating the decrease in plasma leptin levels and
uates Tau hyperphosphorylation in neuronal cells and protects against the onset of AD. Briefly, the blood was obtained at the time of euthanasia
Aβ-induced neurotoxicity in cellular models (Garza et al., 2008; Doherty of the animals and a small sample was placed in tubes at ambient tem­
et al., 2012; Liu et al., 2015a, 2015b). Thus, there is growing evidence perature for 1-2 hours in an upright position. The tubes were then
that leptin-based therapies may be beneficial in AD. centrifuged at 4 ◦ C for 10 minutes at 3000g and the clear serum was
Although the pathway linking neurogenesis and AD is still under removed and stored. The concentrations of leptin levels from non­
debate, mainly due to the different animal models used, it is important transgenic WT and 2xTgAD mice were measured in serum by an enzyme-
that further studies be done to determine the role of leptin in neuro­ linked immunosorbent assay (ELISA) kit Quantikine Mouse Leptin
genesis and whether it can be considered as a therapeutic strategy in AD. Immunoassay (R&D Systems) according to the manufacturer’s recom­
We used the double transgenic APPswe/PS1dE9 (2xTgAD) mouse mendations. Leptin levels were normalized to total protein levels and
model of different ages for investigating the temporal changes of neu­ expressed as picograms of leptin content per milliliters of total proteins
rogenesis and the action of leptin during the disease progress in the two (pg/mL). Values were determined using a four-parameter nonlinear
main neurogenic niches. The 2xTgAD mouse lineage reproduces a subset regression equation.
of neuropathological, biochemical and behavioral changes identified in Tissue Preparation – The animals that received BrdU injections (n = 5
patients with AD, such as massive Aβ accumulation and plaque forma­ per group) were terminally anesthetized with a ketamine cocktail
tion, vascular deficits and memory impairment (Holcomb et al., 1998; (43mg/kg ketamine, 10mg/kg xylazine, and 1.4mg/kg acepromazine)
Jankowsky et al., 2002). and transcardially perfused through the ascending aorta using 0.1M PBS
Our findings demonstrate that leptin plays a role in extra- (pH 7.4) and by 4% paraformaldehyde (PFA, pH 7.4) in 0.1M PBS. The
hippocampal neurogenesis, in regions involved in neural cell replace­ brain was removed from the skull and post-fixed overnight, also in 4%
ment, regardless of age or disease stage. Collectively, we show that PFA, before being transferred to a 30% (w/v) sucrose solution in 0.1M
leptin participates in several events that reduce the pathology and exert PBS for 48h. Brains were embedded in O.C.T. frozen tissue embedding
multiple functions besides neurogenesis, contributing to the Aβ clear­ media (HistoPrep™, Fisher Scientific) and coronally cryosectioned at
ance and decreasing oxidative stress. We hypothesized that strategies 30μm thick sections on a cryostat (Leica CM 1850). The sections were
targeted at stimulating this process could have significant therapeutic ordered in a systematic and uniformly-random manner (SURS) (Gun­
value, suggesting that leptin may be used as an approach for the dersen, 2002) and stored free-floating in anti-freezing solution (30%

2
M.L. Calió et al. Neurobiology of Disease 148 (2021) 105219

sucrose, 30% ethylene glycol, 0.1M PBS) in a multi-well plate until coverslipped using DPX (Dibutylphthalate Polystyrene Xylene)
processing for immunofluorescence. For the molecular biology experi­ mounting medium (Sigma).
ment, hippocampi of the remaining animals (n = 5 per group) were Superoxide anion detection - Superoxide was detected with the
frozen on dry ice, processed for real-time polymerase chain reaction oxidative fluorescent probe DHE (dihydroethidium; Molecular Probes),
(PCR), being stored at -80◦ C for later analysis. which reacts with O.2, resulting in ethidine fluorescence. To remove the
BrdU Immunofluorescence staining - Once BrdU is incorporated into differences in cellular densities, DAPI was used in all of the frozen slices.
the DNA, the material was pretreated with a nucleic acid denaturant to The frozen brain sections were incubated in a light-protected humidified
make it accessible to the BrdU antibody. Brain sections were briefly chamber at room temperature with DHE (5mM) for 5 min and then
washed in 0.1M PBS, incubated in 1% hydrogen peroxide for 10 min, counterstained with the nuclear tracer DAPI (5mM; Sigma). The slides
and treated with 2N HCl solution in 0.1% Triton X-100 for 30 min were immediately analyzed with a computer-based scanning image
succeed by incubation with 0.1M borate buffer (pH 8.5) for 10 min. system (Zeiss Axiovert 100M; Carl Zeiss, Germany) connected to an LSM
Next, the slices were immersed in a blocking solution with 0.1% Triton 810 Confocal Laser Scanning System. Fluorescence was detected with
X-100 and 10% normal goat serum in PBS for 1h and then incubated 510–560nm excitation and 590nm emission filters. The results are
with rat monoclonal anti-BrdU (1:300, Molecular Probes) in the block­ expressed as the DHE/DAPI ratio according to Oudot et al. (2006).
ing solution overnight at 4◦ C. After washing in PBS, the fragments were Cell Quantification and Analyses - The SVZ and hippocampus coronal
then incubated with goat anti-rat Alexa Fluor 647nm (1:300, Molecular sections were mounted on glass slides and were coded. The cell counts
Probes) in blocking solution for 1h at room temperature. A negative were conducted by personnel blinded to the experimental condition of
sample test was performed using serum rather than the primary anti­ each sample. The brain was sectioned at 30μm, and every eight sections
body to establish the specificity of the immunostaining. The slides were were spaced 240μm apart throughout the entire SVZ and the hippo­
examined under a laser scanning confocal microscope (Zeiss LSM 810 campus extent was used to assess the number of labeled cells. This en­
Axiovert; Carl Zeiss GmbH, Germany). sures that the same cell will not be counted twice on consecutive sections
Fluorescence Immunolabeling - To determine the phenotypes of BrdU- and guarantees that the area counted for each brain is constant. A
labeled (BrdU+) cells, brain sections were processed for doublecortin sequence of eight slices per animal was obtained using the Smooth
(DCX) and glial fibrillary acidic protein (GFAP) fluorescent double la­ Fractionator method (Gundersen, 2002), representing the entire volume
beling. The Leptin-receptor (LepRb) antibody was used to check whether of the dorsal hippocampus or anterolateral SVZ. Cell counting was
these cells were responding to leptin treatment and expressing its re­ performed at 40x magnification. In the SVZ, the cells of the anterolateral
ceptor. Briefly, free-floating SVZ and hippocampus sections were region were considered while in the hippocampus cells of the SGZ and
collected and first pretreated as described above, and then incubated the gyrus were counted. The three planes (X, Y and Z) of z-stack images
with rat anti-BrdU antibody (1:300, Molecular Probes) and guinea pig (1.5μm/z-stack) were visualized to confirm the extent of cell colocali­
anti-DCX (1:500, Merk/Millipore) or chicken anti-GFAP (1:500, Merk/ zation using the EzColocalization plugin for ImageJ analysis software.
Millipore). To detect the leptin receptor, Aβ deposits and microglia, Cells single labeled for BrdU, Iba1 and FJB or double-labeled for BrdU/
hippocampus sections from mice were immunostained overnight DCX, BrdU/GFAP or DCX/LepRb were counted. Cell body was defined as
without DNA denaturation step and the following primary antibodies the counting unit. The Cell Counter tool of ImageJ software (National
were used: rabbit anti-LepRb (1:1000, Abcam) or mouse anti-βAmyloid Institute of Health - NIH Image, https://imagej.nih.gov/ij/34) was used
6E10 (1:1000, Covance/Biologend) and goat anti-Iba1 (ionized calcium- to estimate the number of BrdU double stained cells. The percentage of
binding adaptor molecule, 1:1000, Wako). After washing in 0.1 M PBS BrdU cells double-labeled was calculated by dividing the number of
three times, sections were incubated for 1h with fluorescent secondary double-labeled cells by the total number of BrdU cells and multiplying
antibodies conjugated with Alexa Fluor 647nm goat anti-rat IgG to 100. As mentioned, the morphometric analysis was performed using
reveal immunoreactivity of BrdU, Alexa Fluor 488nm goat anti-guinea ImageJ software.
pig IgG to reveal DCX or Alexa Fluor 594nm goat anti-chicken IgG to Microglia Density and morphological Analysis - The number of Iba1
reveal immunoreactivity of GFAP, Alexa Fluor 594nm goat anti-rabbit microglial cells has been reported as the number of somas per acquired
IgG to reveal LepRb, Alexa Fluor 488nm goat anti-mouse IgG to reveal area (300 mm2). To quantify microglia density, the images were
Aβ plaques or Alexa Fluor 594nm rabbit anti-goat IgG to reveal Iba1, analyzed by Optical Fractionator tool of Stereo Investigator software. A
respectively (1:300 for all secondary antibodies, Molecular Probes) and z-axis projection based on the maximal intensity signal throughout the
counterstained with the nuclear tracer 4’6’-diamidino-2-phenylindole thickness of the section was obtained and after threshold setting, the
(DAPI, 5mM; 1:10000, Sigma). Then, the sections were rinsed in PBS, number of cells was recorded. Data are expressed as the density of
ordered and mounted onto coated glass slides, and coverslipped with positive fluorescent cells versus total area (Grimaldi et al., 2019).
Fluoromount G™ (Thermo Fisher Scientific). The slides were analyzed Quantification of the morphological features of the microglia was
in a blind manner using a computer-based digitizing image system (Zeiss carried out by taking z-stacks of 10 ROI per slice which were then
Axiovert 100M; Carl Zeiss, Germany) connected to an LSM 810 Confocal converted to maximum intensity projection tiff files. Microglia
Laser Scanning System. The laser and detector were maintained at morphology was analyzed to identify changes in the resting or activated
constant settings when imaging each staining set. state, such as alterations on cell body and cell ramification between
Fluoro-Jade B labeling – FJB is a fluorochrome derived from fluores­ groups. Photomicrographs of microglia at 40x magnification from all
cein and is commonly used in neuroscience to label degenerating neu­ groups were used. We used ImageJ software and appropriate plugins
rons in brain tissue. Fluoro-Jade B (Histo-Chem Inc) staining was carried (AnalyzeSkeleton v.3.2.2 and FracLac v.2.5) to convert all photomi­
out as described by Schmued and Hopkins (2000). Briefly, PFA-fixed crographs to binary, skeletonized and outline images to analyze cell
brain slices were mounted on 1.5% gelatin-coated slides, dehydrated morphology. In addition, cell somas were manually counted in each
overnight at room temperature and kept into a heater for 30 min at 40◦ C photomicrograph. We summarized the number of process endpoints and
before staining. The slices were dipped for 5 min in a solution of 1% length from the Analyze Skeleton plugin (http://imagej.net/AnalyzeSke
sodium hydroxide in 80% alcohol before placed for 2 min in 70% leton31) data output and normalized all data by the number of microglia
ethanol, and lastly for 1 min in distilled water. Sections were then cell somas in each image to calculate the number of microglia end­
oxidized for 10 min by immersion in 0.06% KMnO4, under slow shaking. points/cell and microglia process length/cell. To compare microglia
After numerous rinses in distilled water, slices were incubated for 30 complexity, we used FracLac plugin for ImageJ (https://imagej.nih.gov/
min in 0.004% FJB dye in 0.1% acetic acid, washed thoroughly in ij/plugins/fraclac/FLHelp/Introduction.htm), which quantifies each
distilled water, and placed into the heater set to 40◦ C until the material cell’s contour bounded by the endpoints and process lengths, calculating
was entirely dry. Lastly, the tissues were cleared in xylene and the fractal dimension. In addition, we evaluated the span ratios as

3
M.L. Calió et al. Neurobiology of Disease 148 (2021) 105219

indicators of cell shape, and density as an indicator of cell size. In variance (ANOVA) with repeated measures followed by Tukey’s
Table S1, we summarize skeleton (endpoints and process length) and correction post hoc. When pertinent, a nonparametric Student t-test was
fractal analysis measures (density, fractal dimension and span ratio) in applied. The data are expressed as mean ± standard error of the mean
terms of measure, unit, sampling and interpretation (Supplemental Data (SEM). Significance was set at *p < 0.05, **p < 0.01, ***p < 0.001.
Table S1). Statistical analyzes were performed using the GraphPad Prism software
Estimations of Amyloid Plaque Density and Volume - Quantitative version 5.0 (GraphPad Software).
image analysis of Aβ plaques was performed as previously described (Liu
et al., 2015a, 2015b; Furcila et al., 2019). The number of Aβ plaques per 3. Results
field (density), diameter and volume of plaque occupied area were
estimated with the aid of Stereo Investigator software (Stereo Investi­ 3.1. Leptin Changes Gene Expression of Proliferation and Immature
gator 11.0, MicroBright Field Inc., MBF Bioscience; Williston, USA), Neural Cells Markers in the Hippocampus
using its Optical and Area Fraction Fractionator tool in the entire hip­
pocampal area of each slice. To estimate the Aβ plaque diameter, the We treated WT and 2xTgAD mice with leptin for 7 consecutive days
edges of the plaque were delineated with the Nucleator tool of Stereo (See Supplemental Data Fig. S1A). Bodyweight of the mice was
Investigator software. This tool provides the volume of each Aβ plaque measured before (Supplemental Data Fig. S1B) and throughout the 7
analyzed, as well as the relative area occupied by them in each examined days of leptin treatment. The percentage of body weight loss are shown
hippocampal subfield to provide the percentage (%) of area coverage by in Fig. S1D. The treatment with leptin did not affect the weight loss of
Aβ plaques. All Aβ plaques with a diameter between 5 -100μm (this cut- animals.
off ensured analysis of discrete plaques - Gowrishankar et al., 2015) We observed that there is a tendency in the decrease of serum leptin
were examined. concentration in 2xTgAD animals, although there was no significant
A minimum of six coronal sections were selected for each animal, at difference. Even after treatment, we did not detect a significant
equal 240-μm intervals with 10x and 40x magnification. Field selection enhancement in leptin levels in serum samples in 2xTgAD mice (See
was performed by choosing 3 evenly-spaced random fields encompass­ Supplemental Data Fig. S1C).
ing all of the hippocampi of each section. These regions of interest were After this, we investigated the expression profile of genes involved in
delineated using 10x objective lenses and plaque counting was per­ neurogenesis in the hippocampus: glial fibrillary acid protein (GFAP -
formed with 40x objective lenses. Images were captured using a digital neural stem cell and astrocyte marker); Nestin (amplifying and neural
camera mounted to a Nikon ECLIPSE 80i inverted microscope (Tokyo, stem cell marker); proliferating cell nuclear antigen (PCNA) and Ki67
Japan) with a motorized stage connected to a computer. All analyses (proliferation and cell-cycle markers); doublecortin (DCX - neuronal
used z projections of 3 optical slices at the center of the plaque. For each progenitor marker); βIII-tubulin, microtubule-associated protein2a
mouse, quantitative analysis was performed on an area of about 3 x 3 (Map2a) and neuronal-specific nuclear protein (NeuN - mature neuron
mm for each image at 40x magnification (Yuan and Grutzendler, 2016). markers).
Real-time quantitative PCR - To determine the action of leptin on gene We observed that treatment with leptin increased the expression of
expression, quantitative reverse transcription PCR (qPCR) was per­ Nestin by young WT, and young and aged 2xTgAD mice (Fig. 1B). The
formed. The hippocampi of five animals of each group were collected, expression of DCX was increased in all treated groups (Fig. 1E) and
and total RNA was extracted using Tryzol® (Invitrogen/Thermo Fisher decreased with aging in both genotypes, whereas the upregulation of
Scientific) according to the manufacturer’s instructions. The comple­ PCNA was observed only in young animals of both strains (Fig. 1C).
mentary DNA (cDNA) was generated from the extracted RNA using the GFAP expression was downregulated in leptin-treated aged 2xTgAD
SuperScript™First-Strand Synthesis System (Thermo Fisher Scientific) animals, but not in the other groups (Fig. 1A). Finally, no difference was
for RT-PCR. Relative quantitation was performed using 25 ng of reverse- observed in the expression of tubulin βIII, Map2a and NeuN with the use
transcribed mRNA (cDNA samples) that were amplified with TaqMan of leptin, but a decrease in the expression of Map2 and NeuN genes was
PCR Master Mix and TaqMan probes. The reactions were run on a 7500 found in aged 2xTgAD mice in comparison to the WT or young animals
Real-Time PCR System (Applied Biosystems) according to the manu­ (Fig. 1F-H).
facturer’s instructions. After amplification, the PCR products were
analyzed with the ABI PRISM sequence detection software (Applied
Biosystems). In addition, melting curve analysis was performed to 3.2. Age and genotype-dependent reduced proliferation of SVZ and SGZ
confirm the authenticity of the PCR products. Quantitative gene cells is increased after leptin treatment
expression was analyzed by the comparative CT method (2-∆∆CT) ac­
cording to Livak and Schmittgen (2001). TaqMan probes to caspase-3, In the present study, we asked whether leptin regulates adult neu­
NeuN, Map2a, DCX, GFAP, βIII Tubulin, Ki67, PCNA, Nestin or rogenesis in the SVZ and the hippocampus. To address this question, we
Custom TaqMan Array Fast Plates to Oxidative stress were used (all labeled newborn cells by administrating BrdU in the last two days of
probes and mixes were purchased from Applied Biosystems). The treatment with leptin and quantified the number of BrdU+ cells in the
expression of each gene was normalized by the mean of the five SGZ and the SVZ of treated and untreated, young and aged, WT and
endogenous controls (β-actin, 18S, Gusb, Gapdh and Hprt). 2xTgAD mice. In the hippocampus, BrdU+ cells usually are present in the
In the analysis Custom TaqMan Array Fast Plates to Oxidative stress, internal layer of the granular cell layer of the DG, while in SVZ, neural
the Student t-test was used for statistical analysis, and the comparison stem cells can be found in several areas, although we have focused on
between the groups was made as follows: the groups treated with leptin the anterolateral region (Fig. 2A). We found a significantly higher
were compared with their respective untreated groups, of the same number of BrdU+ cells in the SVZ of all leptin-treated groups (Fig. 2B).
lineage and age, considering the treatment factor (untreated vs. treated). Analysis of the SGZ revealed that all groups, except the young WT mice,
In another analysis, the untreated groups of aged animals were showed an increased number of BrdU+ cells following treatment with
compared with groups of young animals of the same genotype, taking leptin (Fig. 2C). In addition, in SVZ, both young and aged 2xTgAD an­
into account the age factor (young vs aged). Finally, the remaining imals demonstrated a lower number of proliferative cells compared to
group of young untreated 2xTgAD mice was correlated with the group of WT animals of both ages and a decrease in the number of these cells was
untreated age-matched littermate wild-type animals, where the geno­ also observed in aged WT animals when compared to young WT animals
type factor was considered for comparison purposes (WT vs 2xTgAD). (Fig. 2B). In SGZ, the comparison of both ages or genotypes showed a
Statistical Analysis – Multiple comparisons of treatment vs. age or decrease in aged 2xTgAD animals when compared with aged WT ani­
treatment vs. genotype was performed by one or two-way analysis of mals or young double transgenic animals (Fig. 2C).

4
M.L. Calió et al. Neurobiology of Disease 148 (2021) 105219

Fig. 1. Gene expression analysis by qPCR of proliferative and neural markers in the hippocampus of mice treated or not with leptin in different ages. The animals
were injected i.p. with vehicle (PBS) or leptin (1mg/kg) daily for 7 days. Leptin treatment increased the expression of most genes analyzed. Notice the difference in
the expression of GFAP (A), Ki67 (D) and DCX (E) between young and aged mice or treated mice. The data were normalized using four endogenous controls and are
relative to the PBS control group; n = 5 per group. One-way ANOVA, data are shown as mean ± SEM, represented by error bars;*p < 0.05, **p < 0.01 ***p < 0.001.

5
M.L. Calió et al. Neurobiology of Disease 148 (2021) 105219

Fig. 2. In vivo leptin administration increases proliferation in the SVZ and SGZ neurogenic niches. BrdU labeling was used to evaluate proliferation in both
neurogenic niches. The animals were injected i.p. with vehicle (PBS) or leptin (1mg/kg) daily for 7 days. BrdU+ cells were counted in the SVZ of the lateral ventricle
and in the SGZ of the dentate gyrus of the hippocampus. (A) Representative images showing BrdU+ cells in the SVZ and SGZ from control (PBS) and treated (leptin)
WT and 2xTgAD mice of different ages. Regions used for the analysis and quantification of BrdU+ cells showing increased proliferation in the treated groups from
both genotypes in the SVZ (B) and SGZ (C). Scale bar = 50μm. Data are expressed as mean ± SEM, n = 5 animals per group. *p < 0.05; **p < 0.01; ***p < 0.001.

Fig. 3. Leptin administration decreases astrogliosis in the hippocampus. GFAP labeling was used to evaluate the proliferation of neural stem cells and astrocytes or
astrogliosis in both neurogenic niches. Double positive BrdU+/GFAP+ cells were counted in the SVZ of the lateral ventricle and in the hippocampus. (A) Repre­
sentative images showing BrdU+ (white) and GFAP+ (red) cells in SVZ and SGZ from control (PBS) and treated (leptin) WT and 2xTgAD mice of different ages. (B)
Quantification of BrdU+/GFAP+ cells shows no difference in the proliferation of neural stem cells in the SVZ when mice received leptin. (C) Quantification of BrdU+/
GFAP+ cells shows decreased proliferation of astrocyte in the hippocampus of the leptin treated group. Scale bar = 50μm. Data are expressed as mean ± SEM, n = 5
animals per group. *p < 0.05; **p < 0.01; ***p < 0.001.

6
M.L. Calió et al. Neurobiology of Disease 148 (2021) 105219

3.3. Leptin improves temporal changes during aging and AD by increasing analyzed, when compared to the hippocampus, where the action of
neurogenesis in the SVZ and SGZ and decreasing astrogliosis in the leptin was more significant in the aged 2xTgAD (Fig. 4C). Thus, the
hippocampus increments in the number of neuroblasts were approximately 1.5 fold in
the SVZ and 30 fold in the hippocampus of aged 2xTgAD group.
To explore how leptin affected neurogenesis in AD, we analyzed the
expression of DCX (a marker for immature progenitor neurons), GFAP (a 3.4. Leptin treatment upregulates the expression of LepRb in adult neural
marker for astrocytes), and BrdU (a marker for proliferation) in the SVZ stem/progenitor cells
and DG of mice of different ages and treatment conditions.
Aβ can trigger the neuroinflammatory process to produce a diversity We next sought to elucidate the mechanisms involved in transducing
of inflammatory cytokines, causing neurotoxic effects in the brain. GFAP the effects of leptin in neurogenesis and differentiation. It has been
expression was therefore examined as an indicator of astrogliosis. In described in the literature that LepRb is expressed in the dentate gyrus
SGZ, the presence of proliferative GFAP+ cells was higher in the AD (Scott et al., 2009), and to determine if leptin treatment would affect
model even at 3 months of age and this difference became even more LepRb expression, we used qPCR and immunocytochemistry. qPCR
significant at 12 months of age, compared to wild-type animals. In analysis using RNA isolated from hippocampal tissue of untreated and
addition, an increase in these cells was also observed in aged 2xTgAD treated groups of both ages and genotypes showed that the expression
animals compared to young animals (Fig. 3C). We also observed that level of LepRb was significantly higher in all treated groups compared to
leptin treatment led to a decrease in the number of GFAP+ cells in the control (Fig. 5A). Interestingly, there was no significant change in LepRb
hippocampus of aged 2xTgAD animals (Fig. 3A,C), suggesting that leptin expression as a function of age in either WT or 2xTgAD group (Fig. 5A).
may reduce astrogliosis. The number of GFAP-labeled cells in the SVZ To investigate which type of neural progenitor was expressing leptin
was not significantly altered by leptin treatment in WT and transgenic receptor, the presence of LepRb on the hippocampus and SVZ was
mice (Fig. 3A,B). further confirmed by immunohistochemical staining with antibodies
Representative confocal images also showed that after 7 days of against LepRb and DCX of treated animals of both age and genotype
leptin treatment there was a significant increase in the number of DCX (Fig. 5B). The immunoreactivity for LepRb was observed in a large
coexpressed with BrdU cells in all treated groups of animals (Fig. 4). number of DCX+ cells. As negative control, the primary antibody was
Higher numbers of DCX+ cells were observed in both regions of interest omitted from the reaction and no immunostaining was seen in this case.
including the anterolateral area in SVZ and SGZ in the hippocampal
dentate gyrus (Fig. 4B,C). Regarding age, in SGZ, both genotypes 3.5. Leptin restores Impaired Sod2 and GPx1 expression and leads to
showed a decrease in the number of proliferative neuroblasts in the decreased superoxide production in 2xTgAD mice brains
groups of aged animals. However, aged 2xTgAD animals showed an even
lower number of cells compared to WT animals (Fig. 4C). Curiously, the We used Custom TaqMan Array Fast Plates to Oxidative stress and
presence of neuroblasts is more evident in the SVZ, in all groups Antioxidant defense (See Supplemental Data Table S2 and S3) to

Fig. 4. Leptin administration increases the generation of neuroblasts in the SVZ and hippocampal SGZ. DCX labeling was used to evaluate the production of
neuroblasts by neural stem cells in the two neurogenic niches. Double positive BrdU+/DCX+ cells were counted in the SVZ of the lateral ventricle and in the SGZ of
the dentate gyrus of the hippocampus. (A) Representative images showing BrdU+ (white) and DCX+ (green) cells in the SVZ and SGZ from control (PBS) and treated
(leptin) WT and 2xTgAD mice of different ages. (B) Quantification of BrdU+/DCX+ cells shows an increased generation of neuroblasts in the treated groups in the
SVZ. (C) Quantification of BrdU+/DCX+ cells shows an increased generation of neuroblasts in the treated groups in SGZ. Scale bar = 50μm. Data are expressed as
mean ± SEM, n = 5 animals per group. *p < 0.05; **p < 0.01; ***p < 0.001.

7
M.L. Calió et al. Neurobiology of Disease 148 (2021) 105219

Fig. 5. Neural stem cells and neuroblasts express LepRb. (A) Gene expression analysis by qPCR for LepR in the hippocampus. Leptin treatment increased the
expression of the receptor on all treated groups. (B) Representative images show double staining LepR+/DCX+ cells in SVZ and SGZ of treated animals. Scale bar
= 50μm.

investigate whether neurodegeneration and leptin treatment could 3.6. Leptin reduces Aβ plaque accumulation and neurodegeneration
affect the gene expression pattern of antioxidant enzymes. The number
of upregulated or downregulated genes for each group are listed in To describe the capacity of leptin to reduce brain Aβ levels, we used
Table 1. Most genes related to antioxidant enzymes that could have their transgenic mice at the age of 12 months, treated and untreated with
relative expression (RE) quantified, did not show a significant change in leptin. Our data show that Aβ staining in the hippocampus of aged
the treated groups (Table 2), except for superoxide dismutase-2 (Sod2) 2xTgAD mice was significantly reduced after treatment with leptin
and Glutathione peroxidase-1 (GPx1) in 2xTgAD treated animals (Fig. 8B). The hippocampus of aged 2xTgAD mice treated with leptin
(Fig. 6). Interestingly, even in the younger DA models, the treatment showed a lower frequency of plaques per occupied area (Fig. 8C) and
with leptin-induced an increase in gene expression of these two genes. also demonstrated smaller proportions of plaques of larger sizes,
No significant variation in the level of mRNA was detected in the hip­ although the number of plaques of small diameter was relatively higher
pocampus considering age. Regarding the WT animals, treatment with than in untreated animals of the same age and genotype (Fig. 8E).
leptin was effective only in young animals, increasing the SOD2 gene Taking into account the age and genotype of animals, we observed
expression. that the number of FJB+ cells increases in the hippocampus. An example
Since our results showed significant differences in Sod2 expression in of the contrasting findings between control and leptin groups can be
leptin-treated AD animals, we focused on analyzing whether the anti­ visualized in Fig. 8D. In fact, we observed that FJB labeling in the hip­
oxidant activity of leptin could decrease the level of superoxide pro­ pocampus of 2xTgAD mice decreased significantly after treatment with
duced and attenuate the oxidative stress in AD animals. For that, we leptin as compared with the control group and was similar to WT mice
used dihydroethidium (DHE), which is a detector of intracellular su­ (See Supplemental Data Fig. S2). In addition, the hippocampus of aged
peroxide. Leptin treatment significantly reduced cellular superoxide transgenic mice contains more neurodegenerating cells when compared
levels in the hippocampus (Fig. 7C, D) of 2xTgAD animals. The indicator to young mice and transgenic mice treated with leptin. We also observed
of the presence of superoxide production was four times greater in the FJB staining in other areas, such as cortical regions and thalamus (data
dentate gyrus of untreated aged 2xTgAD than in treated animals, which not shown). Due to the association between brain-derived neurotrophic
exhibited a significant decrease in the DHE/DAPI ratio, confirming the factor (BDNF) levels and the onset of AD, we investigated BDNF mRNA
protective role of this hormone. Curiously in SVZ, leptin decreased su­ expression in the animals. BDNF was detected in all groups, with sig­
peroxide production also in younger animals. nificant increments in aged 2xTgAD leptin-treated group while lower
levels of BDNF expression were associated with the progression of pa­
thology (Fig. 8F).

Table 1
Number of genes related to oxidative stress in the hippocampus that were regulated positively by leptin above a 2.0 ratio (Upregulated genes) or were negatively
regulated by leptin below a 0.5 ratio (Downregulated genes). It was considered that genes that showed value between 0.5 and 2.0 had no difference in genic expression
after treatment with leptin (No difference genes).
Number of genes

Upregulated* Downregulated* No difference*a Undeterminedb Total of genes analyzed #

Young WT + Leptin 4 26 6 55 91
Aged WT + PBS 8 23 5 55 91
Aged WT + Leptin 5 27 5 54 91
Young 2xTgAD + PBS 4 17 7 63 91
Young 2xTgAD + Leptin 13 21 1 56 91
Aged 2xTgAD + PBS 4 24 6 57 91
Aged 2xTgAD + Leptin 4 27 5 55 91
*
Compared to the untreated control group of each age and genotype
a
Genes with values of expression between 0.5 and 2; compared to untreated control across all 3 replicates
b
Genes that did not show any CT value across all 3 replicates
#
Without the five endogenous controls used

8
M.L. Calió et al. Neurobiology of Disease 148 (2021) 105219

Table 2
Fold increase of the relative expression (RE) of genes related to antioxidant enzymes that were upregulated by leptin
Relative Expression

Gene Young WT + Aged WT + Aged WT + Young 2xTgAD + Young 2xTgAD + Aged 2xTgAD + Aged 2xTgAD +
symbol Leptin* PBS# Leptin* PBSα Leptin* PBS# Leptin*

Apc 0,001 0,008 0,002 0,004 0,070 0,004 0,002


Ccs 0,062 0,063 0,032 0,064 0,130 0,064 0,032
Gpx1 0,980 2,047 2,547 2,007 7,943 1,002 3,602
Gpx2 0,002 0,002 0,002 0,004 0,004 0,008 0,002
Gpx3 0,016 0,062 0,004 0,008 0,067 0,008 0,004
Gpx4 0,250 0,516 0,126 0,504 2,075 0,501 0,126
Gpx7 0,001 0,002 0,002 0,004 0,004 0,004 0,002
Gpx8 0,124 0,127 0,126 0,251 0,256 0,248 0,126
Prdx3 0,004 0,008 0,004 0,008 0,033 0,004 0,004
Prdx4 0,246 0,500 0,122 0,499 2,014 0,254 0,122
Ptgs1 0,987 2,045 0,992 0,993 2,049 1,005 0,992
Sod2 2,775 1,962 2,460 0,996 8,165 0,993 2,826
*
Relative to control group: PBS of same age and genotype
#
Relative to control group: Young PBS, of the same genotype
α
Relative to control group: Young WT PBS

Fig. 6. Leptin increases Sod2 and GPx1 and reduces oxidative stress in the hippocampus of 2xTgAD mice. In vivo leptin administration for 7 days upregulated the
expression of Sod2 (A) and GPx1 gene expression (B) in the hippocampus of 2xTgAD but not in wild type. The data were normalized using four endogenous controls
and are relative to the young WT PBS control group. Data are expressed as mean ± SEM, n = 5 animals per group. *p < 0.05; **p < 0.01.

These results suggest a novel mechanism linking leptin action to treated animals. While only 22.84% of Iba1 cells were diminished in the
BDNF expression in AD, while it also implies the neurogenesis in this WT hippocampus, 52.2% of the cells were diminished in the 2xTgAD
pathology. hippocampus (Fig. 9B).
The morphology of Iba1 immunoreactive microglia was estimated in
3.7. Leptin treatment attenuates the increased number and alters the the hippocampus of 12-months-old mice. Several features were analyzed
morphology of microglial cells resulting from AD in the hippocampus of as endpoints, process length, cell size, cell complexity and cell shape.
2xTgAD elderly animals In all the groups examined, microglia exhibited the typical branched
morphology with small cell bodies, multiple long proximal branches
To verify neuroinflammation in the hippocampus, microglia were extending in all directions and extensive ramification of the distal pro­
immunolabeled with anti-Iba1 and counted (Fig. 9A). For this purpose, cesses. In several areas, the microglia of AD animals appeared less
we chose 12-months-old mice, because, at this stage, the 2xTgAD ani­ branched, with an atrophied appearance (Fig. 10 A). The analysis of
mals already have massive amyloid-beta plaque formation in the hip­ microglial cells in the data set showed significant differences between
pocampus and cortex, high levels of microgliosis, and microglial the groups treated with leptin and the untreated groups, in both geno­
activation as well as alterations in microglia cytokine production types in the analysis of process length and density (Fig. 10 C and F). The
(Babcock et al., 2015; Manocha et al., 2016). branching index (endpoints) increased only in the treated WT animals.
Leptin treatment largely depleted the DG of Iba1+ cells in both ge­ The evaluation of this characteristic also showed that the microglia is
notypes (Fig. 8A and B). After 7 days of treatment with leptin, a few more branched in the 2xTgAD animals compared to the WT strain,
randomly distributed Iba1+ cells were observed in WT animals while in whereas in the 2xTgAD mice treated with leptin, the microglial cells
2xTgAD treated mice, the remaining Iba1+ cells were observed gathered exhibited a lower number of branches when compared with the WT
mainly around the sites of amyloid-beta plaques (Fig. 8C). Interestingly, animals that received leptin (Fig. 10 B).
2xTgAD animals started out having higher numbers of Iba1+ cells No difference was observed after statistical comparisons of the data
compared to WT animals (Fig. 8B), and the Iba1 cell reduction in regarding the geometric structure (fractal dimension) (Fig. 10 D) while
2xTgAd animals treated with leptin was more efficient compared to WT only animals 2xTgAD that received leptin showed an increase in the

9
M.L. Calió et al. Neurobiology of Disease 148 (2021) 105219

Fig. 7. Leptin reduces superoxide production in the hippocampus of 2xTgAD mice. (A) Representative confocal images of superoxide staining in the hippocampus of
WT and 2xTgAD animals treated and not treated with leptin for 7 days. Dihydroethidium staining (DHE - red). The nuclei were stained with DAPI (blue). The ratio
intensity of DHE/DAPI fluorescence was used to calculate the oxidative stress rate. Intense levels of DHE indicate the greater occurrence of superoxide anion for­
mation. (B) Quantification of superoxide anion by the analysis of pixilation in SVZ. A decrease in leptin-treated 2xTgAD animals of both ages was observed when
compared to untreated animals. (C) Quantification of superoxide anion by the analysis of pixilation in the hippocampus. A decrease in leptin-treated aged 2xTgAD
animals was observed when compared to untreated animals. Scale bar = 50μm. Data are expressed as mean ± SEM represented by error bars, n = 5 animals per
group. ***p < 0.001.

span ratio when compared to untreated animals (Fig. 10 E). Changes in leptin levels found in age-related diseases are considered
important factors for the origin of cognitive deficits. Several clinical and
4. Discussion epidemiological studies have addressed the association between low
leptin levels and the risk of AD, supporting the concept that a disrupted
Leptin is known to repress appetite and reduce body weight gain leptin metabolism and/or signaling contributes to the pathogenesis of
(Zhang et al., 2007b) and has been shown to enhance hippocampal AD. In this regard, Power et al. (2001) found lower levels of serum leptin
neurogenesis (Garza et al., 2008; Perez-Gonzalez et al., 2011). Leptin is a in patients with AD underweight than in controls, being the first study
pleiotropic hormone and as an adipokine, it plays multiple functions in correlating Alzheimer’s disease with low leptin. Studies have shown a
reproduction, glucose homeostasis, bone formation, tissue remodeling, strong inverse association of dementia and AD with leptin levels both in
inflammation, as well as in other elements of the endocrine and immune plasma and in cerebrospinal fluid (CFS) (Lieb et al., 2009; Theodor­
systems (Paz-Filho et al., 2010). Interestingly, growing evidence shows opoulou et al., 2012; Bonda et al., 2014). Another study showed lower
that leptin, a peptide hormone classically associated with energy bal­ levels of circulating leptin in AD subjects as well as its strong positive
ance and metabolic control, is also importantly implicated in learning correlation with the severity of dementia (Khemka et al., 2014). Simi­
and memory (Morrison, 2009) performing a direct and indirect role as a larly, several other case-control studies have confirmed that lower
cognitive enhancer, especially in the context of metabolic disorders and serum leptin is associated with AD (Bigalke et al., 2011; Karaoulanis
dementia. et al., 2012). Contradicting this, another study has shown no difference
Since dietary limitation and physical activity have been shown to in serum leptin levels in AD patients compared to controls (Theodor­
improve hippocampal neurogenesis, it is possible that the effects of opoulou et al., 2012). Similarly, distinct research including AD patients
leptin on neurogenesis could be mediated by its action in food intake and and normal controls has failed to find any significant alteration in the
metabolism regulation (Trejo et al., 2001; Lee et al., 2002). However, in leptin levels between these groups (Warren et al., 2012). A negative
our study, the treatment with leptin did not affect the weight of treated relationship between leptin levels and severity of dementia has also been
animals. Garza et al. (2008) reported that bodyweight gain decreased observed by Baranowska-Bik et al. (2015). The results of these studies
without a significant effect on hippocampal neurogenesis after acute show that individuals with low leptin levels had a greater decline in their
leptin injection. Furthermore, it was found that chronic leptin admin­ cognitive ability than those with normal levels (Holden et al., 2009).
istration did not alter locomotor activity (Overton et al., 2001). These We observed that there is a tendency in the decrease of serum leptin
data demonstrate that the action of leptin on neurogenesis maybe is concentration in 2xTgAD animals, although there was no significant
dissociated from its effect on energy homeostasis. difference. Even after treatment, we did not detect a significant
Although there is evidence that leptin could be synthesized in the enhancement in leptin levels in serum samples in mice. Importantly, our
brain, it is known that most of the leptin found in the CNS is derived results seem contradictory to the finding of some of these clinical studies
from peripheral white adipose tissue (WAT) (Campfield et al., 1996). showing that higher leptin levels are related to reduced risk of AD and

10
M.L. Calió et al. Neurobiology of Disease 148 (2021) 105219

Fig. 8. Leptin decreases the accumulation of Aβ in the hippocampus and increases BDNF expression. Leptin-treated 2xTgAD mice exhibit reduced levels of Aβ
deposits (green) compared with control (PBS) 2xTgAD mice. (A) Representative images of Aβ staining in the hippocampus of aged WT and 2xTgAD mice treated with
PBS or leptin. The three planes (X, Y and Z) of Z-stack images (1.5μm/z-stack) were visualized to confirm the extent of cell colocalization or the diameter and area
occupied by the amyloid plaque. Scale bar = 100μm. (B) Quantitative analysis of Aβ shows a lower number of plaques in the brain of 2xTgAd treated mice. The
arrows point at the senile plaques, which are magnified in the inset. Data are expressed as mean ± SEM, n = 5 animals per group. **p < 0.01. (C) Quantification of
areas (percentage) of Aβ plaques in the hippocampus of all groups studied. (D) Quantitative analysis of FJB staining revealed a significant reduction in the number of
labeled cells after leptin treatment, indicating less neurodegeneration. (E) Quantitative analysis of Aβ sizes shows a lower number of larger plaques in the brain of
2xTgAd treated mice. (F) Aged 2xTgAD animals show downregulation of BDNF when compared with WT animals, and leptin administration restores the levels of
BDNF. One-way ANOVA, data are expressed as mean ± SEM, n = 5 animals per group. *p < 0.05; **p < 0.01; ***p < 0.001.

Fig. 9. Leptin attenuates microglial cell number in the hippocampus of WT and 2xTgAD mice. (A) Representative images of Iba-1+ cells, in the hippocampus sections
of aged WT and 2xTgAD animals (B) Quantitative analysis of Iba-1+ stained cells. (C) Higher magnification of amyloid plaques seen in 2xTgAD - PBS animal. One-way
ANOVA, data are expressed as mean ± SEM, n = 5 mice per group, 4-8 sections per animal; ** p < 0.01; *** p < 0.001. Scale bar = 100μm in (A) and 20μm in (C).

11
M.L. Calió et al. Neurobiology of Disease 148 (2021) 105219

Fig. 10. Microglia morphology, evidenced by the density and the presence and the length of secondary/tertiary branches in the hippocampus. (A) Pre-processing of
cell digital image. After a random selection of cells from the tissue picture, the noise was removed by filtering the overall background to get a shape extraction. Next,
the image was changed to grayscale and then transformed into a binary image using the same threshold for all pictures. The binary image was edited to clear the
background and to join all branches so that the cell image would be formed by a continuous set of pixels. Finally, a skeletonized shape and its pairwise outline shape
were used for morphological parameters measures shown (B-F). One-way ANOVA, data are expressed as mean ± SEM, n = 5 mice per group, 4-8 sections per animal;
** p < 0.01; *** p < 0.001. Scale bar = 50μm.

low leptin is a risk factor for AD (Power et al., 2001; Lieb et al., 2009; have been influenced by the supposed greater amount of fat present in
Bigalke et al., 2011; Rizoulis et al., 2005; Khemka et al., 2014) and re­ AD animals. Similarly, Peng et al. (2014) also observed a trend toward
sults from animal studies that suggest that lower plasma leptin levels are decreased levels of leptin levels in transgenic AD mice compared with
related to the role of leptin in memory decline in transgenic mice models the control.
overexpressing APP (Greco et al., 2010; Ishii et al., 2014). Indeed It is known that circulating leptin levels are modulated by fasting or
transgenic models that have the same mutations linked to familial AD, cold exposure (Hardie et al., 1996). As leptin is secreted in a circadian
also exhibit reduced serum leptin levels (Fewlass et al., 2004). manner, the amount of circulating leptin is dynamic and changes
Regarding the data observed in our study, both results may be due to throughout the day according to food intake or energy expenditure
leptin half-life in plasma, which is about 40 min. Also considering that (Ahima et al., 1998; Ramos-Lobo et al., 2017). Since in this study the
the levels of circulating leptin correlate directly with the amount of animals had ad libitum access to water and food, this may have influ­
adipose tissue (Bhatia et al., 2017) and that the weight of the 2xTgAD enced the leptin levels observed here. Moreover, as a trend of decreasing
animals was greater than that of the WT animals, we suggest that the leptin levels was observed in 2xTgAD animals compared to age-matched
discrepancy in terms of circulating leptin observed in this study may controls, we believe that by increasing the n we would obtain similar

12
M.L. Calió et al. Neurobiology of Disease 148 (2021) 105219

results to those shown in previous studies. 2009; Sahay et al., 2011). Despite this, the increase in neurogenesis may
Although our findings appear to be contradictory to most previous not be sufficient to compensate the damages that occur in this pathology
studies, a few studies also demonstrate that no difference in leptin levels (Jin et al., 2004a).
was observed between the different genotypes (Peng et al., 2014; Lee Since the hippocampus is one of the first areas to be affected in AD,
et al., 2018; Yeh et al., 2020) and some clinical studies have found no most studies about neuroregeneration examine the involvement of SGZ-
apparent link between plasma leptin levels and AD (McGregor and born neuroblasts in brain repair, and neurogenesis at the SVZ has not
Harvey, 2018). In fact, Maioli et al. (2015) demonstrated alterations in been widely investigated so far mainly in terms of possible regulation of
brain leptin signaling despite unchanged CSF leptin levels in AD. Lee adult neurogenesis by leptin. Therefore, in the present study, one of our
et al. (2018) observed that the level of leptin was comparable between questions was whether the neurogenic action of leptin seen in SGZ could
WT and the same AD model used in our study. Another recent study also occur similarly in SVZ, which could help replace damaged cells and/or
showed that the fasting levels of leptin were not different among WT and prevent neurodegeneration. If the progression of the disease is related to
the transgenic AD model (Yeh et al., 2020). One possible reason for this the loss of the functional capacity of the NSCs, since neuroregeneration
divergence is that the plasma levels of leptin may not be a good indicator could be affected in several regions of the brain and not only in the
of leptin levels in the CNS once altered blood-brain transport of leptin hippocampus, the discovery of therapeutic strategies that increase
has been documented in AD (Dietrich et al., 2008). Besides serum leptin neurogenesis or prevent the decrease of the proliferative process in these
levels may not reflect the intracerebral concentration of that peptide. areas could be a new treatment option for neurodegenerative diseases.
The determination of the levels of these peptides in the CSF requires an In addition, SVZ NSCs are better characterized than SGZ in terms of
invasive procedure and for this reason it was not evaluated in our functional responses such as proliferation and migration, both in phys­
investigation. Despite this, new studies would be helpful in identifying iological and pathological conditions (Fainstein et al., 2018). Besides,
whether changes in leptin levels correlate and track with disease pro­ the neurodegeneration in AD affects the entire neocortex and several
gression and worsening AD pathology. While there is potential for leptin areas of the brain, rather than just the hippocampus, highlighting the
to serve as both a novel therapeutic intervention and diagnostic target, need to study NSCs populations from different niches, including the SVZ,
particularly early in AD (Tezapsidis et al., 2009), additional studies are SGZ, and others.
clearly needed to test the clinical utility of leptin in AD. For this reason, we analyzed two neurogenic niches and evaluated
In the adult brain, neurogenesis occurs in the SGZ of the hippo­ whether leptin would be able to regulate adult neurogenesis both in the
campal dentate gyrus and the SVZ of the lateral ventricles. While SVZ and the SGZ. Since neurogenesis is tightly related to AD, under­
migration of the newly generated cells in the SGZ is restricted to the standing the mechanisms of alterations in this process will contribute to
hippocampus, where they participate in learning and memory forma­ understanding AD pathophysiology.
tion, cells from the SVZ are destined to renew interneurons of the ol­ Here, we have demonstrated that the administration of leptin in­
factory bulb (Kriegstein and Alvarez-Buylla, 2009). SVZ-migrating NSCs creases the proliferation of neural progenitors in the main neurogenic
are thought to participate in the attempt to regenerate an injured area niches and reduces neurodegeneration, even in the transgenic AD mouse
(Conover and Notti, 2007; Clelland et al., 2009; Deng et al., 2009; model of 12 months of age, thereby suggesting by this mechanism that
Garthe et al., 2009; Mundim et al., 2018) including neurological diseases dead or damaged neurons could be replaced.
such as AD (Chen et al., 2012; Fainstein et al., 2018). We studied gene expression of some proliferation and differentiation
Previous studies have shown that neurogenesis happens at a dimin­ markers expressed in the brain, asking whether their expressions were
ished rate with the advancing of age, impacting its physiological role in modified in the transgenic mice and if leptin could alter this pattern.
neuronal replacement and consequently its ability to participate in the Leptin induced an increase in gene expression of DCX, a highly
repair mechanisms triggered by injury or disease (Enwere et al., 2004; expressed gene in neuroblasts, which was found at low levels in all
Katsimpardi and Lledo, 2018; Kuhn et al., 2018). Despite this, the aged groups of 2xTgAD animals prior to treatment, although no difference
brain partially retains its neurogenic capacity through different mech­ was observed between the genotypes. Increased PCNA expression was
anisms (Kempermann et al., 2002; Moreno-Jimenez et al., 2019). In only observed in WT animals of both ages, whereas increased Ki67 and
conditions like AD, which occurs at an increasing frequency with nestin expression was also be observed in transgenic animals. Interest­
advancing age, the ability of the brain to mobilize new neurons opens ingly, some genes related to proliferation showed a lessened response to
novel possibilities for cell-replacement and provides a promising ther­ leptin-evoked expression. This data suggests a possible interaction be­
apeutic methodology. Numerous studies have demonstrated that tween age and genotype factors. We hypothesize that certain prolifera­
increasing neurogenesis might promote functional recovery in the tive genes are more activated in the presence of Aβ plaques, which are
damaged area in the brain (Jin et al., 2002; Jin et al., 2004a; Kern et al., only found in aged AD models. In this sense, it is known that PS1 reg­
2011; Diaz-Moreno et al., 2013). ulates NSCs differentiation both in vitro and in vivo (Demars et al., 2011)
It is known that the neurogenic process can be triggered by physio­ while sAPPα acts as a proliferation factor on NSCs in the adult brain
logical factors, such as growth factors and environmental enrichment, (Demars et al., 2013; Baratchi et al., 2012).
and by pathological processes, including trauma and neurodegeneration Unlike expected by the effect of leptin on cell proliferation, this
(Perez-Gonzalez et al., 2011; Mundim et al., 2018). In vivo and in vitro hormone decreased the gene expression of GFAP, which was twice as
studies have shown that leptin stimulates neurogenesis in the hippo­ high in the aged 2xTgAD animals compared to WT and treated animals.
campal dentate gyrus by increasing cell proliferation (Garza et al., 2008) This fact suggests that the decrease in GFAP is linked to the reduction of
and neuroprotection (Weng et al., 2007; Zhang et al., 2007b). Adult astrogliosis observed in aged transgenic AD animals.
hippocampal neurogenesis is a highly plastic process that responds The decreased levels of neurotrophins have been found in AD in prior
swiftly to neuronal activity. Adult hippocampal neurogenesis can be studies (Stopa et al., 1990; Hock et al., 2000). This environment
regulated at the level of neural stem cell recruitment and activation, impaired the maturation of NSCs into differentiated neurons. Our results
progenitor proliferation, as well as newborn cell survival and differen­ of gene expression contrast with prior findings from Jin et al. (2004b),
tiation (Pineda and Encinas, 2016). from the hippocampus of AD patients, showed increased, rather than
A dysfunction in hippocampal neurogenesis has been observed in impaired, expression of the immature neuronal marker. These dissimi­
multiple models of AD and human AD brain tissue and may be related to larities raise the possibility that different forms of AD may diverge
the cognitive and memory dysfunction (Yamashita et al., 2006; Demars concerning their association with neurogenesis, particularly considering
et al., 2010; Winner et al., 2011; Perry et al., 2012; Moreno-Jimenez the sporadic form of AD.
et al., 2019), whereas the increase of endogenous neurogenesis can However, it is not known whether the newly generated neurons can
improve the spatial memory in experimental models (Mirochnic et al., differentiate into mature neurons. Furthermore, using qPCR, no

13
M.L. Calió et al. Neurobiology of Disease 148 (2021) 105219

differences were observed in the gene expression of the mature neuronal To determine whether the genotype and leptin treatment could also
markers (βIII-tubulin, Map2 and NeuN) when leptin was used, and lower affect differentiation of neural stem cells in neuroblasts or astrocytes, or
expression was observed in the aged 2xTgAD animals compared to even affect the proliferation of neural progenitors, cell differentiation
young 2xTgAD and aged WT mice. These findings are consistent with was assessed using DCX and GFAP labeling.
previous report that showed a decrease in MAP2 expression in the Our results regarding the coexpression of BrdU with DCX in the SGZ
dentate gyrus in AD (Li et al., 2008). indicated that the number of these coexpressing cells decreased with age
This is likely to be due to the fact that the one-week period used for in both genotypes. At the age of 12 months, the number of BrdU/DCX+
treatment is not sufficient for the complete neuronal maturation process. cells decreased in the DG of the AD mice model when compared to the
Thus, although we have observed an increase in neuroproliferation, we WT animals. These results show that there is a decreased generation of
cannot affirm that the newly generated neurons have differentiated into DG neuroblasts during the progression of AD. Namely, AD temporally
mature neurons. In this sense, previous report showed that the decrease declines neurogenesis. On the other hand, leptin treatment has been
in MAP2 expression in the AD dentate gyrus is mainly attributable to an shown to increase the number of BrdU/DCX coexpressing cells in both
interruption in the maturation of developing immature granular cells as genotypes and ages.
a reaction to changes in the AD microenvironment (Li et al., 2008). Concerning SVZ, the quantity of BrdU/DCX+ cells did not vary ac­
Most studies of the pathogenesis of AD have focused on the degen­ cording to age or between genotypes. Also, our data showed increased
eration of existing synapses and neuronal cells and only a few studies expression of the immature neuronal marker DCX in the SVZ of treated
explore the possibility of a defect or decreased capacity in the produc­ mice from both ages and genotypes.
tion of new neural cells from NSCs. Whether enhanced neurogenesis would be beneficial or not fully
Neurogenesis in the hippocampus plays an important role in the understood. It is important to know what occurs when neuronal activity
maintenance and improvement of memory and learning (Shors et al., is increased leading to abnormal hyperexcitatory levels. Pineda and
2001; Snyder et al., 2005). Thus, disruption of neurogenesis in the Encinas (2016) discuss the effects of neuronal hyperexcitation on hip­
dentate gyrus can be involved in the age-associated decline of hippo­ pocampal neurogenesis. In this sense, the contribution of aberrant
campal learning and memory, also observed in AD. The balance between neurogenesis is seen to be linked to the generation of neurons with
neurogenesis and neurodegeneration is thought to be critical for the ectopic location and mossy fiber sprouting (Parent et al., 1997), which
maintenance of these functions (Gould et al., 1999). However, these are features evidenced by epilepsy and show little effect in AD. Several
cognitive abilities require not only the birth of new neurons to replace studies have shown that increased neurogenesis, especially in the hip­
the lost cells but also the differentiation of these newborn cells. The pocampus, is useful in AD cases, improving cognition and memory since
present study demonstrates for the first time the differences between WT adult neurogenesis is a form of hippocampal plasticity with NSCs
and 2xTgAD animals on neurogenesis in different niches that are bearing little resemblance to their mature counterparts (Deng et al.,
compromised in AD. Besides this, we also evaluated the effect of leptin, 2009; Greco et al., 2010; Kim et al., 2010; Maioli et al., 2012; Perez-
addressing the genotype effects. Gonzalez et al., 2014; Akers et al., 2014; Ghasemi et al., 2016). More­
Using immunohistochemistry, we found that the proliferation of cells over, there is significant cross-talk between many of the important
is dramatically decreased in both SVZ and SGZ in AD. Our results indi­ proteins that regulate the progression of familial AD and adult neuro­
cate that NSCs from Alzheimer’s disease mice exhibit decreased func­ genesis (Lazarov and Marr, 2010, 2013). PS1, for example, regulates
tional properties in terms of their ability to proliferate, even in the early- NSCs differentiation both in vitro and in vivo (Demars et al., 2011), while
onset of disease in SVZ, when compared to WT animals. sAPPα acts as a proliferation factor on NSCs in the adult brain (Demars
This loss of proliferative capacity is accentuated with advancing age et al., 2013; Baratchi et al., 2012).
in both genotypes and not only in a late-onset neurodegenerative dis­ Chronic neuroinflammation is also common to nearly all neurode­
ease, as would be expected. In 2xTgAD animals, NSCs proliferative generative diseases, and it contributes to their pathophysiology (Heneka
function has been shown to decrease at the age of 3 months (young et al., 2014). Astrocyte cells are capable of local proliferation under
animals), a time point before Aβ accumulation in the brain. At the age of certain conditions, with indications that their progeny may resume
12 months, a time point with advanced Alzheimer’s pathology, the normal physiological function over time (Bardehle et al., 2013; Buffo
proliferation of NSCs proves to be even more diminished and dysfunc­ et al., 2008). It is known that injury and degeneration can cause astro­
tional. Interestingly, leptin is able to increase cell proliferation in SVZ in gliosis and subsequent astrocyte proliferation, although the exact mo­
both genotypes, regardless of the age of the animals. lecular stimulus of proliferation is currently unknown (Doetsch, 2003).
Concerning the SGZ, dramatically reduced proliferation in the den­ Nevertheless, although anti-inflammatory and immunosuppressive
tate gyrus has been observed in aged transgenic mice for the mutated therapies have demonstrated some efficacy in neurodegenerative dis­
form of APP, although a reduction in proliferation has also been noticed ease models, these treatments have largely failed in the clinic (Arvani­
in aged WT animals when compared to young WT animals but less takis et al., 2008; Wyss-Coray and Rogers, 2012).
markedly. Concerning SGZ, DCX expression gradually decreases with
Here we show that overexpression of mutant APP and PS1 in the advancing age, but GFAP expression increases mainly in 2xTgAD ani­
transgenic mice reduces the proliferation of neural progenitors in the mals, and the two markers seem to have contrary expression patterns
hippocampus of aged animals. However, we observed that leptin in­ with aging. Since GFAP is also an indicator of NSCs, it is uncertain
creases the number of proliferative cells both in the aged WT and young whether the BrdU/GFAP+ cells that we have identified are proliferative
and aged double transgenic animals. Thus, collectively, the fact that we NSCs or reactive astrocytes. Given the strong GFAP staining as well as
observed a decrease in the proliferation of NSCs even before the onset of consistent results from previous studies (Schmitz et al., 2004; Lee et al.,
AD suggests that mutations linked to AD may have deleterious proper­ 2014; Zeng et al., 2016), it is likely that they are the latter, but we cannot
ties in neurogenesis, regardless of the effect of Aβ deposition. These data conclusively classify them as reactive astrocytes without further anal­
suggest that in addition to advancing age, both the expression of ysis. Also, it is known that astrogliosis is found in AD and other neuro­
transgenes and the brain environment of AD can impact the proliferation degenerative diseases (Wright et al., 2013; Hostenbach et al., 2014).
of NSCs. Indeed, the neurogenic process in the adult brain could also be Reactive astrocytes play critical roles in modulating neuroinflammation
viewed as one form of neural plasticity, being triggered by many factors and oxidative stress, which are among the main aspects of the patho­
that are not only due to injury or situations that require cell replace­ genesis of AD (Agostinho et al., 2010). Consequently, targeting neuro­
ment. In fact, several studies summarized the idea that the pathology of inflammation in the early stages is considered to be one of the
AD may reflect the fundamental failure of neural plasticity (Mesulam, methodologies to treat or delay the progression of AD.
1999; Haughey et al., 2002; Arendt, 2005; Kizil and Bhattarai, 2018). In this sense, our data show that leptin treatment may decrease

14
M.L. Calió et al. Neurobiology of Disease 148 (2021) 105219

astrogliosis in aged 2xTgAD animals as seen by the exacerbated prolif­ and memory storage in the hippocampus (Lu et al., 2013). More
eration in the hippocampus of these animals which is a consequence of recently, it has also been described as important in the central control of
the neurodegenerative lesions found in this pathology. Our findings are energy homeostasis (Rios, 2013; Vanevski and Xu, 2013). BDNF is
consistent with the results of Garcia et al. (Garcia et al., 2014), which widely distributed in the brain, being highly expressed in the hippo­
observed a large number of GFAP+ cells in the hippocampus of this campus, cortex, and basal forebrain, where it is important for learning,
animal model at the age of 9 and 12 months, compared to WT mice. In memory, and higher thinking (Huang and Reichardt, 2001; Yamada and
addition, in both the 2xTgAD and WT animals, of both ages, there were Nabeshima, 2003; Voineskos et al., 2011) and in several hypothalamic
no significant changes in the number of BrdU/GFAP+ within the SVZ. nuclei, where it also regulates feeding and body weight (Rios, 2013).
Leptin receptors are highly expressed in the CA1 and CA3 regions of Although BDNF is needed in the developmental stages, BDNF levels have
the hippocampus, as well as in the DG (Mercer et al., 1996; Shanley been shown to decrease in tissues with aging (Tapia-Arancibia et al.,
et al., 2002). Leptin receptor-deficient animal models (db/db mice) 2008). In humans, decreased hippocampal volume is associated with
exhibit intense deficits in spatial memory, learning and insufficient short decreasing BDNF levels, suggesting there is a relationship that might
term memory processing (Li et al., 2002; Farr et al., 2006; Oomura et al., explain some of the cognitive declines that occur during aging (Erickson
2006) et al., 2010; Buchman et al., 2016). Changes of BDNF level and mRNA
To further characterize the association between leptin and neuro­ expression have been also reported in brain samples as well as blood of
genesis in AD, we assessed the mRNA expression and the presence of the AD patients, which also indicates a potential role of BDNF in the path­
long form of leptin receptor (LepRb) in 2xTgAD and wild-type (WT) ogenic process of AD (Huang and Reichardt, 2001; Peng et al., 2009;
leptin-treated mice at different ages and analyzed the co-localization of Angelucci et al., 2010; Laske et al., 2011; Voineskos et al., 2011; Francis
LepRb with the marker of neural differentiation in the neurogenic et al., 2012a; Ventriglia et al., 2013; Nagata et al., 2014; Prakash and
niches. Gene expression analysis by qPCR for LepR in the hippocampus Kumar, 2014; Song et al., 2015; Li et al., 2016; Marsh and Blurton-Jones,
showed no difference between groups, regardless of age or genotype. 2017). Correspondingly, higher blood BDNF levels seem to delay the
Besides, leptin treatment increased the expression of its receptor on all rate of cognitive decline in AD (Bollen et al., 2013). As early as 1991, it
treated groups. The overexpression of LepRb suggests its involvement in was found that BDNF mRNA decreased in the hippocampus of in­
the observed improvement. Double staining with LepR/DCX showed dividuals with AD, which suggested that BDNF may be related and
that some cells coexpressed these markers, in the two areas analyzed. contribute to the progression of cell loss in AD (Phillips et al., 1991).
It is well accepted that the environment of the AD brain is toxic to Compared with aged controls, BDNF protein expression, as well as
neural cells mainly due to the formation of Aβ oligomers, which can mRNA levels, were found decreased in hippocampus and frontal, pari­
trigger AD pathogenesis (Hardy and Selkoe, 2002; Rapoport et al., 2002) etal, and temporal cortex of AD postmortem samples (Connor et al.,
and can be one of the reasons why there is a limitation on the ability to 1997; Hock et al., 2000; Holsinger et al., 2000; Michalski and Fahne­
repair neuronal death via neurogenesis in AD (Haughey et al., 2002; stock, 2003; Peng et al., 2005), meanwhile, similar results are also
Zhang et al., 2007a). 2xTgAD mice brains showed that the Aβ plaques replicated in AD animal models (Francis et al., 2012b; Kim et al., 2010;
were localized in the hippocampus and entorhinal cortex (data not Li et al., 2009; Maioli et al., 2012; Meng et al., 2013; Naert and Rivest,
shown). In addition, Caille et al. (2004) established another theory that 2012; Peng et al., 2009). In addition, another study demonstrated that
sAPP (soluble amyloid precursor protein) regulates the proliferation of the induction of Alzheimer’s disease model leads to a decrease in BDNF
NSCs responsive to EGF in SVZ due to the lack of sAPP-binding sites in mRNA levels in the hippocampus (Smith et al., 1995).
the SGZ, thereby causing regional differences in adult neurogenesis. BDNF functions depend not only on its site of action but also on a
Therefore, it might be probable that this regional difference in neuro­ complex array of metabolic and environmental factors that regulate its
genesis was reliant on the amount of Aβ deposition. expression in a brain region-specific manner. It is currently accepted
Leptin has been shown to reduce brain Aβ levels in 6-months old that peripheral metabolic cues such as leptin increases BDNF mRNA
CRND8 transgenic mice following 8 weeks of treatment (Greco et al., expression in the nucleus of the hypothalamus. Leptin is an adipocyte-
2010). Our findings presented here indicate that leptin administration derived hormone that acts as an anorexigenic stimulus and it is known
reduced the accumulation of Aβ deposits in 12 months-age 2xTgAD to acutely induce BDNF gene expression in the ventromedial nucleus of
mice, coinciding with previously published studies (Perez-Gonzalez the hypothalamus of fasted mice (Komori et al., 2006; Unger et al.,
et al., 2011; Perez-Gonzalez et al., 2014). Recent findings suggest that 2007) but is unclear whether leptin regulates mRNA BDNF in the hip­
the inhibitory effect of this hormone concerning Aβ accumulation is pocampus, as a modulator of neural activation. In our experiments,
presumably attributed to its inhibitory action of APP processing through changes in hippocampal BDNF gene expression were present in the aged
the amyloidogenic pathway, regulating BACE expression, the most treated group in which significantly higher levels were observed.
important β-secretase involved in APP processing (Perez-Gonzalez et al., However, it is unlikely that leptin alone contributed to the increase in
2014). In the present study, we found that AD model animals treated BDNF expression observed in these animals.
with leptin had a greater number of smaller plaques. However, mice in In a previous study, Guo et al. (2008) demonstrated that chronic
this group exhibited larger plaques at a lower frequency than untreated intraventricular leptin infusion is capable of protecting the neurons in
animals. Our treatment with leptin, by inducing the modulation of the the hippocampus from cell death induced by neuronal insults (Guo et al.,
immune response and decreasing the size of senile plaques, demon­ 2008). Besides the formation of Aβ plaques, we evaluated the neuro­
strated the capacity to inhibit Aβ accumulation. Additionally, our data degeneration in leptin-treated and untreated mice. In this sense, our data
support the observations of Greco et al. (2010) who revealed that 8 are contradictory to Garza et al. (2008) who observed that leptin did not
weeks of leptin treatment significantly reduced the amyloid burden in show effects on cell survival, but are consistent with the findings of
the hippocampus of 6-month old TgCRND8 mice, compared to age and Perez-Gonzalez et al. (2014) who used lentivirus gene delivery to
gender-matched saline-treated transgenic mice. This result was parallel overexpress leptin in the brains of AD animals. Curiously, they also
to a decrease in the average size of plaques also analyzed in this study. observed that leptin significantly reduced neuronal degeneration in the
Therapeutic explorations on AD have been constantly conducted on hippocampus of the AD model, also suggesting that leptin has a neuro­
the pathogenesis and subsequently tailored therapeutic intervention, protective effect.
especially in the neurobiological field, and the discovery of neuro­ It is known that oxidative stress can initiate the process of apoptosis
trophins (NTs) was a milestone, which provided a new perspective into and that abnormalities in the regulation and expression of antioxidant
neurogenesis and neuron survival (Bothwell, 2014). Brain-derived enzymes may have a role in mechanisms of CNS aging and neuro­
neurotrophic factor (BDNF) is the most widely distributed NT in the degeneration. In fact, neurons are at particular risk from damage caused
adult brain and has key roles in neuronal survival, synaptic plasticity by free radicals because of high rates of oxygen consumption and high

15
M.L. Calió et al. Neurobiology of Disease 148 (2021) 105219

contents of polyunsaturated fatty acids (Zemlan et al., 1989; Uysal et al., reactive, which could justify the fact that we observed a more evident
2012; Radi et al., 2014). Several lines of evidence point that the mo­ result to treatment with leptin in these animals.
lecular mechanisms that link AB to the onset of neurotoxicity are the Taken together, these results suggest that Sod2 protects the aging
production of reactive oxygen species induced by the presence of this brain against oxidative stress in AD by controlling superoxide over­
oligomer. Actually, the oxidative stress resulting from the production of production and that leptin treatment can restore this antioxidant
ROS is considered a common effector of the cascade of degenerative pattern. Moreover, the significant decrease in superoxide anion (O-2)
events in many neurological disorders (Cimini et al., 2009). production observed after treatment indicates that leptin also has anti­
In the present study, we also evaluated the action of leptin on gene oxidant action in AD.
expression of antioxidant enzymes. Besides, we checked whether leptin Microglial activation has been considered a crucial player in the
was able to decrease previously existing levels of superoxide in the pathological process of multiple human neurodegenerative diseases
neurogenic niches of AD animals. Although the main cause of neuronal (Heneka et al., 2015; Navarro et al., 2018). In this study, we briefly
death in AD is the formation of senile plaques, in vivo evidence showed highlight our data about neuroinflammation in the hippocampal area,
that mitochondrial dysfunction and redox imbalance triggers or worsens including microgliosis and astrogliosis. Recent studies mention that
AD-related neuronal deficits (Calon et al., 2004; Lustbader et al., 2004; Iba1is widely expressed in microglia of the hippocampus and cortex of
Beal, 2005). amyloidogenic transgenic models (Unger et al., 2018; Navarro et al.,
Studies suggest that there are multiple mechanisms by which 2018). In these mice, microglial cells clustered primarily at sites around
oxidative stress may accumulate and create dysfunctional neuronal re­ amyloid-beta plaques (Nimmerjahn et al., 2005; Unger et al., 2018)
sponses in AD and that the development of the AD phenotype requires supporting our interpretation of Iba-1 staining in the hippocampus of
multiple insults (Zhu et al., 2004). Since our previous results demon­ aged 2xTgAD mice. We observed that the number of microglial cells was
strate the ability of leptin to decrease neurodegeneration in animal higher in the 2xTgAD animals compared to WT mice. In this sense, leptin
models of AD, we decided to investigate whether leptin could improve was able to attenuate the number of Iba1+ cells, compared to the PBS
the existing oxidative stress profile in this pathology. In order to obtain a group, and that the number of cells labeled after treatment was similar
better insight into the ROS scavenging defenses of the hippocampus of to the WT animals. On the other hand, in the WT-leptin group, the Iba-1
Tg mice, we studied GPx1 and SOD2. We have demonstrated the exis­ immunoreactive microglia was similar to the PBS group. Total microglia
tence of significantly abnormal increased superoxide generation and the number in both Alzheimer’s patients and animal models is increased and
downregulation of Sod2 expression in the hippocampus of aged 2xTgAD, correlates with disease severity and progression (Olmos-Alonso et al.,
suggesting a negative correlation between cellular superoxide produc­ 2016). This suggests that disease pathology promotes microglial
tion and Sod2 decline in advanced AD. proliferation.
The analysis of gene expression shows relevant differences between This corroborates a previous publication that attests that leptin de­
treated and untreated Tg animals of both ages for the two genes creases the level of activated microglia in the hippocampus 1 day after
evaluated. transient ischemia (Yan et al., 2011), indicating that this hormone is also
The literature shows that there is no difference in the expression of effective in reducing this neuroinflammatory marker.
some genes related to the oxidative response in the hippocampus of AD It is known that AD brains exhibit at least two different morpho­
models. Several studies have shown that there are different reactions in logical microglial phenotypes. Morphologically, microglia are classified
response to the perturbation of the redox status, depending on the as ramified (quiescent/resting), or ameboid/phagocytotic (activated),
specific brain area. In this sense, it is known that the cortex is more (Tremblay et al., 2011). The cells associated with the amyloid plaques
reactive while the hippocampus is more prone and susceptible earlier to display an activated amoeboid-like phenotype while those present in
the oxidative insult (Scheff et al., 2007; Cimini et al., 2009). other regions have a homeostatic-like morphology (Krabbe et al., 2013).
In this perspective, our results corroborate the findings of Cimini Microglia behavior in AD may also depend heavily upon the stage of the
et al. (2009) who observed that there is no difference in the levels of disease (Clayton et al., 2017). In accordance with these studies, in
mRNA of GPx1 or SOD2 in the hippocampus of WT animals and trans­ 2xTgAD animals we observed that the microglia had heterogeneous
genic AD mice. The cortex is physiologically more exposed to ROS, and morphology, from the branched homeostatic state to atrophied and
therefore basally endowed with more effective antioxidant defenses. In amoeboid forms, depending on the observed region. Indeed, regional
this perspective, the increases of SOD2 and GPX1 in the cortex of AD heterogeneity of microglia could be also observed during homeostasis.
animals observed by Cimini et al. (2009) may be interpreted as a During pathology, disease-associated microglia emerge with distinct
compensatory response to an early perturbation of the redox status. This transcriptional profiles that reflect specific activation states (Priller and
response likely consists of stabilization and/or activation of the enzy­ Prinz, 2019).
matic proteins, rather than induction, since the corresponding mRNA Microglial activation plays a neuroprotective role and could improve
levels were unchanged. Differential effects of oxidative stress on tran­ AD pathology by reducing Aβ levels in APP-based models (Michaud
scriptional or post-transcriptional regulation of antioxidant enzymes et al., 2013). Differently, chronic activation that occurs from excessive
have been previously reported (Rohrdanz et al., 2001). neuronal damage in neurological diseases can lead to the constant
We also found that the fluorescence intensity of DHE staining was release of pro-inflammatory molecules and harmful production of ROS
increased in non-treated aged 2xTgAD in comparison to WT and that the which results in adverse effects. Moderate increases in these cytokines
superoxide levels decreased after treatment with leptin in these animals, are generally considered a normal part of aging (Clayton et al., 2017).
in both niches analyzed. Besides, no difference was found in the WT However, large increases, as observed in AD, lead to excessive neuro­
group. Surprisingly, in SVZ, the antioxidant action of leptin was detected toxicity that drives the neurodegenerative process and, in consequence,
in younger animals, whereas in the hippocampus, the same result could could be implicated in the pathology of AD (Spangenberg et al., 2016;
only be observed in the aged animals. Interestingly, the hippocampus Clayton et al., 2017; Navarro et al., 2018).
and the subventricular zone show a different behavior, making it According to Navarro et al. (2018), the high accumulation of Aβ
possible to hypothesize a higher susceptibility of the former area to the produces a fast microglial response. In this same study, it was observed
oxidative insult in AD (Scheff et al., 2007) and also to leptin treatment. that in samples from patients in advanced stages of AD, there was a
We believe that leptin’s role in decreasing DHE is due to its immuno­ degenerative process of microglial cells, which compromised the role of
modulatory capacity through microglia, which is responsible for the the immune response to damage, mainly in the dentate gyrus (Navarro
production of ROS in the hippocampus of aged 2xTgAD mice as well as et al., 2018). Interestingly, our data showed that in aged 2xTgAD mice,
astrocytes (von Bernhardi et al., 2015). With aging and the deposition of leptin also induced some morphological changes in microglia.
Aβ, the microglia’s response becomes exaggeratedly dysregulated and Considering that microglial cells are implicated in many beneficial

16
M.L. Calió et al. Neurobiology of Disease 148 (2021) 105219

functions, such as Aβ phagocytosis, senile plaque compaction and limi­ Severino (Universidade Federal de São Paulo) for technical assistance.
tation of Aβ toxicity and elimination of damaged neurons (Yuan et al.,
2016; Ulland et al., 2017), an impaired microglial response could Appendix A. Supplementary data
exacerbate the progression of pathology. On the other hand, there is also
abundant evidence that microglia can be harmful to neurons by Supplementary data to this article can be found online at https://doi.
secreting factors to injure these cells (Colonna and Butovsky, 2017; org/10.1016/j.nbd.2020.105219.
Liddelow et al., 2017), suggesting a possible participation of this hor­
mone in the modulation of the immune response in AD. Promisingly, References
microglia activation seems to be an important target reached by leptin
treatment. Agostinho, P., Cunha, R.A., Oliveira, C., 2010. Neuroinflammation, oxidative stress and
the pathogenesis of Alzheimer’s disease. Curr Pharm Des 16, 2766–2778. https://
Our results suggest that leptin has several important actions in the doi.org/10.2174/138161210793176572.
brain: first, it has a stimulatory effect on the proliferation and differ­ Ahima, R.S., Prabakaran, D., Flier, J.S., 1998 Mar 1. Postnatal leptin surge and regulation
entiation of neuronal progenitors and may also have an immunomod­ of circadian rhythm of leptin by feeding. Implications for energy homeostasis and
neuroendocrine function. J Clin Invest. 101 (5), 1020–1027. https://doi.org/
ulatory effect. Second, it has neuroprotective and antioxidant capacity 10.1172/JCI1176. PMID: 9486972; PMCID: PMC508653.
by reducing amyloid-beta plaques, neurodegeneration and oxidative Akers, K.G., Martinez-Canabal, A., Restivo, L., Yiu, A.P., De Cristofaro, A., Hsiang, H.L.,
stress. These data demonstrate the utility of leptin as a potential thera­ Wheeler, A.L., Guskjolen, A., Niibori, Y., Shoji, H., Ohira, K., Richards, B.A.,
Miyakawa, T., Josselyn, S.A., Frankland, P.W., 2014. Hippocampal neurogenesis
peutic approach for AD, although essential features are needed to ensure regulates forgetting during adulthood and infancy. Science (80-.) 344, 598–602.
its translation to the clinic in the future. https://doi.org/10.1126/science.1248903.
Altman, J., Das, G.D., 1965. Autoradiographic and histological evidence of postnatal
hippocampal neurogenesis in rats. J Comp Neurol 124, 319–335. https://doi.org/
5. Conclusions
10.1002/cne.901240303.
Alvarez-Buylla, A., Lim, D.A., 2004. For the long run: maintaining germinal niches in the
In summary, our results suggest that leptin increases the production adult brain. Neuron 41, 683–686. https://doi.org/10.1016/s0896-6273(04)00111-
4.
of newborn cells in two neurogenic niches even in the aged AD brain.
Angelucci, F., Spalletta, G., di Iulio, F., Ciaramella, A., Salani, F., Colantoni, L., Varsi, A.
These data sustain an unusual role of leptin in the course of adult neu­ E., Gianni, W., Sancesario, G., Caltagirone, C., Bossu, P., 2010. Alzheimer’s disease
rogenesis and neurodegeneration in other neurogenic niches than the (AD) and Mild Cognitive Impairment (MCI) patients are characterized by increased
hippocampus, providing new understandings into the process of BDNF serum levels. Curr Alzheimer Res 7, 15–20. https://doi.org/10.2174/
156720510790274473.
neurogenic control in dementia. Future efforts are needed to clarify Arendt, T., 2005. Alzheimer’s disease as a disorder of dynamic brain self-organization.
which signaling mechanisms are involved in the neurogenic and neu­ Prog Brain Res 147, 355–378. https://doi.org/10.1016/S0079-6123(04)47025-3.
roprotective function of this hormone. Our data also provide the basis Armstrong, R.A., 2011. The pathogenesis of Alzheimer’s disease: a reevaluation of the
amyloid cascade hypothesis. Int J Alzheimers Dis 2011, 630865. https://doi.org/
for further analysis of the role of leptin, as an alternative or adjuvant, to 10.4061/2011/630865.
slow down or delay the central pathology associated with AD. Arvanitakis, Z., Grodstein, F., Bienias, J.L., Schneider, J.A., Wilson, R.S., Kelly, J.F.,
Evans, D.A., Bennett, D.A., 2008. Relation of NSAIDs to incident AD, change in
cognitive function, and AD pathology. Neurology 70, 2219–2225. https://doi.org/
Funding 10.1212/01.wnl.0000313813.48505.86.
Babcock, A.A., Ilkjaer, L., Clausen, B.H., Villadsen, B., Dissing-Olesen, L., Bendixen, A.T.,
This work was supported by Fundação de Amparo à Pesquisa do Lyck, L., Lambertsen, K.L., Finsen, B., 2015. Cytokine-producing microglia have an
altered beta-amyloid load in aged APP/PS1 Tg mice. Brain Behav Immun 48,
Estado de São Paulo – FAPESP (grant numbers 2014/24341-9 and 2015/
86–101. https://doi.org/10.1016/j.bbi.2015.03.006.
19231-8), Conselho Nacional de Desenvolvimento Científico e Tec­ Baranowska-Bik, A., Bik, W., Styczynska, M., Chodakowska-Zebrowska, M.,
nológico – CNPq (grant number 404646/2012-3), and Coordenação de Barcikowska, M., Wolinska-Witort, E., Kalisz, M., Martynska, L., Baranowska, B.,
2015. Plasma leptin levels and free leptin index in women with Alzheimer’s disease.
Aperfeiçoamento de Pessoal de Nível Superior – CAPES (Finance Code
Neuropeptides 52, 73–78. https://doi.org/10.1016/j.npep.2015.05.006.
001). Baratchi, S., Evans, J., Tate, W.P., Abraham, W.C., Connor, B., 2012. Secreted amyloid
precursor proteins promote proliferation and glial differentiation of adult
Credit And Author Contributions Statement hippocampal neural progenitor cells. Hippocampus 22, 1517–1527. https://doi.org/
10.1002/hipo.20988.
Bardehle, S., Kruger, M., Buggenthin, F., Schwausch, J., Ninkovic, J., Clevers, H.,
Michele L. Calió conceptualized the project, designed methodology, Snippert, H.J., Theis, F.J., Meyer-Luehmann, M., Bechmann, I., Dimou, L., Gotz, M.,
carried out the immunohistochemistry, qPCR experiments, performed 2013. Live imaging of astrocyte responses to acute injury reveals selective
juxtavascular proliferation. Nat Neurosci 16, 580–586. https://doi.org/10.1038/
all formal and statistical analysis, interpreted the data, wrote and edited nn.3371.
the manuscript and the fig.s. Darci S. Marinho performed DNA extrac­ Beal, M.F., 2005. Oxidative damage as an early marker of Alzheimer’s disease and mild
tion and genotyped all the animals, separating them for the experiments. cognitive impairment. Neurobiol Aging 26, 585–586. https://doi.org/10.1016/j.
neurobiolaging.2004.09.022.
Geisa N. Salles carried out serum extraction, ELISA experiments and Becker, M., Lavie, V., Solomon, B., 2007. Stimulation of endogenous neurogenesis by
tissue preparations. Amanda F. Mosini contributed to RNA and cDNA for anti-EFRH immunization in a transgenic mouse model of Alzheimer’s disease. Proc
molecular analysis and qPCR experiments. Fernando H. Massinhani Natl Acad Sci U S A 104, 1691–1696. https://doi.org/10.1073/pnas.0610180104.
Bellefontaine, N., Chachlaki, K., Parkash, J., Vanacker, C., Colledge, W., d’Anglemont de
performed morphometrical data collection and analysis and assisted cell Tassigny, X., Garthwaite, J., Bouret, S.G., Prevot, V., 2014. Leptin-dependent
quantification. Gui M. Ko selected and crossed the heterozygous animals neuronal NO signaling in the preoptic hypothalamus facilitates reproduction. J Clin
and maintained the colony of transgenic mice and assisted the treatment Invest 124, 2550–2559. https://doi.org/10.1172/jci65928.
Bhatia, S., Fu, Y., Hsiao, J.T., Halliday, G.M., Kim, W.S., 2017. Deletion of Alzheimer’s
of animals. Marimélia A. Porcionatto conceptualized the project,
Disease Risk Gene ABCA7 Alters White Adipose Tissue Development and Leptin
designed experiments, interpreted the data, provided guidance and su­ Levels. J Alzheimers Dis Rep. 1, 237–247. https://doi.org/10.3233/ADR-170029.
pervision, was responsible for funding acquisition and reviewed the Bigalke, B., Schreitmuller, B., Sopova, K., Paul, A., Stransky, E., Gawaz, M., Stellos, K.,
manuscript. The work presented here was carried out in collaboration Laske, C., 2011. Adipocytokines and CD34 progenitor cells in Alzheimer’s disease.
PLoS One. 6, e20286 https://doi.org/10.1371/journal.pone.0020286.
between all authors. All authors revised and approved the final version Bollen, E., Vanmierlo, T., Akkerman, S., Wouters, C., Steinbusch, H.M., Prickaerts, J.,
of the manuscript. Conflict of interest: The authors declare no conflict of 2013. 7,8-Dihydroxyflavone improves memory consolidation processes in rats and
interest. mice. Behav Brain Res 257, 8–12. https://doi.org/10.1016/j.bbr.2013.09.029.
Bonda, D.J., Stone, J.G., Torres, S.L., Siedlak, S.L., Perry, G., Kryscio, R., Jicha, G.,
Casadesus, G., Smith, M.A., Zhu, X., Lee, H.G., 2014. Dysregulation of leptin
Acknowledgments signaling in Alzheimer disease: evidence for neuronal leptin resistance.
J Neurochem. 128, 162–172. https://doi.org/10.1111/jnc. 12380.
Bothwell, M., 2014. NGF, BDNF, NT3, and NT4. Handb Exp Pharmacol 220, 3–15.
We would like to thank Professor Beatriz Monteiro (Universidade https://doi.org/10.1007/978-3-642-45106-5_1.
Federal de São Paulo) for the 6E10 and Iba1 antibodies and Clivandir

17
M.L. Calió et al. Neurobiology of Disease 148 (2021) 105219

Buchman, A.S., Yu, L., Boyle, P.A., Schneider, J.A., De Jager, P.L., Bennett, D.A., 2016. Eriksson, P.S., Perfilieva, E., Bjork-Eriksson, T., Alborn, A.M., Nordborg, C., Peterson, D.
Higher brain BDNF gene expression is associated with slower cognitive decline in A., Gage, F.H., 1998. Neurogenesis in the adult human hippocampus. Nat Med 4,
older adults. Neurology 86, 735–741. https://doi.org/10.1212/ 1313–1317. https://doi.org/10.1038/3305.
wnl.0000000000002387. Ernst, A., Alkass, K., Bernard, S., Salehpour, M., Perl, S., Tisdale, J., Possnert, G.,
Buffo, A., Rite, I., Tripathi, P., Lepier, A., Colak, D., Horn, A.P., Mori, T., Gotz, M., 2008. Druid, H., Frisen, J., 2014. Neurogenesis in the striatum of the adult human brain.
Origin and progeny of reactive gliosis: A source of multipotent cells in the injured Cell 156, 1072–1083. https://doi.org/10.1016/j.cell.2014.01.044.
brain. Proc Natl Acad Sci U S A 105, 3581–3586. https://doi.org/10.1073/ Fainstein, N., Dan-Goor, N., Ben-Hur, T., 2018. Resident brain neural precursor cells
pnas.0709002105. develop age-dependent loss of therapeutic functions in Alzheimer’s mice. Neurobiol
Caille, I., Allinquant, B., Dupont, E., Bouillot, C., Langer, A., Muller, U., Prochiantz, A., Aging 72, 40–52. https://doi.org/10.1016/j.neurobiolaging.2018.07.020.
2004. Soluble form of amyloid precursor protein regulates proliferation of Farr, S.A., Banks, W.A., Morley, J.E., 2006. Effects of leptin on memory processing.
progenitors in the adult subventricular zone. Development 131, 2173–2181. https:// Peptides 27, 1420–1425. https://doi.org/10.1016/j.peptides.2005.10.006.
doi.org/10.1242/dev.01103. Feitosa, C.M., da Silva Oliveira, G.L., do Nascimento Cavalcante, A., Morais Chaves, S.K.,
Calon, F., Lim, G.P., Yang, F., Morihara, T., Teter, B., Ubeda, O., Rostaing, P., Triller, A., Rai, M., 2018. Determination of Parameters of Oxidative Stress in vitro Models of
Salem Jr., N., Ashe, K.H., Frautschy, S.A., Cole, G.M., 2004. Docosahexaenoic acid Neurodegenerative Diseases-A Review. Curr Clin Pharmacol 13, 100–109. https://
protects from dendritic pathology in an Alzheimer’s disease mouse model. Neuron doi.org/10.2174/1574884713666180301091612.
43, 633–645. https://doi.org/10.1016/j.neuron.2004.08.013. Fewlass, D.C., Noboa, K., Pi-Sunyer, F.X., Johnston, J.M., Yan, S.D., Tezapsidis, N., 2004.
Campfield, L.A., Smith, F.J., Burn, P., 1996. The OB protein (leptin) pathway–a link Obesity-related leptin regulates Alzheimer’s Abeta. FASEB J. 18, 1870–1878.
between adipose tissue mass and central neural networks. Horm Metab Res 28, https://doi.org/10.3389/fncel.2018.00340.
619–632. https://doi.org/10.1055/s-2007-979867. Folch, J., Pedros, I., Patraca, I., Sureda, F., Junyent, F., Beas-Zarate, C., Verdaguer, E.,
Chen, S.Q., Cai, Q., Shen, Y.Y., Wang, P.J., Teng, G.J., Li, M.H., Zhang, W., Zang, F.C., Pallas, M., Auladell, C., Camins, A., 2012. Neuroprotective and anti-ageing role of
2012. (1)H-MRS evaluation of therapeutic effect of neural stem cell transplantation leptin. J Mol Endocrinol 49, R149–R156. https://doi.org/10.1530/jme-12-0151.
on Alzheimer’s disease in AbetaPP/PS1 double transgenic mice. J Alzheimers Dis 28, Francis, B.M., Kim, J., Barakat, M.E., Fraenkl, S., Yucel, Y.H., Peng, S., Michalski, B.,
71–80. https://doi.org/10.3233/JAD-2010-110893. Fahnestock, M., McLaurin, J., Mount, H.T., 2012a. Object recognition memory and
Cimini, A., Moreno, S., D’Amelio, M., Cristiano, L., D’Angelo, B., Falone, S., Benedetti, E., BDNF expression are reduced in young TgCRND8 mice. Neurobiol Aging 33,
Carrara, P., Fanelli, F., Cecconi, F., Amicarelli, F., Ceru, M.P., 2009. Early 555–563. https://doi.org/10.1016/j.neurobiolaging.2010.04.003.
biochemical and morphological modifications in the brain of a transgenic mouse Francis, B.M., Yang, J., Hajderi, E., Brown, M.E., Michalski, B., McLaurin, J.,
model of Alzheimer’s disease: a role for peroxisomes. J Alzheimers Dis 18, 935–952. Fahnestock, M., Mount, H.T., 2012b. Reduced tissue levels of noradrenaline are
https://doi.org/10.3233/JAD-2009-1199. associated with behavioral phenotypes of the TgCRND8 mouse model of Alzheimer’s
Clayton, K.A., Van Enoo, A.A., Ikezu, T., 2017. Alzheimer’s Disease: The Role of disease. Neuropsychopharmacology 37, 1934–1944. https://doi.org/10.1038/
Microglia in Brain Homeostasis and Proteopathy. Front Neurosci 11, 680. https:// npp.2012.40.
doi.org/10.3389/fnins.2017.00680. Fuentealba, L.C., Obernier, K., Alvarez-Buylla, A., 2012. Adult neural stem cells bridge
Clelland, C.D., Choi, M., Romberg, C., Clemenson Jr., G.D., Fragniere, A., Tyers, P., their niche. Cell Stem Cell 10, 698–708. https://doi.org/10.1016/j.
Jessberger, S., Saksida, L.M., Barker, R.A., Gage, F.H., Bussey, T.J., 2009. stem.2012.05.012.
A functional role for adult hippocampal neurogenesis in spatial pattern separation. Furcila, D., Dominguez-Alvaro, M., DeFelipe, J., Alonso-Nanclares, L., 2019. Subregional
Science (80-.) 325, 210–213. https://doi.org/10.1126/science.1173215. Density of Neurons, Neurofibrillary Tangles and Amyloid Plaques in the
Colonna, M., Butovsky, O., 2017. Microglia Function in the Central Nervous System Hippocampus of Patients With Alzheimer’s Disease. Front Neuroanat 13, 99. https://
During Health and Neurodegeneration. Annu Rev Immunol 35, 441–468. https:// doi.org/10.3389/fnana.2019.00099.
doi.org/10.1146/annurev-immunol-051116-052358. Gage, F.H., 2000. Mammalian neural stem cells. Science (80-.) 287, 1433–1438. https://
Connor, B., Young, D., Yan, Q., Faull, R.L., Synek, B., Dragunow, M., 1997. Brain-derived doi.org/10.1126/science.287.5457.1433.
neurotrophic factor is reduced in Alzheimer’s disease. Brain Res Mol Brain Res 49, Gallaway, P.J., Miyake, H., Buchowski, M.S., Shimada, M., Yoshitake, Y., Kim, A.S.,
71–81. https://doi.org/10.1016/S0169-328X(97)00125-3. Hongu, N., 2017. Physical Activity: A Viable Way to Reduce the Risks of Mild
Conover, J.C., Notti, R.Q., 2007. The neural stem cell niche. Cell Tissue Res 331, Cognitive Impairment, Alzheimer’s Disease, and Vascular Dementia in Older Adults.
211–224. https://doi.org/10.1007/s00441-007-0503-6. Epub 2007 Oct 6. Brain Sci 7. https://doi.org/10.3390/brainsci7020022.
Dai, M.H., Zheng, H., Zeng, L.D., Zhang, Y., 2018. The genes associated with early-onset Garcia, K.O., Ornellas, F.L., Martin, P.K., Patti, C.L., Mello, L.E., Frussa-Filho, R., Han, S.
Alzheimer’s disease. Oncotarget 9, 15132–15143. https://doi.org/10.18632/ W., Longo, B.M., 2014. Therapeutic effects of the transplantation of VEGF
oncotarget.23738. overexpressing bone marrow mesenchymal stem cells in the hippocampus of murine
Demars, M., Hu, Y.S., Gadadhar, A., Lazarov, O., 2010. Impaired neurogenesis is an early model of Alzheimer’s disease. Front Aging Neurosci 6, 30. https://doi.org/10.3389/
event in the etiology of familial Alzheimer’s disease in transgenic mice. J Neurosci fnagi.2014.00030.
Res 88, 2103–2117. https://doi.org/10.1002/jnr.22387. Garthe, A., Behr, J., Kempermann, G., 2009. Adult-generated hippocampal neurons allow
Demars, M.P., Bartholomew, A., Strakova, Z., Lazarov, O., 2011. Soluble amyloid the flexible use of spatially precise learning strategies. PLoS One 4, e5464. https://
precursor protein: a novel proliferation factor of adult progenitor cells of ectodermal doi.org/10.1371/journal.pone.0005464.
and mesodermal origin. Stem Cell Res Ther 2, 36. https://doi.org/10.1186/scrt77. Garza, J.C., Guo, M., Zhang, W., Lu, X.Y., 2008. Leptin increases adult hippocampal
Demars, M.P., Hollands, C., Zhao Kda, T., Lazarov, O., 2013. Soluble amyloid precursor neurogenesis in vivo and in vitro. J Biol Chem 283, 18238–18247. https://doi.org/
protein-alpha rescues age-linked decline in neural progenitor cell proliferation. 10.1074/jbc.M800053200.
Neurobiol Aging 34, 2431–2440. https://doi.org/10.1016/j. Ghasemi, M., Zendehbad, B., Zabihi, H., Hosseini, M., Hadjzadeh, M.A.,
neurobiolaging.2013.04.016. Hayatdavoudi, P., 2016. Beneficial Effect of Leptin on Spatial Learning and Memory
Deng, W., Saxe, M.D., Gallina, I.S., Gage, F.H., 2009. Adult-born hippocampal dentate in Streptozotocin-Induced Diabetic Rats. Balk. Med J 33, 102–107. https://doi.org/
granule cells undergoing maturation modulate learning and memory in the brain. 10.5152/balkanmedj.2015.15084.
J Neurosci 29, 13532–13542. https://doi.org/10.1523/jneurosci.3362-09.2009. Gould, E., Beylin, A., Tanapat, P., Reeves, A., Shors, T.J., 1999. Learning enhances adult
Diaz-Moreno, M., Hortiguela, R., Goncalves, A., Garcia-Carpio, I., Manich, G., Garcia- neurogenesis in the hippocampal formation. Nat Neurosci 2, 260–265. https://doi.
Bermudez, E., Moreno-Estelles, M., Eguiluz, C., Vilaplana, J., Pelegri, C., Vilar, M., org/10.1038/6365.
Mira, H., 2013. Abeta increases neural stem cell activity in senescence-accelerated Gowrishankar, S., Yuan, P., Wu, Y., Schrag, M., Paradise, S., Grutzendler, J., De
SAMP8 mice. Neurobiol Aging 34, 2623–2638. https://doi.org/10.1016/j. Camilli, P., Ferguson, S.M., 2015. Massive accumulation of luminal protease-
neurobiolaging.2013.05.011. deficient axonal lysosomes at Alzheimer’s disease amyloid plaques. Proc Natl Acad
Dietrich, M.O., Spuch, C., Antequera, D., Rodal, I., de Yebenes, J.G., Molina, J.A., Sci U S A 112, E3699–E3708. https://doi.org/10.1073/pnas.1510329112.
Bermejo, F., Carro, E., 2008. Megalin mediates the transport of leptin across the Greco, S.J., Bryan, K.J., Sarkar, S., Zhu, X., Smith, M.A., Ashford, J.W., Johnston, J.M.,
blood-CSF barrier. Neurobiol Aging. 29, 902–912. https://doi.org/10.1016/j. Tezapsidis, N., Casadesus, G., 2010. Leptin reduces pathology and improves memory
neurobiolaging.2007.01.008. in a transgenic mouse model of Alzheimer’s disease. J Alzheimers Dis 19,
Doetsch, F., 2003. The glial identity of neural stem cells. Nat Neurosci 6, 1127–1134. 1155–1167. https://doi.org/10.3233/JAD-2010-1308.
https://doi.org/10.1038/nn1144. Grimaldi, A., Pediconi, N., Oieni, F., Pizzarelli, R., Rosito, M., Giubettini, M., Santini, T.,
Doherty, G.H., Beccano-Kelly, D., Yan, S.D., Gunn-Moore, F.J., Harvey, J., 2012. Leptin Limatola, C., Ruocco, G., Ragozzino, D., Di Angelantonio, S., 2019.
prevents hippocampal synaptic disruption and neuronal cell death induced by Neuroinflammatory Processes, A1 Astrocyte Activation and Protein Aggregation in
amyloid beta. Neurobiol Aging 34, 226–237. https://doi.org/10.1016/j. the Retina of Alzheimer’s Disease Patients, Possible Biomarkers for Early Diagnosis.
neurobiolaging.2012.08.003. Epub 2012 Aug 22. Front Neurosci 13, 925. https://doi.org/10.3389/fnins.2019.00925.
Dosunmu, R., Wu, J., Basha, M.R., Zawia, N.H., 2007. Environmental and dietary risk Gundersen, H.J., 2002. The smooth fractionator. J Microsc 207, 191–210. https://doi.
factors in Alzheimer’s disease. Expert Rev Neurother 7, 887–900. https://doi.org/ org/10.1046/j.1365-2818.2002.01054.x.
10.1586/14737175.7.7.887. Guo, Z., Jiang, H., Xu, X., Duan, W., Mattson, M.P., 2008. Leptin-mediated cell survival
Enwere, E., Shingo, T., Gregg, C., Fujikawa, H., Ohta, S., Weiss, S., 2004. Aging results in signaling in hippocampal neurons mediated by JAK STAT3 and mitochondrial
reduced epidermal growth factor receptor signaling, diminished olfactory stabilization. J Biol Chem 283, 1754–1763. https://doi.org/10.1074/jbc.
neurogenesis, and deficits in fine olfactory discrimination. J Neurosci 24, M703753200.
8354–8365. https://doi.org/10.1523/JNEUROSCI.2751-04.2004. Hamilton, L.K., Dufresne, M., Joppe, S.E., Petryszyn, S., Aumont, A., Calon, F., Barnabe-
Erickson, K.I., Prakash, R.S., Voss, M.W., Chaddock, L., Heo, S., McLaren, M., Pence, B. Heider, F., Furtos, A., Parent, M., Chaurand, P., Fernandes, K.J., 2015. Aberrant
D., Martin, S.A., Vieira, V.J., Woods, J.A., McAuley, E., Kramer, A.F., 2010. Brain- Lipid Metabolism in the Forebrain Niche Suppresses Adult Neural Stem Cell
derived neurotrophic factor is associated with age-related decline in hippocampal Proliferation in an Animal Model of Alzheimer’s Disease. Cell Stem Cell 17, 397–411.
volume. J Neurosci 30, 5368–5375. https://doi.org/10.1523/jneurosci.6251- https://doi.org/10.1016/j.stem.2015.08.001.
09.2010.

18
M.L. Calió et al. Neurobiology of Disease 148 (2021) 105219

Hardie, L.J., Rayner, D.V., Holmes, S., Trayhurn, P., 1996. Circulating leptin levels are Kern, D.S., Maclean, K.N., Jiang, H., Synder, E.Y., Sladek Jr., J.R., Bjugstad, K.B., 2011.
modulated by fasting, cold exposure and insulin administration in lean but not Neural stem cells reduce hippocampal tau and reelin accumulation in aged Ts65Dn
Zucker (fa/fa) rats as measured by ELISA. Biochem Biophys Res Commun. 223, Down syndrome mice. Cell Transpl. 20, 371–379. https://doi.org/10.3727/
660–665. https://doi.org/10.1006/bbrc.1996.0951. 096368910X528085.
Hardy, J., Selkoe, D.J., 2002. The amyloid hypothesis of Alzheimer’s disease: progress Khemka, V.K., Bagchi, D., Bandyopadhyay, K., Bir, A., Chattopadhyay, M., Biswas, A.,
and problems on the road to therapeutics. Science (80) 297, 353–356. https://doi. Basu, D., Chakrabarti, S., 2014. Altered serum levels of adipokines and insulin in
org/10.1126/science.1072994. probable Alzheimer’s disease. J Alzheimers Dis. 41, 525–533. https://doi.org/
Haughey, N.J., Nath, A., Chan, S.L., Borchard, A.C., Rao, M.S., Mattson, M.P., 2002. 10.3233/JAD-140006.
Disruption of neurogenesis by amyloid beta-peptide, and perturbed neural Kim, S.E., Ko, I.G., Kim, B.K., Shin, M.S., Cho, S., Kim, C.J., Kim, S.H., Baek, S.S., Lee, E.
progenitor cell homeostasis, in models of Alzheimer’s disease. J Neurochem 83, K., Jee, Y.S., 2010. Treadmill exercise prevents aging-induced failure of memory
1509–1524. https://doi.org/10.1046/j.1471-4159.2002.01267.x. through an increase in neurogenesis and suppression of apoptosis in rat
Heneka, M.T., Kummer, M.P., Latz, E., 2014. Innate immune activation in hippocampus. Exp Gerontol 45, 357–365. https://doi.org/10.1016/j.
neurodegenerative disease. Nat Rev Immunol 14, 463–477. https://doi.org/ exger.2010.02.005.
10.1038/nri3705. Kizil, C., Bhattarai, P., 2018. Is Alzheimer’s Also a Stem Cell Disease? - The Zebrafish
Heneka, M.T., Carson, M.J., El Khoury, J., Landreth, G.E., Brosseron, F., Feinstein, D.L., Perspective. Front Cell Dev Biol 6, 159. https://doi.org/10.3389/fcell.2018.00159.
Jacobs, A.H., Wyss-Coray, T., Vitorica, J., Ransohoff, R.M., Herrup, K., Frautschy, S. Komori, T., Morikawa, Y., Nanjo, K., Senba, E., 2006. Induction of brain-derived
A., Finsen, B., Brown, G.C., Verkhratsky, A., Yamanaka, K., Koistinaho, J., Latz, E., neurotrophic factor by leptin in the ventromedial hypothalamus. Neuroscience 139,
Halle, A., Petzold, G.C., Town, T., Morgan, D., Shinohara, M.L., Perry, V.H., 1107–1115. https://doi.org/10.1016/j.neuroscience.2005.12.066.
Holmes, C., Bazan, N.G., Brooks, D.J., Hunot, S., Joseph, B., Deigendesch, N., Krabbe, G., Halle, A., Matyash, V., Rinnenthal, J.L., Eom, G.D., Bernhardt, U., Miller, K.
Garaschuk, O., Boddeke, E., Dinarello, C.A., Breitner, J.C., Cole, G.M., Golenbock, D. R., Prokop, S., Kettenmann, H., Heppner, F.L., 2013. Functional impairment of
T., Kummer, M.P., 2015. Neuroinflammation in Alzheimer’s disease. Lancet Neurol microglia coincides with Beta-amyloid deposition in mice with Alzheimer-like
14, 388–405. https://doi.org/10.1016/S1474-4422(15)70016-5. pathology. PLoS One 8, e60921. https://doi.org/10.1371/journal.pone.0060921.
Hock, C., Heese, K., Hulette, C., Rosenberg, C., Otten, U., 2000. Region-specific Kriegstein, A., Alvarez-Buylla, A., 2009. The glial nature of embryonic and adult neural
neurotrophin imbalances in Alzheimer disease: decreased levels of brain-derived stem cells. Annu Rev Neurosci 32, 149–184. https://doi.org/10.1146/annurev.
neurotrophic factor and increased levels of nerve growth factor in hippocampus and neuro.051508.135600.
cortical areas. Arch Neurol 57, 846–851. https://doi.org/10.1001/ Kuhn, H.G., Toda, T., Gage, F.H., 2018. Adult Hippocampal Neurogenesis: A Coming-of-
archneur.57.6.846. Age Story. J Neurosci 38, 10401–10410. https://doi.org/10.1523/
Holcomb, L., Gordon, M.N., McGowan, E., Yu, X., Benkovic, S., Jantzen, P., Wright, K., JNEUROSCI.2144-18.2018.
Saad, I., Mueller, R., Morgan, D., Sanders, S., Zehr, C., O’Campo, K., Hardy, J., Laske, C., Stellos, K., Hoffmann, N., Stransky, E., Straten, G., Eschweiler, G.W., Leyhe, T.,
Prada, C.M., Eckman, C., Younkin, S., Hsiao, K., Duff, K., 1998. Accelerated 2011. Higher BDNF serum levels predict slower cognitive decline in Alzheimer’s
Alzheimer-type phenotype in transgenic mice carrying both mutant amyloid disease patients. Int J Neuropsychopharmacol 14, 399–404. https://doi.org/
precursor protein and presenilin 1 transgenes. Nat Med 4, 97–100. https://doi.org/ 10.1017/s1461145710001008.
10.1038/nm0198-097. Lauritzen, I., Pardossi-Piquard, R., Bauer, C., Brigham, E., Abraham, J.D., Ranaldi, S.,
Holden, K.F., Lindquist, K., Tylavsky, F.A., Rosano, C., Harris, T.B., Yaffe, K., Health, A.B. Fraser, P., St-George-Hyslop, P., Le Thuc, O., Espin, V., Chami, L., Dunys, J.,
C. study, 2009. Serum leptin level and cognition in the elderly: Findings from the Checler, F., 2012. The beta-secretase-derived C-terminal fragment of betaAPP, C99,
Health ABC Study. Neurobiol Aging 30, 1483–1489. https://doi.org/10.1016/j. but not Abeta, is a key contributor to early intraneuronal lesions in triple-transgenic
neurobiolaging.2007.11.024. mouse hippocampus. J Neurosci 32. https://doi.org/10.1523/jneurosci.2775-
Holsinger, R.M., Schnarr, J., Henry, P., Castelo, V.T., Fahnestock, M., 2000. Quantitation 12.2012, 16243–1655a.
of BDNF mRNA in human parietal cortex by competitive reverse transcription- Lazarov, O., Marr, R.A., 2010. Neurogenesis and Alzheimer’s disease: at the crossroads.
polymerase chain reaction: decreased levels in Alzheimer’s disease. Brain Res Mol Exp Neurol 223, 267–281. https://doi.org/10.1016/j.expneurol.2009.08.009.
Brain Res 76, 347–354. https://doi.org/10.1016/S0169-328X(00)00023-1. Lazarov, O., Marr, R.A., 2013. Of mice and men: neurogenesis, cognition and Alzheimer’s
Hormoz, S., 2013. Stem cell population asymmetry can reduce rate of replicative aging. disease. Front Aging Neurosci 5, 43. https://doi.org/10.3389/fnagi.2013.00043.
J Theor Biol 331, 19–27. https://doi.org/10.1016/j.jtbi.2013.04.018. Lazarov, O., Mattson, M.P., Peterson, D.A., Pimplikar, S.W., van Praag, H., 2010. When
Hostenbach, S., Cambron, M., D’Haeseleer, M., Kooijman, R., De Keyser, J., 2014. neurogenesis encounters aging and disease. Trends Neurosci 33, 569–579. https://
Astrocyte loss and astrogliosis in neuroinflammatory disorders. Neurosci Lett 565, doi.org/10.1016/j.tins.2010.09.003.
39–41. https://doi.org/10.1016/j.neulet.2013.10.012. Lee, J., Seroogy, K.B., Mattson, M.P., 2002. Dietary restriction enhances neurotrophin
Huang, E.J., Reichardt, L.F., 2001. Neurotrophins: roles in neuronal development and expression and neurogenesis in the hippocampus of adult mice. J Neurochem 80,
function. Annu Rev Neurosci 24, 677–736. https://doi.org/10.1146/annurev. 539–547. https://doi.org/10.1046/j.0022-3042.2001.00747.x.
neuro.24.1.677. Lee, S., Yang, M., Kim, J., Kim, J., Son, Y., Kwon, S., Kim, S.H., Kim, J.C., Kang, S.S.,
Ishii, M., Wang, G., Racchumi, G., Dyke, J.P., Iadecola, C., 2014. Transgenic mice Wang, H., Shin, T., Moon, C., 2014. Nestin expression and glial response in the
overexpressing amyloid precursor protein exhibit early metabolic deficits and a hippocampus of mice after trimethyltin treatment. Acta Histochem 116, 1276–1288.
pathologically low leptin state associated with hypothalamic dysfunction in arcuate https://doi.org/10.1016/j.acthis.2014.07.009.
neuropeptide Y neurons. J Neurosci. 34, 9096–9106. https://doi.org/10.1523/ Lee, Y.H., Hsu, H.C., Kao, P.C., Shiao, Y.J., Yeh, S.H., Shie, F.S., Hsu, S.M., Yeh, C.W.,
JNEUROSCI.0872-14.2014. Liu, H.K., Yang, S.B., Tsay, H.J., 2018. Augmented Insulin and Leptin Resistance of
Jankowsky, J.L., Savonenko, A., Schilling, G., Wang, J., Xu, G., Borchelt, D.R., 2002. High Fat Diet-Fed APPswe/PS1dE9 Transgenic Mice Exacerbate Obesity and
Transgenic mouse models of neurodegenerative disease: opportunities for Glycemic Dysregulation. Int J Mol Sci. 19. https://doi.org/10.3390/ijms19082333.
therapeutic development. Curr Neurol Neurosci Rep 2, 457–464. https://doi.org/ Li, X.L., Aou, S., Oomura, Y., Hori, N., Fukunaga, K., Hori, T., 2002. Impairment of long-
10.1007/s11910-002-0073-7. term potentiation and spatial memory in leptin receptor-deficient rodents.
Jeong, S., 2017. Molecular and Cellular Basis of Neurodegeneration in Alzheimer’s Neuroscience 113, 607–615. https://doi.org/10.1016/s0306-4522(02)00162-8.
Disease. Mol Cells 40, 613–620. https://doi.org/10.14348/molcells.2017.0096. Li, B., Yamamori, H., Tatebayashi, Y., Shafit-Zagardo, B., Tanimukai, H., Chen, S.,
Jin, K., Mao, X.O., Sun, Y., Xie, L., Greenberg, D.A., 2002. Stem cell factor stimulates Iqbal, K., Grundke-Iqbal, I., 2008. Failure of neuronal maturation in Alzheimer
neurogenesis in vitro and in vivo. J Clin Invest 110, 311–319. https://doi.org/ disease dentate gyrus. J Neuropathol Exp Neurol 67, 78–84. https://doi.org/
10.1172/JCI15251. 10.1097/nen.0b013e318160c5db.
Jin, K., Galvan, V., Xie, L., Mao, X.O., Gorostiza, O.F., Bredesen, D.E., Greenberg, D.A., Li, Q., Zhao, H.F., Zhang, Z.F., Liu, Z.G., Pei, X.R., Wang, J.B., Cai, M.Y., Li, Y., 2009.
2004a. Enhanced neurogenesis in Alzheimer’s disease transgenic (PDGF-APPSw,Ind) Long-term administration of green tea catechins prevents age-related spatial learning
mice. Proc Natl Acad Sci U S A 101, 13363–13367. https://doi.org/10.1073/ and memory decline in C57BL/6 J mice by regulating hippocampal cyclic amp-
pnas.0403678101. response element binding protein signaling cascade. Neuroscience 159, 1208–1215.
Jin, K., Peel, A.L., Mao, X.O., Xie, L., Cottrell, B.A., Henshall, D.C., Greenberg, D.A., https://doi.org/10.1016/j.neuroscience.2009.02.008.
2004b. Increased hippocampal neurogenesis in Alzheimer’s disease. Proc Natl Acad Li, W., Li, X., Xin, X., Kan, P.C., Yan, Y., 2016. MicroRNA-613 regulates the expression of
Sci U S A 101, 343–347. https://doi.org/10.1073/pnas.2634794100. brain-derived neurotrophic factor in Alzheimer’s disease. Biosci Trends 10, 372–377.
Kanemoto, S., Griffin, J., Markham-Coultes, K., Aubert, I., Tandon, A., George-Hyslop, P. https://doi.org/10.5582/bst.2016.01127.
S., Fraser, P.E., 2013. Proliferation, differentiation and amyloid-beta production in Liddelow, S.A., Guttenplan, K.A., Clarke, L.E., Bennett, F.C., Bohlen, C.J., Schirmer, L.,
neural progenitor cells isolated from TgCRND8 mice. Neuroscience 261, 52–59. Bennett, M.L., Munch, A.E., Chung, W.S., Peterson, T.C., Wilton, D.K., Frouin, A.,
https://doi.org/10.1016/j.neuroscience.2013.12.02110.1016/j. Napier, B.A., Panicker, N., Kumar, M., Buckwalter, M.S., Rowitch, D.H., Dawson, V.
neuroscience.2013.12.021. Epub 2013 Dec 20. L., Dawson, T.M., Stevens, B., Barres, B.A., 2017. Neurotoxic reactive astrocytes are
Karaoulanis, S.E., Daponte, A., Rizouli, K.A., Rizoulis, A.A., Lialios, G.A., Theodoridou, C. induced by activated microglia. Nature 541, 481–487. https://doi.org/10.1038/
T., Christakopoulos, C., Angelopoulos, N.V., 2012. The role of cytokines and hot nature21029.
flashes in perimenopausal depression. Ann Gen Psychiatry. 11, 9. https://doi.org/ Lie, D.C., Song, H., Colamarino, S.A., Ming, G.L., Gage, F.H., 2004. Neurogenesis in the
10.1186/1744-859X-11-9. adult brain: new strategies for central nervous system diseases. Annu Rev Pharmacol
Katsimpardi, L., Lledo, P.M., 2018. Regulation of neurogenesis in the adult and aging Toxicol 44, 399–421. https://doi.org/10.1146/annurev.
brain. Curr Opin Neurobiol 53, 131–138. https://doi.org/10.1016/j. pharmtox.44.101802.121631.
conb.2018.07.006. Lieb, W., Beiser, A.S., Vasan, R.S., Tan, Z.S., Au, R., Harris, T.B., Roubenoff, R.,
Kempermann, G., Gast, D., Gage, F.H., 2002. Neuroplasticity in old age: sustained Auerbach, S., DeCarli, C., Wolf, P.A., Seshadri, S., 2009. Association of plasma leptin
fivefold induction of hippocampal neurogenesis by long-term environmental levels with incident Alzheimer disease and MRI measures of brain aging. JAMA 302,
enrichment. Ann Neurol 52, 135–143. https://doi.org/10.1002/ana.10262. 2565–2572. https://doi.org/10.1001/jama.2009.1836.

19
M.L. Calió et al. Neurobiology of Disease 148 (2021) 105219

Lin, R., Iacovitti, L., 2015. Classic and novel stem cell niches in brain homeostasis and Morrison, C.D., 2009. Leptin signaling in brain: A link between nutrition and cognition?
repair. Brain Res 1628, 327–342. https://doi.org/10.1016/j.brainres.2015.04.029. Biochim Biophys Acta 1792, 401–408. https://doi.org/10.1016/j.
Liu, K.X., Edwards, B., Lee, S., Finelli, M.J., Davies, B., Davies, K.E., Oliver, P.L., 2015a. bbadis.2008.12.004.
Neuron-specific antioxidant OXR1 extends survival of a mouse model of Moult, P.R., Cross, A., Santos, S.D., Carvalho, A.L., Lindsay, Y., Connolly, C.N., Irving, A.
amyotrophic lateral sclerosis. Brain 138, 1167–1181. https://doi.org/10.1093/ J., Leslie, N.R., Harvey, J., 2010. Leptin regulates AMPA receptor trafficking via
brain/awv039. PTEN inhibition. J Neurosci 30, 4088–4101. https://doi.org/10.1523/
Liu, S., Shi, D., Wang, H.C., Yu, Y.Z., Xu, Q., Sun, Z.W., 2015b. Co-immunization with jneurosci.3614-09.2010.
DNA and protein mixture: a safe and efficacious immunotherapeutic strategy for Mundim, M.V., Zamproni, L.N., Pinto, A.A.S., Galindo, L.T., Xavier, A.M., Glezer, I.,
Alzheimer’s disease in PDAPP mice. Sci Rep 5, 7771. https://doi.org/10.1038/ Porcionatto, M., 2018. A new function for Prokineticin 2: Recruitment of SVZ-
srep07771. derived neuroblasts to the injured cortex in a mouse model of traumatic brain injury.
Livak, K.J., Schmittgen, T.D., 2001. Analysis of relative gene expression data using real- Mol Cell Neurosci 94, 1–10. https://doi.org/10.1016/j.mcn.2018.10.004.
time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 25, 402–408. Naert, G., Rivest, S., 2012. Age-related changes in synaptic markers and monocyte
https://doi.org/10.1006/meth.2001.1262. subsets link the cognitive decline of APP(Swe)/PS1 mice. Front Cell Neurosci 6, 51.
Lu, B., Nagappan, G., Guan, X., Nathan, P.J., Wren, P., 2013. BDNF-based synaptic repair https://doi.org/10.3389/fncel.2012.00051 eCollection 2012.
as a disease-modifying strategy for neurodegenerative diseases. Nat Rev Neurosci 14, Nagata, T., Kobayashi, N., Shinagawa, S., Yamada, H., Kondo, K., Nakayama, K., 2014.
401–416. https://doi.org/10.1038/nrn3505. Plasma BDNF levels are correlated with aggressiveness in patients with amnestic
Luo, X., McGregor, G., Irving, A.J., Harvey, J., 2015. Leptin Induces a Novel Form of mild cognitive impairment or Alzheimer disease. J Neural Transm 121, 433–441.
NMDA Receptor-Dependent LTP at Hippocampal Temporoammonic-CA1 Synapses. https://doi.org/10.1007/s00702-013-1121-y.
eNeuro 2. https://doi.org/10.1523/eneuro.0007-15.2015. Naumann, N., Alpar, A., Ueberham, U., Arendt, T., Gartner, U., 2009. Transgenic
Lustbader, J.W., Cirilli, M., Lin, C., Xu, H.W., Takuma, K., Wang, N., Caspersen, C., expression of human wild-type amyloid precursor protein decreases neurogenesis in
Chen, X., Pollak, S., Chaney, M., Trinchese, F., Liu, S., Gunn-Moore, F., Lue, L.F., the adult hippocampus. Hippocampus 20, 971–979. https://doi.org/10.1002/
Walker, D.G., Kuppusamy, P., Zewier, Z.L., Arancio, O., Stern, D., Yan, S.S., Wu, H., hipo.20693.
2004. ABAD directly links Abeta to mitochondrial toxicity in Alzheimer’s disease. Navarro, V., Sanchez-Mejias, E., Jimenez, S., Munoz-Castro, C., Sanchez-Varo, R.,
Science (80-.) 304, 448–452. https://doi.org/10.1126/science.1091230. Davila, J.C., Vizuete, M., Gutierrez, A., Vitorica, J., 2018. Microglia in Alzheimer’s
Maioli, S., Puerta, E., Merino-Serrais, P., Fusari, L., Gil-Bea, F., Rimondini, R., Cedazo- Disease: Activated, Dysfunctional or Degenerative. Front Aging Neurosci 10, 140.
Minguez, A., 2012. Combination of apolipoprotein E4 and high carbohydrate diet https://doi.org/10.3389/fnagi.2018.00140.
reduces hippocampal BDNF and arc levels and impairs memory in young mice. Nimmerjahn, A., Kirchhoff, F., Helmchen, F., 2005. Resting microglial cells are highly
J Alzheimers Dis 32, 341–355. https://doi.org/10.3233/jad-2012-120697. dynamic surveillants of brain parenchyma in vivo. Science (80-.) 308, 1314–1318.
Maioli, S., Lodeiro, M., Merino-Serrais, P., Falahati, F., Khan, W., Puerta, E., Codita, A., https://doi.org/10.1126/science.1110647.
Rimondini, R., Ramirez, M.J., Simmons, A., Gil-Bea, F., Westman, E., Cedazo- Olmos-Alonso, A., Schetters, S.T., Sri, S., Askew, K., Mancuso, R., Vargas-Caballero, M.,
Minguez, A., Alzheimer’s Disease Neuroimaging, I., 2015. Alterations in brain leptin Holscher, C., Perry, V.H., Gomez-Nicola, D., 2016. Pharmacological targeting of
signalling in spite of unchanged CSF leptin levels in Alzheimer’s disease. Aging Cell. CSF1R inhibits microglial proliferation and prevents the progression of Alzheimer’s-
14, 122–129. https://doi.org/10.1111/acel.12281. like pathology. Brain 139, 891–907. https://doi.org/10.1093/brain/awv379.
Manocha, G.D., Floden, A.M., Rausch, K., Kulas, J.A., McGregor, B.A., Oomura, Y., Hori, N., Shiraishi, T., Fukunaga, K., Takeda, H., Tsuji, M., Matsumiya, T.,
Rojanathammanee, L., Puig, K.R., Puig, K.L., Karki, S., Nichols, M.R., Darland, D.C., Ishibashi, M., Aou, S., Li, X.L., Kohno, D., Uramura, K., Sougawa, H., Yada, T.,
Porter, J.E., Combs, C.K., 2016. APP Regulates Microglial Phenotype in a Mouse Wayner, M.J., Sasaki, K., 2006. Leptin facilitates learning and memory performance
Model of Alzheimer’s Disease. J Neurosci 36, 8471–8486. https://doi.org/10.1523/ and enhances hippocampal CA1 long-term potentiation and CaMK II
JNEUROSCI.4654-15.2016. phosphorylation in rats. Peptides 27, 2738–2749. https://doi.org/10.1016/j.
Markiewicz, I., Sypecka, J., Domanska-Janik, K., Wyszomirski, T., Lukomska, B., 2011. peptides.2006.07.001.
Cellular environment directs differentiation of human umbilical cord blood-derived Oudot, A., Martin, C., Busseuil, D., Vergely, C., Demaison, L., Rochette, L., 2006. NADPH
neural stem cells in vitro. J Histochem Cytochem 59, 289–301. https://doi.org/ oxidases are in part responsible for increased cardiovascular superoxide production
10.1369/0022155410397997. during aging. Free Radic Biol Med 40, 2214–2222. https://doi.org/10.1016/j.
Marsh, S.E., Blurton-Jones, M., 2017. Neural stem cell therapy for neurodegenerative freeradbiomed.2006.02.020.
disorders: The role of neurotrophic support. Neurochem Int 106, 94–100. https:// Overton, J.M., Williams, T.D., Chambers, J.B., Rashotte, M.E., 2001. Central leptin
doi.org/10.1016/j.neuint.2017.02.006. infusion attenuates the cardiovascular and metabolic effects of fasting in rats.
Matarredona, E.R., Talaveron, R., Pastor, A.M., 2018. Interactions Between Neural Hypertension 37, 663–669.
Progenitor Cells and Microglia in the Subventricular Zone: Physiological Parent, J.M., Yu, T.W., Leibowitz, R.T., Geschwind, D.H., Sloviter, R.S., Lowenstein, D.
Implications in the Neurogenic Niche and After Implantation in the Injured Brain. H., 1997. Dentate granule cell neurogenesis is increased by seizures and contributes
Front Cell Neurosci 12, 268. https://doi.org/10.3389/fncel.2018.00268. to aberrant network reorganization in the adult rat hippocampus. J Neurosci 17,
McGregor, G., Harvey, J., 2018. Regulation of Hippocampal Synaptic Function by the 3727–3738.
Metabolic Hormone, Leptin: Implications for Health and Neurodegenerative Disease. Paz-Filho, G., Wong, M.L., Licinio, J., 2010. The procognitive effects of leptin in the brain
Front Cell Neurosci. 12, 340. https://doi.org/10.3389/fncel.2018.00340. and their clinical implications. Int J Clin Pr. 64, 1808–1812. https://doi.org/
Meng, C., He, Z., Xing, D., 2013. Low-level laser therapy rescues dendrite atrophy via 10.1111/j.1742-1241.2010.02536.x.
upregulating BDNF expression: implications for Alzheimer’s disease. J Neurosci 33, Peng, S., Wuu, J., Mufson, E.J., Fahnestock, M., 2005. Precursor form of brain-derived
13505–13517. https://doi.org/10.1523/jneurosci.0918-13.2013. neurotrophic factor and mature brain-derived neurotrophic factor are decreased in
Mercer, J.G., Hoggard, N., Williams, L.M., Lawrence, C.B., Hannah, L.T., Trayhurn, P., the pre-clinical stages of Alzheimer’s disease. J Neurochem 93, 1412–1421. https://
1996. Localization of leptin receptor mRNA and the long form splice variant (Ob-Rb) doi.org/10.1111/j.1471-4159.2005.03135.x.
in mouse hypothalamus and adjacent brain regions by in situ hybridization. FEBS Peng, S., Garzon, D.J., Marchese, M., Klein, W., Ginsberg, S.D., Francis, B.M., Mount, H.
Lett 387, 113–116. https://doi.org/10.1016/0014-5793(96)00473-5. T., Mufson, E.J., Salehi, A., Fahnestock, M., 2009. Decreased brain-derived
Mesulam, M.M., 1999. Neuroplasticity failure in Alzheimer’s disease: bridging the gap neurotrophic factor depends on amyloid aggregation state in transgenic mouse
between plaques and tangles. Neuron 24, 521–529. https://doi.org/10.1016/s0896- models of Alzheimer’s disease. J Neurosci 29, 9321–9329. https://doi.org/10.1523/
6273(00)81109-5. jneurosci.4736-08.2009.
Meyer, L.R., Zhu, V., Miller, A., Roghair, R.D., 2014. Growth restriction, leptin, and the Peng, Y., Liu, J., Tang, Y., Liu, J., Han, T., Han, S., Li, H., Hou, C., Liu, J., Long, J., 2014.
programming of adult behavior in mice. Behav Brain Res 275, 131–135. https://doi. High-Fat-Diet-Induced Weight Gain Ameliorates Bone Loss without Exacerbating
org/10.1016/j.bbr.2014.08.054. AbetaPP Processing and Cognition in Female APP/PS1 Mice. Front Cell Neurosci. 8,
Michalski, B., Fahnestock, M., 2003. Pro-brain-derived neurotrophic factor is decreased 225. https://doi.org/10.3389/fncel.2014.00225.
in parietal cortex in Alzheimer’s disease. Brain Res Mol Brain Res 111, 148–154. Perez-Gonzalez, R., Antequera, D., Vargas, T., Spuch, C., Bolos, M., Carro, E., 2011.
Michaud, J.P., Halle, M., Lampron, A., Theriault, P., Prefontaine, P., Filali, M., Tribout- Leptin induces proliferation of neuronal progenitors and neuroprotection in a mouse
Jover, P., Lanteigne, A.M., Jodoin, R., Cluff, C., Brichard, V., Palmantier, R., model of Alzheimer’s disease. J Alzheimers Dis 24 (Suppl. 2), 17–25. https://doi.
Pilorget, A., Larocque, D., Rivest, S., 2013. Toll-like receptor 4 stimulation with the org/10.3233/jad-2011-102070.
detoxified ligand monophosphoryl lipid A improves Alzheimer’s disease-related Perez-Gonzalez, R., Alvira-Botero, M.X., Robayo, O., Antequera, D., Garzon, M., Martin-
pathology. Proc Natl Acad Sci U S A 110, 1941–1946. https://doi.org/10.1073/ Moreno, A.M., Brera, B., de Ceballos, M.L., Carro, E., 2014. Leptin gene therapy
pnas.1215165110. attenuates neuronal damages evoked by amyloid-beta and rescues memory deficits
Ming, G.L., Song, H., 2011. Adult neurogenesis in the mammalian brain: significant in APP/PS1 mice. Gene Ther 21, 298–308. https://doi.org/10.1038/gt.2013.85.
answers and significant questions. Neuron 70, 687–702. https://doi.org/10.1016/j. Perry, E.K., Johnson, M., Ekonomou, A., Perry, R.H., Ballard, C., Attems, J., 2012.
neuron.2011.05.001. Neurogenic abnormalities in Alzheimer’s disease differ between stages of
Mirochnic, S., Wolf, S., Staufenbiel, M., Kempermann, G., 2009. Age effects on the neurogenesis and are partly related to cholinergic pathology. Neurobiol Dis 47,
regulation of adult hippocampal neurogenesis by physical activity and 155–162. https://doi.org/10.1016/j.nbd.2012.03.033.
environmental enrichment in the APP23 mouse model of Alzheimer disease. Phillips, H.S., Hains, J.M., Armanini, M., Laramee, G.R., Johnson, S.A., Winslow, J.W.,
Hippocampus 19, 1008–1018. https://doi.org/10.1002/hipo.20560. 1991. BDNF mRNA is decreased in the hippocampus of individuals with Alzheimer’s
Moreno-Jimenez, E.P., Flor-Garcia, M., Terreros-Roncal, J., Rabano, A., Cafini, F., Pallas- disease. Neuron 7, 695–702. https://doi.org/10.1016/0896-6273(91)90273-3.
Bazarra, N., Avila, J., Llorens-Martin, M., 2019. Adult hippocampal neurogenesis is Pineda, J.R., Encinas, J.M., 2016. The Contradictory Effects of Neuronal Hyperexcitation
abundant in neurologically healthy subjects and drops sharply in patients with on Adult Hippocampal Neurogenesis. Front Neurosci 10, 74. https://doi.org/
Alzheimer’s disease. Nat Med 25, 554–560. https://doi.org/10.1038/s41591-019- 10.3389/fnins.2016.00074.
0375-9.

20
M.L. Calió et al. Neurobiology of Disease 148 (2021) 105219

Power, D.A., Noel, J., Collins, R., O’Neill, D., 2001. Circulating leptin levels and weight Alzheimer’s disease. Brain Res 1678, 64–74. https://doi.org/10.1016/j.
loss in Alzheimer’s disease patients. Dement Geriatr Cogn Disord 12, 167–170. brainres.2017.10.012. Epub 2017 Oct 16.
https://doi.org/10.1159/000051252. Taniuchi, N., Niidome, T., Goto, Y., Akaike, A., Kihara, T., Sugimoto, H., 2007. Decreased
Prakash, A., Kumar, A., 2014. Role of nuclear receptor on regulation of BDNF and proliferation of hippocampal progenitor cells in APPswe/PS1dE9 transgenic mice.
neuroinflammation in hippocampus of beta-amyloid animal model of Alzheimer’s Neuroreport 18, 1801–1805. https://doi.org/10.1097/WNR.0b013e3282f1c9e9.
disease. Neurotox Res 25, 335–347. https://doi.org/10.1007/s12640-013-9437-9. Tapia-Arancibia, L., Aliaga, E., Silhol, M., Arancibia, S., 2008. New insights into brain
Priller, J., Prinz, M., 2019. Targeting microglia in brain disorders. Science (80-.) 365, BDNF function in normal aging and Alzheimer disease. Brain Res Rev 59, 201–220.
32–33. https://doi.org/10.1126/science.aau9100. https://doi.org/10.1016/j.brainresrev.2008.07.007.
Radi, E., Formichi, P., Battisti, C., Federico, A., 2014. Apoptosis and oxidative stress in Tezapsidis, N., Johnston, J.M., Smith, M.A., Ashford, J.W., Casadesus, G., Robakis, N.K.,
neurodegenerative diseases. J Alzheimers Dis 42 (Suppl. 3), S125–S152. https://doi. Wolozin, B., Perry, G., Zhu, X., Greco, S.J., Sarkar, S., 2009. Leptin: a novel
org/10.3233/jad-132738. therapeutic strategy for Alzheimer’s disease. J Alzheimers Dis. 16, 731–740. https://
Ramos-Lobo, Angela M., Donato Jr., Jose, 2017. The role of leptin in health and disease. doi.org/10.3233/JAD-2009- 10213375752606620632.
Temperature (Austin) 4 (3), 258–291. https://doi.org/10.1080/ Theodoropoulou, A., Metallinos, I.C., Psyrogiannis, A., Vagenakis, G.A.,
23328940.2017.1327003. Published online 2017 May 26. Kyriazopoulou, V., 2012. Ghrelin and leptin secretion in patients with moderate
Rapoport, M., Dawson, H.N., Binder, L.I., Vitek, M.P., Ferreira, A., 2002. Tau is essential Alzheimer’s disease. J Nutr Heal. Aging 16, 472–477. https://doi.org/10.1007/
to beta -amyloid-induced neurotoxicity. Proc Natl Acad Sci U S A 99, 6364–6369. s12603-012-0058-4.
https://doi.org/10.1073/pnas.092136199. Trejo, J.L., Carro, E., Torres-Aleman, I., 2001. Circulating insulin-like growth factor I
Reitz, C., Mayeux, R., 2014. Alzheimer disease: epidemiology, diagnostic criteria, risk mediates exercise-induced increases in the number of new neurons in the adult
factors and biomarkers. Biochem Pharmacol 88, 640–651. https://doi.org/10.1016/ hippocampus. J Neurosci 21, 1628–1634. https://doi.org/10.1523/JNEUROSCI.21-
j.bcp.2013.12.024. 05-01628.2001.
Rios, M., 2013. BDNF and the central control of feeding: accidental bystander or essential Tremblay, M.E., Stevens, B., Sierra, A., Wake, H., Bessis, A., Nimmerjahn, A., 2011. The
player? Trends Neurosci 36, 83–90. https://doi.org/10.1016/j.tins.2012.12.009. role of microglia in the healthy brain. J Neurosci 31, 16064–16069. https://doi.org/
Rizoulis, A.A., Karaoulanis, S.E., Rizouli, K.A., Papadimitriou, A., 2005. Serum leptin 10.1523/JNEUROSCI.4158-11.2011.
levels in patients with Alzheimer’s disease. Int J Caring Sci. 5, 43–49. Ulland, T.K., Song, W.M., Huang, S.C., Ulrich, J.D., Sergushichev, A., Beatty, W.L.,
Rodriguez, J.J., Jones, V.C., Tabuchi, M., Allan, S.M., Knight, E.M., LaFerla, F.M., Loboda, A.A., Zhou, Y., Cairns, N.J., Kambal, A., Loginicheva, E., Gilfillan, S.,
Oddo, S., Verkhratsky, A., 2008. Impaired adult neurogenesis in the dentate gyrus of Cella, M., Virgin, H.W., Unanue, E.R., Wang, Y., Artyomov, M.N., Holtzman, D.M.,
a triple transgenic mouse model of Alzheimer’s disease. PLoS One 3, e2935. https:// Colonna, M., 2017. TREM2 Maintains Microglial Metabolic Fitness in Alzheimer’s
doi.org/10.1371/journal.pone.0002935. Disease. Cell 170 (649-663), e13. https://doi.org/10.1016/j.cell.2017.07.023.
Rodriguez, J.J., Noristani, H.N., Olabarria, M., Fletcher, J., Somerville, T.D., Yeh, C.Y., Unger, T.J., Calderon, G.A., Bradley, L.C., Sena-Esteves, M., Rios, M., 2007. Selective
Verkhratsky, A., 2011. Voluntary running and environmental enrichment restores deletion of Bdnf in the ventromedial and dorsomedial hypothalamus of adult mice
impaired hippocampal neurogenesis in a triple transgenic mouse model of results in hyperphagic behavior and obesity. J Neurosci 27, 14265–14274. https://
Alzheimer’s disease. Curr Alzheimer Res 8, 707–717. https://doi.org/10.2174/ doi.org/10.1523/jneurosci.3308-07.2007.
156720511797633214. Unger, M.S., Schernthaner, P., Marschallinger, J., Mrowetz, H., Aigner, L., 2018.
Rohrdanz, E., Schmuck, G., Ohler, S., Kahl, R., 2001. The influence of oxidative stress on Microglia prevent peripheral immune cell invasion and promote an anti-
catalase and MnSOD gene transcription in astrocytes. Brain Res 900, 128–136. inflammatory environment in the brain of APP-PS1 transgenic mice.
https://doi.org/10.1016/s0006-8993(01)02277-6. J Neuroinflammation 15, 274. https://doi.org/10.1186/s12974-018-1304-4.
Sahay, A., Scobie, K.N., Hill, A.S., O’Carroll, C.M., Kheirbek, M.A., Burghardt, N.S., Uysal, N., Tugyan, K., Aksu, I., Ozbal, S., Ozdemir, D., Dayi, A., Gonenc, S., Acikgoz, O.,
Fenton, A.A., Dranovsky, A., Hen, R., 2011. Increasing adult hippocampal 2012. Age-related changes in apoptosis in rat hippocampus induced by oxidative
neurogenesis is sufficient to improve pattern separation. Nature 472, 466–470. stress. Biotech Histochem 87, 98–104. https://doi.org/10.3109/
https://doi.org/10.1038/nature09817. 10520295.2011.556665.
Sanabria-Castro, A., Alvarado-Echeverria, I., Monge-Bonilla, C., 2017. Molecular Vanevski, F., Xu, B., 2013. Molecular and neural bases underlying roles of BDNF in the
Pathogenesis of Alzheimer’s Disease: An Update. Ann Neurosci 24, 46–54. https:// control of body weight. Front Neurosci 7, 37. https://doi.org/10.3389/
doi.org/10.1159/000464422. fnins.2013.00037.
Scheff, S.W., Price, D.A., Schmitt, F.A., DeKosky, S.T., Mufson, E.J., 2007. Synaptic Ventriglia, M., Zanardini, R., Bonomini, C., Zanetti, O., Volpe, D., Pasqualetti, P.,
alterations in CA1 in mild Alzheimer disease and mild cognitive impairment. Gennarelli, M., Bocchio-Chiavetto, L., 2013. Serum brain-derived neurotrophic
Neurology 68, 1501–1508. https://doi.org/10.1212/01.wnl.0000260698.46517.8f. factor levels in different neurological diseases. Biomed Res Int 2013, 901082.
Schmitz, C., Rutten, B.P., Pielen, A., Schafer, S., Wirths, O., Tremp, G., Czech, C., https://doi.org/10.1155/2013/901082.
Blanchard, V., Multhaup, G., Rezaie, P., Korr, H., Steinbusch, H.W., Pradier, L., Voineskos, A.N., Lerch, J.P., Felsky, D., Shaikh, S., Rajji, T.K., Miranda, D., Lobaugh, N.
Bayer, T.A., 2004. Hippocampal neuron loss exceeds amyloid plaque load in a J., Mulsant, B.H., Pollock, B.G., Kennedy, J.L., 2011. The brain-derived neurotrophic
transgenic mouse model of Alzheimer’s disease. Am J Pathol 164, 1495–1502. factor Val66Met polymorphism and prediction of neural risk for Alzheimer disease.
https://doi.org/10.1016/S0002-9440(10)63235-X. Arch Gen Psychiatry 68, 198–206. https://doi.org/10.1001/
Schmued, L.C., Hopkins, K.J., 2000. Fluoro-Jade B: a high affinity fluorescent marker for archgenpsychiatry.2010.194.
the localization of neuronal degeneration. Brain Res 874, 123–130. https://doi.org/ von Bernhardi, R., Eugenin-von Bernhardi, L., Eugenin, J., 2015. Microglial cell
10.1016/s0006-8993(00)02513-0. dysregulation in brain aging and neurodegeneration. Front Aging Neurosci 7, 124.
Shanley, L.J., O’Malley, D., Irving, A.J., Ashford, M.L., Harvey, J., 2002. Leptin inhibits https://doi.org/10.3389/fnagi.2015.00124.
epileptiform-like activity in rat hippocampal neurones via PI 3-kinase-driven Warren, M.W., Hynan, L.S., Weiner, M.F., 2012. Lipids and adipokines as risk factors for
activation of BK channels. J Physiol 545, 933–944. https://doi.org/10.1113/ Alzheimer’s disease. J Alzheimers Dis. 29, 151–157. https://doi.org/10.3233/JAD-
jphysiol.2002.029488. 2012-111385.
Shors, T.J., Miesegaes, G., Beylin, A., Zhao, M., Rydel, T., Gould, E., 2001. Neurogenesis Weng, Z., Signore, A.P., Gao, Y., Wang, S., Zhang, F., Hastings, T., Yin, X.M., Chen, J.,
in the adult is involved in the formation of trace memories. Nature 410, 372–376. 2007. Leptin protects against 6-hydroxydopamine-induced dopaminergic cell death
https://doi.org/10.1038/35066584. via mitogen-activated protein kinase signaling. J Biol Chem 282, 34479–34491.
Scott, M.M., Lachey, J.L., Sternson, S.M., Elias, C.F., Friedman, J.M., Elmquist, J.K., 2009 https://doi.org/10.1074/jbc.M705426200.
Jun 10. Leptin targets in the mouse brain. J Comp Neurol. 514 (5), 518–532. https:// Winner, B., Winkler, J., 2015. Adult neurogenesis in neurodegenerative diseases. Cold
doi.org/10.1002/cne.22025. Spring Harb Perspect Biol 7, a021287. https://doi.org/10.1101/cshperspect.
Smith, M.A., Makino, S., Kim, S.Y., Kvetnansky, R., 1995. Stress increases brain-derived a021287.
neurotropic factor messenger ribonucleic acid in the hypothalamus and pituitary. Winner, B., Kohl, Z., Gage, F.H., 2011. Neurodegenerative disease and adult
Endocrinology 136, 3743–3750. https://doi.org/10.1210/endo.136.9.7649080. neurogenesis. Eur J Neurosci 33, 1139–1151. https://doi.org/10.1111/j.1460-
Snyder, J.S., Hong, N.S., McDonald, R.J., Wojtowicz, J.M., 2005. A role for adult 9568.2011.07613.x.
neurogenesis in spatial long-term memory. Neuroscience 130, 843–852. https://doi. Wright, A.L., Zinn, R., Hohensinn, B., Konen, L.M., Beynon, S.B., Tan, R.P., Clark, I.A.,
org/10.1016/j.neuroscience.2004.10.009. Abdipranoto, A., Vissel, B., 2013. Neuroinflammation and neuronal loss precede
Song, J.H., Yu, J.T., Tan, L., 2015. Brain-Derived Neurotrophic Factor in Alzheimer’s Abeta plaque deposition in the hAPP-J20 mouse model of Alzheimer’s disease. PLoS
Disease: Risk, Mechanisms, and Therapy. Mol Neurobiol 52, 1477–1493. https://doi. One 8, e59586. https://doi.org/10.1371/journal.pone.0059586.
org/10.1007/s12035-014-8958-4. Wyss-Coray, T., Rogers, J., 2012. Inflammation in Alzheimer disease-a brief review of the
Spalding, K.L., Bergmann, O., Alkass, K., Bernard, S., Salehpour, M., Huttner, H.B., basic science and clinical literature. Cold Spring Harb Perspect Med 2, a006346.
Bostrom, E., Westerlund, I., Vial, C., Buchholz, B.A., Possnert, G., Mash, D.C., https://doi.org/10.1101/cshperspect.a006346.
Druid, H., Frisen, J., 2013. Dynamics of hippocampal neurogenesis in adult humans. Yamada, K., Nabeshima, T., 2003. Brain-derived neurotrophic factor/TrkB signaling in
Cell 153, 1219–1227. https://doi.org/10.1016/j.cell.2013.05.002. memory processes. J Pharmacol Sci 91, 267–270. https://doi.org/10.1254/
Spangenberg, E.E., Lee, R.J., Najafi, A.R., Rice, R.A., Elmore, M.R., Blurton-Jones, M., jphs.91.267.
West, B.L., Green, K.N., 2016. Eliminating microglia in Alzheimer’s mice prevents Yamashita, T., Ninomiya, M., Hernandez Acosta, P., Garcia-Verdugo, J.M., Sunabori, T.,
neuronal loss without modulating amyloid-beta pathology. Brain 139, 1265–1281. Sakaguchi, M., Adachi, K., Kojima, T., Hirota, Y., Kawase, T., Araki, N., Abe, K.,
https://doi.org/10.1093/brain/aww016. Okano, H., Sawamoto, K., 2006. Subventricular zone-derived neuroblasts migrate
Stopa, E.G., Gonzalez, A.M., Chorsky, R., Corona, R.J., Alvarez, J., Bird, E.D., Baird, A., and differentiate into mature neurons in the post-stroke adult striatum. J Neurosci
1990. Basic fibroblast growth factor in Alzheimer’s disease. Biochem Biophys Res 26, 6627–6636. https://doi.org/10.1523/JNEUROSCI.0149-06.2006.
Commun 171, 690–696. https://doi.org/10.1016/0006-291x(90)91201-3. Yan, B.C., Choi, J.H., Yoo, K.Y., Lee, C.H., Hwang, I.K., You, S.G., Kang, I.J., Kim, J.D.,
Tai, J., Liu, W., Li, Y., Li, L., Holscher, C., 2017. Neuroprotective effects of a triple GLP-1/ Kim, D.J., Kim, Y.M., Won, M.H., 2011. Leptin’s neuroprotective action in
GIP/glucagon receptor agonist in the APP/PS1 transgenic mouse model of experimental transient ischemic damage of the gerbil hippocampus is linked to

21
M.L. Calió et al. Neurobiology of Disease 148 (2021) 105219

altered leptin receptor immunoreactivity. J Neurol Sci 303, 100–108. https://doi. Zeng, Q., Zheng, M., Zhang, T., He, G., 2016. Hippocampal neurogenesis in the APP/
org/10.1016/j.jns.2010.12.025. PS1/nestin-GFP triple transgenic mouse model of Alzheimer’s disease. Neuroscience
Yeh, S.H., Shie, F.S., Liu, H.K., Yao, H.H., Kao, P.C., Lee, Y.H., Chen, L.M., Hsu, S.M., 314, 64–74. https://doi.org/10.1016/j.neuroscience.2015.11.054.
Chao, L.J., Wu, K.W., Shiao, Y.J., Tsay, H.J., 2020. A high-sucrose diet aggravates Zhang, C., McNeil, E., Dressler, L., Siman, R., 2007a. Long-lasting impairment in
Alzheimer’s disease pathology, attenuates hypothalamic leptin signaling, and hippocampal neurogenesis associated with amyloid deposition in a knock-in mouse
impairs food-anticipatory activity in APPswe/PS1dE9 mice. Neurobiol Aging. 90, model of familial Alzheimer’s disease. Exp Neurol 204, 77–87. https://doi.org/
60–74. https://doi.org/10.1016/j.neurobiolaging.2019.11.018. 10.1016/j.expneurol.2006.09.018.
Yin, F., Sancheti, H., Patil, I., Cadenas, E., 2016. Energy metabolism and inflammation in Zhang, F., Wang, S., Signore, A.P., Chen, J., 2007b. Neuroprotective effects of leptin
brain aging and Alzheimer’s disease. Free Radic Biol Med 100, 108–122. https://doi. against ischemic injury induced by oxygen-glucose deprivation and transient
org/10.1016/j.freeradbiomed.2016.04.200. cerebral ischemia. Stroke 38, 2329–2336. https://doi.org/10.1161/
Yuan, P., Grutzendler, J., 2016. Attenuation of beta-Amyloid Deposition and strokeaha.107.482786.
Neurotoxicity by Chemogenetic Modulation of Neural Activity. J Neurosci 36, Zhang, Y.W., Thompson, R., Zhang, H., Xu, H., 2011. APP processing in Alzheimer’s
632–641. https://doi.org/10.1523/JNEUROSCI.2531-15.2016. disease. Mol Brain 4, 3. https://doi.org/10.1186/1756-6606-4-3.
Yuan, P., Condello, C., Keene, C.D., Wang, Y., Bird, T.D., Paul, S.M., Luo, W., Zhang, W., Gu, G.J., Zhang, Q., Liu, J.H., Zhang, B., Guo, Y., Wang, M.Y., Gong, Q.Y.,
Colonna, M., Baddeley, D., Grutzendler, J., 2016. TREM2 Haplodeficiency in Mice Xu, J.R., 2017. NSCs promote hippocampal neurogenesis, metabolic changes and
and Humans Impairs the Microglia Barrier Function Leading to Decreased Amyloid synaptogenesis in APP/PS1 transgenic mice. Hippocampus 27, 1250–1263. https://
Compaction and Severe Axonal Dystrophy. Neuron 90, 724–739. https://doi.org/ doi.org/10.1002/hipo.22794.
10.1016/j.neuron.2016.05.003. Zhu, X., Smith, M.A., Perry, G., Aliev, G., 2004. Mitochondrial failures in Alzheimer’s
Zemlan, F.P., Thienhaus, O.J., Bosmann, H.B., 1989. Superoxide dismutase activity in disease. Am J Alzheimers Dis Other Demen 19, 345–352. https://doi.org/10.1177/
Alzheimer’s disease: possible mechanism for paired helical filament formation. Brain 153331750401900611.
Res 476, 160–162. https://doi.org/10.1016/0006-8993(89)91550-3.

22

You might also like