Elsevier

Neurobiology of Aging

Volume 33, Issue 8, August 2012, Pages 1493-1506
Neurobiology of Aging

Regular paper
Allopregnanolone restores hippocampal-dependent learning and memory and neural progenitor survival in aging 3xTgAD and nonTg mice

https://doi.org/10.1016/j.neurobiolaging.2011.06.008Get rights and content

Abstract

We previously demonstrated that allopregnanolone (APα) increased proliferation of neural progenitor cells and reversed neurogenic and cognitive deficits prior to Alzheimer's disease (AD) pathology (Wang, J.M., Johnston, P.B., Ball, B.G., Brinton, R.D., 2005. The neurosteroid allopregnanolone promotes proliferation of rodent and human neural progenitor cells and regulates cell-cycle gene and protein expression. J. Neurosci. 25, 4706–4718; Wang, J.M., Singh, C., Liu, L., Irwin, R.W., Chen, S., Chung, E.J., Thompson, R.F., Brinton, R.D., 2010. Allopregnanolone reverses neurogenic and cognitive deficits in mouse model of Alzheimer's disease. Proc. Natl. Acad. Sci. U. S. A. 107, 6498–6503). Herein, we determined efficacy of APα to restore neural progenitor cell survival and associative learning and memory subsequent to AD pathology in male 3xTgAD mice and their nontransgenic (nonTg) counterparts. APα significantly increased survival of bromodeoxyuridine positive (BrdU+) cells and hippocampal-dependent associative learning and memory in 3xTgAD mice in the presence of intraneuronal amyloid beta (Aβ) whereas APα was ineffective subsequent to development of extraneuronal Aβ plaques. Restoration of hippocampal-dependent associative learning was maximal by the first day and sustained throughout behavioral training. Learning and memory function in APα-treated 3xTgAD mice was 100% greater than vehicle-treated and comparable to maximal normal nonTg performance. In aged 15-month-old nonTg mice, APα significantly increased survival of bromodeoxyuridine-positive cells and hippocampal-dependent associative learning and memory. Results provide preclinical evidence that APα promoted survival of newly generated cells and restored cognitive performance in the preplaque phase of AD pathology and in late-stage normal aging.

Introduction

Generation of new neurons is well documented to occur throughout adulthood across various species, including rodents (Ma et al., 2009, Zhao et al., 2008) and humans (Eriksson et al., 1998). In most mammals, adult neurogenesis is restricted to the subgranular zone (SGZ) of the dentate gyrus in the hippocampus (Aimone et al., 2010) and subventricular zone (SVZ) of the lateral ventricle (Alvarez-Buylla et al., 2008). Both SGZ and subventricular zone neurogenesis have been shown to play a significant role in various forms of learning and memory (Bath et al., 2008, Kim et al., 2007, Lazarini et al., 2009, Saxe et al., 2006, Waddell and Shors, 2008, Wang et al., 2010). While the regenerative potential of the mammalian brain is sustained throughout the life span, the magnitude of the proliferative efficacy of neural progenitors declines with age and diseases, such as Alzheimer's disease (AD) (Hattiangady and Shetty, 2008, Hattiangady et al., 2007, Kuhn et al., 1996, Lazarov et al., 2010, Rao et al., 2005). Age- and AD-associated declines in hippocampal neurogenesis have been observed in multiple mouse models of AD along with a concomitant decline in cognitive function (Ermini et al., 2008, Niidome et al., 2008, Rodríguez et al., 2008, Rodríguez et al., 2009, Taniuchi et al., 2007, Verret et al., 2007, Zhang et al., 2007). Recent studies have shown that impaired neurogenesis is an early event in the etiology of mouse models of familial AD (Demars et al., 2010, Wang et al., 2010). Malfunctioning presenilin-1, misprocessing of amyloid precursor protein and toxic effects of hyperphosphorylated tau and amyloid beta (Aβ) could be contributing to impaired neurogenesis in AD models.

Neural stem cells have been proposed to be a therapeutic strategy to restore brain function in neurodegenerative disease. Several studies have shown restoration of cognitive function following neural stem cell transplants (Blurton-Jones et al., 2009, Ebert et al., 2008, Lu et al., 2003, Park et al., 2006). Ablated regions can be repopulated by neural stem cells under optimal conditions. Collectively, these data provide preclinical evidence for the potential of neural progenitor cells as a therapeutic avenue to regenerate neuronal circuits and to restore neurological function. While the transplant strategy shows efficacy, the clinical utility of this approach is limited. An alternative approach is to promote the intrinsic regenerative capacity of the brain. The challenge in this approach is the neurochemical and neuropathological milieu of the diseased brain. Changes in local biochemical milieu, including growth factors, cytokines, neurotransmitters, and neurosteroids can contribute to impaired neurogenesis (Brinton, 2009, Brinton et al., 2008, Demars et al., 2010, Wang et al., 2010). Reversing the decline in adult neurogenesis using regenerative factors could be a therapeutic strategy to reverse disease- and age-associated cognitive decline.

Neurosteroids are a class of therapeutic agents with blood-brain barrier-penetrating physicochemical properties (Brinton and Wang, 2006). One such neurosteroid is allopregnanolone (APα), a metabolite of progesterone, which is synthesized de novo in both embryonic and adult central nervous system (CNS) (Baulieu et al., 2001, Mellon and Griffin, 2002a, Mellon and Griffin, 2002b) as well as in pluripotent progenitor cells (Gago et al., 2004, Lauber and Lichtensteiger, 1996) and which exhibits an age- and AD-associated decline (Bernardi et al., 1998, Brinton et al., 2008, Marx et al., 2006, Weill-Engerer et al., 2002). We have recently shown that the triple-transgenic mouse model of Alzheimer's disease (3xTgAD), overexpressing APPSWE, PS1M146V, and tauP301L (Oddo et al., 2003b), has both neurogenic and cognitive deficits beginning at 3 months of age (Wang et al., 2010). Despite the lack of evident AD pathology, basal level of newborn cells in the SGZ of the hippocampus was significantly lower in these mice as compared with the age-matched nontransgenic (nonTg) mice. The concentration of APα was found to be significantly reduced within the brain and serum of 3xTgAD mice as compared with nonTg mice, similar to that observed in human AD Patients (Weill-Engerer et al., 2002). Furthermore, severe cognitive deficits were observed in 3-month-old 3xTgAD male mice as compared with nonTg mice in a hippocampal-dependent associative task. Treatment with APα reversed this cognitive deficit and restored learning and memory performance to the level of normal nonTg mice. APα also induced a significant increase in survival of neural progenitor cells, significantly correlated with APα-induced enhanced memory performance.

3xTgAD mice manifest age-dependent neuropathology of AD in the form of both Aβ plaques and neurofibrillary tangles along with age-dependent learning and memory deficits (Billings et al., 2005, Oddo et al., 2003a, Oddo et al., 2003b). The current study was directed at determining the efficacy of APα to restore associative learning and memory function and to promote neural progenitor generation and survival in the aged 3xTgAD mice at varying degrees of Aβ burden, from intraneuronal accumulation to extraneuronal plaque formation. Parallel analyses were conducted in the age-matched nonTg male mice to investigate the neurogenic and cognitive deficits that accompany aging in normal mice and probe the efficacy of APα to reverse those deficits. Results of our current analyses are further indicative of the potential of APα as a regenerative therapeutic to prevent or delay neurogenic and cognitive deficits associated with mild cognitive impairment, Alzheimer's disease, and late-stage normal aging.

Section snippets

Chemicals

Allopregnanolone (APα; 3α-hydroxy-5α-pregnan-20-1) (also known as AP, Allo, or THP) used for this study was purchased from Steraloids, Inc. (Newport, RI, USA). Chemicals were from MP Biomed (Irvine, CA, USA) unless otherwise noted.

Animals and treatment

Breeding pairs of the triple transgenic Alzheimer's disease mouse (3xTgAD, homozygous mutant of human APPSwe, PS1M146v, and tauP301L) and its background strain (129/Sv × C57BL/6) were obtained from Dr. Frank LaFerla (University of California, Irvine) and the colonies

Rationale for and design of APα treatment experiments

Our previous analyses demonstrated that APα reversed cognitive and neurogenic deficits of male 3xTgAD mice (3-month-old) prior to the onset of AD pathology (Wang et al., 2010). The current study was designed to determine the efficacy of APα to promote cognitive and neurogenic function in 3xTgAD male mice with mild (6-month-old), moderate (9-month-old), and severe (12-month-old) Aβ accumulation. Cognitive performance was assessed using the trace eyeblink conditioning paradigm (Fig. 1A) which

Discussion

Using a triple transgenic mouse model of Alzheimer's disease (3xTgAD) and its background strain control (nonTg), the data indicate that aging and Alzheimer's disease burden diminishes hippocampal neurogenesis and associated cognitive performance. These effects on hippocampal neurogenesis were differentially regulated by aging and AD pathology, with AD pathology affecting cell survival and hippocampal-dependent associative learning and memory more severely than normal aging. APα reversed the

Disclosure statement

Patents pending on allopregnanolone as a therapeutic for mild cognitive impairment and Alzheimer's disease.

All experiments strictly conformed to the Animal Welfare Act, Guide to Use and Care of Laboratory Animals, and the U.S. Government Principles of the Utilization and Care of Vertebrate Animals Used in Testing, Research, and Training guidelines on the ethical use of animals.

Acknowledgements

This research was supported by grants from by the National Institute on Aging U01AG031115, the Alzheimer Drug Discovery Foundation, and the Kenneth T. and Eileen L. Norris Foundation to RDB.

References (71)

  • B. Hattiangady et al.

    Brain-derived neurotrophic factor, phosphorylated cyclic AMP response element binding protein and neuropeptide Y decline as early as middle age in the dentate gyrus and CA1 and CA3 subfields of the hippocampus

    Exp. Neurol

    (2005)
  • B. Hattiangady et al.

    Aging does not alter the number or phenotype of putative stem/progenitor cells in the neurogenic region of the hippocampus

    Neurobiol. Aging

    (2008)
  • O. Lazarov et al.

    When neurogenesis encounters aging and disease

    Trends Neurosci

    (2010)
  • P. Lu et al.

    Neural stem cells constitutively secrete neurotrophic factors and promote extensive host axonal growth after spinal cord injury

    Exp. Neurol

    (2003)
  • C.E. Marx et al.

    The neurosteroid allopregnanolone is reduced in prefrontal cortex in Alzheimer's disease

    Biol. Psychiatry

    (2006)
  • S.H. Mellon et al.

    Neurosteroids: biochemistry and clinical significance

    Trends Endocrinol. Metab

    (2002)
  • J.C. Naylor et al.

    Allopregnanolone levels are reduced in temporal cortex in patients with Alzheimer's disease compared to cognitively intact control subjects

    Biochim. Biophys. Acta

    (2010)
  • S. Oddo et al.

    Amyloid deposition precedes tangle formation in a triple transgenic model of Alzheimer's disease

    Neurobiol. Aging

    (2003)
  • S. Oddo et al.

    Triple-transgenic model of Alzheimer's disease with plaques and tangles: intracellular Abeta and synaptic dysfunction

    Neuron

    (2003)
  • K.I. Park et al.

    Neural stem cells may be uniquely suited for combined gene therapy and cell replacement: Evidence from engraftment of Neurotrophin-3-expressing stem cells in hypoxic-ischemic brain injury

    Exp. Neurol

    (2006)
  • M.S. Rao et al.

    Fetal hippocampal CA3 cell grafts enriched with FGF-2 and BDNF exhibit robust long-term survival and integration and suppress aberrant mossy fiber sprouting in the injured middle-aged hippocampus

    Neurobiol. Dis

    (2006)
  • I. Sayeed et al.

    Allopregnanolone, a progesterone metabolite, is more effective than progesterone in reducing cortical infarct volume after transient middle cerebral artery occlusion

    Ann. Emerg. Med

    (2006)
  • I. Sayeed et al.

    Direct inhibition of the mitochondrial permeability transition pore: a possible mechanism for better neuroprotective effects of allopregnanolone over progesterone

    Brain Res

    (2009)
  • P. Taupin et al.

    FGF-2-responsive neural stem cell proliferation requires CCg, a novel autocrine/paracrine cofactor

    Neuron

    (2000)
  • C. Zhang et al.

    Long-lasting impairment in hippocampal neurogenesis associated with amyloid deposition in a knock-in mouse model of familial Alzheimer's disease

    Exp. Neurol

    (2007)
  • C. Zhao et al.

    Mechanisms and functional implications of adult neurogenesis

    Cell

    (2008)
  • A. Alvarez-Buylla et al.

    The heterogeneity of adult neural stem cells and the emerging complexity of their niche

    Cold Spring Harb Symp. Quant. Biol

    (2008)
  • K.G. Bath et al.

    Variant brain-derived neurotrophic factor (Val66Met) alters adult olfactory bulb neurogenesis and spontaneous olfactory discrimination

    J. Neurosci

    (2008)
  • F. Bernardi et al.

    Aging is associated with changes in allopregnanolone concentrations in brain, endocrine glands and serum in male rats

    Eur. J. Endocrinol

    (1998)
  • L.M. Billings et al.

    Learning decreases A beta*56 and tau pathology and ameliorates behavioral decline in 3xTg-AD mice

    J. Neurosci

    (2007)
  • M. Blurton-Jones et al.

    Neural stem cells improve cognition via BDNF in a transgenic model of Alzheimer disease

    Proc. Natl. Acad. Sci. U. S. A.

    (2009)
  • R.D. Brinton et al.

    Therapeutic potential of neurogenesis for prevention and recovery from Alzheimer's disease: allopregnanolone as a proof of concept neurogenic agent

    Curr. Alzheimer Res

    (2006)
  • S. Chen et al.

    Chronic administration of allopregnanolone modifies progression of Alzheimer's pathology in the triple transgenic Alzheimer's disease mouse

    Society for Neuroscience

    (2008)
  • M. Demars et al.

    Impaired neurogenesis is an early event in the etiology of familial Alzheimer's disease in transgenic mice

    J. Neurosci. Res

    (2010)
  • M. Djebaili et al.

    The neurosteroids progesterone and allopregnanolone reduce cell death, gliosis, and functional deficits after traumatic brain injury in rats

    J. Neurotrauma

    (2005)
  • Cited by (0)

    View full text