Elsevier

Experimental Neurology

Volume 231, Issue 1, September 2011, Pages 72-81
Experimental Neurology

Progesterone treatment normalizes the levels of cell proliferation and cell death in the dentate gyrus of the hippocampus after traumatic brain injury

https://doi.org/10.1016/j.expneurol.2011.05.016Get rights and content

Abstract

Traumatic brain injury (TBI) increases cell death in the hippocampus and impairs hippocampus-dependent cognition. The hippocampus is also the site of ongoing neurogenesis throughout the lifespan. Progesterone treatment improves behavioral recovery and reduces inflammation, apoptosis, lesion volume, and edema, when given after TBI. The aim of the present study was to determine whether progesterone altered cell proliferation and short-term survival in the dentate gyrus after TBI. Male Sprague–Dawley rats with bilateral contusions of the frontal cortex or sham operations received progesterone or vehicle at 1 and 6 h post-surgery and daily through post-surgery Day 7, and a single injection of bromodeoxyuridine (BrdU) 48 h after injury. Brains were then processed for Ki67 (endogenous marker of cell proliferation), BrdU (short-term cell survival), doublecortin (endogenous marker of immature neurons), and Fluoro-Jade B (marker of degenerating neurons). TBI increased cell proliferation compared to shams and progesterone normalized cell proliferation in injured rats. Progesterone alone increased cell proliferation in intact rats. Interestingly, injury and/or progesterone treatment did not influence short-term cell survival of BrdU-ir cells. All treatments increased the percentage of BrdU-ir cells that were co-labeled with doublecortin (an immature neuronal marker in this case labeling new neurons that survived 5 days), indicating that cell fate is influenced independently by TBI and progesterone treatment. The number of immature neurons that survived 5 days was increased following TBI, but progesterone treatment reduced this effect. Furthermore, TBI increased cell death and progesterone treatment reduced cell death to levels seen in intact rats. Together these findings suggest that progesterone treatment after TBI normalizes the levels of cell proliferation and cell death in the dentate gyrus of the hippocampus.

Research highlights

► Progesterone improves outcome following traumatic brain injury in humans and rodents. ► Traumatic brain injury increased cell proliferation and cell death in hippocampus. ► Progesterone normalized the injury-induced increase in cell proliferation and death. ► Improved cognitive outcome could be related to progesterone effects on neurogenesis.

Introduction

Progesterone, a naturally occurring steroid hormone, is present in both male and female brains. Its actions in the central nervous system are not limited to the control of neuroendocrine regulation and reproduction (Baulieu and Schumacher, 2000, Birzniece et al., 2006); it influences several neural survival functions including neuronal and glial differentiation, hippocampal neurogenesis, and synaptic stability (Ghoumari et al., 2005, Smith et al., 1987, Tsutsui et al., 2004, Zhang et al., 2010a, Zhang et al., 2010b). There are now more than 160 publications showing that progesterone is an effective neuroprotective agent against a number of different insults including blunt and penetrating traumatic brain injury (TBI) (De Nicola et al., 2009, Garcia-Estrada et al., 1999, Labombarda et al., 2010, Schumacher et al., 2007, Stein, 2007, Stein and Sayeed, 2010, Stein and Wright, 2010), diffuse TBI (O'Connor et al., 2007), stroke (Ishrat et al., 2010, Sayeed and Stein, 2009), anoxic brain injury, glutamate toxicity (Ogata et al., 1993), and spinal cord injury (Labombarda et al., 2010) in both males and females. Progesterone exerts its beneficial effects in the central nervous system via multiple pathways that are not always dependent on the activation of the classical intranuclear progesterone receptor (Bottino et al., 2010, Falkenstein et al., 2000, Losel et al., 2003). These effects include, among others, protecting the blood–brain barrier, reducing cerebral edema, decreasing the inflammatory cascade, and decreasing cellular necrosis and apoptosis (Djebaili et al., 2005, Gibson et al., 2005, He et al., 2004, Ishrat et al., 2010, Majewska, 1992, Mani, 2006, Roof et al., 1997, Smith and Woolley, 2004).

Treatment with progesterone following TBI also improves functional outcome. For example, cognitive deficits, a hallmark of TBI, are attenuated following progesterone treatment (Roof et al., 1994). Systemic treatment with progesterone facilitates recovery of spatial learning and memory, which involves the hippocampus, in brain-injured male rats compared to placebo-treated male rats (Djebaili et al., 2004, Roof et al., 1994, Shear et al., 2002). The neuroprotective properties of progesterone in response to TBI are also seen in humans. Two recently completed phase IIa single-center clinical trials found a beneficial effect of progesterone as a treatment in moderate to severe TBI in male and female adults, with fewer mortalities and better functional outcome seen in progesterone-treated patients (Wright et al., 2007, Xiao et al., 2008). These encouraging findings have led to two phase III double-blind randomized clinical trials now enrolling patients (ProTECT III: NCT00822900; SyNAPSe: NCT01143064).

Hippocampal neurons may be the most vulnerable to TBI-induced degeneration and are among the first population of neurons in the brain to die following experimental TBI (Ariza et al., 2006, Kotapka et al., 1991). The dramatic upregulation in apoptotic and necrotic cell death following TBI can contribute to cognitive deficits, and drugs that prevent cell death also reduce cognitive impairment (Sinson et al., 1997).

Progenitor cells in the dentate gyrus of the hippocampus retain the ability to proliferate into neurons during adulthood in most mammalian species studied including humans (Eriksson et al., 1998, Gould et al., 1997, Gould et al., 1999, Gould et al., 2001, Huang and Sato, 1998, Lavenex et al., 2000). Hippocampal neurogenesis consists of at least four processes: cell proliferation, cell migration, cell differentiation, and cell survival. Cell proliferation in the dentate gyrus is the production of new cells from the division of progenitor cells within the subgranular zone. Daughter cells migrate from the subgranular zone to the inner layers of the granule cell layer while undergoing a process of fate determination, differentiating into either neurons or glial cells (Brown et al., 2003, Cameron et al., 1993). These new cells continue to mature over a period of weeks to months resulting in new functionally mature neurons (Esposito et al., 2005, van Praag et al., 2002). Cell survival is the number of new cells that survive to maturity with the majority of these new cells becoming neurons (Cameron et al., 1993).

In addition to its effects on cell death and hippocampus-dependent learning and memory, TBI also affects different aspects of adult hippocampal neurogenesis (Chirumamilla et al., 2002, Gao et al., 2009, Lu et al., 2005, Richardson et al., 2007). Different experimental models of TBI, including lateral fluid percussion and controlled cortical impact (CCI) injury, induce a 3- to 4-fold increase in the number of BrdU-ir cells in the dentate gyrus as early as 2 days postinjury that peaks within the first 7 days following injury (Dash et al., 2001, Emery et al., 2005, Kernie et al., 2001, Lu et al., 2003, Lu et al., 2005, Rice et al., 2003, Sun et al., 2005, Yoshimura et al., 2003) and returns to baseline levels by 35 days post-injury (Kleindienst et al., 2005). However it is important to note that in the majority of these studies cell proliferation was not independently assessed from cell survival as multiple BrdU injections were given and thus the number of BrdU-ir cells were an index of both proliferating cells and the short-term survival of newly-produced cells. The capacity of TBI to increase cell proliferation and subsequent neurogenesis in the dentate gyrus may not necessarily be beneficial and could actually contribute to injury-induced cognitive deficits. Some authors have suggested that too much or too little neurogenesis may interfere with proper functioning of the hippocampus, as predicted by computer modeling (Butz et al., 2008). Indeed, prolonged and continuous seizures lead to an increase in neurogenesis which can then directly contribute to cognitive decline seen after the seizures (Jessberger et al., 2007). Here we offer the hypothesis that the long-term beneficial effects of progesterone on cognitive outcome after TBI are associated with progesterone's influence on modulating potentially pathological levels of neurogenesis.

The aim of the present study was to determine the effects of progesterone treatment on cell death, cell proliferation and immature neuron survival in the hippocampus after TBI. We tested the idea that TBI would increase cell proliferation, the survival of immature neurons and cell death and that progesterone treatment would reduce this TBI-induced augmentation.

Section snippets

Animals

A total of twenty-four adult male Sprague–Dawley rats (8 to 12 weeks; Charles River Laboratories, Wilmington, MA) weighing 275–300 g were used according to procedures approved by the Institutional Animal Care and Use Committee, Emory University, Atlanta, GA, USA (protocol 164–2008). The rats were handled for at least 5 days before surgery, and were individually housed. Food and water were provided ad libitum throughout the experiment and the animals lived in a reversed 12-h light/12-h dark cycle

Brain injury increases cell proliferation compared to sham operations. Progesterone decreases cell proliferation in TBI rats but increases cell proliferation in intact rats

The total number of Ki67-ir cells in the granule cell layer and the hilus for all groups are shown in Fig. 3A. A repeated-measures ANOVA conducted on the total number of Ki67-ir cells in the granule cell layer and the hilus showed a significant interaction between group and region [F(3, 16) = 8.05, p < 0.002]. Post-hoc analysis revealed that the sham group had significantly fewer total Ki67-ir cells in the granule cell layer compared to the sham + progesterone group (p < 0.004) and the TBI group (p < 

Discussion

The results from the present study demonstrate that TBI increased cell proliferation and cell death in the dentate gyrus of the adult male rat, while progesterone treatment normalized the injury-induced levels of both cell proliferation and cell death to sham levels. Progesterone's ability to influence cell proliferation was dependent on whether it was administered to a healthy or injured brain. In the present study neither injury nor progesterone treatment influenced short-term cell survival

Conclusions

Severe TBI is often very debilitating and leads to alterations in hippocampal function and structure. Progesterone is an effective neuroprotective agent that improves functional outcome following TBI. The results of our study extend the evidence for the beneficial effects of progesterone following TBI, and show that progesterone administration following contusion reduces the contusion-induced increases in cell proliferation, number of immature neurons, and cell death in the dentate gyrus of

Acknowledgments

We would like to thank Stephanie Lieblich for assistance with this work. Financial support for this research was provided by the Canadian Institutes of Health Research (LAMG) and U01-NSO-62676-02A2 (DGS). CKB and JRE are funded by Michael Smith Foundation for Medical Research senior trainee awards. LAMG is a Michael Smith Senior Scholar. DGS receives research support from BHR Pharma, LLC and Allen & Company.

References (130)

  • A.F. De Nicola et al.

    Progesterone neuroprotection in traumatic CNS injury and motoneuron degeneration

    Front. Neuroendocrinol.

    (2009)
  • M. Djebaili et al.

    Allopregnanolone and progesterone decrease cell death and cognitive deficits after a contusion of the rat pre-frontal cortex

    Neuroscience

    (2004)
  • J.R. Epp et al.

    Hippocampus-dependent strategy choice predicts low levels of cell proliferation in the dentate gyrus

    Neurobiol. Learn. Mem.

    (2009)
  • J.R. Epp et al.

    Strain differences in neurogenesis and activation of new neurons in the dentate gyrus in response to spatial learning

    Neuroscience

    (2011)
  • S.A. Farr et al.

    Effect of ovarian steroids on footshock avoidance learning and retention in female mice

    Physiol. Behav.

    (1995)
  • M.R. Foy et al.

    Regulation of hippocampal synaptic plasticity by estrogen and progesterone

    Vitam. Horm.

    (2010)
  • X. Gao et al.

    Moderate traumatic brain injury promotes proliferation of quiescent neural progenitors in the adult hippocampus

    Exp. Neurol.

    (2009)
  • J. Garcia-Estrada et al.

    Dehydroepiandrosterone, pregnenolone and sex steroids down-regulate reactive astroglia in the male rat brain after a penetrating brain injury

    Int. J. Dev. Neurosci.

    (1999)
  • A.M. Ghoumari et al.

    Progesterone increases oligodendroglial cell proliferation in rat cerebellar slice cultures

    Neuroscience

    (2005)
  • C.L. Gibson et al.

    Progesterone suppresses the inflammatory response and nitric oxide synthase-2 expression following cerebral ischemia

    Exp. Neurol.

    (2005)
  • R. Guennoun et al.

    The membrane-associated progesterone-binding protein 25-Dx: expression, cellular localization and up-regulation after brain and spinal cord injuries

    Brain Res. Rev.

    (2008)
  • L.L. Harburger et al.

    Post-training progesterone dose-dependently enhances object, but not spatial, memory consolidation

    Behav. Brain Res.

    (2008)
  • J. He et al.

    Progesterone and allopregnanolone reduce inflammatory cytokines after traumatic brain injury

    Exp. Neurol.

    (2004)
  • S. Huang et al.

    Progenitor cells in the adult zebrafish nervous system express a Brn-1-related POU gene, tai-ji

    Mech. Dev.

    (1998)
  • T. Ishrat et al.

    Effects of progesterone administration on infarct volume and functional deficits following permanent focal cerebral ischemia in rats

    Brain Res.

    (2009)
  • T. Ishrat et al.

    Progesterone and allopregnanolone attenuate blood–brain barrier dysfunction following permanent focal ischemia by regulating the expression of matrix metalloproteinases

    Exp. Neurol.

    (2010)
  • I.M. Johansson et al.

    Allopregnanolone inhibits learning in the Morris water maze

    Brain Res.

    (2002)
  • N. Kee et al.

    The utility of Ki-67 and BrdU as proliferative markers of adult neurogenesis

    J. Neurosci. Methods

    (2002)
  • J.J. Lacapere et al.

    Peripheral-type benzodiazepine receptor: structure and function of a cholesterol-binding protein in steroid and bile acid biosynthesis

    Steroids

    (2003)
  • S.K. Mani

    Signaling mechanisms in progesterone-neurotransmitter interactions

    Neuroscience

    (2006)
  • D. Meffre et al.

    3beta-Hydroxysteroid dehydrogenase/5-ene-4-ene isomerase mRNA expression in rat brain: effect of pseudopregnancy and traumatic brain injury

    J. Steroid Biochem. Mol. Biol.

    (2007)
  • C.A. O'Connor et al.

    Effects of progesterone on neurologic and morphologic outcome following diffuse traumatic brain injury in rats

    Exp. Neurol.

    (2007)
  • T. Ogata et al.

    Steroid hormones protect spinal cord neurons from glutamate toxicity

    Neuroscience

    (1993)
  • B.K. Ormerod et al.

    Reproductive status influences cell proliferation and cell survival in the dentate gyrus of adult female meadow voles: a possible regulatory role for estradiol

    Neuroscience

    (2001)
  • B.K. Ormerod et al.

    Estradiol enhances neurogenesis in the dentate gyri of adult male meadow voles by increasing the survival of young granule neurons

    Neuroscience

    (2004)
  • P.T. Orr et al.

    Dorsal hippocampal progesterone infusions enhance object recognition in young female mice

    Pharmacol. Biochem. Behav.

    (2009)
  • V. Papadopoulos et al.

    Peripheral benzodiazepine receptor in cholesterol transport and steroidogenesis

    Steroids

    (1997)
  • A. Reyna-Neyra et al.

    Estradiol and progesterone modify microtubule associated protein 2 content in the rat hippocampus

    Brain Res. Bull.

    (2002)
  • A.C. Rice et al.

    Proliferation and neuronal differentiation of mitotically active cells following traumatic brain injury

    Exp. Neurol.

    (2003)
  • R.M. Richardson et al.

    Neurogenesis after traumatic brain injury

    Neurosurg. Clin. N. Am.

    (2007)
  • R. Rola et al.

    Alterations in hippocampal neurogenesis following traumatic brain injury in mice

    Exp. Neurol.

    (2006)
  • R.L. Roof et al.

    Progesterone facilitates cognitive recovery and reduces secondary neuronal loss caused by cortical contusion injury in male rats

    Exp. Neurol.

    (1994)
  • R. Rupprecht et al.

    Progesterone receptor-mediated effects of neuroactive steroids

    Neuron

    (1993)
  • I. Sayeed et al.

    Progesterone as a neuroprotective factor in traumatic and ischemic brain injury

    Prog. Brain Res.

    (2009)
  • Barha, C.K., Galea, L.A., in press. Motherhood alters the cellular response to estrogens in the hippocampus later in...
  • C.K. Barha et al.

    Regulation of adult hippocampal neurogenesis in the mammalian brain

  • C.K. Barha et al.

    Different forms of oestrogen rapidly upregulate cell proliferation in the dentate gyrus of adult female rats

    J. Neuroendocrinol.

    (2009)
  • Barha, C.K., Brummelte, S., Lieblich, S.E., Galea, L.A., in press. Chronic restraint stress in adolescence...
  • E.E. Baulieu et al.

    Progesterone as a neuroactive neurosteroid, with special reference to the effect of progesterone on myelination

    Hum. Reprod.

    (2000)
  • H.A. Bimonte-Nelson et al.

    Ovarian hormones and cognition in the aged female rat: II. progesterone supplementation reverses the cognitive enhancing effects of ovariectomy

    Behav. Neurosci.

    (2004)
  • Cited by (94)

    • Cellular regeneration treatments for traumatic brain injury

      2022, Medicine in Novel Technology and Devices
    • Molecular mechanisms of sex hormones in the development and progression of Alzheimer's disease

      2021, Neuroscience Letters
      Citation Excerpt :

      Progesterone activates various pathways in the nervous system that can perform different tasks. Progesterone can affect the proliferation, survival, and death of the cells using MAPK [6]. Progesterone can also activate the ERK pathway to protect nerve cells with estradiol help [29,30].

    • Sexual dimorphism in pre-clinical studies of depression

      2021, Progress in Neuro-Psychopharmacology and Biological Psychiatry
    View all citing articles on Scopus
    View full text