Skip to content
Publicly Available Published by De Gruyter February 14, 2019

Neurogenesis and brain aging

  • Nickolay K. Isaev EMAIL logo , Elena V. Stelmashook and Elisaveta E. Genrikhs

Abstract

Human aging affects the entire organism, but aging of the brain must undoubtedly be different from that of all other organs, as neurons are highly differentiated postmitotic cells, for the majority of which the lifespan in the postnatal period is equal to the lifespan of the entire organism. In this work, we examine the distinctive features of brain aging and neurogenesis during normal aging, pathological aging (Alzheimer’s disease), and accelerated aging (Hutchinson-Gilford progeria syndrome and Werner syndrome).

Introduction

Aging is associated with a gradual reduction in the effectiveness of mechanisms involved in the maintenance of homeostasis of the organism and its organs and tissues, which leads to an increased risk of various pathologies and death. Currently, there are several dozen theories of aging, but all of them can be divided into two groups: aging is a genetically programmed process and aging is a random process caused by a gradual damage of the organism over time as a result of its vital activities. Human aging affects the entire organism, but aging of the brain must undoubtedly be different from that of all other organs, as neurons are highly differentiated postmitotic cells, for the majority of which the lifespan in the postnatal period is equal to the lifespan of the entire organism (Isaev et al., 2013). Using single-cell whole genome sequencing, the accumulation of somatic mutations with age has been observed in single postmitotic human neurons of the prefrontal cortex and hippocampus (Lodato et al., 2018). There is no large loss of neurons in the process of normal aging, but the number, diameter, length, and branching of the dendrites as well as the density of the dendritic spines in the neurons may decrease with aging (Page et al., 2002; Duan et al., 2003). It should be taken into account that the structure of these changes is very heterogeneous in different regions of the brain, and the decrease in the number of neurons is very local (Smith et al., 2004). As the intensity of neuronal death is varied in different regions of the brain, the decrease in volume of the brain also varies in its different departments and structures. The greatest shrinkage occurs in the frontal and temporal cortex, putamen, thalamus, and nucleus accumbens (Fjell and Walhovd, 2010). It should be noted that, in the hippocampus (perhaps in other regions of the brain as well), neuronal death could be partially compensated as a result of neurogenesis. However, with age, neurogenesis is significantly impaired. In recent years, accumulated evidence has shown that neurogenesis can restore a more youthful state during aging. An important objective for the coming years will be to find ways to intervene in this process to slow the aging of the brain. There are already some data that suggest that it may be possible to influence lifespan and aging through genetic manipulations. One of the genes that are of interest in this regard is the Klotho gene. The Klotho protein is involved in the regulation of phosphate and calcium metabolism, protects cells from oxidative stress and apoptosis (Duce et al., 2008), and suppresses the intracellular signal transduction pathways of insulin and insulin-like growth factor-I (IGF-I; Kurosu et al., 2005). The overexpression of this protein increases the lifespan of female mice by 19% and that of males by 30% compared to normal mice. The disrupted expression of the Klotho gene in mice causes a syndrome that resembles human aging and results in a shortened lifespan, development of infertility, arteriosclerosis, skin atrophy, osteoporosis, and emphysema (Kuro-o et al., 1997). Notably, the expression of the Klotho protein in the mammalian brain decreases during normal aging (Duce et al., 2008), whereas mice with a mutation of this gene exhibit cognitive impairment and accumulation of lipid peroxide in the brain (Nagai et al., 2003). A high level of expression of the Klotho gene was detected in the brain, which suggests that the Klotho gene is involved in the regulation of brain aging. This assumption is supported by the fact that the Klotho protein is involved in the regulation of neurogenesis in the brain of adult animals. The overexpression of the Klotho gene has been found to enhance neurogenesis at least in early adulthood, whereas Klotho-deficient brains undergo a rapid collapse of the hippocampal neurogenic niche (Laszczyk et al., 2017). It should be noted that accelerated aging of the organism is observed not only as a result of mutation of the Klotho gene but also accompanies a number of other genetic pathologies. Therefore, one of the approaches to the problem of aging is studying genetic pathologies leading to accelerated aging, such as the Hutchinson-Gilford progeria syndrome (HGPS), Werner syndrome (WS), and Down syndrome. This approach can also be used to investigate the neuronal mechanisms underlying normal and pathological brain aging (Isaev et al., 2018).

Brain neurogenesis in normal aging

Neurogenesis is the process of the formation of new neurons from their precursors – stem cells. Stem cells in various tissues are located in a special microenvironment – niche of stem cells. The concept of neurogenic niches is becoming widely accepted due to the growing evidence of the important role of the microenvironment created in the close vicinity of stem cells in establishing adequate control of cell proliferation, differentiation, and apoptosis (Pozhilenkova et al., 2017). All cells that are in the niches of stem cells and in their immediate vicinity affect the level of neurogenesis. The main components of the niche are mature neurons, neuronal progenitors, endothelial cells, ependymal cells, astrocytes, and microglia. This microenvironment is necessary not only for the normal functioning of the stem cells but also for the coordination of their behavior and interaction with the environment of the organism.

Neural stem cells (NSCs) transplanted beyond the neurogenic niche lose their ability for self-renewal and formation of new neurons (Suhonen et al., 1996). This is likely to happen because of the lack of necessary signals from the microenvironment (Apple et al., 2017). In adult primitive vertebrates, such as zebrafish, the zones of NSC niches are located along the entire rostrocaudal axis of the brain (Kizil et al., 2012). The ability of fish, frogs, and salamanders for regeneration in the brain is apparently associated with the presence of extensive germinative regions in their central nervous system (Tincer et al., 2016). In the brain and spinal cord of zebrafish, stem cells retain their ability to reactivate the molecular programs required for central nervous system regeneration (Ceci et al., 2019). However, in the adult mammalian brain, NSC niches are located only in the subventricular zone (SVZ) of the lateral ventricles and in the subgranular zone (SGZ) of the dentate gyrus of the hippocampus (Ming and Song, 2011). It should be noted that whereas the neurogenic ability of stem cells in SVZ niches in rodents is retained throughout the adult life, the formation of new neurons in SVZ in humans is greatly reduced 2 years after birth (Sanai et al., 2011; Conover and Todd, 2017). New neurons not only maintain the structural integrity and regeneration of brain tissue, but they are also necessary for learning and memory (Snyder et al., 2005; Aimone et al., 2006; Lledo et al., 2006). However, it should be noted that there is still a lot of controversy about the direct involvement of newly formed neurons in learning and memory. According to one hypothesis, neurogenesis is more crucial for more complex tasks, such as pattern separation and cognitive flexibility, rather than for direct learning and direct recollection. But undoubtedly, a reduction of neurogenesis in the adult brain results in the disruption of vital physiological processes (Cameron and Glover, 2015). Neurogenesis in mammals goes on throughout the entire life, but as the animals age this process is significantly diminished (Enwere et al., 2004; Ahlenius et al., 2009; Schouten et al., 2012; Cameron and Glover, 2015); however, the ability for synaptogenesis in newly formed hippocampal neurons varies little with age (Schouten et al., 2012). A number of studies have shown that a reduction in the level of hippocampal neurogenesis correlates with the development of deficiency in memory and learning (Drapeau et al., 2003; Driscoll et al., 2006). It should be noted that the administration of fibroblast growth factor-2 and heparin-binding epidermal growth factor to old mice restores the number of newly formed neurons in SVZ and SGZ to the level observed in young animals (Jin et al., 2003). In addition, an understanding of the age-dependent changes in proteostasis in the NSC pool could help identify ways to enhance the function of old NSCs and maintain the brain health. In the adult brain, the NSC pool comprises quiescent and activated populations with distinct roles. During aging, quiescent NSCs accumulate defects in their lysosomes, which leads to an increased formation of protein aggregates and the ability of NSCs for activation decreases. Restoring the lysosomal pathway in old quiescent NSCs helps clear cells from protein aggregates and improve the ability of NSCs to activate, allowing them to regain a more youthful state (Leeman et al., 2018).

The level of neurogenesis in old animals can be maintained by means of enriching their environment. In mice that lived in an enriched environment from age 10 to 20 months, adult hippocampal neurogenesis was fivefold higher than in the controls. Such cellular plasticity was observed in the context of significant improvements in learning parameters, exploratory behavior, and locomotor activity. Mice that lived in an enriched environment also had a reduced lipofuscin load in the dentate gyrus, which indicates decreased nonspecific age-dependent degeneration. Therefore, signs of neuronal aging in mice can be diminished by a prolonged active and challenging lifestyle even if this stimulation started only at the middle age. The activity exerts not only a short-term but also a long-term effect on brain plasticity (Kempermann et al., 2002). These data support the hypothesis that the rate of aging is determined by a number of factors and in particular by psychological and social aspects of people behavior; these factors affect the brain, which, depending on the signals received, can accelerate or slow down the work of the aging program (Skulachev et al., 2014; Isaev et al., 2015).

It should be noted that recent studies have demonstrated that brain injury is capable of activating an endogenous program of neurogenesis that results in the replacement of neurons in various cerebral regions of rodents and primates (Yamashima et al., 2007). Brain ischemia may be an example of such injury. In ischemia, endogenous neural precursors located in different regions of the adult primate brain are differentially activated. In the adult macaque monkey brain, a limited endogenous potential for postischemic neuronal repair exists in the neocortex and striatum but not in the hippocampus per se (Tonchev et al., 2007). The authors believed that the presence of putative parenchymal progenitors and sustained progenitors in germinative centers provides new opportunities for precursor cell recruitment to sites of injury. The molecular manipulation of this process could make it possible to employ brain progenitor cells for the treatment of human neurological diseases. The possibility of stimulation of early stages of neurogenic response was also shown in a diffuse brain injury model (Bye et al., 2011).

Very interesting data have been obtained by studying cultured progenitor cells derived from murine SVZ. The authors have shown that primary NSC cultures from adult and old animals form fewer neurospheres than those from embryonic ones, but with prolonged cultivation this difference disappears. Moreover, aging results in the reduction of the proliferation of NSCs and their survival during their differentiation in vitro. However, with further cultivation, the remaining living cells have retained their ability to actively proliferate and differentiate. Thus, old NSCs can differentiate into functional neurons that are indistinguishable in their electrophysiological properties from neurons derived from NSCs of young animals (Ahlenius et al., 2009). These results demonstrate that the age-related mechanisms leading to a reduction in neurogenesis do not affect the morphofunctional properties of newly formed neurons. This is also confirmed by the fact that the density of dendritic spines in new neurons in young and old animals is the same despite the decrease in neurogenesis in the hippocampus of old mice (Morgenstern et al., 2008). At the same time, the pyramidal neurons of the cerebral cortex of old animals are characterized by not only age-related selective elimination of small, thin spines but also by a decrease in the density of axo-spine synapses (Dumitriu et al., 2010).

Apparently, unlike stem cells that can differentiate into other specialized cells (not neurons), the functions of NSCs of old animals are largely restored at the conditions in vitro (Ahlenius et al., 2009), which implies the influence of epigenetic factors on the formation of the age-related phenotype of these stem cells. Indeed, heterochronic parabiosis restores levels of IGF-I, growth hormone, Wnt3, transforming growth factor-β, or GDF11 in old mice and improves neurogenesis (Schultz and Sinclair, 2016).

Stem cells of the lateral ventricle of the brain are in contact with the cerebrospinal fluid and bloodstream that can affect the state of these cells. One of the negative regulators of adult neurogenesis and an inducer of microglial reactivity is the vascular cell adhesion molecule 1; its systemic blockade with antibodies prevents the inhibitory effect of the old environment on neurogenesis and microglial reactivity (Yousef et al., 2018).

Summarizing, the function of NSCs of old animals is largely restored not only when they are transferred to the conditions in vitro but also in vivo, when the animals are kept in the enriched environment or subjected to heterochronic parabiosis or the treatment resulting in the intensification of the lysosome work. This implies that the age-related mechanisms leading to a decrease in neurogenesis in normal aging do not affect the morphological and functional properties of newly formed neurons.

Brain neurogenesis in pathological aging: Alzheimer’s disease (AD)

In the contemporary aging society, cognitive dysfunction is one of the most serious issues that need to be urgently addressed (Iwamoto and Ouchi, 2014). Aging of the brain can follow the pathological pathway with the development of AD. Diagnostic signs of AD arise very slowly and sequentially, as if the disease develops according to a specific program (Isaev et al., 2015). The population frequency of this disease steadily increases with age and comprises 0.7, 4.6, 16.5, and 18.2% in the age groups 60–69, 70–79, 80–89, and 90+ years, respectively (Kalyn and Bratsun, 1999). In contrast to normal aging, AD is accompanied by a progressive impairment of cognitive functions, synaptic degeneration, an increase in the expression of the active form of the apoptotic enzyme caspase-3 and procaspase-3 in the synapses, and the massive death of neurons (Louneva et al., 2008). Especially significant is the loss of neurons in the cerebral cortex, cerebellum, and cholinergic basal nuclei and degeneration of synaptic contacts (Bertoni-Freddari et al., 1996; Kerbler et al., 2014). The main initiators of cognitive impairment and synaptic degeneration followed by neuronal loss in patients with AD are currently believed to be the β-amyloid peptide and the hyperphosphorylated intracellular protein τ, the accumulation of which leads to mitochondrial damage and development of oxidative stress (Schmitt et al., 2012). Mass death of neurons in this neurodegenerative disease causes the disintegration of various parts of the brain and the complete disruption of its functions, whereas in normal aging the disruption of connections in the brain can be partially compensated by the delocalization of activity, i.e. by the involvement of additional brain regions in cognitive activities, which can significantly prevent the deterioration of cognitive abilities (Bishop et al., 2010).

In recent years, accumulated evidence has shown that the symptoms of AD may be partially associated with the impairment of the neuron’s formation from NSCs, which is necessary to maintain the function of learning and memory (Li et al., 2016). However, a complete clarity about the changes in the process of neurogenesis in the human brain in AD is still missing. Rather contradictory data have been obtained in the animal models of this disease as well (Chuang, 2010). For most mouse AD models that are based on mutations of the APP protein, the precursor of the β-amyloid peptide, a decrease in neurogenesis has been shown, whereas in transgenic mice with different mutations of preselenin, neurogenesis could either increase or decrease (Zhao et al., 2008). Enriching the environment with various devices to provide extensive physical and learning activities (tunnels, houses, hammocks, stairs, boxes, and wheels) restores neurogenesis affected by the toxic action of β-amyloid peptide in animal AD models (Salmin et al., 2017).

It should be noted that the modeling of sporadic form of AD by the administration of streptozotocin into the lateral ventricles of the rodent brain (Mayer et al., 1990; Hoyer et al., 1994) is accompanied not only by the impairment of animals’ ability to learn and memorize but also by a significant decrease in neurogenesis in the lateral ventricles of the brain and the hippocampus (Sun et al., 2015; Mishra et al., 2017). It is interesting that when modeling AD by administering β-amyloid peptide into the brains of old zebrafish, the activation of NSC proliferation and neurogenesis has been observed (Bhattarai et al., 2017). According to some data, in the hippocampus of patients with AD, there is an increase in cell proliferation as well, but this process is apparently associated with glial cells rather than neurogenesis (Boekhoorn et al., 2006). It is interesting that the dentate gyrus is the most resilient area of the hippocampus to many pathological conditions including AD (Parent et al., 2013).

Summarizing the available data, we can conclude that, despite the large amount of research performed in animal models of AD, it remains unclear how this pathology affects hippocampal neurogenesis (Martinez-Canabal, 2014). At the present time, possible approaches to AD therapy, such as transplantation of various types of stem cells, are being actively discussed and developed, but it is too early to talk about the actual progress made in this direction.

Brain neurogenesis in accelerated aging

Another form of pathological or rather accelerated aging is progeria, or the HGPS, in which the typical signs of aging begin to appear in early childhood. The average life expectancy of patients with this disease is 14.6 years (Gordon et al., 2003). In children with this pathology, growth retardation after birth, loss of subcutaneous fat, sclerotic skin with pigmentation spots, alopecia, atherosclerosis, and anomalies in the development of the skeleton are observed (Coutinho et al., 2009). The development of the disease is associated with a mutation of the LMNA gene on the chromosome 1 encoding lamin A, a fibrillar structural protein that is part of the nuclear lamina and is involved in the regulation of transcription in the cell nucleus. At the ultrastructural level, the nuclear lamina is a protein network adjacent to the inner membrane of the nuclear envelope and involved in maintaining the shape, size, and integrity of the nucleus and the pore complex of the nuclear envelope. Mutant lamin A, also called progerin, is capable of binding to the transcription factor NRF2 that is activated by the exposure of the cell to oxidative stress and induces the expression of antioxidant enzymes. Binding of NRF2 leads to a disruption of its transcriptional activity, which in turn causes chronic oxidative stress (Kubben et al., 2016). Despite accelerated aging, patients with HGPS do not have cognitive impairments and symptoms typical of AD (Ullrich and Gordon, 2015; Isaev et al., 2018).

It is possible that progerin is involved in the process of normal aging as well, as nuclei of fibroblasts from old people express progerin and have defects similar to those in cell nuclei of patients with HGPS (Scaffidi and Misteli, 2006; Cao et al., 2007). Moreover, the content of this protein in the coronary vessels has been shown to increase with age (Olive et al., 2010; Graziotto et al., 2012).

There is currently no evidence of changes in neurogenesis in patients with HGPS. However, interesting results have been obtained in the modeling of this disease in transgenic mice expressing a mutation of the LMNA gene (Baek et al., 2015). The mutant progerin protein has been found in the skin, heart, brain, and kidneys of the animals, but the adverse effect of progerin in the brain has been less pronounced than expected. Although the synthesis of ATP by the mitochondria of fibroblasts of mice expressing progerin or prelamin A was significantly reduced, no such impairments were found in the mitochondria of the brain. Long-term expression of the mutant gene did not cause pronounced neuropathological defects despite serious disruptions in the ultrastructure of the nuclei of hippocampal neurons. A study of the process of neurogenesis in the brain of animals with LMNA gene mutation showed that the fractions of newly formed and immature neurons as well as the numbers of proliferating cells in the SGZ of the dentate gyrus of the hippocampus are similar in mutant and wild-type animals (Baek et al., 2015). The authors concluded that nerve cells are less sensitive to progerin expression, although it has a long-term effect on the adult organism.

It should be noted that, in comparison to peripheral tissues, neurons and glial cells in the murine brain have rather low levels of lamin A and its precursor prelamin A, whereas the C form of lamin is predominant. Moreover, in knockout mice expressing progerin in peripheral tissues, the level of this mutant protein in the brain is also very low and the regulation of its decrease at the transcriptional level is associated with brain-specific miR-9 microRNA (Jung et al., 2012; Nissan et al., 2012). Recent studies of induced pluripotent stem cells derived from patients with HGPS confirmed the absence of lamin A expression in neurons derived from these cells (Nissan et al., 2012). In addition, these studies showed that protection of neurons from the negative effect of progerin in HGPS is realized via a physiological limitation of expression of this protein by means of miR-9 microRNA.

To explain the selectivity of the manifestation of HGPS in different tissues, a hypothesis has been proposed that progeria syndromes manifest themselves mainly in tissues that are more susceptible to mechanical stress (for example, blood vessels and joints) or are necessary to maintain continuous growth (hair follicles and nails) and in which the replacement of cells and depletion of the pool of progenitors occur more rapidly. Therefore, diseases associated with tissues that are protected against mechanical stress (the brain) are mostly absent in HGPS. However, there are some facts that contradict this hypothesis. For example, there is practically no change in skeletal muscles in progeria (Halaschek-Wiener and Brooks-Wilson, 2007).

It is interesting that patients with HGPS have normal cognition and do not show signs of memory impairment or cognitive problems that are often observed during normal aging. However, vascular changes occurring in HGPS cause neurological disorders. Thus, although neuronal cells do not appear to be negatively affected by progerin expression, many patients with HGPS have problems such as headaches, muscle weakness, or convulsions resulting from circulatory disorders (Ullrich and Gordon, 2015).

Another disease with signs of premature aging is WS or ‘adult progeria’ caused by mutations of the WRN gene. This gene encodes a multifunctional nuclear protein with exonuclease and ATP-dependent helicase activities. The mean age of death is 40–50 years; the mean age of diagnosis is 15–20 years (Huang et al., 2006). Based on presently available limited data, it was suggested that the symptoms of accelerated aging in WS are much less pronounced in the brain than in other organs (Isaev et al., 2018). This is confirmed by the fact that WS patients exhibit premature aging predominantly in mesenchyme-derived tissues, but not in neural lineages, which may be a consequence of telomere dysfunction and accelerated senescence. Indeed, telomerase activity was found to be higher in neural progenitor cells (NPCs) than in mesenchymal stem cells derived from induced pluripotent stem cells. The activity of telomerase in normal NPCs and in NPCs from patients with WS did not vary significantly. Similarly, the length of telomeres in WS NPCs was comparable to that in normal NPCs. WS NPCs actively proliferate in cell culture and include bromodeoxyuridine at a speed similar to that of normal NPCs. In the cultures of WS NPCs, premature aging was not observed. Based on these data, there is no impairment of proliferation and telomere function in WS, which is consistent with the clinical phenotype of WS (Cheung et al., 2014). These data demonstrate that in pathologies associated with accelerated aging, NPCs and the whole brain are generally less affected than other tissues and organs.

Conclusion

Apparently, aging is not a simple accumulation of errors; rather, it is a gradual impairment of the work of a multifactorial program responsible for the prevention and repair of the damages in genome, proteins, and lipids. Therefore, mutations of individual genes involved in the implementation of this program can cause pathologies manifested in accelerated aging. Human aging affects the entire organism; however, the above data demonstrate that the brain has a number of distinctive features and the age-related mechanisms leading to a decrease in neurogenesis do not have an effect on the morphological properties of the newly formed neurons. In addition, unlike other stem cells, the function of NSCs of old animals can largely be restored in conditions in vitro and in vivo. Probably, one of the factors that protect the brain during accelerated and normal aging is a low level of lamin A and its precursor prelamin A in neurons and glia. All of this suggests that the brain has additional antiaging systems; without them, normal aging could have a much higher rate.

Thus, the presented facts indicate that aging is not necessarily a permanent state but may be susceptible to treatment and that finding ways to interfere in the intrinsic cell machinery to slow down or even reverse this process will be the challenge for years to come (Katsimpardi and Lledo, 2018).

Award Identifier / Grant number: 14-24-00107

Funding statement: This work was done with financial support from the Russian Science Foundation, Funder Id: 10.13039/501100006769 (project nos. 14-24-00107, section ‘Normal Aging’, and 16-15-10108, other sections).

References

Ahlenius, H., Visan, V., Kokaia, M., Lindvall, O., and Kokaia, Z. (2009). Neural stem and progenitor cells retain their potential for proliferation and differentiation into functional neurons despite lower number in aged brain. J. Neurosci. 29, 4408–4419.10.1523/JNEUROSCI.6003-08.2009Search in Google Scholar PubMed PubMed Central

Aimone, J.B., Wiles, J., and Gage, F.H. (2006). Potential role for adult neurogenesis in the encoding of time in new memories. Nat. Neurosci. 9, 723–727.10.1038/nn1707Search in Google Scholar PubMed

Apple, D.M., Solano-Fonseca, R., and Kokovay, E. (2017). Neurogenesis in the aging brain. Biochem. Pharm. 141, 77–85.10.1016/j.bcp.2017.06.116Search in Google Scholar PubMed

Baek, J.H., Schmidt, E., Viceconte, N., Strandgren, C., Pernold, K., Richard, T.J., Van Leeuwen, F.W., Dantuma, N.P., Damberg, P., Hultenby, K., et al. (2015). Expression of progerin in aging mouse brains reveals structural nuclear abnormalities without detectible significant alterations in gene expression, hippocampal stem cells or behavior. Hum. Mol. Genet. 24, 1305–1321.10.1093/hmg/ddu541Search in Google Scholar PubMed PubMed Central

Bertoni-Freddari, C., Fattoretti, P., Casoli, T., Caselli, U., and Meier-Ruge, W. (1996). Deterioration threshold of synaptic morphology in aging and senile dementia of Alzheimer’s type. Anal. Quant. Cytol. Histol. 18, 209–213.Search in Google Scholar

Bhattarai, P., Thomas, A.K., Zhang, Y., and Kizil, C. (2017). The effects of aging on amyloid-β42-induced neurodegeneration and regeneration in adult zebrafish brain. Neurogenesis Austin 4, e1322666.10.1080/23262133.2017.1322666Search in Google Scholar PubMed PubMed Central

Bishop, N.A., Lu, T., and Yankner, B.A. (2010). Neural mechanisms of ageing and cognitive decline. Nature 464, 529–535.10.1038/nature08983Search in Google Scholar PubMed PubMed Central

Boekhoorn, K., Joels, M., and Lucassen, P.J. (2006). Increased proliferation reflects glial and vascular-associated changes, but not neurogenesis in the presenile Alzheimer hippocampus. Neurobiol. Dis. 24, 1–14.10.1016/j.nbd.2006.04.017Search in Google Scholar PubMed

Bye, N., Carron, S., Han, X., Agyapomaa, D., Ng, S.Y., Yan, E., Rosenfeld, J.V., and Morganti-Kossmann, M.C. (2011). Neurogenesis and glial proliferation are stimulated following diffuse traumatic brain injury in adult rats. J. Neurosci. Res. 89, 986–1000.10.1002/jnr.22635Search in Google Scholar PubMed

Cameron, H.A. and Glover, L.R. (2015). Adult neurogenesis: beyond learning and memory. Annu. Rev. Psychol. 66, 53–81.10.1146/annurev-psych-010814-015006Search in Google Scholar PubMed PubMed Central

Cao, K., Capell, B.C., Erdos, M.R., Djabali, K., and Collins, F.S. (2007). A lamin A protein isoform overexpressed in Hutchinson-Gilford progeria syndrome interferes with mitosis in progeria and normal cells. Proc. Natl. Acad. Sci. USA 104, 4949–4954.10.1073/pnas.0611640104Search in Google Scholar PubMed PubMed Central

Ceci, M., Mariano, V., and Nicla Romano, N. (2019). Zebrafish as a translational regeneration model to study the activation of neural stem cells and role of their environment. Rev. Neurosci. 30, 45–66.10.1515/revneuro-2018-0020Search in Google Scholar PubMed

Cheung, H.H., Liu, X., Canterel-Thouennon, L., Li, L., Edmonson, C., and Rennert, O.M. (2014). Telomerase protects Werner syndrome lineage-specific stem cells from premature aging. Stem Cell Rep. 2, 534–546.10.1016/j.stemcr.2014.02.006Search in Google Scholar PubMed PubMed Central

Chuang, T.T. (2010). Neurogenesis in mouse models of Alzheimer’s disease. Biochim. Biophys. Acta Mol. Basis Dis. 1802, 872–880.10.1016/j.bbadis.2009.12.008Search in Google Scholar PubMed

Conover, J.C. and Todd, K.L. (2017). Development and aging of a brain neural stem cell niche. Exp. Gerontol. 94, 9–13.10.1016/j.exger.2016.11.007Search in Google Scholar PubMed PubMed Central

Coutinho, H.D., Falcão-Silva, V.S., Gonçalves, G.F., and da Nóbrega, R.B. (2009). Molecular ageing in progeroid syndromes: Hutchinson-Gilford progeria syndrome as a model. Immun. Ageing 6, 4.10.1186/1742-4933-6-4Search in Google Scholar PubMed PubMed Central

Drapeau, E., Mayo, W., Aurousseau, C., Le Moal, M., Piazza, P.V., and Abrous, D.N. (2003). Spatial memory performances of aged rats in the water maze predict levels of hippocampal neurogenesis. Proc. Natl. Acad. Sci. USA 100, 14385–14390.10.1073/pnas.2334169100Search in Google Scholar PubMed PubMed Central

Driscoll, I., Howard, S.R., Stone, J.C., Monfils, M.H., Tomanek, B., Brooks, W.M., and Sutherland, R.J. (2006). The aging hippocampus: a multi-level analysis in the rat. Neuroscience 139, 1173–1185.10.1016/j.neuroscience.2006.01.040Search in Google Scholar PubMed

Duan, H., Wearne, S.L., Rocher, A.B., Macedo, A., Morrison, J.H., and Hof, P.R. (2003). Age-related dendritic and spine changes in corticocortically projecting neurons in macaque monkeys. Cereb. Cortex 13, 950–961.10.1093/cercor/13.9.950Search in Google Scholar PubMed

Duce, J.A., Podvin, S., Hollander, W., Kipling, D., Rosene, D.L., and Abraham, C.R. (2008). Gene profile analysis implicates Klotho as an important contributor to aging changes in brain white matter of the rhesus monkey. Glia 56, 106–117.10.1002/glia.20593Search in Google Scholar PubMed

Dumitriu, D., Hao, J., Hara, Y., Kaufmann, J., Janssen, W.G., Lou, W., Rapp, P.R., and Morrison, J.H. (2010). Selective changes in thin spine density and morphology in monkey prefrontal cortex correlate with aging-related cognitive impairment. J. Neurosci. 30, 7507–7515.10.1523/JNEUROSCI.6410-09.2010Search in Google Scholar PubMed PubMed Central

Enwere, E., Shingo, T., Gregg, C., Fujikawa, H., Ohta, S., and Weiss, S. (2004). Aging results in reduced epidermal growth factor receptor signaling, diminished olfactory neurogenesis, and deficits in fine olfactory discrimination. J. Neurosci. 24, 8354–8365.10.1523/JNEUROSCI.2751-04.2004Search in Google Scholar PubMed PubMed Central

Fjell, A.M. and Walhovd, K.B. (2010). Structural brain changes in aging: courses, causes and cognitive consequences. Rev. Neurosci. 21, 187–221.10.1515/REVNEURO.2010.21.3.187Search in Google Scholar PubMed

Gordon, L.B., Brown, W.T., and Collins, F.S. (2003). Hutchinson-Gilford progeria syndrome. In: Pagon, R.A., Adam, M.P., Ardinger, H.H., Wallace, S.E., Amemiya, A., Bean, L.J.H., Bird, T.D., Fong, C.T., Mefford, H.C., Smith, R.J.H., et al., editors. GeneReviews® [Internet]. Seattle (WA): University of Washington, 1993–2016. Available at: http://www.ncbi.nlm.nih.gov/books/NBK1121/.Search in Google Scholar

Graziotto, J.J., Cao, K., Collins, F.S., and Krainc, D. (2012). Rapamycin activates autophagy in Hutchinson-Gilford progeria syndrome: implications for normal aging and age-dependent neurodegenerative disorders. Autophagy 8, 147–151.10.4161/auto.8.1.18331Search in Google Scholar PubMed PubMed Central

Halaschek-Wiener, J. and Brooks-Wilson, A. (2007). Progeria of stem cells: stem cell exhaustion in Hutchinson-Gilford progeria syndrome. J. Gerontol. A Biol. Sci. Med. Sci. 62, 3–8.10.1093/gerona/62.1.3Search in Google Scholar PubMed

Hoyer, S., Müller, D., and Plaschke, K. (1994). Desensitization of brain insulin receptor. Effect on glucose/energy and related metabolism. J. Neural Transm. Suppl. 44, 259–268.10.1007/978-3-7091-9350-1_20Search in Google Scholar PubMed

Huang, S., Lee, L., Hanson, N.B., Lenaerts, C., Hoehn, H., Poot, M., Rubin, C.D., Chen, D.F., Yang, C.C., Juch, H., et al. (2006). The spectrum of WRN mutations in Werner syndrome patients. Hum. Mutat. 27, 558–567.10.1002/humu.20337Search in Google Scholar PubMed PubMed Central

Isaev, N.K., Stelmashook, E.V., Stelmashook, N.N., Sharonova, I.N., and Skrebitsky, V.G. (2013). Brain aging and mitochondria targeted plastoquinone antioxidants of SkQ-type. Biochemistry Mosc. 78, 295–300.10.1134/S0006297913030127Search in Google Scholar PubMed

Isaev, N.K., Stelmashook, E.V., Genrikhs, E.E., Oborina, M.V., Kapkaeva, M.R., and Skulachev, V.P. (2015). Alzheimer’s disease: an exacerbation of senile phenoptosis. Biochemistry Mosc. 80, 1578–1581.10.1134/S0006297915120056Search in Google Scholar PubMed

Isaev, N.K., Genrikhs, E.E., Oborina, M.V., and Stelmashook, E.V. (2018). Accelerated aging and aging process in the brain. Rev. Neurosci. 29, 233–240.10.1515/revneuro-2017-0051Search in Google Scholar PubMed

Iwamoto, T. and Ouchi, Y. (2014). Emerging evidence of insulin-like growth factor 2 as a memory enhancer: a unique animal model of cognitive dysfunction with impaired adult neurogenesis. Rev. Neurosci. 25, 559–574.10.1515/revneuro-2014-0010Search in Google Scholar PubMed

Jin, K., Sun, Y., Xie, L., Batteur, S., Mao, X.O., Smelick, C., Logvinova, A., and Greenberg, D.A. (2003). Neurogenesis and aging: FGF-2 and HB-EGF restore neurogenesis in hippocampus and subventricular zone of aged mice. Aging Cell 2, 175–183.10.1046/j.1474-9728.2003.00046.xSearch in Google Scholar PubMed

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. (2012). Regulation of prelamin A but not lamin C by miR-9, a brain-specific microRNA. Proc. Natl. Acad. Sci. USA 109, E423–E431.10.1073/pnas.1111780109Search in Google Scholar PubMed PubMed Central

Kalyn, Ya.B. and Bratsun, A.L. (1999). Distribution and risk factors in developing Alzheimer like dementia. In: Alzheimer’s Disease and Aging: From Neurobiology to Therapy: Materials of Second Russian Conference [in Russian], Moscow, p. 5258.Search in Google Scholar

Katsimpardi, L. and Lledo, P.M. (2018). Regulation of neurogenesis in the adult and aging brain. Curr. Opin. Neurobiol. 53, 131–138.10.1016/j.conb.2018.07.006Search in Google Scholar PubMed

Kempermann, G., Gast, D., and Gage, F.H. (2002). Neuroplasticity in old age: sustained fivefold induction of hippocampal neurogenesis by long-term environmental enrichment. Ann. Neurol. 52, 135–143.10.1002/ana.10262Search in Google Scholar PubMed

Kerbler, G.M., Fripp, J., Rowe, C.C., Villemagne, V.L., Salvado, O., Rose, S., and Coulson, E.J. (2014). Alzheimer’s disease neuroimaging initiative. Basal forebrain atrophy correlates with amyloid β burden in Alzheimer’s disease. Neuroimage Clin. 7, 105–113.10.1016/j.nicl.2014.11.015Search in Google Scholar PubMed PubMed Central

Kizil, C., Kaslin, J., Kroehne, V., and Brand, M. (2012). Adult neurogenesis and brain regeneration in zebrafish. Dev. Neurobiol. 72, 429–461.10.1002/dneu.20918Search in Google Scholar PubMed

Kubben, N., Zhang, W., Wang, L., Voss, T.C., Yang, J., Qu, J., Liu, G.H., and Misteli, T. (2016). Repression of the antioxidant NRF2 pathway in premature aging. Cell 165, 1361–1374.10.1016/j.cell.2016.05.017Search in Google Scholar PubMed PubMed Central

Kurosu, H., Yamamoto, M., Clark, J.D., Pastor, J.V., Nandi, A., Gurnani, P., McGuinness, O.P., Chikuda, H., Yamaguchi, M., Kawaguchi, H., et al. (2005). Suppression of aging in mice by the hormone Klotho. Science 309, 1829–1833.10.1126/science.1112766Search in Google Scholar PubMed PubMed Central

Kuro-o, M., Matsumura, Y., Aizawa, H., Kawaguchi, H., Suga, T., Utsugi, T., Ohyama, Y., Kurabayashi, M., Kaname, T., Kume, E., et al. (1997). Mutation of the mouse Klotho gene leads to a syndrome resembling ageing. Nature 6, 45–51.10.1038/36285Search in Google Scholar PubMed

Laszczyk, A.M., Fox-Quick, S., Vo, H.T., Nettles, D., Pugh, P.C., Overstreet-Wadiche, L., and King G.D. (2017). Klotho regulates postnatal neurogenesis and protects against age-related spatial memory loss. Neurobiol. Aging 59, 41–54.10.1016/j.neurobiolaging.2017.07.008Search in Google Scholar PubMed PubMed Central

Leeman, D.S., Hebestreit, K., Ruetz, T., Webb, A.E., McKay, A., Pollina, E.A., Dulken, B.W., Zhao, X., Yeo, R.W., Ho, T.T., et al. (2018). Lysosome activation clears aggregates and enhances quiescent neural stem cell activation during aging. Science 359, 1277–1283.10.1126/science.aag3048Search in Google Scholar

Li, X., Bao, X., and Wang, R. (2016). Neurogenesis-based epigenetic therapeutics for Alzheimer’s disease. Mol. Med. Rep. 14, 1043–1053.10.3892/mmr.2016.5390Search in Google Scholar

Lledo, P-M., Alonso, M., and Grubb, M.S. (2006). Adult neurogenesis and functional plasticity in neuronal circuits. Nat. Rev. Neurosci. 7, 179–193.10.1038/nrn1867Search in Google Scholar

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. (2018). Aging and neurodegeneration are associated with increased mutations in single human neurons. Science 359, 555–559.10.1126/science.aao4426Search in Google Scholar

Louneva, N., Cohen, J. W., Han, L.-Y., Talbot, K., Wilson, R.S., Bennett, D.A., Trojanowski, J.Q., and Arnold, S.E. (2008). Caspase-3 is enriched in postsynaptic densities and increased in Alzheimer’s disease. Am. J. Pathol. 173, 1488–1495.10.2353/ajpath.2008.080434Search in Google Scholar

Martinez-Canabal, A. (2014). Reconsidering hippocampal neurogenesis in Alzheimer’s disease. Front. Neurosci. 8, 147.10.3389/fnins.2014.00147Search in Google Scholar

Mayer, G., Nitsch, R., and Hoyer, S. (1990). Effects of changes in peripheral and cerebral glucose metabolism on locomotor activity, learning and memory in adult male rats. Brain Res 532, 95–100.10.1016/0006-8993(90)91747-5Search in Google Scholar

Ming, G.-L. and Song, H. (2011). Adult neurogenesis in the mammalian brain: significant answers and significant questions. Neuron 70, 687–702.10.1016/j.neuron.2011.05.001Search in Google Scholar PubMed PubMed Central

Mishra, S.K., Singh, S., Shukla, S., and Shukla, R. (2017). Intracerebroventricular streptozotocin impairs adult neurogenesis and cognitive functions via regulating neuroinflammation and insulin signaling in adult rats. Neurochem. Int. 113, 56–68.10.1016/j.neuint.2017.11.012Search in Google Scholar PubMed

Morgenstern, N.A., Lombardi, G., and Schinder, A.F. (2008). Newborn granule cells in the ageing dentate gyrus. J. Physiol. 586, 3751–3757.10.1113/jphysiol.2008.154807Search in Google Scholar PubMed PubMed Central

Nagai, T., Yamada, K., Kim, H.C., Kim, Y.S., Noda, Y., Imura, A., Nabeshima, Y., and Nabeshima, T. (2003). Cognition impairment in the genetic model of aging Klotho gene mutant mice: a role of oxidative stress. FASEB J. 17, 50–52.10.1096/fj.02-0448fjeSearch in Google Scholar

Nissan, X., Blondel, S., Navarro, C., Maury, Y., Denis, C., Girard, M., Martinat, C., De Sandre-Giovannoli, A., Levy, N., and Peschanski, M. (2012). Unique preservation of neural cells in Hutchinson-Gilford progeria syndrome is due to the expression of the neural-specific miR-9 microRNA. Cell Rep. 2, 1–9.10.1016/j.celrep.2012.05.015Search in Google Scholar

Olive, M., Harten, I., Mitchell, R., Beers, J.K., Djabali, K., Cao, K., Erdos, M.R., Blair, C., Funke, B., Smoot, L., et al. (2010). Cardiovascular pathology in Hutchinson-Gilford progeria: correlation with the vascular pathology of aging. Arterioscler. Thromb. Vasc. Biol. 30, 2301–2309.10.1161/ATVBAHA.110.209460Search in Google Scholar

Page, T.L., Einstein, M., Duan, H., He, Y., Flores, T., Rolshud, D., Erwin, J.M., Wearne, S.L., Morrison, J.H., and Hof, P.R. (2002). Morphological alterations in neurons forming corticocortical projections in the neocortex of aged Patas monkeys. Neurosci. Lett. 317, 37–41.10.1016/S0304-3940(01)02428-4Search in Google Scholar

Parent, M.J., Bedard, M.A., Aliaga, A., Minuzzi, L., Mechawar, N., Soucy, J.P., Schirrmacher, E., Kostikov, A., Gauthier, S.G., and Rosa-Neto, P. (2013). Cholinergic depletion in Alzheimer’s disease shown by [18F]FEOBV autoradiography. Int. J. Mol. Imaging 2013, 205045.10.1155/2013/205045Search in Google Scholar PubMed PubMed Central

Pozhilenkova, E.A., Lopatina, O.L., Komleva, Y.K., Salmin, V.V., and Salmina, A.B. (2017). Blood-brain barrier-supported neurogenesis in healthy and diseased brain. Rev. Neurosci. 28, 397–415.10.1515/revneuro-2016-0071Search in Google Scholar PubMed

Salmin, V.V., Komleva, Y.K., Kuvacheva, N.V., Morgun, A.V., Khilazheva, E.D., Lopatina, O.L., Pozhilenkova, E.A., Shapovalov, K.A., Uspenskaya, Y.A., and Salmina, A.B. (2017). Differential roles of environmental enrichment in Alzheimer’s type of neurodegeneration and physiological aging. Front. Aging Neurosci. 9, 245.10.3389/fnagi.2017.00245Search in Google Scholar PubMed PubMed Central

Sanai, N., Nguyen, T., Ihrie, R.A., Mirzadeh, Z., Tsai, H.H., Wong, M., Gupta, N., Berger, M.S., Huang, E., Garcia-Verdugo, J.M., et al. (2011). Corridors of migrating neurons in the human brain and their decline during infancy. Nature 478, 382–386.10.1038/nature10487Search in Google Scholar PubMed PubMed Central

Scaffidi, P. and Misteli, T. (2006). Lamin A-dependent nuclear defects in human aging. Science 312, 1059–1063.10.1126/science.1127168Search in Google Scholar PubMed PubMed Central

Schmitt, K., Grimm, A., Kazmierczak, A., Strosznajder, J.B., Gotz, J., and Eckert, A. (2012). Insights into mitochondrial dysfunction: aging, amyloid-β and τ – a deleterious trio. Antioxid. Redox Signal 16, 1456–1466.10.1089/ars.2011.4400Search in Google Scholar PubMed

Schouten, M., Buijink, M.R., Lucassen, P.J., and Fitzsimons, C.P. (2012). New neurons in aging brains: molecular control by small non-coding RNAs. Front. Neurosci. 6, 25.10.3389/fnins.2012.00025Search in Google Scholar PubMed PubMed Central

Schultz, M.B. and Sinclair, D.A. (2016). When stem cells grow old: phenotypes and mechanisms of stem cell aging. Development 143, 3–14.10.1242/dev.130633Search in Google Scholar PubMed PubMed Central

Skulachev, M.V., Severin, F.F., and Skulachev, V.P. (2014). Receptor mediated regulation of senile phenoptosis, Biochemistry Mosc. 79, 994–1003.10.1134/S0006297914100022Search in Google Scholar PubMed

Smith, D.E., Rapp, P.R., McKay, H.M., Roberts, J.A., and Tuszynski, M.H. (2004). Memory impairment in aged primates is associated with focal death of cortical neurons and atrophy of subcortical neurons. J. Neurosci. 24, 4373–4381.10.1523/JNEUROSCI.4289-03.2004Search in Google Scholar PubMed PubMed Central

Snyder, J.S., Hong, N.S., McDonald, R.J., and Wojtowicz, J.M. (2005). A role for adult neurogenesis in spatial long-term memory. Neuroscience 130, 843–852.10.1016/j.neuroscience.2004.10.009Search in Google Scholar PubMed

Suhonen, J.O., Peterson, D.A., Ray, J., and Gage, F.H. (1996). Differentiation of adult hippocampus-derived progenitors into olfactory neurons in vivo. Nature 383, 624–627.10.1038/383624a0Search in Google Scholar PubMed

Sun, P., Knezovic, A., Parlak, M., Cuber, J., Karabeg, M.M., Deckert, J., Riederer, P., Hua, Q., Salkovic-Petrisic, M., and Schmitt, A.G. (2015). Long-term effects of intracerebroventricular streptozotocin treatment on adult neurogenesis in the rat hippocampus. Curr. Alzheimer Res. 12, 772–784.10.2174/1567205012666150710112147Search in Google Scholar PubMed

Tincer, G., Mashkaryan, V., Bhattarai, P., and Kizil, C. (2016). Neural stem/progenitor cells in Alzheimer’s disease. Yale J. Biol. Med. 89, 23–35.Search in Google Scholar

Tonchev, A.B., Yamashima, T., and Chaldakov, G.N. (2007). Distribution and phenotype of proliferating cells in the forebrain of adult macaque monkeys after transient global cerebral ischemia. Adv. Anat. Embryol. Cell Biol. 191, 1–106.10.1007/978-3-540-39617-8Search in Google Scholar

Ullrich, N.J. and Gordon, L.B. (2015). Hutchinson-Gilford progeria syndrome. Handb. Clin. Neurol. 132, 249–264.10.1016/B978-0-444-62702-5.00018-4Search in Google Scholar PubMed

Yamashima, T., Tonchev, A.B., and Yukie, M. (2007). Adult hippocampal neurogenesis in rodents and primates: endogenous, enhanced, and engrafted. Rev. Neurosci. 18, 67–82.10.1515/REVNEURO.2007.18.1.67Search in Google Scholar

Yousef, H., Czupalla, C.J., Lee, D., Burke, A., Chen, M., Zandstra, J., Berber, E., Lehallier, B., Mathur, V., Nair, R.V., et al. (2018). Aged blood inhibits hippocampal neurogenesis and activates microglia through VCAM1 at the blood-brain barrier. bioRxiv Doi: 10.1101/242198.10.1101/242198Search in Google Scholar

Zhao, B., Zhong, M., and Jin, K. (2008). Neurogenesis and neurodegenerative diseases in human. Panminerva Med. 50, 55–64.Search in Google Scholar

Received: 2018-08-14
Accepted: 2018-11-18
Published Online: 2019-02-14
Published in Print: 2019-07-26

©2019 Walter de Gruyter GmbH, Berlin/Boston

Downloaded on 20.4.2024 from https://www.degruyter.com/document/doi/10.1515/revneuro-2018-0084/html
Scroll to top button