Next Article in Journal
CircTCF4 Suppresses Proliferation and Differentiation of Goat Skeletal Muscle Satellite Cells Independent from AGO2 Binding
Next Article in Special Issue
Amyloid Beta in Aging and Alzheimer’s Disease
Previous Article in Journal
Boosted Activity of g-C3N4/UiO-66-NH2 Heterostructures for the Photocatalytic Degradation of Contaminants in Water
Previous Article in Special Issue
Amyloid β, Lipid Metabolism, Basal Cholinergic System, and Therapeutics in Alzheimer’s Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Pathological Nuclear Hallmarks in Dentate Granule Cells of Alzheimer’s Patients: A Biphasic Regulation of Neurogenesis

by
Laura Gil
1,
Erika Chi-Ahumada
2,
Sandra A. Niño
3,
Gabriela Capdeville
4,
Areli M. Méndez-Torres
2,
Carmen Guerrero
5,
Ana B. Rebolledo
5,
Isabel M. Olazabal
1 and
María E. Jiménez-Capdeville
2,*
1
Facultad de Medicina, Universidad Alfonso X el Sabio (UAX), Avenida de la Universidad, 1, 28691 Villanueva de la Cañada, Spain
2
Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, Av. Venustiano Carranza 2405, San Luis Potosí 78210, Mexico
3
Geroscience Center for Brain Health and Metabolism (GERO), Santiago de Chile 7750000, Chile
4
Escuela de Medicina, Universidad Panamericana, Ciudad de Mexico 03920, Mexico
5
Banco de Cerebros (Biobanco), Hospital Universitario Fundación Alcorcón, 28922 Alcorcón, Spain
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(21), 12873; https://doi.org/10.3390/ijms232112873
Submission received: 24 September 2022 / Revised: 14 October 2022 / Accepted: 18 October 2022 / Published: 25 October 2022
(This article belongs to the Special Issue Frontier on Alzheimer's Disease)

Abstract

:
The dentate gyrus (DG) of the human hippocampus is a complex and dynamic structure harboring mature and immature granular neurons in diverse proliferative states. While most mammals show persistent neurogenesis through adulthood, human neurogenesis is still under debate. We found nuclear alterations in granular cells in autopsied human brains, detected by immunohistochemistry. These alterations differ from those reported in pyramidal neurons of the hippocampal circuit. Aging and early AD chromatin were clearly differentiated by the increased epigenetic markers H3K9me3 (heterochromatin suppressive mark) and H3K4me3 (transcriptional euchromatin mark). At early AD stages, lamin B2 was redistributed to the nucleoplasm, indicating cell-cycle reactivation, probably induced by hippocampal nuclear pathology. At intermediate and late AD stages, higher lamin B2 immunopositivity in the perinucleus suggests fewer immature neurons, less neurogenesis, and fewer adaptation resources to environmental factors. In addition, senile samples showed increased nuclear Tau interacting with aged chromatin, likely favoring DNA repair and maintaining genomic stability. However, at late AD stages, the progressive disappearance of phosphorylated Tau forms in the nucleus, increased chromatin disorganization, and increased nuclear autophagy support a model of biphasic neurogenesis in AD. Therefore, designing therapies to alleviate the neuronal nuclear pathology might be the only pathway to a true rejuvenation of brain circuits.

1. Introduction

Maintaining hippocampal synaptic plasticity is crucial for the storage and persistence of memories through brain aging [1,2,3]. Neuronal aging is directly related to DNA damage and disruption of genomic integrity [4,5,6,7,8,9,10,11,12,13] and it is the main cause of neurodegenerative diseases such as Alzheimer’s disease (AD) [14,15,16,17,18,19,20,21]. Considering that hippocampal atrophy is one of the earliest morphological alterations detected in AD by imaging techniques [22,23,24,25], many studies are interested in the progression of these hippocampal changes.
The hippocampus has many different functions, which are carried out through the trisynaptic pathway described by Cajal [26]. This pathway is a complex and dynamic circuit from the entorhinal cortex to the dentate gyrus (DG) and two hippocampal areas, CA1 and CA3. Although these areas are physically close, each presents specific cellular and molecular structure and function [27]. It has recently been described that, under AD conditions, pyramidal CA1 neurons undergo a nuclear transformation that distinguishes them from the neurons of neurologically healthy senile individuals [28,29,30]. These nuclear changes are associated with the aberrant re-entry of post-mitotic pyramidal neurons into the cell cycle [31] and the later development of neurofibrillary tangles (NFTs), toxic cytoplasmic aggregates that characterize intermediate and late AD stages [32]. The pathological hallmarks of aberrant genomic expression in the nucleus of AD neurons are the absence of the chromatin-modifier Tau protein [33,34], the instability of heterochromatin blocs constituting the domains associated with lamin (LADs) and the nucleolus (NADs) [28,35], histone modifications, and a dysfunctional nucleus-cytoplasm restructuration, whereby lamin A expression and altered nucleus-cytoplasm transport through the nuclear pore play an essential role [30,36].
Along with lamin dysfunction [29,30,31] and cytoplasmic Tau pathology [37,38,39], the aberrant chromatin structure of pyramidal AD neurons in humans, rodents, and Drosophila AD models is characterized by the dysregulated expression of epigenetic markers H4K20me3 [40], H3K9me3 [41] H3K9me2 [42], and H3K9ac [39]. An altered chromatin pattern has also been reported in cell cultures of fibroblast-derived neurons from AD patients (iNs) with MAPT overexpression [19]. In all cases, nuclear Tau has been closely related to chromatin organization [43,44,45]. In contrast to the regions occupied by pyramidal post-mitotic neurons, the subgranular zone (SGZ) of the DG is one of the few brain areas where neurogenesis persists through adulthood in most mammal species [46]. However, whether this occurs in humans is still under debate [47,48,49,50,51,52,53,54,55]. It has been proposed that new neurons generated in adulthood are involved in adaptive behavioral responses to cognitive and emotional challenges [56,57,58].
The granule cell layer (GCL) displays great cellular heterogeneity determined by its ontogenic origin [59,60,61,62,63]. In rodents, the first granule cells are born in the last embryogenesis stage (embryo granule cells) and the highest rates of postnatal neurogenesis are reached during the first two weeks of life (postnatal granule cells). Though persistent in adult life, the rate of neurogenesis decreases with age [64,65,66]. Neural adult stem cells (NSCs) or quiescent neural progenitors progress to neurons through the proliferation of several intermediate progenitors (amplifiers type 2A and 2B), which lead to neuroblasts or type-3 progenitors after mitotic stages. Neuroblasts differentiate into immature adult-born neurons, which develop axons and ramified dendrites in 4–6 weeks [67,68,69]. Although functionally connected to the tri-synaptic pathway, NSCs are electrophysiologically different from mature granule cells [70]. Their higher excitability and lower threshold for synaptic plasticity suggest that the new and immature adult-born neurons play a unique role in learning and memory within the hippocampal circuit [71].
In rodents, immature adult-born neurons reach morphologic and functional maturity at eight weeks of age, becoming indistinguishable from embryo- and early postnatal-born neurons [72,73,74,75]. In contrast, the maturation process of adult-born neurons of the rhesus monkeys, a species more closely related to humans, lasts up to 24 months [59,76,77]. This extended maturation in primates results in a longer period of favored synaptic plasticity of immature adult-born neurons and an earlier decay of neurogenesis [78,79,80].
Describing nuclear changes throughout senile and AD stages in DG is challenging because this dynamic brain region harbors many immature granular cells in diverse proliferative states [81,82], as opposed to the quiescent pyramidal neurons from the CA1 region [27,83]. In murine models, brain activity related to the exploration of novel environments causes multiple double-strand breaks (DSBs) in DG neurons [84]. However, DSBs are usually repaired within 24 h, especially during sleep [84,85]. Interestingly, DSBs can accumulate in murine AD models, cell cultures, and post-mortem human brain samples obtained from early AD patients [86,87]. These findings have important consequences for DG neurons as the accumulation of DNA damage and failure to repair DNA in proliferating cells cause permanent cell-cycle arrest, senescence, or apoptosis, as predetermined antitumoral mechanisms [88]. In this context, a recent study employing phase-contrast X-ray computed tomography of post-mortem AD human hippocampi showed important nuclear alterations in granular DG cells [89].
This study analyzed the progression of nuclear pathology in DG neurons at early, intermediate, and late AD stages compared with healthy aged and senile conditions. Similar to previous studies in pyramidal CA1 neurons [28], we examined the expression of the chromatin markers phospho-Tau AT100 y AT8, the components of nuclear lamin (NL), and epigenetic histone modifications related to chromatin condensation. In light of recent reports of lamin degradation by autophagy as a response to genotoxic insult [90], we also explored autophagy as a component of nuclear AD pathogeny [91,92,93,94].

2. Results

2.1. Nuclear Changes in the Entorhinal Cortex at Early AD Stages

Layer II pyramidal neurons of the entorhinal cortex are the main input to DG granular neurons. We observed that these pyramidal neurons displayed AD-associated changes that followed a similar time course to those reported in pyramidal hippocampal cells. Figure 1 shows the redistribution of Lamin B2 (Table 1) to the nucleoplasm, which started in the senile stage and increased in AD I–II stages (A–C). Increased lamin B2 expression was accompanied by the expression of Lamin A (Table 1) from early to late AD stages (H–J). The shift of phospho-Tau from the nucleus to the cytoplasm preceded NFT formation. The presence of AT100 (Table 1) in the nucleus increased in senile subjects (L) and constituted cytoplasmic NFTs in AD III–VI (N–O). Moreover, AT8 (Table 1) first appeared in the nucleus of senile neurons (Q) and formed NFTs from I to VI AD stages (R–T).

2.2. The Distribution of Lamin B2 Uncovers Two Populations of DG Granular Neurons

Lamin B2 plays an important role in chromatin organization and nuclear architecture. It forms a protein meshwork on the nucleoplasmic side of the nuclear membrane. Lamin B2 staining in granular DG neurons was found in thin layers surrounding the nucleus in some cells to the whole nucleoplasm in others. The proportion of these two types of nuclear Lamin B2 neurons varied drastically across age and AD disease progression. In healthy adults, perinuclear Lamin B2 predominated (Figure 2A), but senile and AD I–II stages were characterized by immunopositive nucleoplasms (Figure 2, senile, AD I–II). In contrast, intermediate and late AD stages showed mostly perinuclear Lamin B2 staining around clear nucleoplasms (Figure 2, AD III–IV, AD V–VI). As expected, no Lamin A expression was detected in any condition, given its normal regulation through miR-9 in healthy neurons [11,12].

2.3. Phosphorylated Tau (AT100 and AT8) Is Absent in the DG Cell Nucleus at Intermediate and Late AD Stages

Nuclear immunopositivity to AT100 was present in adult and senile DG cells and increased notably at early AD stages (Figure 3A–C). At AD III–IV, AT100 staining was no longer homogenous; instead, it faded and concentrated in nuclear puncta. Moreover, extranuclear AT100 was observed in the neuropile and some cells still showed nuclear positivity (Figure 3D). Scarce nuclear positivity and increased extracellular AT100 staining were accentuated at late AD stages (Figure 3E).
Strong AT8 nuclear positivity appeared only in senile DG neurons and started fading in AD I–II (Figure 3F,G). From AD III to AD VI, AT8 nuclear positivity was no longer observed. Furthermore, neuropile aggregates and even a few NFTs were found at late AD stages (Figure 3H–J). The quantification of nuclear AT100 and AT8 immunopositivity showed the lowest levels in late AD stages (Figure 3C,D).

2.4. AD Granular Neurons Show Drastic Changes in Epigenetic Chromatin Markers

As reported in the literature, the suppressive mark H3K9me3 (Table 1) increased sharply at early AD stages and remained elevated until late stages (Figure 4A,B). A similar pattern was observed in H4K20me3 (Figure 4E,F and Table 1). Chromatin markers H3K4me3 (Figure 4C,D) and H3K36me3 (Figure 4G,H) (Table 1), which are associated with increased genic expression, also changed from senile to AD I–II, indicating complete chromatin remodeling of DG neurons.

2.5. Intermediate and Late AD Stages Are Characterized by Increased Nuclear and Perinuclear Autophagy Marker (LC3)

We found a striking spatial rearrangement of LC3 (Table 1) from the healthy to the AD condition and across AD stages. In healthy and early AD stages, LC3 was mainly localized in the cytoplasm of DG cells (Figure 5A–C); however, this autophagy marker increased in the nucleus and perinucleus from AD III to AD VI (Figure 5D,E), suggesting a nucleophagy phenomenon. The increased nuclear LC3 immunopositivity is concomitant with the disappearance of nucleoplasmic Lamin B2 (Figure 5E,F). In agreement with this, nuclear L3 increases in a similar manner to perinuclear Lamin B2 (Figure 6A,B).

3. Discussion

This study describes the nuclear pathology of DG neurons throughout AD stages as compared with aging-related changes. We found nuclear alterations in granular cells different from those reported in pyramidal neurons from CA1 and CA3 hippocampal regions. However, pyramidal neurons from layer II of the entorhinal cortex, which are the main input to the DG, also present the nuclear pathology previously described in hippocampal subfields of AD brains [28]. We describe important changes in nuclear lamin B2 distribution across different AD stages compared with healthy DG cells. These changes occur alongside the disappearance of phosphorylated Tau forms from the nuclei and, interestingly, with increased nuclear autophagy markers in intermediate and late AD stages. As a hallmark of chromatin transformation from a healthy to a pathologic state, the remarkable increase in the epigenetic markers H3K9me3 and H3K4me3 clearly delineates the limit between aging and AD in granular cells. In the context of the abundant data on adult human neurogenesis (AHN), these results obtained in the dynamic niche of DG neurons suggest a biphasic model for the regulation of neuronal survival through AD.

3.1. Similarities between Changes in CA1 and Entorhinal Cortex Pyramidal Cells in AD

The tri-synaptic circuit contains three regions with post-mitotic pyramidal neurons containing big nuclei and extensively ramified dendritic trees [22]. However, neuropathological data suggest that they have a different vulnerability to aging and AD [23,95]. Pyramidal neurons from these three regions accumulate nuclear Tau (phosphoepitope AT100) associated with aging, which constitutes a hallmark of aged chromatin [96]. Nuclear Tau is a global genome stabilizer, especially of heterochromatin blocs NADs and LADs [33,97,98]. Oxidative DNA damage associated with neuronal aging triggers the exit from the quiescent state and the aberrant re-entry into the cell cycle [99,100]. The transformation of a healthy senile brain to AD pathology inaugurates the coexistence of two types of pyramidal neurons. One population expresses nucleoplasmic lamin B2, a sign of active transcription and abortive cell cycle that leads to neuronal death [31,101,102,103,104], and the population of AD neurons, characterized by an anomalous lamin A expression [28,29,30,105], which stops the cell cycle, thus protecting the nucleus and generating a new cytoskeleton [106,107,108]. Early AD stages are also distinguished by the gradual disappearance of nuclear AT100 [34], which is strongly associated with chromatin disorganization [29].

3.2. The Dynamic Neuronal Nature of DG through Development and Aging

The DG is considered the “gateway” to the hippocampus that introduces different cellular and functional characteristics into the tri-synaptic circuit. The GCL includes (i) small post-mitotic mature neurons with short dendritic trees and different ages [109]; (ii) immature newborn granule neurons in different proliferative stages [110]; and (iii) quiescent radial glia cells, which generate the latter group and are located in the subgranular zone (SGZ) niche [111]. Notably, the number of immature granule newborn cells that the GCL incorporates is not constant; rather, it changes depending on internal and external factors [112,113,114,115,116]. In humans, the persistence of AHN [54,55,56] is supported by BrdU (5-bromo-2′-deoxyuridine) incorporation and 14C quantification in proliferating neurons [48,49]. However, the human GCL shows more immature neurons than other non-primate mammals [52,76,117].
Aging is one of the endogenous factors that account for the progressively slower incorporation of newborn mature neurons into the GCL [67,118]. These cells gradually replace neurons generated throughout development [49]. Compared with other hippocampal regions, the DG shows less aging-associated neuronal loss [54,55,68] and a lower decline rate [119,120,121,122,123,124]. DG neurogenesis occurs in five phases characterized by the expression of specific cell markers [27,84,85,86,87,88]: proliferative (stage 1), differentiation (stage 2), migration (stage 3), targeting (stage 4), and synaptic integration (stage 5) [50]. From the proliferative point of view [110], immature newborn granular neurons in stages 1, 2, and 3 are in a replicative cycle. In contrast, stage 4 granular neurons have differentiated into a post-mitotic state, associated with the temporal expression of calcium-binding protein calretinin [125,126,127]. Finally, stage 5 synaptically mature neurons express calbindin [50].
Our results indicate that nuclear phenotypes of senile GCL neurons are slightly different from those of adult brains regarding nuclear lamin structure and chromatin organization. In rat GCL, lamin B2 positivity increases throughout neurogenesis, reaching its highest level in mature neurons [128]. In human GCL, however, B2 positivity can be found in the perinucleus, similar to senile pyramidal neurons [28], as well as in the nucleus. B-type lamins redistribute from the nuclear envelope to replication factories during S-phase [129] and modulate nucleolar function [130]; therefore, the presence of B2 lamin in the nucleus suggests that the cells are in a replicative cycle, probably as newborn granular neurons in the first maturation stages (1, 2, and 3). In contrast, the cells with strong perinuclear positivity [106] would be either (i) newborn post-mitotic granular neurons undergoing different axonal and dendritic development (stages 4 and 5) [71,131] or mature neurons already structurally and functionally integrated into the hippocampus. The significantly higher proportion of perinuclear positivity in intermediate and late AD stages indicates fewer immature neurons, lower neurogenesis, and fewer adaptations to environmental factors.

3.3. Heterochromatin Markers and Nuclear Tau: Hallmarks of Early AD in Granular Cells

DG cells with aged chromatin cannot properly repair DSBs [132]. DSBs are involved in the synaptic activity required to explore novel environments [84,85]; thus, the accumulation of DNA mutations affects genome integrity [20,133]. Our results show that phosphorylated Tau is increased in the nuclei of senile GCL cells, similar to previous reports of pyramidal neurons from CA1, CA3 [28,96], and EC (Figure 1). Tau plays an important role in the protection against DNA damage [134,135]. Moreover, DSBs’ induction immediately increases Tau in the nuclear fraction of primary mouse cortical neurons [136]. The phosphorylation of the AT8 site is specific for senile chromatin [28] and has been directly related to DSBs’ induction in cortical neurons [136]. In granular neurons, the double AT100 and AT8 presence seems to determine the nuclear Tau function interacting with aged chromatin, favoring DSBs’ repair [136,137] and maintaining genomic stability [43,138]. This stabilization would compensate for the decrease in the constitutive heterochromatin marker H3K9me3 [139,140,141], which is fundamental for the structural organization of NADs and LADs [142] and genomic integrity [143].
In this context, as senile GCL preserves the number of functional neurons [144,145,146,147], the regression of dendritic trees and synaptic dysfunction would be more closely related to the observed age-related cognitive decline [55,147,148,149]. What molecular events in the GCL dynamic niche could explain the dysfunctional communication progressing through AD?
Using three-dimensional histology based on phase-contrast computed tomography, Eckermann and collaborators recently reported a decreased nuclear volume of GCL cells in AD as compared with healthy brains, which was related to a higher ratio of heterochromatin to euchromatin [89]. In agreement with these observations, we found histological evidence of the upregulation of the constitutive heterochromatin epigenetic marker H3K9me3 [139,150]. Furthermore, H3K4me3 is a hallmark of the GCL neuronal transformation from senile to AD. Similar increments of H3K9me3 have been reported in cortex motor neurons of AD and Huntington’s disease patients [151,152]. H3K9 trimethylation maintains the structural heterochromatin domains and contributes to silencing repetitive sequences of peri- and centromeric regions [150] and stabilizing most of the long interspersed nuclear elements and endogenous retroviruses [153]. Conversely, the absence of H3K9me3 dissolves heterochromatin [139], leading to genomic instability and chromatin disorganization [143]. Based on an integrated analysis of genome-wide ChiP- and mRNA-sequencing, abnormal heterochromatin remodeling by increased neuronal H3K9me3 expression results in down-regulation of (i) synaptic function, (ii) neuronal differentiation, and (iii) cell motility-related genes [114]. In the human GCL, the transformation of senile granular neurons to the AD phenotype implies high H3K9me3 expression along disease progression, reaching its highest level at early AD stages (AD I–II). Nevertheless, the exact role of increased H3K9me3 is still controversial as it may be affecting populations in different maturity states. In vivo reprogramming of murine aging models to elevate H3K9me3 in the DG showed that it increases the survival of newborn DG neurons and improves memory functions [154].
Nuclear alterations, including NL reinforcing and heterochromatin stabilization in the absence of cytoplasmic pathology, characterize early AD in CA1, CA3, EC pyramidal neurons, and DG cells. These hippocampal functional changes are associated with learning and memory alterations and AHN as an adaptation to hippocampal functioning [127]. This AHN is similarly stimulated by pathologies like hippocampal or cortical damage associated with ischemic insults or epileptic seizures [155,156,157,158,159,160]. In this context, several groups reported increased neurogenesis and proliferation markers in murine models and human AD, such as doublecortin (DCX-labeled immature neurons) [161,162,163], calretinin (CR), and Ki67 [164]. Given that the levels of DCX and CR increase in early AD compared with late AD stages, an AHN increment might be a characteristic response to the transformation from senile to AD neurons [55]. Our results support AHN at early AD stages because we observed the upregulation of the epigenetic silencer H3K9me3 along with an abrupt increase in H3K4me3 expression. As H3K4me3 is considered a sign of actively transcribed promoters of cellular function/identity, we believe that these cells are under active transcription [165].
Both post-transductional trimethylations of H3 are the highest at the early AD stages and slightly decline at the late stages (Figure 4). In this context, it is remarkable that, during the intermediate and late AD stages, the DG shows more neurons with perinuclear lamin B2 than in adult, senile, and even AD I–II stages. These nuclei with reinforced NL and less active transcription might correspond to the pool of newborn granular post-mitotic neurons with a suboptimal development of their dendritic tree and affected synaptic connections [114,129]. Further experiments will confirm the identity of this cell population with increased perinuclear lamin B2. Nevertheless, the increase in post-mitotic cells in AD III–IV would result from a higher rate of cells entering the cell cycle, coinciding with the upregulation of AT100 at early AD stages. As mentioned before, nuclear Tau protects the structural integrity of condensed chromatin [100,101] and has been widely reported in the nucleolus of human hippocampal neurons and cultured cells [28,34,96,166,167].
Hence, we propose that the synchronic nuclear pathology throughout the tri-synaptic circuit profoundly affects the regulation of the neurogenic niche, leading to a robust increase in immature cells, DCX+, and CR+, which could represent compensation for neuronal damage and loss [28].

3.4. Increased Nuclear Autophagy in Late AD Stages Supports Biphasic Neurogenesis Changes in AD

The higher numbers of DCX+ and CR+ granular cells detected at the III–IV AD stages [55,161,162,164] have also been reported in other neuropathologies such as epilepsy, schizophrenia, and Creutzfeld–Jacobs disease [164,168,169,170,171]. This situation has led pathologists to coin a new term to define this peculiar state: “immature dentate gyrus” (iDG) [168]. However, this proliferation boost does not generate a net increase in granular post-mitotic neurons in maturation stages at intermediate and late AD stages [55,161,162]; rather, the number of granular DG cells remains relatively stable [164]. In epilepsy, for example, this “partial rejuvenation” process is followed by aberrant maturation and integration into the tri-synaptic circuit, which contributes to the further development of spontaneous and frequent seizures [171,172]. Contrarily, our results show, for the first time, that intermediate and terminal AD stages are characterized by nuclear degeneration through autophagy-based lamin B2 degradation, known as nucleophagy [90,173].
Autophagy is essential to induce senescence after oncogenic activation derived from DNA damage [174]; this mechanism involves lamin B1 nucleophagy [175]. Autophagy is also considered a common trait of neurodegenerative diseases like AD [176,177]. Accordingly, increased nucleophagy has been reported in patients with dentatorubral-pallidoluysian atrophy (DRPLA), one of the diseases associated with poliglutamine (polyQ) repeats, in which affected individuals develop symptoms of ataxia, epilepsy, and dementia [178]. Functional autophagy seems to prevent neuronal senescence [179]. In our study, increased nucleophagy in the DG overlaps with nuclear depletion of AT 100 at intermediate AD stages. In fact, the loss of nuclear Tau function and the consequent heterochromatin disruption have been suggested as the actual triggering factors for AD [138]. Chromatin disorganization is initiated by nuclear Tau downregulation and lamin B2 disruption, which involves the disappearance of the nucleoplasmic NL structure [180]. Under these conditions, the heterochromatin markers H3K9me3 and H4K20me3 still hold back the phenomenon, and transcription is still activated, as indicated by increased H3K4me3 and H3K36me3 markers. However, in late AD stages (V–VI), chromatin becomes significantly disorganized, as indicated by the decreased epigenetic markers, putting GCL neurons to the same end as pyramidal cells of the tri-synaptic circuit [29] and neurons from the transgenic Drosophila and mouse AD models [37,38].

4. Materials and Methods

4.1. Human Brain Samples and Immunohistochemistry

Human brain tissue samples were collected at the Tissue Biobank of the Hospital Universitario Fundación Alcorcón (HUFA), C/ Budapest 1, 28922 Alcorcón, Madrid, España. The protocol was reviewed and approved by the institutional Clinical Research Ethics Committee of HUFA (47/2018, in October 2018). Samples were obtained from four healthy subjects of different ages (37, 42, 65, and 76 years) with no evidence of cognitive impairment or dementia and six AD cases diagnosed post-mortem according to Braak’s classification. To classify the specimens, we used the characteristics of I–VI AD stages updated for paraffin sections and immunocytochemistry by Braak and collaborators in 2006 [180]. Three samples presented early stages I–II (ages 68 and 77), two intermediate stages III–IV (72 and 82), and two late stages (79 and 89). Paraffin sections obtained at the DG level of the hippocampus were processed for immunohistochemistry with the following antibodies.
The immunohistochemical assessment was performed on 4 µm thick dewaxed sections. After boiling the sections in a pressure cooker with DIVA decloaking solution (Biocare Medical, LLC, Concord, CA, USA) for epitope recovery, endogenous peroxidases were blocked with Dako Peroxidase Blocking Reagent (DAKO, Glostrup, Denmark). Next, diluted primary antibodies were incubated overnight at 4 °C. After incubation with the primary antibody and buffered phosphate rinses (PBS), sections were exposed to the streptavidin-biotin marked secondary antibody. The peroxidase reaction was visualized with 3′3-diaminobenzidine. The sections were finally counterstained with hematoxylin (HE), dehydrated, and cover-slipped for microscopic observation. The sections were observed on an Olympus microscope equipped with a digital camera (Amscope, Irvine, CA, USA).

4.2. Image Acquisition and Analysis

DG section and image serial photomicrographs at 20X, 40X, and 100X were obtained all along the DG structure. The total number of microphotographs (16 bit) inspected ranged from 40 to 60 per subject. All image analyses were performed on raw data.
Image blinding: For each antibody, all images were taken on the same day. Collected images were stitched together using Stitching plug-in of Fiji/Image J2 version 2.9.0/1.53t; Java 1.8.0_202 (Wayne Rasband, Bethesda, MD, USA). Image files were each assigned a random alphanumeric code to ensure that the subsequent steps in image segmentation were blind to diagnosis (control vs. AD) and donor identity.
Background subtraction: To minimize background differences across sections, the contrast and brightness were adjusted equally for all images within a series. The rolling ball Fiji algorithm was used with a radius of 200 pixels. Non-specific background staining was subtracted from the measured values.
Region of interest segmentation and processing: A specific built-in algorithm called color deconvolution was used [181]. This algorithm separated the staining of HE and DAB into three different panels, namely, HE (panel 1), DAB-only image (panel 2), and background (panel 3). Panel 2 was converted to grayscale for threshold selection and was auto-thresholded using the Otsu Fiji method and converted into a mask [28]. The area was selected (region of interest, ROI) by adjusting the brush size to the neuron soma, with a minimum cell size of 100 pixels. Masks were then eroded to separate the nucleus. The mean optical density of the immunopositive cells was calculated and pixel intensities were maintained within a linear range to ensure accurate quantification.
To quantify the LC3 nuclear area, the threshold was established in the images and particles were evaluated within defined circular ROIs of 10 µm in diameter per picture/field/subject and compared to the global average. Only particles larger than 0.5–1.5 pixels were selected by the algorithm CellProfiler 2.2.0 (https://cellprofileranalyst.org/, accessed on 9 June 2021). Aggregate analysis was verified by manual inspection of each neuron at 100X magnification. Finally, images containing errors in focus, tracing, or other artifacts, such as lint fragments or dye precipitates, were omitted from the analysis.

4.3. Data Analysis

All data were analyzed for normality and variance homogeneity and several tests were used for statistical comparison. Statistical calculations were carried out in Prism 9.4.1 (GraphPad Software, San Diego, CA USA, www.graphpad.com, accessed on 9 June 2021) Adult, senile, and AD groups were compared with either a one-way ANOVA followed by Dunnett’s test or a Kruskal–Wallis test followed by the Mann−Whitney U test in order to identify significant differences (* p < 0.05) versus the control group.

5. Conclusions

Similar to prion disease [164], our results suggest a biphasic model of neurogenesis regulation in AD [182]. Nuclear alterations in all regions of the tri-synaptic circuit induce a strong neurogenesis reactivation in DG. These findings, together with the evidence of substantial transcriptional heterogeneity within the hippocampus [183], could be related to the lower vulnerability of DG to oxidative stress, caloric restriction, and different insults [1,184,185,186,187,188,189].
The elevated number of granular cells, immature and without functional chromatin, do not integrate into the GCL, but suffer nucleophagy at intermediate and late AD stages. Although somehow in disagreement with the data presented by Tobin and collaborators [55], the laminar and chromatin pathology affecting all granular neurons (namely, old mature, newborn immature, and those generated during “AD neurogenesis”) has negative consequences on DG gene transcription, synaptic plasticity, and integrated cognitive functions [13]. In summary, our results suggest that generalized nuclear pathology plays a central role in the neurogenic state of the DG through aging and early AD stages, which later progresses into increasingly severe genomic dysfunction. This dysfunction generates progressive cognitive deficits determined by the neuron’s different vulnerability and connectivity, whereby cytoplasmic pathology plays a delayed role.

6. Future Perspectives

Dysfunction of all areas of the tri-synaptic hippocampal circuit converges in impaired AHN, leading to severe cognitive deficits in AD brains. The idea that an increased number of adult-generated neurons could ameliorate cognitive impairment [190] becomes questionable in light of the aberrant neurogenesis observed at early AD stages [191], perhaps as a result of neuron subtype 2 (SOX-) proliferation [192]. Regardless of the quiescent or replicative nature of granular GCL neurons, EC, CA1, and CA3 pyramidal neurons, their nuclear pathology appears simultaneously in the damaged hippocampus. All neurons generate different versions of a closed chromatin structure and a rigid nucleoskeleton. Therefore, the crucial point is that aging induces chromatin transformations that drive cells to an aberrant cell-cycle re-entry in order to repair their genome. This process finally leads to cancer or degeneration. Designing therapies to alleviate the neuronal nuclear pathology might be the only pathway to a true rejuvenation of brain circuits.

Author Contributions

L.G.: Conceptualization, investigation, and writing; E.C.-A.: Methodology and visualization; S.A.N.: Formal analysis, software, and visualization; G.C.: Analysis and visualization; A.M.M.-T.: Analysis and visualization; C.G.: Validation and resources; A.B.R.: Investigation and resources; I.M.O.: Funding acquisition and project administration; M.E.J.-C.: Conceptualization, methodology, supervision, investigation, and writing. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by the Fundación “Alfonso X el Sabio”-Banco Santander, Spain, grant 1.400.001. Special thanks to the Biobanco Hospital Universitario Fundación Alcorcón.

Informed Consent Statement

The study was conducted in accordance with the Declaration of Helsinki, and approved by the Clinical Research Ethics Committee of Hospital Universitario Fundación Alcorcón (47/2018, in October 2018). Informed consent was obtained from all subjects involved in the study.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ADAlzheimer’s disease
AHNadult human neurogenesis
AT8Tau protein phosphorylated at Ser202/Thr205
AT100Tau protein phosphorylated at Thr212/Ser214
BrDU5-bromo-2′-deoxyuridine, proliferation marker
CA1Ammon’s horn region 1 (hippocampal region with pyramidal cells)
CA3Ammon’s horn region 3 (hippocampal region with pyramidal cells)
CRcalretinin protein, neurogenesis marker
ChIPchromatin immunoprecipitation
DCXdoublecortin protein, neurogenesis marker
DGdentate gyrus
DRPLAdentatorubral-pallidoluysian atrophy
DSBsdouble strand breaks
ECentorhinal cortex
GCLgranule cell layer
H3K9me3trymethylated Histone 3 protein, heterochromatin marker
H4K20me3trymethylated Histone 4 protein, heterochromatin marker
H3K4me3trymethylated Histone 3 protein euchromatin marker
H3K36me3trymethylated Histone 3 protein euchromatin marker
iDGimmature dentate gyrus
iNsinduced neurons
Ki67proliferation marker
LADslamin associated domains
LC3autophagy marker (protein MAP1LC3B)
MAPTTau protein
NADsnucleolus-associated domains
NLnuclear lamin
NFTneurofibrillary tangles
NSCneural stem cells
PolyQpolyglutamine repeats-associated disease
SGZsubgranular zone

References

  1. McEwen, B.S. Plasticity of the hippocampus: Adaptation to chronic stress and allostatic load. Ann. N. Y. Acad. Sci. 2001, 933, 265–277. [Google Scholar] [CrossRef] [PubMed]
  2. Daugherty, A.; Bender, A.R. Age differences in hippocampal subfield volumes from childhood to late adulthood. Hippocampus 2016, 26, 220–228. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Delgorio, P.L.; Hiscox, L.V. Effect of Aging on the Viscoelastic Properties of Hippocampal Subfields Assessed with High-Resolution MR Elastography. Cereb. Cortex 2021, 31, 2799–2811. [Google Scholar] [CrossRef] [PubMed]
  4. Gensler, H.L.; Bernstein, H. DNA damage as the primary cause of aging. Q. Rev. Biol. 1981, 56, 279–303. [Google Scholar] [CrossRef] [PubMed]
  5. Gaubatz, J.W.; Tan, B.H. Aging affects the levels of DNA damage in postmitotic cells. Ann. N. Y. Acad. Sci. 1994, 719, 97–107. [Google Scholar] [CrossRef]
  6. Hamilton, M.L.; Van Remmen, H. Does oxidative damage to DNA increase with age? Proc. Natl. Acad. Sci. USA 2001, 98, 10469–10474. [Google Scholar] [CrossRef] [Green Version]
  7. Karanjawala, Z.E.; Lieber, M.R. DNA damage and aging. Mech. Ageing Dev. 2004, 125, 405–416. [Google Scholar]
  8. Rutten, B.P.; Schmitz, C. The aging brain: Accumulation of DNA damage or neuron loss? Neurobiol. Aging. 2007, 28, 91–98. [Google Scholar] [CrossRef]
  9. Feser, J.; Tyler, J. Chromatin structure as a mediator of aging. FEBS Lett. 2011, 585, 2041–2048. [Google Scholar] [CrossRef] [Green Version]
  10. Niedernhofer, L.J.; Gurkar, A.U.; Wang, Y.; Vijg, J.; Hoeijmakers, J.H.J.; Robbins, P.D. Nuclear Genomic Instability and Aging. Annu. Rev. Biochem. 2018, 87, 295–322. [Google Scholar] [CrossRef]
  11. Verheijen, B.M.; Vermulst, M.; van Leeuwen, F.W. Somatic mutations in neurons during aging and neurodegeneration. Acta Neuropathol. 2018, 135, 811–826. [Google Scholar] [CrossRef] [PubMed]
  12. Chow, H.M.; Herrup, K. Genomic integrity and the ageing brain. Nat. Rev. Neurosci. 2015, 16, 672–684. [Google Scholar] [CrossRef] [PubMed]
  13. Barrio-Alonso, E.; Hernández-Vivanco, A.; Walton, C.C.; Perea, G.; Frade, J.M. Cell cycle reentry triggers hyperploidization and synaptic dysfunction followed by delayed cell death in differentiated cortical neurons. Sci. Rep. 2018, 8, 14316. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Christen, Y. Oxidative stress and Alzheimer disease. Am. J. Clin. Nutr. 2000, 71, 621S–629S. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Herrup, K. Reimagining Alzheimer’s disease--an age-based hypothesis. J. Neurosci. 2010, 30, 16755–16762. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Ferrer, I. Defining Alzheimer as a common age-related neurodegenerative process not inevitably leading to dementia. Prog. Neurobiol. 2012, 97, 38–51. [Google Scholar] [CrossRef]
  17. Frade, J.M.; Ovejero-Benito, M.C. Neuronal cell cycle: The neuron itself and its circumstances. Cell Cycle 2015, 14, 712–720. [Google Scholar] [CrossRef] [Green Version]
  18. Ionescu-Tucker, A.; Cotman, C.W. Emerging roles of oxidative stress in brain aging and Alzheimer’s disease. Neurobiol. Aging 2021, 107, 86–95. [Google Scholar] [CrossRef]
  19. Mertens, J.; Herdy, J.R. Age-dependent instability of mature neuronal fate in induced neurons from Alzheimer’s patients. Cell Stem Cell 2021, 28, 1533–1548. [Google Scholar] [CrossRef]
  20. Liu, R.M. Aging, Cellular Senescence, and Alzheimer’s Disease. Int. J. Mol. Sci. 2022, 23, 1989. [Google Scholar] [CrossRef]
  21. Alzheimer, A.; Stelzmann, R.A.; Schnitzlein, H.N.; Murtagh, F.R. An English translation of Alzheimer’s 1907 paper, “Uber eine eigenartige Erkankung der Hirnrinde”. Clin. Anat. 1995, 8, 429–431. [Google Scholar] [PubMed]
  22. de Flores, R.; La Joie, R.; Chételat, G. Structural imaging of hippocampal subfields in healthy aging and Alzheimer’s disease. Neuroscience 2015, 309, 29–50. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Mueller, S.G.; Schuff, N. Hippocampal atrophy patterns in mild cognitive impairment and Alzheimer’s disease. Hum. Brain Mapp. 2010, 31, 1339–1347. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Schröder, J.; Pantel, J. Neuroimaging of hippocampal atrophy in early recognition of Alzheimer’s disease—A critical appraisal after two decades of research. Psychiatry Res. Neuroimaging 2016, 247, 71–78. [Google Scholar] [CrossRef]
  25. Madusanka, N.; Choi, H.K.; So, J.H.; Choi, B.K.; Park, H.G. One-year Follow-up Study of Hippocampal Subfield Atrophy in Alzheimer’s Disease and Normal Aging. Curr. Med. Imaging Rev. 2019, 15, 699–709. [Google Scholar] [CrossRef]
  26. Knowles, W.D. Normal anatomy and neurophysiology of the hippocampal formation. J. Clin. Neurophysiol. 1992, 9, 252–263. [Google Scholar] [CrossRef]
  27. Alkadhi, K.A. Cellular and Molecular Differences Between Area CA1 and the Dentate Gyrus of the Hippocampus. Mol. Neurobiol. 2019, 56, 6566–6580. [Google Scholar] [CrossRef]
  28. Gil, L.; Niño, S.A.; Chi-Ahumada, E.; Rodríguez-Leyva, I.; Guerrero, C.; Rebolledo, A.B.; Arias, J.A.; Jiménez-Capdeville, M.E. Perinuclear Lamin A and Nucleoplasmic Lamin B2 Characterize Two Types of Hippocampal Neurons through Alzheimer’s Disease Progression. Int. J. Mol. Sci. 2020, 21, 1841. [Google Scholar] [CrossRef] [Green Version]
  29. Gil, L.; Niño, S.A.; Capdeville, G.; Jiménez-Capdeville, M.E. Aging and Alzheimer’s disease connection: Nuclear Tau and lamin A. Neurosci. Lett. 2021, 749, 135741. [Google Scholar] [CrossRef]
  30. Méndez-López, I.; Blanco-Luquin, I.; Sánchez-Ruiz de Gordoa, J.; Urdánoz-Casado, A.; Roldán, M.; Acha, B.; Echavarri, C.; Zelaya, V.; Jericó, I.; Mendioroz, M. Hippocampal LMNA Gene Expression is Increased in Late-Stage Alzheimer’s Disease. Int. J. Mol. Sci. 2019, 20, 878. [Google Scholar] [CrossRef] [Green Version]
  31. Nagy, Z.; Esiri, M.M.; Cato, A.M.; Smith, A.D. Cell cycle markers in the hippocampus in Alzheimer’s disease. Acta Neuropathol. 1997, 94, 6–15. [Google Scholar] [CrossRef] [PubMed]
  32. Regalado-Reyes, M.; Furcila, D.; Hernández, F.; Ávila, J.; DeFelipe, J.; León-Espinosa, G. Phospho-Tau Changes in the Human CA1 During Alzheimer’s Disease Progression. J. Alzheimer’s Dis. 2019, 69, 277–288. [Google Scholar] [CrossRef] [Green Version]
  33. Antón-Fernández, A.; Vallés-Saiz, L.; Avila, J.; Hernández, F. Neuronal nuclear tau and neurodegeneration. Neuroscience 2022, 22. [Google Scholar] [CrossRef] [PubMed]
  34. Hernandez-Ortega, K.; Garcia-Esparcia, P.; Gil, L.; Lucas, J.J.; Ferrer, I. Altered machinery of protein synthesis in Alzheimer’s: From the nucleolus to the ribosome. Brain Pathol. 2016, 26, 593–605. [Google Scholar] [CrossRef] [PubMed]
  35. Frost, B.; Götz, J.; Feany, M.B. Connecting the dots between tau dysfunction and neurodegeneration. Trends Cell Biol. 2015, 25, 46–53. [Google Scholar] [CrossRef] [Green Version]
  36. Cornelison, G.L.; Levy, S.A.; Jenson, T.; Frost, B. Tau-induced nuclear envelope invagination causes a toxic accumulation of mRNA in Drosophila. Aging Cell 2019, 18, e12847. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Frost, B.; Bardai, F.H. Lamin Dysfunction Mediates Neurodegeneration in Tauopathies. Curr. Biol. 2016, 26, 129–136. [Google Scholar] [CrossRef] [Green Version]
  38. Frost, B. Alzheimer’s disease: An acquired neurodegenerative laminopathy. Nucleus 2016, 7, 275–283. [Google Scholar] [CrossRef] [Green Version]
  39. Klein, H.U.; McCabe, C. Epigenome-wide study uncovers large-scale changes in histone acetylation driven by tau pathology in aging and Alzheimer’s human brains. Nat. Neurosci. 2019, 22, 37–46. [Google Scholar] [CrossRef]
  40. Gil, L.; Niño, S.A. Phospho-Tau and Chromatin Landscapes in Early and Late Alzheimer’s Disease. Int. J. Mol. Sci. 2021, 22, 10283. [Google Scholar] [CrossRef]
  41. Nativio, R.; Donahue, G. Dysregulation of the epigenetic landscape of normal aging in Alzheimer’s disease. Nat. Neurosci. 2018, 21, 497–505. [Google Scholar] [CrossRef] [PubMed]
  42. Frost, B.; Hemberg, M.; Lewis, J.; Feany, M.B. Tau promotes neurodegeneration through global chromatin relaxation. Nat. Neurosci. 2014, 17, 357–366. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Mansuroglu, Z.; Benhelli-Mokrani, H. Loss of Tau protein affects the structure, transcription and repair of neuronal pericentromeric heterochromatin. Sci. Rep. 2016, 6, 33047. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Benhelli-Mokrani, H.; Mansuroglu, Z. Genome-wide identification of genic and intergenic neuronal DNA regions bound by Tau protein under physiological and stress conditions. Nucleic Acids Res. 2018, 46, 11405–11422. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Rico, T.; Gilles, M.; Chauderlier, A.; Comptdaer, T.; Magnez, R.; Chwastyniak, M.; Drobecq, H.; Pinet, F.; Thuru, X.; Buée, L.; et al. Tau Stabilizes Chromatin Compaction. Front. Cell Dev. Biol. 2021, 9, 740550. [Google Scholar] [CrossRef]
  46. Patzke, N.; Spocter, M.A.; Karlsson, K.E.; Bertelsen, M.F.; Haagensen, M.; Chawana, R.; Streicher, S.; Kaswera, C.; Gilissen, E.; Alagaili, A.N.; et al. In contrast to many other mammals, cetaceans have relatively small hippocampi that appear to lack adult neurogenesis. Brain Struct. Funct. 2015, 220, 361–383. [Google Scholar] [CrossRef]
  47. Eriksson, P.S.; Perfilieva, E.; Björk-Eriksson, T.; Alborn, A.M.; Nordborg, C.; Peterson, D.A.; Gage, F.H. Neurogenesis in the adult human hippocampus. Nat. Med. 1998, 4, 1313–1317. [Google Scholar] [CrossRef]
  48. Spalding, K.L.; Bergmann, O.; Alkass, K.; Bernard, S.; Salehpour, M.; Huttner, H.B.; Boström, E.; Westerlund, I.; Vial, C.; Buchholz, B.A.; et al. Dynamics of hippocampal neurogenesis in adult humans. Cell 2013, 153, 1219–1227. [Google Scholar] [CrossRef] [Green Version]
  49. Arellano, J.I.; Harding, B.; Thomas, J.L. Adult Human Hippocampus: No New Neurons in Sight. Cereb. Cortex 2018, 28, 2479–2481. [Google Scholar] [CrossRef]
  50. Kempermann, G.; Gage, F.H.; Aigner, L.; Song, H.; Curtis, M.A.; Thuret, S.; Kuhn, H.G.; Jessberger, S.; Frankland, P.W.; Cameron, H.A.; et al. Human Adult Neurogenesis: Evidence and Remaining Questions. Cell Stem Cell 2018, 23, 25–30. [Google Scholar] [CrossRef] [Green Version]
  51. Cipriani, S.; Ferrer, I.; Aronica, E.; Kovacs, G.G.; Verney, C.; Nardelli, J.; Khung, S.; Delezoide, A.L.; Milenkovic, I.; Rasika, S.; et al. Hippocampal Radial Glial Subtypes and Their Neurogenic Potential in Human Fetuses and Healthy and Alzheimer’s Disease Adults. Cereb. Cortex 2018, 28, 2458–2478. [Google Scholar] [CrossRef] [PubMed]
  52. Sorrells, S.F.; Paredes, M.F.; Cebrian-Silla, A.; Sandoval, K.; Qi, D.; Kelley, K.W.; James, D.; Mayer, S.; Chang, J.; Auguste, K.I.; et al. Human hippocampal neurogenesis drops sharply in children to undetectable levels in adults. Nature 2018, 555, 377–381. [Google Scholar] [CrossRef] [PubMed]
  53. Boldrini, M.; Fulmore, C.A.; Tartt, A.N.; Simeon, L.R.; Pavlova, I.; Poposka, V.; Rosoklija, G.B.; Stankov, A.; Arango, V.; Dwork, A.J.; et al. Human Hippocampal Neurogenesis Persists throughout Aging. Cell Stem Cell 2018, 22, 589–599.e5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Moreno-Jiménez, E.P.; Flor-García, M.; Terreros-Roncal, J.; Rábano, A.; Cafini, F.; Pallas-Bazarra, N.; Ávila, J.; Llorens-Martín, M. Adult hippocampal neurogenesis is abundant in neurologically healthy subjects and drops sharply in patients with Alzheimer’s disease. Nat. Med. 2019, 25, 554–560. [Google Scholar] [CrossRef]
  55. Tobin, M.K.; Musaraca, K.; Disouky, A.; Shetti, A.; Bheri, A.; Honer, W.G.; Kim, N.; Dawe, R.J.; Bennett, D.A.; Arfanakis, K.; et al. Human hippocampal neurogenesis persists in aged adults and alzheimer’s disease patients. Cell Stem Cell 2019, 24, 974–982.e3. [Google Scholar] [CrossRef]
  56. Opendak, M.; Gould, E. Adult neurogenesis: A substrate for experience-dependent change. Trends Cogn. Sci. 2015, 19, 151–161. [Google Scholar] [CrossRef]
  57. Anacker, C.; Hen, R. Adult hippocampal neurogenesis and cognitive flexibility—linking memory and mood. Nat. Rev. Neurosci. 2017, 18, 335–346. [Google Scholar] [CrossRef]
  58. Tronel, S.; Lemaire, V. Influence of ontogenetic age on the role of dentate granule neurons. Brain Struct. Funct. 2015, 220, 645–661. [Google Scholar] [CrossRef]
  59. Laplagne, D.A.; Espósito, M.S.; Piatti, V.C.; Morgenstern, N.A.; Zhao, C.; van Praag, H.; Gage, F.H.; Schinder, A.F. Functional convergence of neurons generated in the developing and adult hippocampus. PLoS Biol. 2006, 4, e409. [Google Scholar] [CrossRef] [Green Version]
  60. Nakashiba, T.; Cushman, J.D.; Pelkey, K.A.; Renaudineau, S.; Buhl, D.L.; McHugh, T.J.; Rodriguez Barrera, V.; Chittajallu, R.; Iwamoto, K.S.; McBain, C.J.; et al. Young dentate granule cells mediate pattern separation, whereas old granule cells facilitate pattern completion. Cell 2012, 149, 188–201. [Google Scholar] [CrossRef] [Green Version]
  61. Kerloch, T.; Clavreul, S.; Goron, A.; Abrous, D.N.; Pacary, E. Dentate Granule Neurons Generated During Perinatal Life Display Distinct Morphological Features Compared with Later-Born Neurons in the Mouse Hippocampus. Cereb. Cortex 2019, 29, 3527–3539. [Google Scholar] [CrossRef] [PubMed]
  62. Cole, J.D.; Espinueva, D.F.; Seib, D.R.; Ash, A.M.; Cooke, M.B.; Cahil, S.P.; O’Leary, T.P.; Kwan, S.S.; Snyder, J.S. Adult-Born Hippocampal Neurons Undergo Extended Development and Are Morphologically Distinct from Neonatally-Born Neurons. J. Neurosci. 2020, 40, 5740–5756. [Google Scholar] [CrossRef] [PubMed]
  63. Masachs, N.; Charrier, V.; Farrugia, F.; Lemaire, V.; Blin, N.; Mazier, W.; Tronel, S.; Montaron, M.F.; Ge, S.; Marsicano, G.; et al. The temporal origin of dentate granule neurons dictates their role in spatial memory. Mol. Psychiatry 2021, 26, 7130–7140. [Google Scholar] [CrossRef] [PubMed]
  64. Altman, J.; Bayer, S.A. Migration and distribution of two populations of hippocampal granule cell precursors during the perinatal and postnatal periods. J. Comp. Neurol. 1990, 301, 365–381. [Google Scholar] [CrossRef] [PubMed]
  65. Bayer, S.A.; Altman, J. Timetables of neurogenesis in the human brain based on experimentally determined patterns in the rat. Neurotoxicology 1993, 14, 83–144. [Google Scholar] [PubMed]
  66. Lopez-Rojas, J.; Kreutz, M.R. Mature granule cells of the dentate gyrus—Passive bystanders or principal performers in hippocampal function? Neurosci. Biobehav. Rev. 2016, 64, 167–174. [Google Scholar] [CrossRef] [Green Version]
  67. Kuhn, H.G.; Dickinson-Anson, H.; Gage, F.H. Neurogenesis in the dentate gyrus of the adult rat: Age-related decrease of neuronal progenitor proliferation. J. Neurosci. 1996, 16, 2027–2033. [Google Scholar] [CrossRef] [Green Version]
  68. Knoth, R.; Singec, I.; Ditter, M.; Pantazis, G.; Capetian, P.; Meyer, R.P.; Horvat, V.; Volk, B.; Kempermann, G. Murine features of neurogenesis in the human hippocampus across the lifespan from 0 to 100 years. PLoS ONE 2010, 5, e8809. [Google Scholar] [CrossRef] [Green Version]
  69. Ben Abdallah, N.M.; Slomianka, L.; Vyssotski, A.L.; Lipp, H.P. Early age-related changes in adult hippocampal neurogenesis in C57 mice. Neurobiol. Aging 2010, 31, 151–161. [Google Scholar] [CrossRef]
  70. Lazic, S.E. Modeling hippocampal neurogenesis across the lifespan in seven species. Neurobiol. Aging 2012, 33, 1664–1671. [Google Scholar] [CrossRef] [Green Version]
  71. Tanapat, P.; Hastings, N.B.; Reeves, A.J.; Gould, E. Estrogen stimulates a transient increase in the number of new neurons in the dentate gyrus of the adult female rat. J. Neurosci. 1999, 19, 5792–5801. [Google Scholar] [CrossRef]
  72. Brown, J.; Cooper-Kuhn, C.M.; Kempermann, G.; Van Praag, H.; Winkler, J.; Gage, F.H.; Kuhn, H.G. Enriched environment and physical activity stimulate hippocampal but not olfactory bulb neurogenesis. Eur. J. Neurosci. 2003, 17, 2042–2046. [Google Scholar] [CrossRef] [PubMed]
  73. Gonçalves, J.T. In vivo imaging of dendritic pruning in dentate granule cells. Nat. Neurosci. 2016, 19, 788–791. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Toda, T.; Parylak, S.L.; Linker, S.B.; Gage, F.H. The role of adult hippocampal neurogenesis in brain health and disease. Mol. Psychiatry 2019, 24, 67–87. [Google Scholar] [CrossRef] [PubMed]
  75. Seki, T. Expression patterns of immature neuronal markers PSA-NCAM, CRMP-4 and NeuroD in the hippocampus of young adult and aged rodents. J. Neurosci. Res. 2002, 70, 327–334. [Google Scholar] [CrossRef]
  76. Kohler, S.J.; Williams, N.I.; Stanton, G.B.; Cameron, J.L.; Greenough, W.T. Maturation time of new granule cells in the dentate gyrus of adult macaque monkeys exceeds six months. Proc. Natl. Acad. Sci. USA 2011, 108, 10326–10331. [Google Scholar] [CrossRef] [Green Version]
  77. Ngwenya, L.B.; Heyworth, N.C.; Shwe, Y.; Moore, T.L.; Rosene, D.L. Age-related changes in dentate gyrus cell numbers, neurogenesis, and associations with cognitive impairments in the rhesus monkey. Front. Syst. Neurosci. 2015, 9, 102. [Google Scholar] [CrossRef] [Green Version]
  78. Geinisman, Y.; de Toledo-Morrell, L.; Morrell, F. Loss of perforated synapses in the dentate gyrus: Morphological substrate of memory deficit in aged rats. Proc. Natl. Acad. Sci. USA 1986, 83, 3027–3031. [Google Scholar] [CrossRef] [Green Version]
  79. Yassa, M.A.; Stark, S.M.; Bakker, A.; Albert, M.S.; Gallagher, M.; Stark, C.E. High-resolution structural and functional MRI of hippocampal CA3 and dentate gyrus in patients with amnestic Mild Cognitive Impairment. Neuroimage 2010, 51, 1242–1252. [Google Scholar] [CrossRef] [Green Version]
  80. Seki, T. Understanding the Real State of Human Adult Hippocampal Neurogenesis from Studies of Rodents and Non-human Primates. Front. Neurosci. 2020, 14, 839. [Google Scholar] [CrossRef]
  81. Zhao, C.; Teng, E.M. Distinct morphological stages of dentate granule neuron maturation in the adult mouse hippocampus. J. Neurosci. 2006, 26, 3–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Denoth-Lippuner, A.; Jessberger, S. Formation and integration of new neurons in the adult hippocampus. Nat. Rev. Neurosci. 2021, 22, 223–236. [Google Scholar] [CrossRef] [PubMed]
  83. Herrup, K. Post-mitotic role of the cell cycle machinery. Curr. Opin. Cell Biol. 2013, 25, 711–716. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Suberbielle, E.; Sanchez, P.E.; Kravitz, A.V.; Wang, X.; Ho, K.; Eilertson, K.; Devidze, N.; Kreitzer, A.C.; Mucke, L. Physiologic brain activity causes DNA double-strand breaks in neurons, with exacerbation by amyloid-β. Nat. Neurosci. 2013, 16, 613–621. [Google Scholar] [CrossRef] [Green Version]
  85. Zada, D.; Bronshtein, I.; Lerer-Goldshtein, T.; Garini, Y.; Appelbaum, L. Sleep increases chromosome dynamics to enable reduction of accumulating DNA damage in single neurons. Nat. Commun. 2019, 10, 895. [Google Scholar] [CrossRef] [Green Version]
  86. Shanbhag, N.M.; Evans, M.D.; Mao, W.; Nana, A.L.; Seeley, W.W.; Adame, A.; Rissman, R.A.; Masliah, E.; Mucke, L. Early neuronal accumulation of DNA double strand breaks in Alzheimer’s disease. Acta Neuropathol. Commun. 2019, 7, 77. [Google Scholar] [CrossRef] [Green Version]
  87. Thadathil, N.; Delotterie, D.F.; Xiao, J.; Hori, R.; McDonald, M.P.; Khan, M.M. DNA Double-Strand Break Accumulation in Alzheimer’s Disease: Evidence from Experimental Models and Postmortem Human Brains. Mol. Neurobiol. 2021, 58, 118–131. [Google Scholar] [CrossRef]
  88. Jackson, S.P.; Bartek, J. The DNA-damage response in human biology and disease. Nature 2009, 461, 1071–1078. [Google Scholar] [CrossRef] [Green Version]
  89. Eckermann, M.; Schmitzer, B.; van der Meer, F.; Franz, J.; Hansen, O.; Stadelmann, C.; Salditt, T. Three-dimensional virtual histology of the human hippocampus based on phase-contrast computed tomography. Proc. Natl. Acad. Sci. USA 2021, 118, e2113835118. [Google Scholar] [CrossRef]
  90. Dou, Z.; Xu, C.; Donahue, G.; Shimi, T.; Pan, J.A.; Zhu, J.; Ivanov, A.; Capell, B.C.; Drake, A.M.; Shah, P.P.; et al. Autophagy mediates degradation of nuclear lamina. Nature 2015, 527, 105–109. [Google Scholar] [CrossRef] [Green Version]
  91. Nixon, R.A.; Wegiel, J.; Kumar, A.; Yu, W.H.; Peterhoff, C.; Cataldo, A.; Cuervo, A.M. Extensive involvement of autophagy in Alzheimer disease: An immuno-electron microscopy study. J. Neuropathol. Exp. Neurol. 2005, 64, 113–122. [Google Scholar] [CrossRef] [PubMed]
  92. Wolfe, D.M.; Lee, J.; Kumar, A.; Lee, S.; Orenstein, S.J.; Nixon, R.A. Autophagy failure in Alzheimer’s disease and the role of defective lysosomal acidification. Eur. J. Neurosci. 2013, 37, 1949–1961. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Bordi, M.; Berg, M.J.; Mohan, P.S.; Peterhoff, C.M.; Alldred, M.J.; Che, S.; Ginsberg, S.D.; Nixon, R.A. Autophagy flux in CA1 neurons of Alzheimer hippocampus: Increased induction overburdens failing lysosomes to propel neuritic dystrophy. Autophagy 2016, 12, 2467–2483. [Google Scholar] [CrossRef] [PubMed]
  94. Chung, K.M.; Hernández, N.; Sproul, A.A.; Yu, W.H. Alzheimer’s disease and the autophagic-lysosomal system. Neurosci. Lett. 2019, 697, 49–58. [Google Scholar] [CrossRef] [PubMed]
  95. Feng, X.; Guo, J.; Sigmon, H.C.; Sloan, R.P.; Brickman, A.M.; Provenzano, F.A.; Small, S.A. Brain regions vulnerable and resistant to aging without Alzheimer’s disease. PLoS ONE 2020, 15, e0234255. [Google Scholar] [CrossRef] [PubMed]
  96. Gil, L.; Federico, C.; Pinedo, F.; Bruno, F.; Rebolledo, A.B.; Montoya, J.J.; Olazabal, I.M.; Ferrer, I.; Saccone, S. Aging dependent effect of nuclear tau. Brain Res. 2017, 1677, 129–137. [Google Scholar] [CrossRef] [Green Version]
  97. Bukar Maina, M.; Al-Hilaly, Y.K.; Serpell, L.C. Nuclear Tau and Its Potential Role in Alzheimer’s Disease. Biomolecules 2016, 6, 9. [Google Scholar] [CrossRef] [Green Version]
  98. Galas, M.C.; Bonnefoy, E.; Buee, L.; Lefebvre, B. Emerging Connections Between Tau and Nucleic Acids. Adv. Exp. Med. Biol. 2019, 1184, 135–143. [Google Scholar]
  99. Kruman, I.I.; Wersto, R.P.; Cardozo-Pelaez, F.; Smilenov, L.; Chan, S.L.; Chrest, F.J.; Emokpae, R., Jr.; Gorospe, M.; Mattson, M.P. Cell cycle activation linked to neuronal cell death initiated by DNA damage. Neuron 2004, 41, 549–561. [Google Scholar] [CrossRef] [Green Version]
  100. Yang, Y.; Herrup, K. Cell division in the CNS: Protective response or lethal event in post-Mitotic neurons? Biochim. Biophys. Acta Mol. Basis Dis. 2007, 1772, 457–466. [Google Scholar] [CrossRef] [Green Version]
  101. Raina, A.K.; Zhu, X.; Rottkamp, C.A.; Monteiro, M.; Takeda, A.; Smith, M.A. Cyclin’ toward dementia: Cell cycle abnormalities and abortive oncogenesis in Alzheimer disease. J. Neurosci. Res. 2000, 61, 128–133. [Google Scholar] [CrossRef]
  102. Yang, Y.; Geldmacher, D.S.; Herrup, K. DNA replication precedes neuronal cell death in Alzheimer’s disease. J. Neurosci. 2001, 21, 2661–2668. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Yang, Y.; Mufson, E.J.; Herrup, K. Neuronal cell death is preceded by cell cycle events at all stages of Alzheimer’s disease. J. Neurosci. 2003, 23, 2557–2563. [Google Scholar] [CrossRef] [Green Version]
  104. Currais, A.; Hortobágyi, T.; Soriano, S. The neuronal cell cycle as a mechanism of pathogenesis in Alzheimer’s disease. Aging 2009, 1, 363–371. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Jung, H.J.; Coffinier, C.; Choe, Y.; Beigneux, A.P.; Davies, B.S.; Yang, S.H.; Barnes, R.H., 2nd; Hong, J.; Sun, T.; Pleasure, S.J.; et al. Regulation of prelamin A but not lamin C by miR-9, a brain-specific microRNA. Proc. Natl. Acad. Sci. USA 2012, 109, E423–E431. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Schirmer, E.C.; Gerace, L. The stability of the nuclear lamina polymer changes with the composition of lamin subtypes according to their individual binding strengths. J. Biol. Chem. 2004, 279, 42811–44281. [Google Scholar] [CrossRef] [Green Version]
  107. Simon, D.N.; Wilson, K.L. The nucleoskeleton as a genome-associated dynamic ‘network of networks’. Nat. Rev. Mol. Cell Biol. 2011, 12, 695–708. [Google Scholar] [CrossRef]
  108. Cho, S.; Vashisth, M.; Abbas, A.; Majkut, S.; Vogel, K.; Xia, Y.; Ivanovska, I.L.; Irianto, J.; Tewari, M.; Zhu, K.; et al. Mechanosensing by the Lamina Protects against Nuclear Rupture, DNA Damage, and Cell-Cycle Arrest. Dev. Cell. 2019, 49, 920–935.e5. [Google Scholar] [CrossRef]
  109. Muramatsu, R.; Ikegaya, Y.; Matsuki, N.; Koyama, R. Neonatally born granule cells numerically dominate adult mice dentate gyrus. Neuroscience 2007, 148, 593–598. [Google Scholar] [CrossRef]
  110. von Bohlen und Halbach, O. Immunohistological markers for proliferative events, gliogenesis, and neurogenesis within the adult hippocampus. Cell Tissue Res. 2011, 345, 1–19. [Google Scholar] [CrossRef]
  111. Altman, J.; Das, G.D. Autoradiographic and histological evidence of postnatal hippocampal neurogenesis in rats. J. Comp. Neurol. 1965, 124, 319–335. [Google Scholar] [CrossRef] [PubMed]
  112. Olson, A.K.; Eadie, B.D.; Ernst, C.; Christie, B.R. Environmental enrichment and voluntary exercise massively increase neurogenesis in the adult hippocampus via dissociable pathways. Hippocampus 2006, 16, 250–260. [Google Scholar] [CrossRef] [PubMed]
  113. Redila, V.A.; Olson, A.K.; Swann, S.E.; Mohades, G.; Webber, A.J.; Weinberg, J.; Christie, B.R. Hippocampal cell proliferation is reduced following prenatal ethanol exposure but can be rescued with voluntary exercise. Hippocampus 2006, 16, 305–311. [Google Scholar] [CrossRef]
  114. Monje, M.L.; Toda, H.; Palmer, T.D. Inflammatory blockade restores adult hippocampal neurogenesis. Science 2003, 302, 1760–1765. [Google Scholar] [CrossRef] [PubMed]
  115. Mizumatsu, S.; Monje, M.L.; Morhardt, D.R.; Rola, R.; Palmer, T.D.; Fike, J.R. Extreme sensitivity of adult neurogenesis to low doses of X-irradiation. Cancer Res. 2003, 63, 4021–4027. [Google Scholar]
  116. Belarbi, K.; Arellano, C.; Ferguson, R.; Jopson, T.; Rosi, S. Chronic neuroinflammation impacts the recruitment of adult-born neurons into behaviorally relevant hippocampal networks. Brain Behav. Immun. 2012, 26, 18–23. [Google Scholar] [CrossRef] [Green Version]
  117. Yuan, T.F.; Li, J.; Arias-Carrion, O. Evidence of adult neurogenesis in non-human primates and human. Cell Tissue Res. 2014, 358, 17–23. [Google Scholar] [CrossRef]
  118. Seki, T.; Arai, Y. Age-related production of new granule cells in the adult dentate gyrus. Neuroreport 1995, 6, 2479–2482. [Google Scholar] [CrossRef]
  119. West, M.J. Regionally specific loss of neurons in the aging human hippocampus. Neurobiol. Aging 1993, 14, 287–293. [Google Scholar] [CrossRef]
  120. Mani, R.B.; Lohr, J.B.; Jeste, D.V. Hippocampal pyramidal cells and aging in the human: A quantitative study of neuronal loss in sectors CA1 to CA4. Exp. Neurol. 1986, 94, 29–40. [Google Scholar] [CrossRef]
  121. Fukuda, S.; Kato, F.; Tozuka, Y.; Yamaguchi, M.; Miyamoto, Y.; Hisatsune, T. Two distinct subpopulations of nestin-positive cells in adult mouse dentate gyrus. J. Neurosci. 2003, 23, 9357–9366. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Maslov, A.Y.; Barone, T.A.; Plunkett, R.J.; Pruitt, S.C. Neural stem cell detection, characterization, and age-related changes in the subventricular zone of mice. J. Neurosci. 2004, 24, 1726–1733. [Google Scholar] [CrossRef] [Green Version]
  123. Kronenberg, G.; Reuter, K.; Steiner, B.; Brandt, M.D.; Jessberger, S.; Yamaguchi, M.; Kempermann, G. Subpopulations of proliferating cells of the adult hippocampus respond differently to physiologic neurogenic stimuli. J. Comp. Neurol. 2003, 467, 455–463. [Google Scholar] [CrossRef] [PubMed]
  124. Seri, B.; García-Verdugo, J.M.; Collado-Morente, L.; McEwen, B.S.; Alvarez-Buylla, A. Cell types, lineage, and architecture of the germinal zone in the adult dentate gyrus. J. Comp. Neurol. 2004, 478, 359–378, Erratum in J. Comp. Neurol. 2004, 20, 427. [Google Scholar] [CrossRef]
  125. Brandt, M.D.; Jessberger, S.; Steiner, B.; Kronenberg, G.; Reuter, K.; Bick-Sander, A.; von der Behrens, W.; Kempermann, G. Transient calretinin expression defines early postmitotic step of neuronal differentiation in adult hippocampal neurogenesis of mice. Mol. Cell Neurosci. 2003, 24, 603–613. [Google Scholar] [CrossRef]
  126. Llorens-Martín, M.; Torres-Alemán, I.; Trejo, J.L. Pronounced individual variation in the response to the stimulatory action of exercise on immature hippocampal neurons. Hippocampus 2006, 16, 480–490. [Google Scholar] [CrossRef]
  127. Kempermann, G.; Jessberger, S.; Steiner, B.; Kronenberg, G. Milestones of neuronal development in the adult hippocampus. Trends Neurosci. 2004, 27, 447–452. [Google Scholar] [CrossRef]
  128. Takamori, Y.; Tamura, Y.; Kataoka, Y.; Cui, Y.; Seo, S.; Kanazawa, T.; Kurokawa, K.; Yamada, H. Differential expression of nuclear lamin, the major component of nuclear lamina, during neurogenesis in two germinal regions of adult rat brain. Eur. J. Neurosci. 2007, 25, 1653–1662. [Google Scholar] [CrossRef]
  129. Kill, I.R.; Hutchison, C.J. S-phase phosphorylation of lamin B2. FEBS Lett. 1995, 377, 26–30. [Google Scholar] [CrossRef] [Green Version]
  130. Sen Gupta, A.; Sengupta, K. Lamin B2 Modulates Nucleolar Morphology, Dynamics, and Function. Mol. Cell Biol. 2017, 37, e00274-17. [Google Scholar] [CrossRef] [Green Version]
  131. Ehninger, D.; Kempermann, G. Neurogenesis in the adult hippocampus. Cell Tissue Res. 2008, 331, 243–250. [Google Scholar] [CrossRef] [PubMed]
  132. Lombard, D.B.; Chua, K.F.; Mostoslavsky, R.; Franco, S.; Gostissa, M.; Alt, F.W. DNA repair, genome stability, and aging. Cell 2005, 120, 497–512. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Lodato, M.A.; Rodin, R.E.; Bohrson, C.L.; Coulter, M.E.; Barton, A.R.; Kwon, M.; Sherman, M.A.; Vitzthum, C.M.; Luquette, L.J.; Yandava, C.N.; et al. Aging and neurodegeneration are associated with increased mutations in single human neurons. Science 2018, 359, 555–559. [Google Scholar] [CrossRef]
  134. Baquero, J.; Varriano, S.; Ordonez, M.; Kuczaj, P.; Murphy, M.R.; Aruggoda, G.; Lundine, D.; Morozova, V.; Makki, A.E.; Alonso, A.D.C.; et al. Nuclear Tau, p53 and Pin1 Regulate PARN-Mediated Deadenylation and Gene Expression. Front. Mol. Neurosci. 2019, 12, 242. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Farmer, K.M.; Ghag, G.; Puangmalai, N.; Montalbano, M.; Bhatt, N.; Kayed, R. P53 aggregation, interactions with tau, and impaired DNA damage response in Alzheimer’s disease. Acta Neuropathol. Commun. 2020, 8, 132. [Google Scholar] [CrossRef] [PubMed]
  136. Asada-Utsugi, M.; Uemura, K.; Ayaki, T.; TUemura, M.; Minamiyama, S.; Hikiami, R.; Morimura, T.; Shodai, A.; Ueki, T.; Takahashi, R.; et al. Failure of DNA double-strand break repair by tau mediates Alzheimer’s disease pathology in vitro. Commun. Biol. 2022, 5, 358. [Google Scholar] [CrossRef]
  137. Brasnjevic, I.; Hof, P.R.; Steinbusch, H.W.; Schmitz, C. Accumulation of nuclear DNA damage or neuron loss: Molecular basis for a new approach to understanding selective neuronal vulnerability in neurodegenerative diseases. DNA Repair 2008, 7, 1087–1097. [Google Scholar] [CrossRef] [Green Version]
  138. Montavon, T.; Shukeir, N.; Erikson, G.; Engist, B.; Onishi-Seebacher, M.; Ryan, D.; Musa, Y.; Mittler, G.; Meyer, A.G.; Genoud, C.; et al. Complete loss of H3K9 methylation dissolves mouse heterochromatin organization. Nat. Commun. 2021, 12, 4359. [Google Scholar] [CrossRef]
  139. Ocampo, A.; Reddy, P.; Martinez-Redondo, P.; Platero-Luengo, A.; Hatanaka, F.; Hishida, T.; Li, M.; Lam, D.; Kurita, M.; Beyret, E.; et al. In Vivo Amelioration of Age-Associated Hallmarks by Partial Reprogramming. Cell 2016, 167, 1719–1733.e12. [Google Scholar] [CrossRef] [Green Version]
  140. Sarkar, T.J.; Quarta, M.; Mukherjee, S.; Colville, A.; Paine, P.; Doan, L.; Tran, C.M.; Chu, C.R.; Horvath, S.; Qi, L.S.; et al. Transient non-integrative expression of nuclear reprogramming factors promotes multifaceted amelioration of aging in human cells. Nat. Commun. 2020, 11, 1545. [Google Scholar] [CrossRef] [Green Version]
  141. Padeken, J.; Zeller, P.; Towbin, B.; Katic, I.; Kalck, V.; Methot, S.P.; Gasser, S.M. Synergistic lethality between BRCA1 and H3K9me2 loss reflects satellite derepression. Genes Dev. 2019, 33, 436–451. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Janssen, A.; Colmenares, S.U.; Karpen, G.H. Heterochromatin: Guardian of the Genome. Annu. Rev. Cell Dev. Biol. 2018, 34, 265–288. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. West, M.J.; Gundersen, H.J. Unbiased stereological estimation of the number of neurons in the human hippocampus. J. Comp. Neurol. 1990, 296, 1–22. [Google Scholar] [CrossRef] [PubMed]
  144. Rapp, P.R.; Gallagher, M. Preserved neuron number in the hippocampus of aged rats with spatial learning deficits. Proc. Natl. Acad. Sci. USA 1996, 93, 9926–9930. [Google Scholar] [CrossRef] [Green Version]
  145. Gazzaley, A.H.; Benson, D.L.; Huntley, G.W.; Morrison, J.H. Differential subcellular regulation of NMDAR1 protein and mRNA in dendrites of dentate gyrus granule cells after perforant path transection. J. Neurosci. 1997, 17, 2006–2017. [Google Scholar] [CrossRef] [Green Version]
  146. Merrill, D.A.; Chiba, A.A.; Tuszynski, M.H. Conservation of neuronal number and size in the entorhinal cortex of behaviorally characterized aged rats. J. Comp. Neurol. 2001, 438, 445–456. [Google Scholar] [CrossRef]
  147. Scheibel, M.E.; Lindsay, R.D.; Tomiyasu, U.; Scheibel, A.B. Progressive dendritic changes in aging human cortex. Exp. Neurol. 1975, 47, 392–403. [Google Scholar] [CrossRef]
  148. Barnes, C.A. Normal aging: Regionally specific changes in hippocampal synaptic transmission. Trends Neurosci. 1994, 17, 13–18. [Google Scholar] [CrossRef]
  149. Wu, R.; Terry, A.V.; Singh, P.B.; Gilbert, D.M. Differential subnuclear localization and replication timing of histone H3 lysine 9 methylation states. Mol. Biol. Cell 2005, 16, 2872–2881. [Google Scholar] [CrossRef] [Green Version]
  150. Lee, M.Y.; Lee, J.; Hyeon, S.J.; Cho, H.; Hwang, Y.J.; Shin, J.Y.; McKee, A.C.; Kowall, N.W.; Kim, J.I.; Stein, T.D.; et al. Epigenome signatures landscaped by histone H3K9me3 are associated with the synaptic dysfunction in Alzheimer’s disease. Aging Cell 2020, 19, e13153. [Google Scholar] [CrossRef]
  151. Ryu, H.; Lee, J.; Hagerty, S.W.; Soh, B.Y.; McAlpin, S.E.; Cormier, K.A.; Smith, K.M.; Ferrante, R.J. ESET/SETDB1 gene expression and histone H3 (K9) trimethylation in Huntington’s disease. Proc. Natl. Acad. Sci. USA 2006, 103, 19176–19181. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Bulut-Karslioglu, A.; De La Rosa-Velázquez, I.A.; Ramirez, F.; Barenboim, M.; Onishi-Seebacher, M.; Arand, J.; Galán, C.; Winter, G.E.; Engist, B.; Gerle, B.; et al. Suv39h-dependent H3K9me3 marks intact retrotransposons and silences LINE elements in mouse embryonic stem cells. Mol. Cell. 2014, 55, 277–290. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Rodríguez-Matellán, A.; Alcazar, N.; Hernández, F.; Serrano, M.; Ávila, J. In vivo reprogramming ameliorates aging features in dentate gyrus cells and improves memory in mice. Stem Cell Rep. 2020, 15, 1056–1066. [Google Scholar] [CrossRef] [PubMed]
  154. Blümcke, I.; Schewe, J.C.; Normann, S.; Brüstle, O.; Schramm, J.; Elger, C.E.; Wiestler, O.D. Increase of nestin-immunoreactive neural precursor cells in the dentate gyrus of pediatric patients with early-onset temporal lobe epilepsy. Hippocampus 2001, 11, 311–321. [Google Scholar] [CrossRef]
  155. Dempsey, R.J.; Kalluri, H.S. Ischemia-induced neurogenesis: Role of growth factors. Neurosurg. Clin. 2007, 18, 183–190. [Google Scholar] [CrossRef]
  156. Kokaia, Z.; Lindvall, O. Neurogenesis after ischaemic brain insults. Curr. Opin. Neurobiol. 2003, 13, 127–132. [Google Scholar] [CrossRef]
  157. Cho, K.O.; Lybrand, Z.R.; Ito, N.; Brulet, R.; Tafacory, F.; Zhang, L.; Good, L.; Ure, K.; Kernie, S.G.; Birnbaum, S.G.; et al. Aberrant hippocampal neurogenesis contributes to epilepsy and associated cognitive decline. Nat. Commun. 2015, 6, 6606. [Google Scholar] [CrossRef] [Green Version]
  158. Ribak, C.E.; Dashtipour, K. Neuroplasticity in the smashed dentate gyrus of the epileptic brain. Prog. Brain Res. 2002, 136, 319–328. [Google Scholar] [PubMed]
  159. Kunze, A.; Grass, S.; Witte, O.W.; Yamaguchi, M.; Kempermann, G.; Redecker, C. Proliferative response of distinct hippocampal progenitor cell populations after cortical infarcts in the adult brain. Neurobiol. Dis. 2006, 21, 324–332. [Google Scholar] [CrossRef]
  160. Jin, K.; Peel, A.L.; Mao, X.O.; Xie, L.; Cottrell, B.A.; Henshall, D.C.; Greenberg, D.A. Increased hippocampal neurogenesis in Alzheimer’s disease. Proc. Natl. Acad. Sci. USA 2004, 101, 343–347. [Google Scholar] [CrossRef] [Green Version]
  161. Perry, E.K.; Johnson, M.; Ekonomou, A.; Perry, R.H.; Ballard, C.; Attems, J. Neurogenic abnormalities in Alzheimer’s disease differ between stages of neurogenesis and are partly related to cholinergic pathology. Neurobiol. Dis. 2012, 47, 155–162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Yu, Y.; He, J.; Zhang, Y.; Luo, H.; Zhu, S.; Yang, Y.; Zhao, T.; Wu, J.; Huang, Y.; Kong, J.; et al. Increased hippocampal neurogenesis in the progressive stage of Alzheimer’s disease phenotype in an APP/PS1 double transgenic mouse model. Hippocampus 2009, 19, 1247–1253. [Google Scholar] [CrossRef] [PubMed]
  163. Gomez-Nicola, D.; Suzzi, S.; Vargas-Caballero, M.; Fransen, N.L.; Al-Malki, H.; Cebrian-Silla, A.; Garcia-Verdugo, J.M.; Riecken, K.; Fehse, B.; Perry, V.H. Temporal dynamics of hippocampal neurogenesis in chronic neurodegeneration. Brain 2014, 137, 2312–2328. [Google Scholar] [CrossRef] [PubMed]
  164. Benayoun, B.A.; Pollina, E.A.; Ucar, D.; Mahmoudi, S.; Karra, K.; Wong, E.D.; Devarajan, K.; Daugherty, A.C.; Kundaje, A.B.; Mancini, E.; et al. H3K4me3 breadth is linked to cell identity and transcriptional consistency. Cell 2014, 158, 673–688. [Google Scholar] [CrossRef] [Green Version]
  165. Teixeira, C.M.; Pallas-Bazarra, N.; Bolós, M.; Terreros-Roncal, J.; Ávila, J.; Llorens-Martín, M. Untold New Beginnings: Adult Hippocampal Neurogenesis and Alzheimer’s Disease. J. Alzheimer’s Dis. 2018, 64, S497–S505. [Google Scholar] [CrossRef] [PubMed]
  166. Thurston, V.C.; Pena, P.; Pestell, R.; Binder, L.I. Nucleolar localization of the microtubule-associated protein tau in neuroblastomas using sense and anti-sense transfection strategies. Cell Motil. Cytoskelet. 1997, 38, 100–110. [Google Scholar] [CrossRef]
  167. Walton, N.M.; Shin, R.; Tajinda, K.; Heusner, C.L.; Kogan, J.H.; Miyake, S.; Chen, Q.; Tamura, K.; Matsumoto, M. Adult neurogenesis transiently generates oxidative stress. PLoS ONE 2012, 7, e35264. [Google Scholar] [CrossRef] [Green Version]
  168. Hester, M.S.; Danzer, S.C. Accumulation of abnormal adult-generated hippocampal granule cells predicts seizure frequency and severity. J. Neurosci. 2013, 33, 8926–8936. [Google Scholar] [CrossRef]
  169. Parent, J.M.; Yu, T.W.; Leibowitz, R.T.; Geschwind, D.H.; Sloviter, R.S.; Lowenstein, D.H. Dentate granule cell neurogenesis is increased by seizures and contributes to aberrant network reorganization in the adult rat hippocampus. J. Neurosci. 1997, 17, 3727–3738. [Google Scholar] [CrossRef] [Green Version]
  170. Shetty, A.K.; Hattiangady, B.; Rao, M.S.; Shuai, B. Neurogenesis response of middle-aged hippocampus to acute seizure activity. PLoS ONE 2012, 7, e43286. [Google Scholar] [CrossRef]
  171. Scharfman, H.E.; Gray, W.P. Relevance of seizure-induced neurogenesis in animal models of epilepsy to the etiology of temporal lobe epilepsy. Epilepsia 2007, 48, 33–41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Park, Y.E.; Hayashi, Y.K.; Bonne, G.; Arimura, T.; Noguchi, S.; Nonaka, I.; Nishino, I. Autophagic degradation of nuclear components in mammalian cells. Autophagy 2009, 5, 795–804. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Galluzzi, L.; Bravo-San Pedro, J.M.; Kroemer, G. Autophagy Mediates Tumor Suppression via Cellular Senescence. Trends Cell Biol. 2016, 26, 1–3. [Google Scholar] [CrossRef] [PubMed]
  174. Dou, Z.; Ivanov, A.; Adams, P.D.; Berger, S.L. Mammalian autophagy degrades nuclear constituents in response to tumorigenic stress. Autophagy 2016, 12, 1416–1417. [Google Scholar] [CrossRef] [Green Version]
  175. Frake, R.A.; Ricketts, T.; Menzies, F.M.; Rubinsztein, D.C. Autophagy and neurodegeneration. J. Clin. Investig. 2015, 125, 65–74. [Google Scholar] [CrossRef] [Green Version]
  176. Reddy, P.H.; Oliver, D.M. Amyloid Beta and Phosphorylated Tau-Induced Defective Autophagy and Mitophagy in Alzheimer’s Disease. Cells 2019, 8, 488. [Google Scholar] [CrossRef] [Green Version]
  177. Baron, O.; Boudi, A.; Dias, C.; Schilling, M.; Nölle, A.; Vizcay-Barrena, G.; Rattray, I.; Jungbluth, H.; Scheper, W.; Fleck, R.A.; et al. Stall in Canonical Autophagy-Lysosome Pathways Prompts Nucleophagy-Based Nuclear Breakdown in Neurodegeneration. Curr. Biol. 2017, 27, 3626–3642.e6. [Google Scholar] [CrossRef] [Green Version]
  178. Moreno-Blas, D.; Gorostieta-Salas, E.; Pommer-Alba, A.; Muciño-Hernández, G.; Gerónimo-Olvera, C.; Maciel-Barón, L.A.; Konigsberg, M.; Massieu, L.; Castro-Obregón, S. Cortical neurons develop a senescence-like phenotype promoted by dysfunctional autophagy. Aging 2019, 11, 6175–6198. [Google Scholar] [CrossRef]
  179. Ranade, D.; Koul, S.; Thompson, J.; Prasad, K.B.; Sengupta, K. Chromosomal aneuploidies induced upon Lamin B2 depletion are mislocalized in the interphase nucleus. Chromosoma 2017, 126, 223–244. [Google Scholar] [CrossRef] [Green Version]
  180. Braak, H.; Alafuzoff, I. Staging of Alzheimer disease-associated neurofibrillary pathology using paraffin sections and immunocytochemistry. Acta Neuropathol. 2006, 112, 389–404. [Google Scholar] [CrossRef] [Green Version]
  181. Ruifrok, A.C.; Johnston, D.A. Quantification of histochemical staining by color deconvolution. Anal. Quant. Cytol. Histol. 2001, 23, 291–299. [Google Scholar] [PubMed]
  182. Waldau, B.; Shetty, A.K. Behavior of neural stem cells in the Alzheimer brain. Cell Mol. Life Sci. 2008, 65, 2372–2384. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Greene, J.G.; Borges, K.; Dingledine, R. Quantitative transcriptional neuroanatomy of the rat hippocampus: Evidence for wide-ranging, pathway-specific heterogeneity among three principal cell layers. Hippocampus 2009, 19, 253–264. [Google Scholar] [CrossRef]
  184. Zeier, Z.; Madorsky, I.; Xu, Y.; Ogle, W.O.; Notterpek, L.; Foster, T.C. Gene expression in the hippocampus: Regionally specific effects of aging and caloric restriction. Mech. Ageing Dev. 2011, 132, 8–19. [Google Scholar] [CrossRef] [Green Version]
  185. Shimizu, H.; Mizuguchi, A.; Aoki, M. Differential responses between CA1 pyramidal cells and granule cells to ischemic insult in rat hippocampal slices. Neurosci. Lett. 1996, 203, 195–198. [Google Scholar] [CrossRef]
  186. Gerges, N.Z.; Stringer, J.L.; Alkadhi, K.A. Combination of hypothyroidism and stress abolishes early LTP in the CA1 but not dentate gyrus of hippocampus of adult rats. Brain Res. 2001, 922, 250–260. [Google Scholar] [CrossRef]
  187. Yao, H.; Huang, Y.H.; Liu, Z.W.; Wan, Q.; Ding, A.S.; Zhao, B.; Fan, M.; Wang, F.Z. The different responses to anoxia in cultured CA1 and DG neurons from newborn rats. Sheng Li Xue Bao Acta Physiol. Sin. 1998, 50, 61–66. [Google Scholar]
  188. Daval, J.L.; Pourié, G.; Grojean, S.; Lièvre, V.; Strazielle, C.; Blaise, S.; Vert, P. Neonatal hypoxia triggers transient apoptosis followed by neurogenesis in the rat CA1 hippocampus. Pediatr. Res. 2004, 55, 561–567. [Google Scholar] [CrossRef] [Green Version]
  189. Hsu, J.C.; Zhang, Y.; Takagi, N.; Gurd, J.W.; Wallace, M.C.; Zhang, L.; Eubanks, J.H. Decreased expression and functionality of NMDA receptor complexes persist in the CA1, but not in the dentate gyrus after transient cerebral ischemia. J. Cereb. Blood Flow Metab. 1998, 18, 768–775. [Google Scholar] [CrossRef] [Green Version]
  190. Choi, S.H.; Tanzi, R.E. Is Alzheimer’s Disease a Neurogenesis Disorder? Cell Stem Cell 2019, 25, 7–8. [Google Scholar] [CrossRef]
  191. Cuartero, M.I.; de la Parra, J. Abolition of aberrant neurogenesis ameliorates cognitive impairment after stroke in mice. J. Clin. Investig. 2019, 129, 1536–1550. [Google Scholar] [CrossRef] [PubMed]
  192. Briley, D.; Ghirardi, V. Preserved neurogenesis in non-demented individuals with AD neuropathology. Sci. Rep. 2016, 6, 27812. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Changes in pyramidal neurons of layer II of the entorhinal cortex. Perinuclear and nucleoplasmic (arrow) Lamin B2 (AE). Lamin A absence in adult and senile stages (F,G) and presence in AD I–IV stages (H,I) and absence in AD V–VI stages (J). Nuclear Tau, AT100 (KM), AT8 (Q) cytoplasmic and extracellular AT100 and AT8 (N,O,RT). Absence of nuclear Tau AT8 (P). Scale bar: 20 µm.
Figure 1. Changes in pyramidal neurons of layer II of the entorhinal cortex. Perinuclear and nucleoplasmic (arrow) Lamin B2 (AE). Lamin A absence in adult and senile stages (F,G) and presence in AD I–IV stages (H,I) and absence in AD V–VI stages (J). Nuclear Tau, AT100 (KM), AT8 (Q) cytoplasmic and extracellular AT100 and AT8 (N,O,RT). Absence of nuclear Tau AT8 (P). Scale bar: 20 µm.
Ijms 23 12873 g001
Figure 2. Lamin B2 distribution in the nuclei of DG neurons. Center: Panoramic view of human DG indicating fields (15) of microphotographs and digital quantification of immunopositivity. Scale bar: 100 µm. Five representative microphotographs of adult, senile, and AD stages I–VI. Scale bar: 20 µm. Intermediate and late AD stages (III–VI) show the highest amount of Lamin B2 positivity limited to perinuclear borders.
Figure 2. Lamin B2 distribution in the nuclei of DG neurons. Center: Panoramic view of human DG indicating fields (15) of microphotographs and digital quantification of immunopositivity. Scale bar: 100 µm. Five representative microphotographs of adult, senile, and AD stages I–VI. Scale bar: 20 µm. Intermediate and late AD stages (III–VI) show the highest amount of Lamin B2 positivity limited to perinuclear borders.
Ijms 23 12873 g002
Figure 3. AT100 and AT8 in DG granule cells. Detail of AT100 transition from nuclei (AC) to cytoplasm and extracellular sites in AD III–VI (D,E). Scale bar: 10 μm. Lower magnification microphotographs (upper panels) show the panoramic view of AT100 immunopositivity. Scale bar: 20 μm. AT8 is scarce in adult DG (F) and shows a strong nuclear increase in senile DG (G). Nuclear positivity gradually disappears from AD I–II to AD V–VI (HJ), and even an NFT can be observed (J). Lower magnification microphotographs (lower panels) show panoramic views of AT8 immunopositivity.
Figure 3. AT100 and AT8 in DG granule cells. Detail of AT100 transition from nuclei (AC) to cytoplasm and extracellular sites in AD III–VI (D,E). Scale bar: 10 μm. Lower magnification microphotographs (upper panels) show the panoramic view of AT100 immunopositivity. Scale bar: 20 μm. AT8 is scarce in adult DG (F) and shows a strong nuclear increase in senile DG (G). Nuclear positivity gradually disappears from AD I–II to AD V–VI (HJ), and even an NFT can be observed (J). Lower magnification microphotographs (lower panels) show panoramic views of AT8 immunopositivity.
Ijms 23 12873 g003
Figure 4. Representative microphotographs of chromatin markers at senile and AD I–II stages. The greatest difference is observed with the H3K9me3 marker (A,B). A slight increase is detected with the rest of the markers: H3K4me3 (C,D); H4K20me3 (E,F) and H3K36me3 (G,H). Scale bar: 10 μm. Quantification bars (mean + SD) show statistically significant differences versus adult (A, p < 0.001, a, p < 0.05) and senile subjects (B, p < 0.001).
Figure 4. Representative microphotographs of chromatin markers at senile and AD I–II stages. The greatest difference is observed with the H3K9me3 marker (A,B). A slight increase is detected with the rest of the markers: H3K4me3 (C,D); H4K20me3 (E,F) and H3K36me3 (G,H). Scale bar: 10 μm. Quantification bars (mean + SD) show statistically significant differences versus adult (A, p < 0.001, a, p < 0.05) and senile subjects (B, p < 0.001).
Ijms 23 12873 g004
Figure 5. Increased nucleophagy in intermediate and late AD stages. Transition from predominantly cytoplasmic (AC) to nuclear and perinuclear LC3 immunopositivity (D,E). Scale bar: 20 µm. Right panels show higher magnification of the nuclear LC3 autophagy marker, noticeable in intermediate and late AD stages. Scale bar: 10 µm.
Figure 5. Increased nucleophagy in intermediate and late AD stages. Transition from predominantly cytoplasmic (AC) to nuclear and perinuclear LC3 immunopositivity (D,E). Scale bar: 20 µm. Right panels show higher magnification of the nuclear LC3 autophagy marker, noticeable in intermediate and late AD stages. Scale bar: 10 µm.
Ijms 23 12873 g005
Figure 6. Quantification of AD hallmarks in DG cells. Perinuclear Lamin B2 (A) and nuclear LC3 immunopositivity (B) show significant increases in intermediate and late AD stages. These changes concur with decreased AT8 nuclear levels (C) and AT100 (D). Statistically significant differences versus adult (A, p < 0.001, a, p < 0.05), senile subjects (B, p < 0.001), and AD I-II (C, p < 0.001, c, p < 0.05). Each bar represents the mean + SD.
Figure 6. Quantification of AD hallmarks in DG cells. Perinuclear Lamin B2 (A) and nuclear LC3 immunopositivity (B) show significant increases in intermediate and late AD stages. These changes concur with decreased AT8 nuclear levels (C) and AT100 (D). Statistically significant differences versus adult (A, p < 0.001, a, p < 0.05), senile subjects (B, p < 0.001), and AD I-II (C, p < 0.001, c, p < 0.05). Each bar represents the mean + SD.
Ijms 23 12873 g006
Table 1. List of employed antibodies.
Table 1. List of employed antibodies.
Antibody (Clone)/Supplier/Catalog Number/Manufacture
  • Anti-Lamin A antibody/Abcam/ab26300/Cambridge, UK
    Species: Rabbit Polyclonal
    Dilution: 1:500
  • Anti-Lamin B2 antibody [EPR9701(B)]/Abcam/ab151735/Cambridge, UK
    Species: Rabbit Monoclonal
    Dilution: 1:200
  • Anti-Human Phospho-PHF-Tau pSer202/Thr205 (AT8)/Thermo Fisher Scientific/MN1020/Rockford, IL, USA
    Species: Mouse monoclonal
    Dilution: 1:20
  • Phospho-Tau (Thr212,Ser214) (AT100)/Invitrogen, Thermo Fisher Scientific/MN1060/Rockford, IL, USA
    Species: Mouse Monoclonal
    Dilution: 1:100
  • Anti-Histone H3 (tri methyl K9) antibody [6F12-H4]/Abcam/ab184677/Cambridge, UK
    Species: Mouse Monoclonal
    Dilution: 1:100
  • Anti-Histone H4 (tri methyl K20) antibody/Abcam/ab195479/Cambridge, UK
    Species: Rabbit Polyclonal
    Dilution: 1:100
  • Anti-Histone H3 (tri methyl K4) antibody/Abcam/ab213224/Cambridge, UK
    Species: Rabbit Monoclonal
    Dilution: 1:100
  • Anti-Histone H3 (tri methyl K36) antibody/Abcam/ab9050/Cambridge, UK
    Species: Rabbit Polyclonal
    Dilution: 1:100
  • Anti-LC3B antibody [EPR18709]—Autophagosome Marker/Abcam/ab192890/Cambridge, UK
    Species: Rabbit Monoclonal
    Dilution: 1:100
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Gil, L.; Chi-Ahumada, E.; Niño, S.A.; Capdeville, G.; Méndez-Torres, A.M.; Guerrero, C.; Rebolledo, A.B.; Olazabal, I.M.; Jiménez-Capdeville, M.E. Pathological Nuclear Hallmarks in Dentate Granule Cells of Alzheimer’s Patients: A Biphasic Regulation of Neurogenesis. Int. J. Mol. Sci. 2022, 23, 12873. https://doi.org/10.3390/ijms232112873

AMA Style

Gil L, Chi-Ahumada E, Niño SA, Capdeville G, Méndez-Torres AM, Guerrero C, Rebolledo AB, Olazabal IM, Jiménez-Capdeville ME. Pathological Nuclear Hallmarks in Dentate Granule Cells of Alzheimer’s Patients: A Biphasic Regulation of Neurogenesis. International Journal of Molecular Sciences. 2022; 23(21):12873. https://doi.org/10.3390/ijms232112873

Chicago/Turabian Style

Gil, Laura, Erika Chi-Ahumada, Sandra A. Niño, Gabriela Capdeville, Areli M. Méndez-Torres, Carmen Guerrero, Ana B. Rebolledo, Isabel M. Olazabal, and María E. Jiménez-Capdeville. 2022. "Pathological Nuclear Hallmarks in Dentate Granule Cells of Alzheimer’s Patients: A Biphasic Regulation of Neurogenesis" International Journal of Molecular Sciences 23, no. 21: 12873. https://doi.org/10.3390/ijms232112873

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop