Elsevier

Neurobiology of Disease

Volume 95, November 2016, Pages 102-110
Neurobiology of Disease

Nicotinamide mononucleotide inhibits post-ischemic NAD+ degradation and dramatically ameliorates brain damage following global cerebral ischemia

https://doi.org/10.1016/j.nbd.2016.07.018Get rights and content

Highlights

  • Effect of treatment with nicotinamide mononucleotide on forebrain ischemia is examined.

  • Nicotinamide mononucleotide administration ameliorates both histologic and neurologic outcome.

  • Treated animals show normalized post-ischemic NAD levels and inhibition of poly-ADP-ribosylation.

Abstract

Nicotinamide adenine dinucleotide (NAD+) is an essential cofactor for multiple cellular metabolic reactions and has a central role in energy production. Brain ischemia depletes NAD+ pools leading to bioenergetics failure and cell death. Nicotinamide mononucleotide (NMN) is utilized by the NAD+ salvage pathway enzyme, nicotinamide adenylyltransferase (Nmnat) to generate NAD+. Therefore, we examined whether NMN could protect against ischemic brain damage. Mice were subjected to transient forebrain ischemia and treated with NMN or vehicle at the start of reperfusion or 30 min after the ischemic insult. At 2, 4, and 24 h of recovery, the proteins poly-ADP-ribosylation (PAR), hippocampal NAD+ levels, and expression levels of NAD+ salvage pathway enzymes were determined. Furthermore, animal's neurologic outcome and hippocampal CA1 neuronal death was assessed after six days of reperfusion. NMN (62.5 mg/kg) dramatically ameliorated the hippocampal CA1 injury and significantly improved the neurological outcome. Additionally, the post-ischemic NMN treatment prevented the increase in PAR formation and NAD+ catabolism. Since the NMN administration did not affect animal's temperature, blood gases or regional cerebral blood flow during recovery, the protective effect was not a result of altered reperfusion conditions. These data suggest that administration of NMN at a proper dosage has a strong protective effect against ischemic brain injury.

Introduction

One of the pathologic outcomes after ischemic insult is the free radical induced DNA damage that activates the nuclear enzyme poly(ADP-ribose) polymerase 1 (PARP1) (Endres et al., 1997, Strosznajder et al., 2003), for review see (Chiarugi, 2005). This enzyme utilizes NAD+ as a substrate to form the poly-ADP-ribose (PAR) polymer. It has been proposed that uncontrolled PARP1 activation can deplete intracellular NAD+ and consequently ATP, leading to cell death (Lo et al., 1998, Szabo and Dawson, 1998). NAD+ is an important cofactor involved in multiple metabolic reactions (Brennan et al., 2006). NAD+ and NADH have central roles in cellular energy production as electron-accepting and electron-donating cofactors. Therefore, maintenance of normal cellular NAD+ levels is essential for tissue bioenergetic metabolism and several cell functions. A prominent role for NAD+ catabolism in cell death mechanisms is supported by the observation that following excitotoxic insult or in vivo models of brain ischemia, epilepsy and Alzheimer's disease, a significant decrease in total cellular NAD+ levels occurs prior to neuronal death (Endres et al., 1997, Liu et al., 2009).

NAD+ can be generated in cells either by de novo synthesis from tryptophan or it is re-synthesized from nicotinamide (Nam) via a salvage pathway (Belenky et al., 2007, Owens et al., 2013b). Majority of the NAD+ is replenished by the salvage pathway since Nam is the byproduct of the NAD+ catabolizing enzymes (Magni et al., 1999, Imai, 2009, Owens et al., 2013b). The salvage pathway represents two enzymatic reactions. In the first step, Nam is converted to nicotinamide mononucleotide (NMN) by nicotinamide phosphoribosyltransferase (Nampt) (Revollo et al., 2004), also known as pre-B-cell colony-enhancing factor (PBEF). NMN is then adenylylated to form NAD+ by nicotinamide nucleotide adenylyltransferase (Nmnat) (Belenky et al., 2007).

To maintain cellular NAD+ levels following an ischemic insult, one can either inhibit the NAD+ catabolizing enzymes (PARP1, CD38) or facilitate NAD+ generation by the salvage pathway via administration of a NAD+ precursor. It was shown that administration of Nam increases tissue NAD+ levels and improves bioenergetics following ischemia (Yang et al., 2002a). Alternatively, one can directly stimulate the NAD+ synthesis by administration of NMN that will not require the activity of Nampt, the rate-limiting enzyme in NAD+ synthesis. Additionally, NMN does not cause the same unfavorable flushing side effect associated with Nam activating the GPR109A receptor (Canto et al., 2012, Benyo et al., 2005).

Application of NMN leads to an increase in cellular NAD+ levels by a one-step enzymatic reaction where NMN is converted to NAD+ by Nmnat (Belenky et al., 2007). Incubation of brain sections with NMN prevented a rapid NAD+ catabolism in the tissue (Balan et al., 2010). Furthermore, decreased activity of Nampt enzyme can significantly aggravate post-ischemic brain damage (Zhang et al., 2010). Thus, heterozygous Nampt knockdown animals manifested aggravated brain damage following photothrombosis-induced focal ischemia (Zhang et al., 2010). Transgenic mice with neuron-specific overexepression of Nampt show reduced infarct size when compared to wild type animals (Jing et al., 2014). Similarly, the adverse effect of Nampt inhibitor FK866 was reversed by intraventricular NMN injection (Wang et al., 2011). Taken together, these data suggest that post-ischemic NMN administration could improve bioenergetic metabolism of the post-ischemic tissue and ameliorate the brain damage. Therefore, the objective of this study was to examine the effect of NMN on brain tissue NAD+ catabolism, PAR formation, and histologic and neurologic outcomes following transient global cerebral ischemia.

Section snippets

Animals and experimental groups

All the experimental procedures and treatment protocols were performed according to the laboratory practices described in (Lapchak et al., 2013), and (Landis et al., 2012). All mice procedures were conducted in AAALAC approved facilities following current ethical regulations from the Guide for the Care and use of Laboratory Animals of the National Institutes of Health international guidelines and were approved by the Animal Care and Use Committee of the University of Maryland Baltimore. Adult, 3

Assessment of physiological parameters

To assure that the blood gases are at physiological levels before and during induction of forebrain ischemia six mice were subjected to physiological study. The ventilator was adjusted to give pO2 and pCO2 values within normal levels. As Table 1 shows, the blood gas data, including pCO2, pO2, and pH showed normal physiological levels with a ventilator setting of 120 strokes per min and about 0.3 cm3 of stroke volume. There were no significant changes in blood gas levels during the ischemic

Discussion

We show here that administration of NMN following transient forebrain ischemia has a dramatic protective effect against the cell death of CA1 pyramidal neurons. This was reflected in complete recovery of post-ischemic neurological scores and in prevention of the ischemia-induced NAD+ catabolism. Furthermore, NMN treated animals did not show any increase in poly-ADP-ribosylation of hippocampal proteins.

Our model of forebrain ischemia utilizes increased levels of isoflurane as tool to reduce the

Conflict of interest

TK has a pending patent application for use of NMN to modulate NAD+ activity in neuropathological conditions. JHP, KO and AL declare no conflict of interest.

Ethical approval

All applicable international, national, and/or institutional guidelines for the care and use of animals were followed.

Acknowledgments

This work was supported by U.S. Veterans Affairs Merit grant BX000917 to TK.

References (73)

  • P. Ellen et al.

    Hippocampal lesions and spontaneous alternation behavior in the rat

    Physiol. Behav.

    (1968)
  • M.D. Ginsberg

    Neuroprotection for ischemic stroke: past, present and future

    Neuropharmacology

    (2008)
  • F. Hua

    The development of a novel mouse model of transient global cerebral ischemia

    Neurosci. Lett.

    (2006)
  • T.M. Kauppinen et al.

    The role of poly(ADP-ribose) polymerase-1 in CNS disease

    Neuroscience

    (2007)
  • Y. Kiyota

    Relationship between brain damage and memory impairment in rats exposed to transient forebrain ischemia

    Brain Res.

    (1991)
  • L.K. Klaidman

    Nicotinamide as a precursor for NAD + prevents apoptosis in the mouse brain induced by tertiary-butylhydroperoxide

    Neurosci. Lett.

    (1996)
  • L.K. Klaidman

    Oxidative changes in brain pyridine nucleotides and neuroprotection using nicotinamide

    Biochim. Biophys. Acta

    (2001)
  • T. Kristian et al.

    A fluorescence-based technique for screening compounds that protect against damage to brain mitochondria

    Brain Res. Brain Res. Protoc.

    (2004)
  • E. Larsson

    Stereological assessment of vulnerability of immunocytochemically identified striatal and hippocampal neurons after global cerebral ischemia in rats

    Brain Res.

    (2001)
  • M.A. Moskowitz

    The science of stroke: mechanisms in search of treatments

    Neuron

    (2010)
  • M. Onken

    Simple model of forebrain ischemia in mouse

    J. Neurosci. Methods

    (2012)
  • K. Owens

    Utilizing commercial microwave for rapid and effective immunostaining

    J. Neurosci. Methods

    (2013)
  • N. Panahian

    Attenuated hippocampal damage after global cerebral ischemia in mice mutant in neuronal nitric oxide synthase

    Neuroscience

    (1996)
  • H. Sheng

    Characterization of a recovery global cerebral ischemia model in the mouse

    J. Neurosci. Methods

    (1999)
  • C.S. Siegel et al.

    NAD + and nicotinamide: sex differences in cerebral ischemia

    Neuroscience

    (2013)
  • C. Szabo et al.

    Role of poly(ADP-ribose) synthetase in inflammation and ischaemia-reperfusion

    Trends Pharmacol. Sci.

    (1998)
  • J. Yang

    Nicotinamide therapy protects against both necrosis and apoptosis in a stroke model

    Pharmacol. Biochem. Behav.

    (2002)
  • J. Yang

    The effects of nicotinamide on energy metabolism following transient focal cerebral ischemia in Wistar rats

    Neurosci. Lett.

    (2002)
  • J. Yoshino

    Nicotinamide mononucleotide, a key NAD(+) intermediate, treats the pathophysiology of diet- and age-induced diabetes in mice

    Cell Metab.

    (2011)
  • G. Zhen et al.

    Optimized protocol to reduce variable outcomes for the bilateral common carotid artery occlusion model in mice

    J. Neurosci. Methods

    (2007)
  • V.L. Baughman

    Neurologic outcome in rats following incomplete cerebral ischemia during halothane, isoflurane, or N2O

    Anesthesiology

    (1988)
  • Z. Benyo

    GPR109A (PUMA-G/HM74A) mediates nicotinic acid-induced flushing

    J. Clin. Invest.

    (2005)
  • T.J. Blanck

    Isoflurane pretreatment ameliorates postischemic neurologic dysfunction and preserves hippocampal Ca2 +/calmodulin-dependent protein kinase in a canine cardiac arrest model

    Anesthesiology

    (2000)
  • A.M. Brennan

    NAD(P)H fluorescence transients after synaptic activity in brain slices: predominant role of mitochondrial function

    J. Cereb. Blood Flow Metab.

    (2006)
  • P.H. Chan

    Overexpression of SOD1 in transgenic rats protects vulnerable neurons against ischemic damage after global cerebral ischemia and reperfusion

    J. Neurosci.

    (1998)
  • C.U. Choe

    CD38 exacerbates focal cytokine production, postischemic inflammation and brain injury after focal cerebral ischemia

    PLoS One

    (2011)
  • Cited by (88)

    • Research progress on Sirtuins (SIRTs) family modulators

      2024, Biomedicine and Pharmacotherapy
    View all citing articles on Scopus
    View full text