+ All Categories
Home > Documents > Aust N Z J Psychiatry 2013 Nierenberg 26 42

Aust N Z J Psychiatry 2013 Nierenberg 26 42

Date post: 26-Nov-2015
Category:
Upload: kthreopus
View: 12 times
Download: 2 times
Share this document with a friend
Popular Tags:
18
http://anp.sagepub.com/ Psychiatry Australian and New Zealand Journal of http://anp.sagepub.com/content/47/1/26 The online version of this article can be found at: DOI: 10.1177/0004867412449303 2013 47: 26 originally published online 18 June 2012 Aust N Z J Psychiatry Andrew A Nierenberg, Christine Kansky, Brian P Brennan, Richard C Shelton, Roy Perlis and Dan V Iosifescu development Mitochondrial modulators for bipolar disorder: A pathophysiologically informed paradigm for new drug Published by: http://www.sagepublications.com On behalf of: The Royal Australian and New Zealand College of Psychiatrists can be found at: Australian and New Zealand Journal of Psychiatry Additional services and information for http://anp.sagepub.com/cgi/alerts Email Alerts: http://anp.sagepub.com/subscriptions Subscriptions: http://www.sagepub.com/journalsReprints.nav Reprints: http://www.sagepub.com/journalsPermissions.nav Permissions: What is This? - Jun 18, 2012 OnlineFirst Version of Record - Jan 4, 2013 Version of Record >> at Fund Diag.Est Imstico PARENT on August 7, 2013 anp.sagepub.com Downloaded from
Transcript
Page 1: Aust N Z J Psychiatry 2013 Nierenberg 26 42

http://anp.sagepub.com/Psychiatry

Australian and New Zealand Journal of

http://anp.sagepub.com/content/47/1/26The online version of this article can be found at:

 DOI: 10.1177/0004867412449303 2013 47: 26 originally published online 18 June 2012Aust N Z J Psychiatry

Andrew A Nierenberg, Christine Kansky, Brian P Brennan, Richard C Shelton, Roy Perlis and Dan V Iosifescudevelopment

Mitochondrial modulators for bipolar disorder: A pathophysiologically informed paradigm for new drug  

Published by:

http://www.sagepublications.com

On behalf of: 

  The Royal Australian and New Zealand College of Psychiatrists

can be found at:Australian and New Zealand Journal of PsychiatryAdditional services and information for    

  http://anp.sagepub.com/cgi/alertsEmail Alerts:

 

http://anp.sagepub.com/subscriptionsSubscriptions:  

http://www.sagepub.com/journalsReprints.navReprints:  

http://www.sagepub.com/journalsPermissions.navPermissions:  

What is This? 

- Jun 18, 2012OnlineFirst Version of Record  

- Jan 4, 2013Version of Record >>

at Fund Diag.Est Imstico PARENT on August 7, 2013anp.sagepub.comDownloaded from

Page 2: Aust N Z J Psychiatry 2013 Nierenberg 26 42

Australian & New Zealand Journal of Psychiatry47(1) 26 –42DOI: 10.1177/0004867412449303

© The Royal Australian and New Zealand College of Psychiatrists 2012 Reprints and permission: sagepub.co.uk/journalsPermissions.navanp.sagepub.com

Australian & New Zealand Journal of Psychiatry, 47(1)

Introduction

Patients with bipolar disorder frequently experience multiple episodes of depression, mania, or hypomania, even if treated with the best available pharmacotherapy. Prospective longitudinal studies suggest that relapse rates within a year approach 67% for a cohort of bipolar patients with mostly inpatient index episodes (Judd et al., 2008). The latest effectiveness trial information from the STEP-BD study showed about 50% of bipolar patients will relapse within 12 months of recovering from an outpatient index mood episode (Perlis et al., 2006). Better continuation and maintenance treatments are needed.

We review potential mitochondrial modulators (MM) for the treatment of bipolar disorder that builds on recent findings on the pathophysiology of bipolar disorder. We

focus on: (a) mitochondrial dysfunction in bipolar disorder (Clay et al., 2011; Konradi et al., 2004; Munakata et al., 2004, 2007; Naydenov et al., 2007; Quiroz et al., 2008; Shao et al., 2008b; Stork and Renshaw, 2005; Wang, 2007); (b) altered brain energy metabolism (Frey et al., 2007a); and (c) an important convergent genetic finding about the alpha-1C subunit of the L-type calcium channel (Sklar et al., 2008) along with calcium channel physiology

Mitochondrial modulators for bipolar disorder: A pathophysiologically informed paradigm for new drug development

Andrew A Nierenberg, Christine Kansky, Brian P Brennan, Richard C Shelton, Roy Perlis and Dan V Iosifescu

Abstract

Objectives: Bipolar patients frequently relapse within 12 months of their previous mood episode, even in the con-text of adequate treatment, suggesting that better continuation and maintenance treatments are needed. Based on recent research of the pathophysiology of bipolar disorder, we review the evidence for mitochondrial dysregulation and selected mitochondrial modulators (MM) as potential treatments.

Methods: We reviewed the literature about mitochondrial dysfunction and potential MMs worthy of study that could improve the course of bipolar disorder, reduce subsyndromal symptoms, and prevent subsequent mood episodes.

Results: MM treatment targets mitochondrial dysfunction, oxidative stress, altered brain energy metabolism and the dysregulation of multiple mitochondrial genes in patients with bipolar disorder. Several tolerable and readily available candidates include N-acetyl-cysteine (NAC), acetyl-L-carnitine (ALCAR), S-adenosylmethionine (SAMe), coenzyme Q10 (CoQ10), alpha-lipoic acid (ALA), creatine monohydrate (CM), and melatonin. The specific metabolic pathways by which these MMs may improve the symptoms of bipolar disorder are discussed and combinations of selected MMs could be of interest as well.

Conclusions: Convergent data implicate mitochondrial dysfunction as an important component of the pathophysiology of bipolar disorder. Clinical trials of individual MMs as well as combinations are warranted.

KeywordsBipolar disorder, mitochondria, modulators

Massachusetts General Hospital, Boston, MA, USA

Corresponding author:Andrew A Nierenberg, Massachusetts General Hospital, Suite 580, 50 Staniford Street, Boston, MA 02114, USA. Email: [email protected]

449303 ANP47110.1177/0004867412449303Nierenberg et al.ANZJP Articles2012

Review

at Fund Diag.Est Imstico PARENT on August 7, 2013anp.sagepub.comDownloaded from

Page 3: Aust N Z J Psychiatry 2013 Nierenberg 26 42

Nierenberg et al. 27

Australian & New Zealand Journal of Psychiatry, 47(1)

associated with bipolar disorder, mitochondrial function and apoptosis. While each MM has the potential for treat-ing bipolar disorder, it is possible that a combination of MMs (which target multiple points in the mitochondrial electron transport chain as well as oxidative stress) added to standard mood stabilizer treatment could reduce bipolar relapse or resurgence of mood symptoms. The purpose of this paper is to review the spectrum of MMs that could be considered for clinical trials. Of note, all MMs reviewed here are endogenous substances, currently available com-mercially as ‘nutraceuticals’ or dietary supplements.

Background

Bipolar disorder, with a prevalence of about 1% for BPI and 1.1% for BPII, is associated with substantial lifelong disease burden and dysfunction and tends to be highly recurrent (Merikangas et al., 2007; Weissman et al., 1996). The NIMH Collaborative Depression Study followed a group of unipolar and bipolar patients for almost 25 years. Of the bipolar patients, most of who were recruited from inpatient settings, almost 67% experienced at least one relapse within a year of recovery, with a higher risk of relapse if they experienced residual mood symptoms (Judd et al., 2008). A subset of bipolar participants in STEP-BD, who were followed prospectively after recovery from a mood episode, had recurrence rates of about 50% within 12 months, despite measurement-based, guideline-informed, systematic care with a fully available complement of mod-ern psychopharmacological treatments – and these partici-pants also had higher relapse rates if they had residual manic symptoms (Perlis et al., 2006). Furthermore, subsyn-dromal symptoms of depression, mania and hypomania with associated dysfunction frequently persist between epi-sodes (Judd et al., 2008). Better treatments are needed to keep bipolar patients well, reduce subsyndromal symp-toms, and prevent subsequent mood episodes.

Because the pathophysiology of bipolar disorder has yet to be determined, developing rational novel therapies pre-sents a formidable challenge (Mathew et al., 2008). Exciting new research that converges on mitochondrial dysfunction, however, could lead to a new drug development paradigm for the treatment of bipolar disorder (Kato, 2008; Kazuno et al., 2008; Munakata et al., 2004, 2007). We postulate that MMs are candidates for novel treatments based on the lat-est studies of mitochondrial abnormalities (Clay et al., 2011) and alterations of brain energy metabolism (Frey et al., 2007a; Stork and Renshaw, 2005) in bipolar disorder, as well as an important whole genome-wide association study that shows altered single nucleotide polymorphism (SNP) for L-type calcium channels (Sklar et al., 2008). The strength of the approach is that it can address a range of dysfunctional mitochondrial targets, especially if combinations of MMs are used. It is possible that the effects of several MMs could be synergistic or target

different abnormalities across the electron transport chain within mitochondria; alternatively, using several modula-tors may not be any better than using one. The potential use of multiple MMs is also consistent with the need for rational polypharmacy in bipolar patients and the finding that par-ticipants in STEP-BD took a median of three medications (Ghaemi et al., 2006).

Mitochondrial dysfunction in bipolar disorder

Multiple lines of evidence converge to strongly implicate mitochondrial dysfunction and oxidative stress in bipolar disorder (Clay et al., 2011). While the degree of mitochon-drial dysfunction may not be severe enough to manifest a systemic disorder, it may be enough to cause neuropsychi-atric symptoms as the brain requires significantly more energy than any other organ in the body (Peters et al., 2004). Mitochondria regulate energy production and gen-eration of adenosine-5'-triphosphate (ATP) through the mitochondrial electron transport chain (ETC), with associ-ated production of reactive oxygen species (ROS) that can result in oxidative stress and cellular damage, especially in the absence of sufficient antioxidant defenses (Ng et al., 2008). In addition to oxidative processes, mitochondria regulate calcium and apoptotic processes, and are central to facilitating neuronal plasticity. Dysfunctional mitochondria can result in neuronal damage via multiple mechanisms: decreased ATP production, oxidative damage of mem-branes and DNA, abnormal calcium sequestration, and apoptosis via activation of caspase proteases (Ng et al., 2008; Wang, 2007). As reviewed below, mitochondrial dys-function in bipolar disorder is associated with decreased ATP production, indicated by decreased brain energy metabolism and a shift from oxidative phosphorylation to anaerobic glycolysis (Cui et al., 2007; Frey et al., 2007a; Naydenov et al., 2007; Stork and Renshaw, 2005) (as shown by decreased phosphocreatine (PCr) and ATP in magnetic resonance spectroscopy (MRS) studies), upregu-lation of genes involved in apoptosis compared to patients with schizophrenia (Benes et al., 2006), downregulation of mitochondrial genes regulating OXPHOS and proteas-ome degradation in bipolar disorder as compared to those with schizophrenia and healthy controls (Konradi et al., 2004), decreased antioxidant defenses, including decreased superoxide dismutase and glutathione-peroxi-dase-1 and -4 (Konradi et al., 2004), and increased lipid peroxidation, as well as abnormal calcium metabolism (Kato, 2008; Munakata et al., 2004). Moreover, abnor-malities in the structure and distribution of mitochondria have been identified in brain and peripheral cells obtained from patients with bipolar disorder (Cataldo et al., 2010). In summary, substantial evidence supports the hypothesis that mitochondrial dysfunction may play a central role in the pathophysiology of bipolar disorder.

at Fund Diag.Est Imstico PARENT on August 7, 2013anp.sagepub.comDownloaded from

Page 4: Aust N Z J Psychiatry 2013 Nierenberg 26 42

28 ANZJP Articles

Australian & New Zealand Journal of Psychiatry, 47(1)

Oxidative phosphorylation and glycolysis

A study by Naydenov et al. (2007), in which they obtained lymphocytes from bipolar patients and healthy controls and examined differences in the pattern of expression of ETC genes in response to energy stress, provides insight into the possible nature of mitochondria dysregulation in bipolar disorder. This study showed no difference between bipolar patients and healthy controls in the expression of ETC genes under normal glucose concentrations. When cultured lymphocytes were exposed to glucose deprivation, how-ever, bipolar patients displayed reduced expression of ETC genes while healthy controls uniformly increased ETC gene expression. This pattern suggests an inability of mito-chondria in bipolar patients to adapt to energy stress.

The impaired response to energy stress observed in bipolar disorder described above is associated with several measurable effects of mitochondrial dysfunction including lower intracellular pH levels, increased lactate levels, and a decrease in overall energy production (Dager et al., 2004; Stork and Renshaw, 2005). Studies utilizing MRS showed that compared to normal controls, bipolar patients not taking medication had increased cerebral lactate levels, specifically in the gray matter (Dager et al., 2004). Moreover, lower pH levels have been reported in the basal ganglia region and whole brain of euthymic bipolar sub-jects in comparison to individuals without bipolar disorder (Hamakawa et al., 2004).

Under energy stress, mitochondria usually rely on glyco-lysis for a quick source of ATP energy. During this anaero-bic cellular respiration process, nicotine adenine dinucleotide (NADH) is converted to NAD, thereby reducing pyruvate to lactate and causing a subsequent decrease in intracellular pH levels. While glycolysis is a readily available source of energy, it only manufactures 2 ATP per glucose molecule in contrast to the 36 ATP produced through oxidation phospho-rylation (Ng et al., 2008). In bipolar disorder, MRS studies have documented increased lactate (Dager et al., 2004; Kato et al., 1998) and lowered pH levels (Hamakawa et al., 2004; Kato et al., 1998) suggesting that neurons are constantly relying on the largely inefficient process of glycolysis to produce energy. Overall, these findings provide evidence that, compared to normal controls, the total energy output of mitochondria in bipolar disorder is inhibited.

Brain energy metabolism in bipolar disorder

Multiple reports using MRS have also suggested that bipo-lar disorder is characterized by abnormalities of brain energy metabolism, consistent with mitochondrial dysfunc-tion. Proton MRS (1H-MRS) studies show decreased cere-bral N-acetyl-aspartate (NAA) in bipolar subjects compared to normal controls (Bertolino et al., 2003; Deicken et al.,

2003). Decreased levels of NAA, a marker of neuronal viability, are consistent with impaired mitochondrial energy production (Clark, 1998) as NAA is of mitochondrial ori-gin: NAA is synthesized in mitochondria by the membrane-bound enzyme L-aspartate N-acetyltransferase, a catalyst that is found only in the brain (Patel and Clark, 1979).

Phosphorus MRS (31P-MRS) studies allow measure-ments of high-energy compounds such as ATP and PCr – a reservoir for the generation of ATP that has been found to be altered in bipolar patients (Kato et al., 1994, 1995). During periods of acute neuronal activity, as ATP is utilized and depleted, PCr is rapidly broken down in order to main-tain the overall concentration of ATP (Erecinska and Silver, 1989). Long-term abnormalities in PCr levels generally reflect much larger alterations in cellular metabolism and, in particular, an insufficient supply of the ATP needed for normal cellular function (Rothman, 1994). Therefore, continually decreased levels of PCr are suggestive of hypo-metabolism, possibly due to mitochondrial dysfunction (Modica-Napolitano and Renshaw, 2004). Persistently low brain PCr values have also been found in patients with a variety of mitochondrial disorders (e.g. mitochondrial encephalomyopathy, lactic acidosis and stroke-like episodes (MELAS), Leigh’s disease, progressive external ophthalmoplegia (PEO), and Leber’s hereditary optic neu-ropathy) (Barbiroli et al., 1993; Eleff et al., 1990).

Indirectly, abnormalities of lipid metabolism, an energy-intensive process (Purdon and Rapoport, 1998), are also supportive of the mitochondrial dysfunction and associated bioenergetic deficits in bipolar disorder. Multiple studies have suggested that total choline levels are elevated in bipolar disorder (Hamakawa et al., 1998; Lafer et al., 1994; Moore et al., 2000). Phosphomonoester (PME) levels (which primarily include the lipid membrane precursors phosphocholine and phosphoethanolamine) are also altered in bipolar disorder. Euthymic bipolar patients show decreased PME levels compared with healthy volunteers (Yildiz et al., 2001). Both series of findings suggest impaired lipid membrane metabolism in bipolar disorder. This is important, as lipid membrane turnover represents 10–15% of total energetic requirements in the cell (Purdon and Rapoport, 1998).

Mitochondrial gene expression in bipolar disorder

Multiple mitochondrial genes have been found to be upreg-ulated or downregulated in bipolar disorder compared to normal controls (Benes et al., 2006; Munakata et al., 2004; Naydenov et al., 2007; Sun et al., 2006; Wang, 2007). Mitochondrial DNA (mtDNA) contains 37 genes, 13 encoding for subunits of the respiratory chain complexes I, III, IV and V, 22 encoding for transfer RNAs, and two for ribosomal RNAs (Taylor and Turnbull, 2005). MtDNA dif-fers somewhat from autosomal (nuclear) DNA: it is not

at Fund Diag.Est Imstico PARENT on August 7, 2013anp.sagepub.comDownloaded from

Page 5: Aust N Z J Psychiatry 2013 Nierenberg 26 42

Nierenberg et al. 29

Australian & New Zealand Journal of Psychiatry, 47(1)

protected by any repair mechanism or histones, does not undergo any recombination during meiosis, and is mater-nally inherited. Notably, polymorphisms are estimated to occur up to 10 times more frequently than in autosomal DNA, probably owing to closeness to free radical produc-tion and absence of an effective DNA repair mechanism (Taylor and Turnbull, 2005).

Because of the compact nature of mtDNA, it has been demonstrated that genotyping a small number of single-nucleotide polymorphisms can capture nearly all common variation in the mitochondrial genome. Specifically, Saxena and colleagues reported a set of SNPs that efficiently ‘tag’ common mitochondrial variation (Saxena et al., 2006). In light of the evidence presented above, such variations would have a high prior probability of association with dif-ferential treatment response.

Mitochondrial variation is also of particular interest in light of the recent association between calcium channel variation and bipolar disorder (Ferreira et al., 2008; Sklar et al., 2008). Specifically, mitochondria and endoplasmic reticulum (ER) function in concert to sequester Ca2+ ions after their temporary diffusion in subcellular regions. A massive movement of positively charged Ca2+ into the mitochondrion could exceed its capacity to export cati-ons, impairing ATP synthesis and promoting apoptosis (Bernardi et al., 1998). Of note, animal, in vitro and mRNA studies demonstrate that chronic lithium treatment, as well as therapeutic doses of valproic acid and electroconvulsive stimulus, increase the expression of Bcl-2 (Chang et al., 2009; Chen et al., 1999). In turn, one of the main effects of the anti-apoptotic protein Bcl-2 is increasing the stability of the mitochondrial permeability transition pore (MPTP) (Weeber et al., 2002).

Bcl-2 , BDNF, neuroprotection and mitochondria as pathophysiology and targets of treatments

Mitochondria play important roles in the regulation of intracellular calcium (Ca2+), and in the general process of synaptic plasticity (Yang et al., 2003). Excessive opening of N-methyl-d-aspartate (NMDA) glutamate receptors can trigger large influxes of Ca2+, which in turn can overwhelm the ability of mitochondria to export protons and cations (Nicholls and Ward, 2000) and can lead to drastic reduc-tion in ATP synthesis (Weeber et al., 2002); at the extreme, this can lead to initiation of apoptosis (Bernardi et al., 1998). Activation of mitochondrial apoptotic mechanisms may lead to selective destruction of synapses (Culmsee and Mattson, 2005). This may be related to neuroplasticity and synaptic pruning in normal conditions, but is also involved with extensive loss of synapses in pathological conditions associated with mitochondrial dysfunction (Mattson and Liu, 2003).

In bipolar disorder, patients with the val66met polymor-phism of the BDNF gene (which may be associated with rapid cycling (Müller et al., 2006)) also have low levels of PCr + Cr in the dorsolateral prefrontal cortex (suggestive of low bioenergetic metabolism (Frey et al., 2007b)). Rats treated with therapeutic doses of lithium and valproic acid have increased levels of Bcl-2 (Manji et al., 2000). Bcl-2 and other neuroprotective agents from its family (Bcl-xl) exert their anti-apoptotic and neuroprotective activity by increasing the ability of mitochondria to resist the toxic effect of elevated intracellular Ca2+ (Murphy et al., 1996).

The presence of mitochondrial dysfunction in bipolar disorder and the link between current effective treatments in bipolar disorder (lithium, valproic acid), major neuropro-tective factors (Bcl-2, BDNF and GSK-3) and their impact on mitochondrial activity may justify enhancing mitochon-drial function as a putative target in the search of future mood stabilizers.

Candidate mitochondrial modulators

Several readily available dietary supplements (or nutraceu-ticals) have been tested as potential treatments in bipolar disorder (for a review see Sarris et al., 2011). Among them, a subset of substances has the potential to improve mito-chondrial function and brain energy metabolism; those are grouped here under the term ‘mitochondrial modulators’. Candidates include N-acetyl-cysteine (NAC), acetyl-L-car-nitine (ALCAR), S-adenosylmethionine (SAMe), coen-zyme Q10 (CoQ10), alpha-lipoic acid (ALA), creatine monohydrate (CM), and melatonin. As pointed out by Hagen and colleagues, increased mitochondrial metabo-lism can result in decreased antioxidant status – and the decreased antioxidant status may need to be offset by pro-viding an additional antioxidant such as ALA (Hagen et al., 2002a) – alternatively, NAC may provide the sufficient antioxidant properties (by increasing glutathione) without ALA. Furthermore, allostatic load in bipolar disorder has been reported to be increased, as evidenced by high levels of ROS (Kapczinski et al., 2008) and MMs could prevent ROS damage.

N-acetyl-cysteine (NAC)

NAC, by supplying cysteine, increases synthesis of the free radical scavenger glutathione (GSH) which, in turn, reduces oxidative stress (Atkuri et al., 2007). Glutathione (GSH), a tripeptide composed of glutamate, cysteine and glycine, is the most abundant thiol antioxidant. Glutamate-cysteine ligase (GCL) is the rate-limiting enzyme for GSH produc-tion – GCL ligates glutamate with cysteine to form gamma-glutamyl-cysteine which is then combined with glycine and catalyzed by GSH synthase to form GSH (Cui et al., 2007). As an antioxidant, GSH reacts with hydrogen peroxide

at Fund Diag.Est Imstico PARENT on August 7, 2013anp.sagepub.comDownloaded from

Page 6: Aust N Z J Psychiatry 2013 Nierenberg 26 42

30 ANZJP Articles

Australian & New Zealand Journal of Psychiatry, 47(1)

H2O2 to form H2O with the byproduct of GSH disulfide. Also, GSH can conjugate with oxidized products, catalyzed by glutathione-S-transferase (GST) to further reduce oxida-tive stress.

Lithium and valproate have well-known neuroprotective effects that may be mediated by increasing GSH and GCL levels. In cultured rat cerebral cortical cell cultures, the neuroprotective effects of lithium and valproate against oxidative damage from H2O2 is abolished by depletion of GSH (Cui et al., 2007). Furthermore, one mechanism of neuroprotection of lithium may be through GSH, since lith-ium increases the gene expression of GST isoenzymes (Shao et al., 2008a). NAC can increase GSH further (and potentially synergistically with lithium or valproate) by driving synthesis of glutathione to the right with increased cysteine availability.

Oral NAC prevents glutathione depletion in the brain by increasing cysteine and synthesis of glutathione systemi-cally (Berk et al., 2008a/b). Furthermore, NAC has been found to be neuroprotective and can prevent oxidative dam-age in complex 1 in the mitochondrial electron transport chain (Mayer and Noble, 1994; Nicoletti et al., 2005). Clinically, NAC augmentation reduced negative symptoms and improved auditory processing in schizophrenia (Berk et al., 2008a/b), improved pathological gambling in a pilot study (Grant et al., 2007), and is a prime candidate to improve mitochondrial function, and perhaps the long-term course, in bipolar disorder.

In a preliminary, double-blind study of bipolar patients (n = 75) who had experienced a mood episode in the past 6 months and who had been stable for at least 1 month, NAC 2 grams per day or placebo was added to ongoing medica-tions (Berk et al., 2008a/b). At baseline, half the patients were euthymic, about a third were depressed, and about 15% had euphoric or dysphoric mania. Measures of depres-sion, quality of life, and functioning were all better with NAC compared to placebo beginning at week 12 and con-tinuing until week 24; no differences were found in manic or hypomanic symptoms, but these symptoms were mini-mal at baseline and during the study. The delayed effect of NAC (observed first at week 12) may be a result of the study design, as most patients were euthymic at baseline. Improvements in mood that occurred during the trial stopped after discontinuation of NAC.

Acetyl-L-carnitine (ALCAR)

L-Carnitine, composed of lysine and methionine, can be endogenously generated or obtained through diet (Hoppel, 2003). Carnitines transport fatty acids into mitochondria for beta-oxidation and energy generation, and scavenge ROS (Al-Majed et al., 2006; Rebouche, 2004). These fatty acids enter mitochondria as acyl-carnitines, and when oxi-dized, release energy and form acyl-coenzyme A, which then enters the citric acid cycle (Hoppel, 2003). ALCAR,

absorbed better than L-carnitine and more able to cross the blood–brain barrier than L-carnitine (Ames and Liu, 2004a), has neuroprotective and anti-apoptotic properties (Al-Majed et al., 2006; Virmani et al., 2005) and has been shown to block glutamate-induced over-expression of glu-tamic acid decarboxylase GAD67 (Hao et al., 2004), the main enzyme that converts glutamate to gamma-amino-butyric acid (GABA). ALCAR appears to reverse age-related degeneration in animal models (Aliev et al., 2009; Ames and Liu, 2004a, 2004b; Hagen et al., 2002a; Liu et al., 2002b) and may slow down or reverse age-related cognitive and motoric decline in rats, as well as reverse diminished reactivity to the environment (Hagen et al., 2002a) – effects thought to result from enhanced mitochon-drial functioning. Notably, adult and aged rats administered ALCAR both show an increase in brain levels of ATP and PCr as measured by 1H nuclear magnetic resonance spec-troscopy (NMR) (Aureli et al., 1990). However, evidence that ALCAR can decrease oxidative stress is mixed (Ames and Liu, 2004a), suggesting that it may be more clinically effective when administered with a potent antioxidant such as NAC or ALA (see below).

Clinically, ALCAR has shown benefit in clinical trials for a number of neuropsychiatric disorders including dementia (Bonavita, 1986; Passeri et al., 1990; Pettegrew et al., 1995; Salvioli and Neri, 1994; Sano et al., 1992; Spagnoli et al., 1991), geriatric depression (Bella et al., 1990; Fulgente et al., 1990; Garzya et al., 1990; Gecele et al., 1991; Nasca et al., 1989; Pettegrew et al., 2002; Tempesta et al., 1987; Villardita et al., 1983), dysthymia (Zanardi and Smeraldi, 2006), hyperactivity associated with fragile X syndrome (Torrioli et al., 1999, 2008), ADHD, inattentive type (Arnold et al., 2007), degenerative cerebellar ataxia (Sorbi et al., 2000), minimal hepatic encephalopathy (Malaguarnera et al., 2008), and hepatic coma secondary to cirrhosis (Malaguarnera et al., 2006), and has demonstrated benefit in disorders thought to be associated with deficits in neuronal mitochondrial energy production such as diabetic peripheral neuropathy (De Grandis and Minardi, 2002; Sima et al., 2005) and HIV-associated antiretroviral toxic neuropathy (Hart et al., 2004; Youle and Osio, 2007). One study investigating the weight-reducing effects of ALCAR in bipolar patients who had gained weight while taking valproate found no difference in weight between those taking ALCAR with diet com-pared to diet alone (Elmslie et al., 2006). Interestingly, valproate-induced hepatotoxicity may be mediated by inhi-bition of mitochondrial fatty acid beta-oxidation and deple-tion of carnitine, suggesting that carnitine supplementation, perhaps through administration of ALCAR, may either pre-vent or reverse valproate-induced hepatotoxicity (Silva et al., 2008).

Importantly, ALCAR has demonstrated efficacy in a variety of depressive spectrum disorders in several placebo-controlled studies (Bella et al., 1990; Fulgente et al., 1990;

at Fund Diag.Est Imstico PARENT on August 7, 2013anp.sagepub.comDownloaded from

Page 7: Aust N Z J Psychiatry 2013 Nierenberg 26 42

Nierenberg et al. 31

Australian & New Zealand Journal of Psychiatry, 47(1)

Garzya et al., 1990; Gecele et al., 1991; Nasca et al., 1989; Pettegrew et al., 2002; Tempesta et al., 1987; Villardita et al., 1983; Zanardi and Smeraldi, 2006). In addition, a preliminary study in depressed geriatric patients demon-strated improvement in depression symptoms that were associated with an increase in PCr as measured by 31P-MRS after monotherapy treatment with ALCAR (Pettegrew et al., 2002). However, no clinical trials, to our knowledge, have yet examined the effect of ALCAR supplementation in individuals with bipolar disorder.

S-Adenosylmethionine (SAMe)

SAMe, formed from methionine and ATP, is a naturally occurring biological component of all living cells, and plays a critical role in cellular metabolism as a major source of methyl groups for key biochemical reactions (Bottiglieri, 1996; Bottiglieri et al., 2000). Specifically as a methyl donor, SAMe is a precursor molecule for pathways cata-lyzed by methyltransferase enzymes, such as methylation, transulfuration and aminopropylation.

Methylation via SAMe is a particularly important pro-cess in the central nervous system. SAMe is integral in the degradation and repair of proteins with aberrant levels of d-aspartate that can accumulate during protein aging and lead to cellular abnormalities and dysfunction (Bottiglieri, 1996; Bottiglieri et al., 2000). SAMe-mediated methylation reactions also are important in the monoamine metabolism. As a methyl donor, SAMe activates tyrosine hydroxylase, an essential enzyme for the synthesis as well as the excita-tory and inhibitory regulation of various monoamine neuro-transmitters, including dopamine, serotonin, adrenaline and noradrenaline.

SAMe is also a precursor molecule for GSH production through transulfuration. S-Adenosyl-homocysteine (SAH), formed when SAMe donates its methyl group, is rapidly metabolized to homocysteine. Homocysteine, when paired with several B vitamin cofactors, including pyridoxal phos-phate (vitamin B-6), is converted into cystathionine and eventually, through transulfuration, converted to GSH. As discussed earlier in the section on NAC, GSH plays an essential role in reducing oxidative stress. Homocysteine can also regenerate SAMe when a sufficient concentration of B vitamins triggers its remethylation.

Clinically, SAMe evidence supports the antidepressant effects for individuals with depressive disorders, with fewer side effects than standard medications such as imipramine, chlorimipramine, nomifensive and minaprine (Bottiglieri, 1996; Bottiglieri et al., 2000). The results of an open label trial of SAMe treatment in depressed inpatients supported the clinical value of SAMe supplementation such that patients experienced remission or improvement of their symptoms with minimal side effects after a 2-week period (Lipinski et al., 1984). More recent research has investi-gated the efficacy and tolerability of SAMe when combined

with other traditional antidepressant medication. When SAMe was added to selective serotonin reuptake inhibitors (SSRI) antidepressants or venlafaxine, individuals with unremitted depression showed significant improvements, including a decrease in both depression-severity and clini-cal global impressions severity scores as assessed by the HAM-D and CGI scales, respectively (Alpert et al., 2004). In a recent randomized controlled trial, SAMe augmenta-tion of antidepressants was more efficacious than placebo augmentation in unipolar depressed subjects not respond-ing to a course of selective serotonin reuptake inhibitors (SSRIs) or serotonin norepinephrine reuptake inhibitors (SNRIs) (Papakostas et al., 2010). While evidence for effi-cacy exists when SAMe is combined with imipramine (Berlanga et al., 1992), the results for SAMe augmentation with older antidepressants, including phenelzine, mianserin and maprotiline (Alvarez et al., 1987) showed inconsistent results. Of note, SAMe may also carry a risk of manic switch in bipolar disorder. In an earlier study, nine of 11 bipolar patients treated with SAMe switched into elevated mood state (hypomania, mania and euphoria) while the other two did not respond (Carney et al., 1989). To the best of our knowledge, no systematic randomized controlled studies of SAMe for bipolar disorder have been conducted to date.

Coenzyme Q10 (CoQ10)

Coenzyme Q10 (CoQ10) consists of polyisopropyl units (usually 9 or 10) attached to a benzoquinone ring that can alternate between free radical ubisemiquinone (*QH), and fully reduced ubiquinol (QH2) (Kwong et al., 2002). The ability to assume multiple redox states allows for CoQ10 to play several functional roles in the cells, not all of which are well understood. CoQ10 acts as a lipid-soluble antioxidant that can prevent cell damage by neutralizing free radicals directly or by reducing an alpha-tocopheroxyl radical to alpha-tocopherol, an absorbable form of vita-min E. Another important function of CoQ10, present in high concentrations within the inner mitochondrial layer, is its ability to generate an electrochemical gradient along the transmembrane of mitochondria. The quinine head of CoQ transfers electrons in the mitochondrial electron transport chain from complexes I and II to complex III while releasing protons into the intermembrane.

While CoQ10 naturally decreases with aging in various tissues, abnormal levels of CoQ10 are also associated with conditions such as hypertension, diabetes, cardiovas-cular disease, and neurodegenerative disorders such as Parkinson’s disease.

Plasma and mitochondrial brain levels of CoQ10 in indi-viduals with Parkinson’s disease were significantly lower than non-parkinsonian controls of the same gender and age (Shults and Haas, 2005). While the total level of plasma CoQ10 was reduced in subjects with Parkinson’s disease,

at Fund Diag.Est Imstico PARENT on August 7, 2013anp.sagepub.comDownloaded from

Page 8: Aust N Z J Psychiatry 2013 Nierenberg 26 42

32 ANZJP Articles

Australian & New Zealand Journal of Psychiatry, 47(1)

Table 1. Evidence for mitochodrial dysfunction in bipolar (BP) disorder.

Bipolar disorder Implications for mitochondrial function References

Ca2+ 1. Elevated levels in mitochondria leads to apoptosis and impaired ATP synthesis

2. D2 subunit of the L-type of the voltage-gated calcium channel

1. Mitochondria regulate Ca2+; excessive Ca2+ interferes with ability to export cations and protons

2. Dysregulation of Ca2+ influx which increases energetic requirements

Bernardi et al., 1998; Weeber et al., 2002

Sklar et al., 2008

pH Levels 1. Lower intracellular pH levels2. Lower pH in basal ganglia region and

whole brain of euthymic BP versus NC

A metabolic shift towards anaerobic glycolysis for energy production leads to increased lactate levels and reduced pH levels

1. Dager et al., 2004; Ng et al., 2008; Stork and Renshaw, 2005

2. Hamakawa et al., 2004

N-acetyl- aspartate (NAA)/ phosphocreatine (PCr)

1. BPI associated with decreased cerebral NAA in the right hippocampus vs NC

2. Regional reduction of NAA relative signals, suggesting neuronal damage of the hippocampus

3. PCr altered in BP; low PCr found in BPII versus NC in all three psychiatric states (hypomanic, euthymic, depressed)

1. NAA is synthesized in mitochondria by the membrane-bound enzyme L-aspartate N-acetyltransferase, only found in brain

2. Decreased NAA are consistent with impaired mitochondrial energy production

3. PCr is a reservoir for ATP generation; breakdown to maintain ATP concentration levels; low levels associated with hypometabolism

1. Deicken et al., 20032. Bertolino et al., 20033. Kato et al., 1994;

Kato et al., 1995

Bcl-2 1. Increased levels with lithium and/or valproic acid treatment

1. Neuroprotetic and anti-apoptotic effect on mitochondria; increase resistance to toxic effects of elevated intracellular Ca2+

2. Increases stability of the PTP

1. Chen et al., 1999; Manji et al., 2000; Murphy et al., 1996; Perera et al., 2007

2. Weeber et al., 2002

Effects of mood stabilizers

1. Therapeutic doses of lithium and valproic acid increase levels of Bcl-2 in rats

2. Neuroprotective effects against oxidative damage from H2O2

1. Neuroprotective effects mediated by increasing GSH and GCL levels

2. Lithium increases gene expression of GST isoenzymes, neuroprotective; if GSH-depleted, neuroprotective effects destroyed

1. Cui et al., 20072. Shao et al., 2008a

Mitochondrial gene expression

1. SNPs ‘tag’ common mitochondrial variations that may be associated with differential treatment response

2. Val66met polymorphism of the BDNF gene is associated with rapid cycling

1. Polymorphisms approximately 10 times more frequent in mitochondrial DNA versus autosomal DNA

2. Low levels of PCr + Cr in DLPFC suggests low bioenergetic metabolism; BDNF may be a neuroprotective factor for mitochondrial dysfunction

1. Richter et al., 1988; Saxena et al., 2006

2. Frey et al., 2007b; Müller et al., 2006

Brain energy metabolism

1. Decreased cerebral NAA, synthesized by brain-specific catalyst L-aspartate N-acetyltransferase

2. Decreased brain PCr levels

1. Impaired mitochondrial energy production

2. Insufficient supply of ATP generated due to hypometabolism and possibly mitochondrial dysfunction

1. Bertolino et al., 2003; Clark, 1998; Patel and Clark, 1979

2. Kato et al., 1994; Kato et al., 1995

Abnormal mitochondrial structure and distribution

1. Mitochondria of smaller area observed in postmortem brain tissue of bipolar disorder patients

2. Atypically shaped mitochondria observed in peripheral cells from bipolar disorder patients

3. Perinuclear clustering of mitochondria observed in peripheral cells from bipolar disorder patients

1. Abnormal morphology and distribution of mitochondria may have significant effects on neuroplasticity and cell resilience and survival

1. Cataldo et al., 2010

ATP: adenosine-5'-triphosphate; DLPFC: dorsolateral prefrontal cortex; GCL: glutamate-cysteine ligase; GSH: glutathione; GST: glutathione-S-transferase; NC: normal controls; PTP: permeability transition pore; SNP: single nucleotide polymorphism.

at Fund Diag.Est Imstico PARENT on August 7, 2013anp.sagepub.comDownloaded from

Page 9: Aust N Z J Psychiatry 2013 Nierenberg 26 42

Nierenberg et al. 33

Australian & New Zealand Journal of Psychiatry, 47(1)

the percentage of oxidized plasma CoQ10 (ubiquinone-10) was increased (Sohmiya et al., 2004). Because CoQH2

–10 (e.g. ubiquinol-10) is oxidized to CoQ10 with early plasma oxidation, high levels of CoQ10 may serve as a biomarker of oxidative stress in the mitochondria. A significant cor-relation was also found between the level of CoQ10 and the activity levels of complex I and II/III. Impaired activity of complex I, II and III was observed in platelet mitochondria of patients with early, untreated Parkinson’s disease (Shults et al., 1997).

Evidence of CoQ10’s role in oxidative stress suggests that oral administration of coenzyme Q10 has the potential to be part of the treatment of such neurodegenerative dis-eases. For example, individuals with Huntington’s disease showed a decrease in elevated lactate levels in the basal ganglia, as well as an increase in complex I activity after treatment with CoQ10 (Feigin et al., 1996). Patients with early, untreated Parkinson’s disease showed a slower decline in functioning when CoQ10 was given at a dose of 1200 mg/day (Shults et al., 2002), a dose that has been

Table 2. Potential mitochondrial modulators and their effects on mitochondria and oxidative stress.

Electron transport chainEffects on fatty acids

Effect on level of ROS

Clinical evidence in neurodegenerative disorder I II III IV V

N-acetyl- cysteine (NAC)

X – – – – – ↓ NAC augmentation reduces negative symptoms and improved auditory processing in schizophrenia

Acetyl-L-carnitine (ALCAR)

X X – X – Transports fatty acid into mitochondria for beta-oxidaton and energy generation

↓ Improves selective areas of cognition for Alzheimer’s disease, ADHD inattentive type (Arnold et al., 2007)Improves fatigue and peripheral neuropathy associated with HIV infection

S-adenosyl-methionine (SAMe)

– – – – – – ↓ Reduces severity of depression (e.g. CGI, HAM-D scores) when added to SSRI antidepressants or venlafaxine (Alpert, 2004)

CoQ10 X X X – – – ↓ Reduced functional decline of untreated, early Parkinson’s disease at 1200 mg/day (Shultz, 2002)Tolerated at high doses (Shultz, 2008)Decreased elevated lactate levels of the basal ganglia and increased complex I activity in Huntington’s disease (Feigin et al., 1996)

Alpha-lipoic acid (ALA)

– – – – – – ↓ Significantly slowed the increase of hepatocellular oxidants , MDA levels, and lipid peroxidation in old rats (Hagen, 1999)Reversed the age-related decline of intracellular GSH and ascorbic acid levels (Hagen, 1999)

Creatine – – – – – – ↓ Oral consumption of CM can increase brain creatine and PCr concentrations (Dechent et al., 1999; Lyoo et al., 2003)

Melatonin X X X X – – ↓ Improved sleep in individuals with seasonal affective disorder (Leppamaki et al., 2003)Open-label administration had sleep-enhancing and antimanic effects in bipolar patients, with no effects on mood or sleep (Bersani and Garavini, 2000)Resulted in a significant reduction of depression symptoms compared to placebo in patients with ‘winter depression’ (Lewy et al., 1998)

ADHD: attention deficit disorder; CGI: Clinical Global Impression Scale; CM: creatine monohydrate; GSH: glutathione; HAM-D: Hamilton Depression Rating Scale; MDA: malondialdehyde; PCr: phosphocreatine; ROS: reactive oxygen species; SSRI: selective serotonin re-uptake inhibitor.

at Fund Diag.Est Imstico PARENT on August 7, 2013anp.sagepub.comDownloaded from

Page 10: Aust N Z J Psychiatry 2013 Nierenberg 26 42

34 ANZJP Articles

Australian & New Zealand Journal of Psychiatry, 47(1)

found to have a good safety profile (Hidaka et al., 2008). A more recent study investigating a higher dosage of CoQ10 in conjunction with vitamin E suggested that CoQ10 is safe and tolerated at doses up to 3000 mg/day for 2 weeks, but should not exceed 2400 mg/day since any dosage beyond this amount does not increase plasma levels of alpha-tocopherol (Shults et al., 2004). Other evidence found that while CoQ10 supplementation increased plasma levels of CoQ10 in patients with Parkinson’s disease, it had no effect on Parkinsonian symptoms after a 3-month period (Storch et al., 2007). Similarly, in an open label trial of 200 mg of daily CoQ10 supplementation, idiopathic parkinsonian patients demonstrated no significant clinical effect in a 3-month period (Strijks et al., 1997).

Clinical trials of CoQ10 in other health conditions also indicate evidence of clinical improvement with CoQ10 supplementation. In a recent meta-analysis of 12 clinical trials of the efficacy of CoQ10 supplementation in hyper-tensive patients, it was suggested that CoQ10 may lower both systolic blood pressure and diastolic blood pressure (Rosenfeldt et al., 2007). A 3-month supplementation of CoQ10 in patients with migraine showed a reduction in the frequency and disability of headaches (Hershey et al., 2007). Similarly, individuals randomized to CoQ10 supple-mentation (300 mg/day) in a double-blind, randomized, placebo-controlled trial showed improvement in symptoms of migraine including reduced attack-frequency, reduced nausea and vomiting associated with migraine, and fewer headache days over a 4-month period (Sandor et al., 2005). Such evidence suggests that the level of CoQ10 may be helpful in both understanding and treating disorders related to dysfunctional energy metabolism, particularly mood dis-orders. To our knowledge, no clinical trials have been com-pleted that have investigated the clinical value of CoQ10 for individuals with major depressive disorder, bipolar dis-order, or anxiety disorders.

Alpha-lipoic acid (ALA)

Alpha-lipoic acid (ALA) is a naturally occurring mitochon-drial coenzyme for pyruvate and alpha-ketoglutarate dehy-drogenase. It is a potent antioxidant (Packer et al., 1995) and carries out an essential role in mitochondrial energy production by recruiting glucose transporters to the cell membrane, thereby increasing the cellular uptake of glu-cose for ATP synthesis (Estrada et al., 1996).

ALA can be obtained through diet in foods such as spinach, yeast and red meats, and has been shown to have antioxidant properties when administered as a dietary sup-plement to increase the unbound fraction of the compound. Both in vitro and in vivo studies of ALA supplementation have demonstrated potent antioxidant effects (Packer et al., 1995). For example, one study found that ALA supplemen-tation slowed the increase of hepatocellular oxidants, malondialdehyde (MDA) levels and lipid peroxidation in

old rats, and reversed the age-related decline of intracellular GSH and ascorbic acid levels (Hagen et al., 1999).

In addition to improving cellular antioxidant status, ALA has been shown to reverse metabolic deficits, oxidative stress, and damage associated with aging when administered as a dietary supplement (Hagen et al., 2002a). Moreover, ALA has been shown to effectively reduce oxidative stress and ameliorate energy-related metabolic impairments, spe-cifically in type II diabetes, liver disease or age-related oxi-dative damage (Hagen et al., 2002a). Specifically, high-dose ALA administered intravenously to diabetic and control rats induced a rapid reduction of blood glucose levels and acetyl CoA content in the liver (Rudich et al., 1999), and oral doses of ALA lowered elevated hepatic and cerebral malondialde-hyde levels of older rats (Hagen et al., 2002b).

ALA supplementation has also demonstrated neuropro-tective effects against peripheral oxidative stress and dam-age from d-galactose exposure (Cui et al., 2006). Toxic levels of d-galactose induced several neurobiological changes in mice, rats, and Drosophilia, including inhibited neural progenitor cell migration, enhanced caspase-medi-ated cell apoptosis and decreased total antioxidative capa-bility (T-AOC), MDA and glutathione peroxidase (GSH-Px) activities. Treatment with ALA reversed this oxidative damage, which subsequently improved cognitive function, slowed hippocampal neurodegeneration and reversed oxi-dative damage (Cui et al., 2006).

ALA has been investigated as a potential treatment in a number of diseases thought to be associated with abnormal mitochondrial functioning and impaired energy utilization, including type II diabetes (Jacob et al., 1999) and diabetic polyneuropathy (Ametov et al., 2003; Hahm et al., 2004; Negrisanu et al., 1999; Reljanovic et al., 1999; Ruhnau et al., 1999; Tankova et al., 2004; Ziegler et al., 1995, 1999, 2006). It has also been studied in the treatment of alcohol-related liver disease (Marshall et al., 1982), burning mouth syndrome (Femiano and Scully, 2002), diabetes-related cardiac autonomic neuropathy (Ziegler et al., 1997), HIV-associated cognitive impairment (Anonymous, 1998), mul-tiple sclerosis (Yadav et al., 2005), migraine (Magis et al., 2007), peripheral arterial disease (Vincent et al., 2007) and Alzheimer’s disease (Hager et al., 2007).

Although the specific mechanism by which ALA improves mitochondrial function has not been identified, several have been discussed in the literature including: (1) increased recycling of endogenous antioxidants includ-ing CoQ10, vitamins C and E, and GSH (Liu, 2008); (2) reduction to the highly potent antioxidant dihydrolipoic acid which reacts with superoxide radicals, hypochlorous acid, peroxyl radicals, and singlet oxygen (Liu et al., 2002a); (3) chelation of heavy metals such as iron and cop-per (Liu, 2008); (4) stimulation of glucose uptake into cells leading to enhanced energy metabolism (Estrada et al., 1996); (5) anti-apoptotic and neuroprotective effects sec-ondary to prevention of glutamate-induced increases in

at Fund Diag.Est Imstico PARENT on August 7, 2013anp.sagepub.comDownloaded from

Page 11: Aust N Z J Psychiatry 2013 Nierenberg 26 42

Nierenberg et al. 35

Australian & New Zealand Journal of Psychiatry, 47(1)

intracellular calcium (Liu, 2008; Liu et al., 2002a); and (6) stimulation of mitochondrial biogenesis through upregula-tion of mitochondrial transcription factors such as peroxi-some proliferator-activated receptor-gamma coactivator-1α (PGC-1α) (Liu, 2008).

Creatine monohydrate (CM)

Creatine is a non-essential dietary element that is found in meat and fish and is synthesized endogenously by the liver and kidneys (Juhn and Tarnopolsky, 1998; Terjung et al., 2000). Exogenous creatine is widely available as a dietary supplement in the form of creatine monohydrate (CM).

Creatine is the precursor of PCr, which plays an integral role in brain and muscle energy metabolism. The primary role of PCr is to function as a reservoir of inorganic phos-phate to be drawn upon to produce ATP by the catalyzing agent creatine kinase (Ames, 2000). During periods of transient high-energy demands, PCr is rapidly converted to creatine in order to donate a high-energy phosphate to adenosine diphosphate (ADP) to maintain the overall intra-cellular concentration of ATP (Erecinska and Silver, 1989; Sauter and Rudin, 1993). While short-term decreases in PCr may reflect typical physiologic responses to acute muscular or neuronal activity, long-term abnormalities in PCr generally reflect an insufficient metabolic supply of ATP needed for normal cellular function, possibly due to underlying mitochondrial dysfunction, leading to deficits in oxidative phosphorylation (Rothman, 1994). This concept is reflected in studies of patients with mitochondrial cytopa-thies, in which reduced brain PCr concentrations have been observed (Barbiroli et al., 1993). Decreased PCr concentra-tions have also been identified in patients with bipolar dis-order (Stork and Renshaw, 2005). Notably, several studies have shown that oral consumption of CM can increase brain creatine and PCr concentrations (Dechent et al., 1999; Lyoo et al., 2003), suggesting that CM supplementation may mitigate bioenergetic abnormalities by providing addi-tional substrate for the production of ATP through the cre-atine kinase reaction.

In addition to enhancing energy production, creatine has been shown to have direct and indirect antioxidant proper-ties (Lawler et al., 2002; Tarnopolsky, 2008; Tarnopolsky and Beal, 2001). For example, creatine has been shown to reduce markers of oxidative stress in animal models of Huntington’s disease (Andreassen et al., 2001) and amyo-trophic lateral sclerosis (ALS) (Klivenyi et al., 1999). Moreover, creatine supplementation was found to have indirect antioxidant properties in an animal model of trau-matic brain injury that were associated with reduced apop-tosis, prevention of opening of the MPTP, and better maintenance of the mitochondrial membrane potential (Lawler et al., 2002; Sullivan et al., 2000).

Several clinical trials with CM in the treatment of mitochondrial cytopathies (Tarnopolsky et al., 1997),

neuromuscular disorders (Tarnopolsky et al., 2004; Walter et al., 2000) and neurodegenerative disorders such as Parkinson’s disease (Bender et al., 2006; NINDS NET-PD Investigators, 2006), Huntington’s disease (Tabrizi et al., 2005; Verbessem et al., 2003) and ALS (Groeneveld et al., 2003; Rosenfeld et al., 2008) have been performed with mixed results. CM supplementation demonstrated benefit in one small, open-label study in patients with post- traumatic stress disorder (PTSD) (Amital et al., 2006a) and was associated with improvement in depression symptoms in a patient with comorbid depression and fibromyalgia (Amital et al., 2006b). In contrast, there was no evidence of efficacy following CM treatment in a small controlled trial in schizophrenia (Kaptsan et al., 2007). A small open-label study suggested a beneficial effect of CM augmentation in patients with treatment-resistant depression (Roitman et al., 2007). Interestingly, this study enrolled two bipolar depressed patients – both of whom developed hypomania/mania after CM treatment. All in all, preclinical and clinical evidence to date encourages further exploration of the potential benefit of CM supplementation in disorders asso-ciated with mitochondrial dysfunction, including bipolar disorder.

Melatonin

Melatonin was long conceived as a hormone secreted in a tightly regulated circadian rhythm by the pineal gland, which served a primary role in reproduction. However, the recent recognition that melatonin is synthesized in many organs and tissues in the body, including the brain, has sparked exploration into its potential role in a variety of physiological processes.

Growing evidence suggests that melatonin has potent antioxidant properties that result from direct free radical scavenging properties as well as effects on mitochondrial physiology. Several studies have demonstrated that mela-tonin and several of its metabolites are major scavengers of both oxygen- and nitrogen-based ROS (Korkmaz et al., 2009; Lopez-Burillo et al., 2003; Reiter et al., 2002a, 2002b, 2003), and melatonin has been shown to stimulate the pro-duction of GSH (Albarran et al., 2001; Winiarska et al., 2006). Interestingly, melatonin has also been shown to have direct genomic effects. For example, melatonin increases the mRNA expression of genes involved in antioxidant pro-duction such as glutathione peroxidase and superoxide dis-mutase (SOD) under physiological conditions and during elevated oxidative stress (Acuna-Castroviejo et al., 2007; Rodriguez et al., 2004; Tan et al., 1998).

In addition to its antioxidant effects, evidence suggests that melatonin may have a direct benefit on mitochondrial functioning. Both in vitro and in vivo animal studies have demonstrated multiple mitochondrial enhancing effects following melatonin administration, including increased oxidative phosphorylation through activation of ETC

at Fund Diag.Est Imstico PARENT on August 7, 2013anp.sagepub.comDownloaded from

Page 12: Aust N Z J Psychiatry 2013 Nierenberg 26 42

36 ANZJP Articles

Australian & New Zealand Journal of Psychiatry, 47(1)

complexes, increased mitochondrial membrane fluidity, and closing of the MPTP (Acuna-Castroviejo et al., 2001, 2007; Garcia et al., 1997; Leon et al., 2004, 2005). Moreover, melatonin has direct effects on mitochondrial genome expression. For example, studies have shown that melatonin administration prevents oxidative degradation of mtDNA and reduction of mtDNA transcripts in several tis-sues including brain (Acuna-Castroviejo et al., 2005) and increases expression of mtDNA-coded polypeptide subu-nits I, II and III of ETC complex IV (Acuna-Castroviejo et al., 2003).

Recent exploration into the potential clinical utility of melatonin, primarily in the treatment of neurodegenerative disorders hypothesized to result, in part, from mitochon-drial dysfunction, has provided some encouraging results. Melatonin prevented neurodegenerative changes in several experimental models of Alzheimer’s disease (Lahiri et al., 2004; Pappolla et al., 1998; Reiter et al., 1999) and Huntington’s disease (Reiter et al., 1999). Furthermore, preliminary studies in Alzheimer’s disease patients have demonstrated a slowing of cognitive impairment and a reduction in sundowning following melatonin treatment (Cardinali et al., 2002). In addition, melatonin has demon-strated neuroprotective properties in animal models of Parkinson’s disease hypothesized to result from ROS scav-enging, enhancement of ETC activity and increased mito-chondrial membrane potential (Acuna-Castroviejo et al., 2007; Reiter et al., 1999).

Several small studies have evaluated the effect of mela-tonin administration in mood disorders with mixed results. A controlled study of melatonin in seasonal affective disor-der demonstrated improvement in sleep and vitality, but no reduction in atypical depression symptoms (Leppamaki et al., 2003). Patients with ‘winter depression’ demon-strated significantly reduced depression symptoms follow-ing melatonin treatment compared to placebo (Lewy et al., 1998). In contrast, melatonin was associated with worsen-ing dysphoria and reduced sleep and weight in a crossover study in patients with severe depression (Carman et al., 1976). With regard to bipolar disorder, open-label mela-tonin had no significant effects on mood or sleep in a small study of rapid-cycling bipolar disorder (Leibenluft et al., 1997) but was found to have sleep-enhancing and anti-manic effects in a small open-label study in bipolar mania (Bersani and Garavini, 2000).

Interestingly, agomelatine, a potent agonist of melatonin MT1 and MT2 receptors, has demonstrated preliminary evi-dence of efficacy in bipolar depression (Calabrese et al., 2007). However, the antioxidant and mitochondrial-enhanc-ing properties of melatonin are speculated to result primarily from direct free radical scavenging and actions linked to cytosolic proteins rather than receptor-mediated mechanisms (Acuna-Castroviejo et al., 2007). Nevertheless, further exploration into the potential for melatonin-receptor modula-tors to enhance mitochondrial functioning is warranted.

Rationale for considering combinations of mitochondrial modulators

While convergent data strongly support the hypothesis that mitochondrial dysfunction occurs in bipolar disorder, mul-tiple abnormalities along the mitochondrial ETC can occur, making it improbable that, for any given patient, the correct abnormality can be practically identified with currently available technology. In addition, while compounds such as ALCAR, SAMe and CM may increase energy production, this increase may also cause an increase in the production of ROS (Hagen et al., 2002a). Mitochondrial DNA lack his-tones and are therefore extremely susceptible to damage from ROS. This feature sets up a vicious cycle in which increased ROS generation leads to further damage to mito-chondrial DNA, proteins and lipids, which can further exacerbate defects in mitochondrial energy production.

Therefore, effective mitochondrial enhancement strate-gies may require several agents that both modulate multiple mitochondrial targets and increase mitochondrial metabolic activity without a concomitant increase in oxidative stress – a strategy that has been proposed previously in the treat-ment of mitochondrial disorders (Rodriguez et al., 2007; Tarnopolsky, 2008).

This ‘mitochondrial cocktail’ could include several of the agents described above. For example, whereas NAC targets complex I in the ETC and reduces oxidative stress, ALCAR targets complexes I, II and IV, as well as enhanc-ing oxidation of fatty acids. Therefore, we reason that the combination treatment of NAC with ALCAR is more likely to modulate multiple mitochondrial targets while mitigat-ing the resulting oxidative stress from ALCAR-induced mitochondrial energy production (Hagen et al., 2002a) by increasing GSH via NAC supplementation. One study found improved cognitive functions and decreased aggres-sion after treatment with a combination of NAC, ALCAR and SAMe in mouse models (Shea, 2007), providing sup-port for this strategy.

Given the enhancing effects of ALCAR on cerebral energy metabolism and the potent antioxidant properties of ALA, we theorize that a combination of ALCAR and ALA might also be effective in increasing mitochondrial meta-bolic activity without a concomitant increase in oxidative stress – as others have proposed (Hagen et al., 2002a; Soczynska et al., 2008). Animal studies have demonstrated that the combination treatment of ALCAR and ALA improves mitochondrial functioning by increasing metabo-lism and lowering oxidative stress more than either com-pound alone (Hagen et al., 2002a; Liu et al., 2002a, 2002b), providing preclinical support for this rationale.

The aim of this review is to support the rationale for rigorous studies of MMs (and combinations) in bipolar dis-order. While clinicians could recommend such treatments currently, rigorous data are lacking in most instances to do

at Fund Diag.Est Imstico PARENT on August 7, 2013anp.sagepub.comDownloaded from

Page 13: Aust N Z J Psychiatry 2013 Nierenberg 26 42

Nierenberg et al. 37

Australian & New Zealand Journal of Psychiatry, 47(1)

so. Of note, combinations of MMs are likely to be well tolerated. For all the individual dietary supplements reviewed here, the available literature suggests adequate tolerability and low side-effect burden. While some of these dietary supplements can be costly (and none is reim-bursed by insurance), it is unlikely that cost considerations will limit the availability of such treatments, especially after proof of clinical efficacy that will be obtained through rigorous clinical trials.

Summary

While mitochondrial disorders can present in any system, the central nervous system is particularly vulnerable to dys-regulations of mitochondrial function since it has high energy demands. Because convergent data implicate subtle mitochondrial dysfunctions as an important component of the pathophysiology of bipolar disorder, it makes sense to consider clinical trials of substances that have the poten-tial to ameliorate those dysfunctions. We reviewed reason-able candidates for modulating mitochondrial function: N-acetyl-cysteine (NAC), acetyl-L-carnitine (ALCAR), S-adenosylmethionine (SAMe), coenzyme Q10 (CoQ10), alpha-lipoic acid (ALA), creatine monohydrate (CM) and melatonin, and postulated that combinations of two or even more of these MMs could be of interest. These MMs should be considered for evaluation in rigorous, well-designed, well-powered clinical trials to improve the long-term course of bipolar disorder.

Funding

This research received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.

Declaration of interest

The authors report no conflicts of interest. The authors alone are responsible for the content and writing of the paper.

ReferencesAcuna-Castroviejo D, Escames G, Leon J, et al. (2003) Mitochondrial

regulation by melatonin and its metabolites. Advances in Experimental Medicine and Biology 527: 549–557.

Acuna-Castroviejo D, Escames G, Lopez LC, et al. (2005) Melatonin and nitric oxide: Two required antagonists for mitochondrial homeostasis. Endocrine 27: 159–168.

Acuna-Castroviejo D, Escames G, Rodriguez MI, et al. (2007) Melatonin role in the mitochondrial function. Frontiers in Bioscience 12: 947–963.

Acuna-Castroviejo D, Martin M, Macias M, et al. (2001) Melatonin, mito-chondria, and cellular bioenergetics. Journal of Pineal Research 30: 65–74.

Albarran MT, Lopez-Burillo S, Pablos MI, et al. (2001) Endogenous rhythms of melatonin, total antioxidant status and superoxide dis-mutase activity in several tissues of chick and their inhibition by light. Journal of Pineal Research 30: 227–233.

Aliev G, Liu J, Shenk JC, et al. (2009) Neuronal mitochondrial ameliora-tion by feeding acetyl-L-carnitine and lipoic acid to aged rats. Journal of Cellular and Molecular Medicine 13: 320–333.

Al-Majed AA, Sayed-Ahmed MM, Al-Omar FA, et al. (2006) Carnitine esters prevent oxidative stress damage and energy depletion follow-ing transient forbrain ischaemia in the rat hippocampus. Clinical and Experimental Pharmacology and Physiology 33: 725–733.

Alpert JE, Papakostas G, Mischoulon D, et al. (2004) S-adenosyl-L-methionine (SAMe) as an adjunct for resistant major depressive disorder – An open trial following partial or nonresponse to selec-tive serotonin reuptake inhibitors or venlafaxine. Journal of Clinical Psychopharmacology 24: 661–664.

Alvarez E, Udina C and Guillamat R (1987) Shortening of latency period in depressed patients treated with SAMe and other antidepressant drugs. Cellular Biological Review S1: 103–110.

Ames A 3rd (2000) CNS energy metabolism as related to function. Brain Research. Brain Research Reviews 34: 42–68.

Ames BN and Liu J (2004) Delaying the mitochondrial decay of aging with acetylcarnitine. Annals of the New York Academy of Sciences 1033: 108–116.

Ametov AS, Barinov A, Dyck PJ, et al. (2003) The sensory symptoms of diabetic polyneuropathy are improved with alpha-lipoic acid: The SYDNEY trial. Diabetes Care 26: 770–776.

Amital D, Vishne T, Roitman S, et al. (2006a) Open study of creatine monohydrate in treatment-resistant posttraumatic stress disorder. Journal of Clinical Psychiatry 67: 836–837.

Amital D, Vishne T, Rubinow A, et al. (2006b) Observed effects of cre-atine monohydrate in a patient with depression and fibromyalgia. American Journal of Psychiatry 163: 1840–1841.

Andreassen OA, Dedeoglu A, Ferrante RJ, et al. (2001) Creatine increase survival and delays motor symptoms in a transgenic animal model of Huntington’s disease. Neurobiology of Disease 8: 479–491.

Anonymous (1998) A randomized, double-blind, placebo-controlled trial of deprenyl and thioctic acid in human immunodeficiency virus-associ-ated cognitive impairment. Dana Consortium on the Therapy of HIV Dementia and Related Cognitive Disorders. Neurology 50: 645–651.

Arnold LE, Amato A, Bozzolo H, et al. (2007) Acetyl-L-carnitine (ALC) in attention-deficit/hyperactivity disorder: A multi-site, placebo-controlled pilot trial. Journal of Child and Adolescent Psychopharmacology 17: 791–802.

Atkuri KR, Mantovani JJ, Herzenberg LA, et al. (2007) N-Acetylcysteine—A safe antidote for cysteine/glutathione deficiency. Current Opinion in Pharmacology 7: 355–359.

Aureli T, Miccheli A, Ricciolini R, et al. (1990) Aging brain: Effect of acetyl-L-carnitine treatment on rat brain energy and phospholipid metabolism. A study by 31P and 1H NMR spectroscopy. Brain Research 526: 108–112.

Barbiroli B, Montagna P, Martinelli P, et al. (1993) Defective brain energy metabolism shown by in vivo 31P MR spectroscopy in 28 patients with mitochondrial cytopathies. Journal of Cerebral Blood Flow and Metabolism 13: 469–474.

Bella R, Biondi R, Raffaele R, et al. (1990) Effect of acetyl-L-carnitine on geriatric patients suffering from dysthymic disorders. International Journal of Clinical Pharmacology Research 10: 355–360.

Bender A, Koch W, Elstner M, et al. (2006) Creatine supplementation in Parkinson disease: A placebo-controlled randomized pilot trial. Neurology 67: 1262–1264.

Benes FM, Matzilevich D, Burke RE, et al. (2006) The expression of proa-poptosis genes is increased in bipolar disorder, but not in schizophre-nia. Molecular Psychiatry 11: 241–251.

Berk M, Copolov D, Dean O, et al. (2008a) N-Acetyl cysteine as a glu-tathione precursor for schizophrenia—A double-blind, randomized, placebo-controlled trial. Biological Psychiatry 64: 361–368.

Berk M, Copolov DL, Dean O, et al. (2008b) N-Acetyl cysteine for depres-sive symptoms in bipolar disorder—A double-blind randomized pla-cebo-controlled trial. Biological Psychiatry 64: 468–475.

Berlanga C, Ortegasoto HA, Ontiveros M, et al. (1992) Efficacy of S-adenosyl-L-methionine in speeding the onset of action of imipra-mine. Psychiatry Research 44: 257–262.

at Fund Diag.Est Imstico PARENT on August 7, 2013anp.sagepub.comDownloaded from

Page 14: Aust N Z J Psychiatry 2013 Nierenberg 26 42

38 ANZJP Articles

Australian & New Zealand Journal of Psychiatry, 47(1)

Bernardi P, Colonna R, Costantini P, et al. (1998) The mitochondrial per-meability transition. Biofactors 8: 273–281.

Bersani G and Garavini A (2000) Melatonin add-on in manic patients with treatment resistant insomnia. Progress in Neuro-Psychopharmacology & Biological Psychiatry 24: 185–191.

Bertolino A, Frye M, Callicott JH, et al. (2003) Neuronal pathology in the hippocampal area of patients with bipolar disorder: A study with pro-ton magnetic resonance spectroscopic imaging. Biological Psychiatry 53: 906−913.

Bonavita E (1986) Study of the efficacy and tolerability of L-acetylcarnitine therapy in the senile brain. International Journal of Clinical Pharmacology, Therapy, and Toxicology 24: 511–516.

Bottiglieri T (1996) Folate, vitamin B-12, and neuropsychiatric disorders. Nutrition Reviews 54: 382–390.

Bottiglieri T, Laundy M, Crellin R, et al. (2000) Homocysteine, folate, methylation, and monoamine metabolism in depression. Journal of Neurology Neurosurgery and Psychiatry 69: 228–232.

Calabrese JR, Guelfi JD and Perdrizet-Chevallier C; Agomelatine Bipolar Study Group (2007) Agomelatine adjunctive therapy for acute bipolar depression: Preliminary open data. Bipolar Disorders 9: 628–635.

Cardinali DP, Brusco LI, Liberczuk C, et al. (2002) The use of melatonin in Alzheimer’s disease. Neuro Endocrinology Letters 23 Suppl 1: 20–23.

Carman JS, Post RM, Buswell R, et al. (1976) Negative effects of mela-tonin on depression. American Journal of Psychiatry 133: 1181–1186.

Carney MW, Chary TK, Bottiglieri T, et al. (1989) The switch mechanism and the bipolar/unipolar dichotomy. British Journal of Psychiatry 154: 48–51.

Cataldo AM, McPhie DL, Lange NT, et al. (2010) Abnormalities in mitochondrial structure in cells from patients with bipolar disorder. American Journal of Pathology 177: 575–585.

Chang YYC, Rapoport SI and Rao JS (2009) Chronic administration of mood stabilizers upregulates BDNF and Bcl-2 expression levels in rat frontal cortex. Neurochemical Research 34: 536–541.

Chen G, Zeng WZ, Yuan PX, et al. (1999) The mood-stabilizing agents lithium and valproate robustly increase the levels of the neuropro-tective protein bcl-2 in the CNS. Journal of Neurochemistry 72: 879–882.

Clark J (1998) N-Acetyl aspartate: A marker for neuronal loss or mito-chondrial dysfunction. Developmental Neuroscience 20: 271–276.

Clay HB, Sillivan S and Konradi C (2011) Mitochondrial dysfunction and pathology in bipolar disorder and schizophrenia. International Journal of Developmental Neuroscience 29: 311–324.

Cui J, Shao L, Young LT, et al. (2007) Role of glutathione in neuropro-tective effects of mood stabilizing drugs lithium and valproate. Neuroscience 144: 1447–1453.

Cui X, Zuo P, Zhang Q, et al. (2006) Chronic systemic d-galactose expo-sure induces memory loss, neurodegeneration, and oxidative damage in mice: Protective effects of R-a-lipoic acid. Journal of Neuroscience Research 83: 1584–1590.

Culmsee C and Mattson M (2005) p53 in neuronal apoptosis. Biochemical and Biophysical Research Communications 331: 761–777.

Dager SR, Friedman SD, Parow A, et al. (2004) Brain metabolic altera-tions in medication-free patients with bipolar disorder. Archives of General Psychiatry 61: 450–458.

Dechent P, Pouwels PJ, Wilken B, et al. (1999) Increase of total creatine in human brain after oral supplementation of creatine-monohydrate. American Journal of Physiology 277: R698–R704.

De Grandis D and Minardi C (2002) Acetyl-L-carnitine (levacecarnine) in the treatment of diabetic neuropathy. A long-term, randomised, double-blind, placebo-controlled study. Drugs in R & D 3: 223–231.

Deicken R, Pegues M, Anzalone S, et al. (2003). Lower concentration of hippocampal N-acetylaspartate in familial bipolar I disorder. American Journal of Psychiatry 160: 873–882.

Eleff S, Barker P, Blackband S, et al. (1990) Phosphorus magnetic res-onance spectroscopy of patients with mitochondrial cytopathies

demonstrates decreased levels of brain phosphocreatine. Annals of Neurology 27: 626–630.

Elmslie JL, Porter RJ, Joyce PR, et al. (2006) Carnitine does not improve weight loss outcomes in valproate-treated bipolar patients consuming an energy-restricted, low-fat diet. Bipolar Disorders 8: 503–507.

Erecinska M and Silver IA (1989) ATP and brain function. Journal of Cerebral Blood Flow and Metabolism 9: 2–19.

Estrada DE, Ewart HS, Tsakiridis T, et al. (1996) Stimulation of glu-cose uptake by the natural coenzyme alpha-lipoic acid/thioctic acid: Participation of elements of the insulin signaling pathway. Diabetes 45: 1798–1804.

Feigin A, Kieburtz K, Como P, et al. (1996) Assessment of coenzyme Q10 tolerability in Huntington’s disease. Movement Disorders 11: 321–323.

Femiano F and Scully C (2002) Burning mouth syndrome (BMS): Double blind controlled study of alpha-lipoic acid (thioctic acid) therapy. Journal of Oral Pathology & Medicine 31: 267–269.

Ferreira MAR, O’Donovan MC, Meng YA, et al. (2008) Collaborative genome-wide association analysis supports a role for ANK3 and CACNA1C in bipolar disorder. Nature Genetics 40: 1056–1058.

Frey BN, Stanley JA, Nery FG, et al. (2007a) Abnormal cellular energy and phospholipid metabolism in the left dorsolateral prefrontal cortex of medication-free individuals with bipolar disorder: An in vivo 1H MRS study. Bipolar Disorders 9: 119–127.

Frey BN, Walss-Bass C, Stanley JA, et al. (2007b) Brain-derived neu-rotrophic factor val66met polymorphism affects prefrontal energy metabolism in bipolar disorder. Neuroreport 18: 1567–1570.

Fulgente T, Onofrj M, Del Re ML, et al. (1990) Laevo-acetylcarnitine (Nicetile) treatment of senile depression. Clinical Trials Journal 27: 155–163.

Garcia JJ, Reiter RJ, Guerrero JM, et al. (1997) Melatonin prevents changes in microsomal membrane fluidity during induced lipid per-oxidation. FEBS Letters 408: 297–300.

Garzya G, Corallo D, Fiore A, et al. (1990) Evaluation of the effects of L-acetylcarnitine on senile patients suffering from depression. Drugs Under Experimental and Clinical Research 16: 101–106.

Gecele M, Francesetti G and Meluzzi A (1991) Acetyl-L-carnitine in aged subjects with major depression: Clinical efficacy and effects on the circadian rhythym of cortisol. Dementia 2: 333–337.

Ghaemi SN, Hsu DJ, Thase ME, et al. (2006) Pharmacological treat-ment patterns at study entry for the first 500 STEP-BD participants. Psychiatric Services 57: 660–665.

Grant JE, Kim SW and Odlaug BL (2007) N-Acetyl cysteine, a glutamate-modulating agent, in the treatment of pathological gambling: A pilot study. Biological Psychiatry 62: 652–657.

Groeneveld GJ, Veldink JH, van der Tweel I, et al. (2003) A randomized sequential trial of creatine in amyotrophic lateral sclerosis. Annals of Neurology 53: 437–445.

Hagen T, Liu J, Lykkesfeldt J, et al. (2002a) Feeding acetyl-L-carnitine and lipoic acid to old rats significantly improves metabolic func-tion while decreasing oxidative stress. Proceedings of the National Academy of Sciences of the United States of America 99: 1870–1875.

Hagen TM, Ingersoll RT, Lykkesfeldt J, et al. (1999) (R)-Alpha-lipoic acid-supplemented old rats have improved mitochondrial func-tion, decreased oxidative damage, and increased metabolic rate. FASEB Journal 13: 411–418.

Hagen TM, Moreau R, Suh JH, et al. (2002b) Mitochondrial decay in the aging rat heart. Annals of the New York Academy of Sciences 959: 491–507.

Hager K, Kenklies M, McAfoose J, et al. (2007) Alpha-lipoic acid as a new treatment option for Alzheimer’s disease—A 48 months follow-up analysis. Journal of Neural Transmission. Supplement: 189–193.

Hahm JR, Kim BJ and Kim KW (2004) Clinical experience with thioctacid (thioctic acid) in the treatment of distal symmetric polyneuropathy in Korean diabetic patients. Journal of Diabetes and its Complications 18: 79–85.

at Fund Diag.Est Imstico PARENT on August 7, 2013anp.sagepub.comDownloaded from

Page 15: Aust N Z J Psychiatry 2013 Nierenberg 26 42

Nierenberg et al. 39

Australian & New Zealand Journal of Psychiatry, 47(1)

Hamakawa H, Kato T, Murashita J, et al. (1998) Quantitative proton mag-netic resonance spectroscopy of the basal ganglia in patients with affective disorders. European Archives of Psychiatry and Clinical Neuroscience 248: 53–58.

Hamakawa H, Murashita J, Yamada N, et al. (2004) Reduced intracellular pH in the basal ganglia and whole brain measured by P-MRS in bipo-lar disorder. Psychiatry Clinical Neuroscience 58: 53–59.

Hao Y, Basile AS, Chen G, et al. (2004) Glutamate-induced over-expres-sion of GAD is down-regulated by acetyl-L-carnitine in rat islet cells. Endocrine Research 30: 107–116.

Hart AM, Wilson AD, Montovani C, et al. (2004) Acetyl-L-carnitine: A pathogenesis based treatment for HIV-associated antiretroviral toxic neuropathy. AIDS 18: 1549–1560.

Hershey AD, Powers SW, Vockell ALB, et al. (2007) Coenzyme Q10 defi-ciency and response to supplementation in pediatric and adolescent migraine. Headache 47: 73–80.

Hidaka T, Fujii K, Funahashi I, et al. (2008) Safety assessment of coen-zyme Q_10 (CoQ_10). Biofactors 32: 199–208.

Hoppel C (2003) The role of carnitine in normal and altered fatty acid metabolism. American Journal of Kidney Diseases 41: S4–S12.

Jacob S, Ruus P, Hermann R, et al. (1999) Oral administration of RAC-alpha-lipoic acid modulates insulin sensitivity in patients with type-2 diabetes mellitus: A placebo-controlled pilot trial. Journal of Free Radicals in Biology & Medicine 27: 309–314.

Judd LL, Schettler PJ, Akiskal HS, et al. (2008) Residual symptom recov-ery from major affective episodes in bipolar disorders and rapid epi-sode relapse/recurrence. Archives of General Psychiatry 65: 386–394.

Juhn MS and Tarnopolsky M (1998) Oral creatine supplementation and athletic performance: A critical review. Clinical Journal of Sport Medicine 8: 286–297.

Kapczinski F, Vieta E, Andreazza AC, et al. (2008) Allostatic load in bipolar disorder: Implications for pathophysiology and treatment. Neuroscience and Biobehavioral Reviews 32: 675–692.

Kaptsan A, Odessky A, Osher Y, et al. (2007) Lack of efficacy of 5 grams daily of creatine in schizophrenia: A randomized, double-blind, pla-cebo-controlled trial. Journal of Clinical Psychiatry 68: 881–884.

Kato T (2008) Role of mitochondrial DNA in calcium signaling abnormal-ity in bipolar disorder. Cell Calcium 44: 92–102.

Kato T, Murashita J, Kamiya A, et al. (1998) Decreased brain intracellular pH measured by P-31-MRS in bipolar disorder: A confirmation in drug-free patients and correlation with white matter hyperintensity. European Archives of Psychiatry and Clinical Neuroscience 248: 301–306.

Kato T, Shioiri T, Murashita J, et al. (1995) Lateralized abnormality of high energy phosphate metabolism in the frontal lobes of patients with bipolar disorder detected by phase-encoded 31P-MRS. Psychological Medicine 25: 557−566.

Kato T, Takahashi S, Shioiri T, et al. (1994) Reduction of brain phospho-creatine in bipolar II disorder detected by phosphorus-31 magnetic resonance spectroscopy. Journal of Affective Disorders 31: 125–133.

Kazuno A, Munakata K, Kato N, et al. (2008) Mitochondrial DNA-dependent effects of valproate on mitochondrial calcium lev-els in transmitochondrial cybrids. International Journal of Neuropsychopharmacology 11: 71–78.

Klivenyi P, Ferrante RJ, Matthews RT, et al. (1999) Neuroprotective effects of creatine in a transgenic animal model of amyotrophic lateral sclerosis. Nature Medicine 5: 347–350.

Konradi C, Eaton M, MacDonald ML, et al. (2004) Molecular evidence for mitochondrial dysfunction in bipolar disorder. Archives of General Psychiatry 61: 301–308.

Korkmaz A, Reiter RJ, Topal T, et al. (2009) Melatonin: An established antioxidant worthy of use in clinical trials. Molecular Medicine 15: 43–50.

Kwong LK, Kamzalov S, Rebrin I, et al. (2002) Effects of coenzyme Q10 administration on its tissue concentrations, mitochondrial oxidant generation, and oxidative stress in the rat. Free Radical Biology and Medicine 33: 627–638.

Lafer B, Renshaw PF, Sachs G, et al. (1994) Proton MRS of the basal ganglia in bipolar disorder. Biological Psychiatry 35: 685.

Lahiri DK, Chen D, Ge YW, et al. (2004) Dietary supplementation with melatonin reduces levels of amyloid beta-peptides in the murine cer-ebral cortex. Journal of Pineal Research 36: 224–231.

Lawler JM, Barnes WS, Wu G, et al. (2002) Direct antioxidant properties of creatine. Biochemical and Biophysical Research Communications 290: 47–52.

Leibenluft E, Feldman-Naim S, Turner EH, et al. (1997) Effects of exog-enous melatonin administration and withdrawal in five patients with rapid-cycling bipolar disorder. Journal of Clinical Psychiatry 58: 383–388.

Leon J, Acuña-Castroviejo D, Escames G, et al. (2005) Melatonin miti-gates mitochondrial malfunction. Journal of Pineal Research 38: 1–9.

Leon J, Acuña-Castroviejo D, Sainz RM, et al. (2004) Melatonin and mitochondrial function. Life Sciences 75: 765–790.

Leppamaki S, Partonen T, Vakkuri O, et al. (2003) Effect of controlled-release melatonin on sleep quality, mood, and quality of life in subjects with seasonal or weather-associated changes in mood and behaviour. European Neuropsychopharmacology 13: 137–145.

Lewy AJ, Bauer VK, Cutler NL, et al. (1998) Melatonin treatment of win-ter depression: A pilot study. Psychiatry Research 77: 57–61.

Lipinski JF, Cohen BM, Frankenburg F, et al. (1984) Open trial of S-adenosylmethionine for treatment of depression. American Journal of Psychiatry 141: 448–450.

Liu J (2008) The effects and mechanisms of mitochondrial nutrient alpha-lipoic acid on improving age-associated mitochondrial and cognitive dysfunction: An overview. Neurochemical Research 33: 194–203.

Liu J, Atamna H, Kuratsune H, et al. (2002a) Delaying brain mito-chondrial decay and aging with mitochondrial antioxidants and metabolites. Annals of the New York Academy of Sciences 959: 133–166.

Liu J, Head E, Gharib AM, et al. (2002b) Memory loss in old rats is associated with brain mitochondrial decay and RNA/DNA oxida-tion: Partial reversal by feeding acetyl-L-carnitine and/or R-alpha-lipoic acid. Proceedings of the National Academy of Sciences of the United States of America 99: 2356–2361.

Lopez-Burillo S, Tan DX, Mayo JC, et al. (2003) Melatonin, xanthurenic acid, resveratrol, EGCG, vitamin C and alpha-lipoic acid differen-tially reduce oxidative DNA damage induced by Fenton reagents: A study of their individual and synergistic actions. Journal of Pineal Research 34: 269–277.

Lyoo IK, Kong SW, Sung SM, et al. (2003) Multinuclear magnetic reso-nance spectroscopy of high-energy phosphate metabolites in human brain following oral supplementation of creatine-monohydrate. Psychiatry Research 123: 87–100.

Magis D, Ambrosini A, Sandor P, et al. (2007) A randomized double-blind placebo-controlled trial of thioctic acid in migraine prophylaxis. Headache 47: 52–57.

Malaguarnera M, Gargante MP, Cristaldi E, et al. (2008) Acetyl-L-carnitine treatment in minimal hepatic encephalopathy. Digestive Diseases and Sciences 53: 3018–3025.

Malaguarnera M, Pistone G, Astuto M, et al. (2006) Effects of L-acetylcarnitine on cirrhotic patients with hepatic coma: Randomized double-blind, placebo-controlled trial. Digestive Diseases and Sciences 51: 2242–2247.

Manji H, Moore G and Chen G (2000) Clinical and preclinical evi-dence for the neurotrophic effects of mood stabilizers: Implications for the pathophysiology and treatment of manic-depressive illness. Biological Psychiatry 48: 740–754.

Marshall AW, Graul RS, Morgan MY, et al. (1982) Treatment of alco-hol-related liver disease with thioctic acid: A six month randomised double-blind trial. Gut 23: 1088–1093.

Mathew SJ, Manji HK and Charney DS (2008) Novel drugs and thera-peutic targets for severe mood disorders. Neuropsychopharmacology 33: 2080–2092.

at Fund Diag.Est Imstico PARENT on August 7, 2013anp.sagepub.comDownloaded from

Page 16: Aust N Z J Psychiatry 2013 Nierenberg 26 42

40 ANZJP Articles

Australian & New Zealand Journal of Psychiatry, 47(1)

Mattson M and Liu D (2003) Mitochondrial potassium channels and uncoupling proteins in synaptic plasticity and neuronal cell death. Biochemical and Biophysical Research Communications 304: 539–549.

Mayer M and Noble M (1994) N-acetyl-L-cysteine is a pluripotent pro-tector against cell death and enhancer of trophic factor-mediated cell survival in vitro. Proceedings of the National Academy of Sciences of the United States of America 91: 7496–7500.

Merikangas KR, Akiskal HS, Angst J, et al. (2007) Lifetime and 12-month prevalence of bipolar spectrum disorder in the National Comorbidity Survey replication. Archives of General Psychiatry 64: 543–552.

Modica-Napolitano J and Renshaw PF (2004) Ethanolamine and phos-phoethanolamine inhibit mitochondrial function in vitro: Implications for mitochondrial dysfunction hypothesis in depression and bipolar disorder. Biological Psychiatry 55: 273–277.

Moore CM, Breeze JL, Gruber SA, et al. (2000) Choline, myo-inositol and mood in bipolar disorder: A proton magnetic resonance spectroscopic imaging study of the anterior cingulate cortex. Bipolar Disorders 2: 207–216.

Müller D, de Luca V, Sicard T, et al. (2006) Brain-derived neurotrophic factor (BDNF) gene and rapid-cycling bipolar disorder: Family-based association study. British Journal of Psychiatry 189: 317–323.

Munakata K, Fujii K, Nanko S, et al. (2007) Sequence and functional anal-yses of mtDNA in a maternally inherited family with bipolar disor-der and depression. Mutation Research/Fundamental and Molecular Mechanisms of Mutagenesis 617: 119–124.

Munakata K, Tanaka M, Mori K, et al. (2004) Mitochondrial DNA 3644T→C mutation associated with bipolar disorder. Genomics 84: 1041–1050.

Murphy A, Bredesen D, Cortopassi G, et al. (1996) Bcl-2 potentiates the maximal calcium uptake capacity of neural cell mitochondria. Proceedings of the National Academy of Sciences of the United States of America 93: 9893–9898.

Nasca D, Zurria G and Aguglia E (1989) Action of acetyl-L-carnitine in association with mianserine on depressed old people. New Trends in Clinical Neuropharmacology 3: 225–230.

Naydenov AV, MacDonald ML, Ongur D, et al. (2007) Differences in lym-phocyte electron transport gene expression levels between subjects with bipolar disorder and normal controls in response to glucose dep-rivation stress. Archives in General Psychiatry 64: 555–564.

Negrişanu G, Roşu M, Bolte B, et al. (1999) Effects of 3-month treatment with the antioxidant alpha-lipoic acid in diabetic peripheral neuropa-thy. Romanian Journal of Internal Medicine 37: 297–306.

Ng F, Berk M, Dean O, et al. (2008) Oxidative stress in psychiatric dis-orders: Evidence base and therapeutic implications. International Journal Neuropsychopharmacology 11: 851–876.

Nicholls D and Ward M (2000) Mitochondrial membrane potential and neuronal glutamate excitotoxicity: Mortality and millivolts. Trends in Neuroscience 23: 166–174.

Nicoletti VG, Marino VM, Cuppari C, et al. (2005) Effect of antioxidant diets and mitochondrial gene expression in rat brain during aging. Neurochemical Research 30: 737–752.

NINDS NET-PD Investigators (2006) A randomized, double-blind, futility clinical trial of creatine and minocycline in early Parkinson disease. Neurology 66: 664–671.

Packer L, Witt EH and Tritschler HJ (1995) alpha-Lipoic acid as a biologi-cal antioxidant. Free Radical Biology & Medicine 19: 227–250.

Papakostas GI, Mischoulon D, Shyu I, et al. (2010) S-adenosyl methio-nine (SAMe) augmentation of serotonin reuptake inhibitors for antidepressant nonresponders with major depressive disorder: A dou-ble-blind, randomized clinical trial. American Journal of Psychiatry 167: 942–948.

Pappolla M, Bozner P, Soto C, et al. (1998) Inhibition of Alzheimer beta-fibrillogenesis by melatonin. Journal of Biological Chemistry 273: 7185–7188.

Passeri M, Cucinotta D, Bonati PA, et al. (1990) Acetyl-L-carnitine in the treatment of mildly demented elderly patients. International Journal of Clinical Pharmacology Research 10: 75–79.

Patel T and Clark J (1979) Synthesis of N-acetyl-L-aspartate by rat brain mitochondria and its involvement in mitochondrial/ cytosolic carbon transport. Biochemical Journal 184: 539–546.

Perlis RH, Ostacher MJ, Patel JK, et al. (2006) Predictors of recurrence in bipolar disorder: Primary outcomes from the systematic treatment enhancement program for bipolar disorder (STEP-BD). American Journal of Psychiatry 163: 217–224.

Peters A, Schweiger U, Pellerin L, et al. (2004) The selfish brain: Competition for energy resources. Neuroscience and Biobehavioral Reviews 28: 143–180.

Pettegrew JW, Klunk WE, Panchalingam K, et al. (1995) Clinical and neurochemical effects of acetyl-L-carnitine in Alzheimer’s disease. Neurobiology of Aging 16: 1–4.

Pettegrew JW, Levine J, Gershon S, et al. (2002) 31P-MRS study of acetyl-L-carnitine treatment in geriatric depression: Preliminary results. Bipolar Disorders 4: 61–66.

Purdon AD and Rapoport SI (1998) Energy requirements for two aspects of phospholipid metabolism in mammalian brain. Biochemical Journal 335: 313–318.

Quiroz JA, Gray NA, Kato T, et al. (2008) Mitochondrially mediated plasticity in the pathophysiology and treatment of bipolar disorder. Neuropsychopharmacology 33: 2551–2565.

Rebouche CJ (2004) Kinetics, pharmacokinetics, and regulation of L-carnitine and acetyl-L-carnitine metabolism. Annals of the New York Academy of Sciences 1033: 30–41.

Reiter RJ, Cabrera J, Sainz RM, et al. (1999) Melatonin as a pharma-cological agent against neuronal loss in experimental models of Huntington’s disease, Alzheimer’s disease and parkinsonism. Annals of the New York Academy of Sciences 890: 471–485.

Reiter RJ, Tan DX and Allegra M (2002a) Melatonin: Reducing molecular pathology and dysfunction due to free radicals and associated reac-tants. Neuro Endocrinology Letters 23 Suppl 1: 3–8.

Reiter RJ, Tan DX and Burkhardt S (2002b) Reactive oxygen and nitro-gen species and cellular and organismal decline: Amelioration with melatonin. Mechanisms of Ageing and Development 123: 1007–1019.

Reiter RJ, Tan DX, Manchester LC, et al. (2003) Melatonin: Detoxification of oxygen and nitrogen-based toxic reactants. Advances in Experimental Medicine and Biology 527: 539–548.

Reljanovic M, Reichel G, Rett K, et al. (1999) Treatment of diabetic polyneuropathy with the antioxidant thioctic acid (alpha-lipoic acid): A two year multicenter randomized double-blind placebo-controlled trial (ALADIN II). Alpha Lipoic Acid in Diabetic Neuropathy. Free Radical Research 31: 171–179.

Richer C, Park J-W, Ames BN (1988) Normal oxidative damage to mito-chondrial and nuclear DNA is extensive. Proceedings of the National Academy of Sciences of the United States of America 85: 6465–6467.

Rodriguez C, Mayo JC, Sainz RM, et al. (2004) Regulation of antioxidant enzymes: A significant role for melatonin. Journal of Pineal Research 36: 1–9.

Rodriguez MC, MacDonald JR, Mahoney DJ, et al. (2007) Beneficial effects of creatine, CoQ10, and lipoic acid in mitochondrial disorders. Muscle & Nerve 35: 235–242.

Roitman S, Green T, Osher Y, et al. (2007) Creatine monohydrate in resist-ant depression: A preliminary study. Bipolar Disorders 9: 754–758.

Rosenfeld J, King RM, Jackson CE, et al. (2008) Creatine monohydrate in ALS: Effects on strength, fatigue, respiratory status and ALSFRS. Amyotrophic Lateral Sclerosis 9: 266–272.

Rosenfeldt FL, Haas SJ, Krum H, et al. (2007) Coenzyme Q(10) in the treatment of hypertension: A meta-analysis of the clinical trials. Journal of Human Hypertension 21: 297–306.

Rothman DL (1994) 1H NMR studies of human brain metabolism and physiology. In: Gilles RJ (ed) NMR in Physiology and Biomedicine. San Diego, CA: Academic Press, pp.353–372.

at Fund Diag.Est Imstico PARENT on August 7, 2013anp.sagepub.comDownloaded from

Page 17: Aust N Z J Psychiatry 2013 Nierenberg 26 42

Nierenberg et al. 41

Australian & New Zealand Journal of Psychiatry, 47(1)

Rudich A, Tirosh A, Potashnik R, et al. (1999) Lipoic acid protects against oxidative stress induced impairment in insulin stimulation of protein kinase B and glucose transport in 3T3-L1 adipocytes. Diabetologia 42: 949–957.

Ruhnau KJ, Meissner HP, Finn JR, et al. (1999) Effects of 3-week oral treatment with the antioxidant thioctic acid (alpha-lipoic acid) in symptomatic diabetic polyneuropathy. Diabetic Medicine 16: 1040–1043.

Salvioli G and Neri M (1994) L-Acetylcarnitine treatment of men-tal decline in the elderly. Drugs Under Experimental and Clinical Research 20: 169–176.

Sandor PS, Di Clemente L, Coppola G, et al. (2005) Efficacy of coen-zyme Q10 in migraine prophylaxis: A randomized controlled trial. Neurology 64: 713–715.

Sano M, Bell K, Cote L, et al. (1992) Double-blind parallel design pilot study of acetyl levocarnitine in patients with Alzheimer’s disease. Archives of Neurology 49: 1137–1141.

Sarris J, Mischoulon D and Schweitzer I (2011) Adjunctive nutraceuticals with standard pharmacotherapies in bipolar disorder: A systematic review of clinical trials. Bipolar Disorders 13: 454–465.

Sauter A and Rudin M (1993) Determination of creatine kinase kinetic parameters in rat brain by NMR magnetization transfer. Correlation with brain function. Journal of Biological Chemistry 268: 13,166–13,171.

Saxena R, de Bakker PIW, Singer K, et al. (2006) Comprehensive asso-ciation testing of common mitochondrial DNA variation in metabolic disease. American Journal of Human Genetics 79: 54–61.

Shao L, Cui J, Young LT, et al. (2008a) The effect of mood stabilizer lithium on expression and activity of glutathione s-transferase isoen-zymes. Neuroscience 151: 518–524.

Shao L, Martin MV, Watson SJ, et al. (2008b) Mitochondrial involvement in psychiatric disorders. Annals of Medicine 40: 281–295.

Shea TB (2007) Effects of dietary supplementation with N-acetyl cysteine, acetyl-L-carnitine and S-adenosyl methionine on cognitive perfor-mance and aggression in normal mice and mice expressing human ApoE4. Neuromolecular Medicine 9: 264–269.

Shults CW and Haas R (2005) Clinical trials of coenzyme Q10 in neuro-logical disorders. Biofactors 25: 117–126.

Shults CW, Beal MF, Song D, et al. (2004) Pilot trial of high dosages of coenzyme Q(10) in patients with Parkinson’s disease. Experimental Neurology 188: 491–494.

Shults CW, Haas RH, Passov D, et al. (1997) Coenzyme Q10 levels corre-late with the activities of complexes I and II/III in mitochondria from parkinsonian and nonparkinsonian subjects. Annals of Neurology 42: 261–264.

Shults CW, Oakes D, Kieburtz K, et al. (2002) Effects of coenzyme Q10 in early Parkinson disease: Evidence of slowing of the functional decline. Archives of Neurology 59: 1541–1550.

Silva M, Aires C, Luis P, et al. (2008) Valproic acid metabolism and its effects on mitochondrial fatty acid oxidation: A review. Journal of Inherited Metabolic Disease 31: 205–216.

Sima AA, Calvani M, Mehra M, et al. (2005) Acetyl-L-carnitine improves pain, nerve regeneration, and vibratory perception in patients with chronic diabetic neuropathy: An analysis of two randomized placebo-controlled trials. Diabetes Care 28: 89–94.

Sklar P, Smoller JW, Fan J, et al. (2008) Whole-genome association study of bipolar disorder. Molecular Psychiatry 13: 558–569.

Soczynska JK, Kennedy SH, Chow CS, et al. (2008) Acetyl-L-carnitine and alpha-lipoic acid: Possible neurotherapeutic agents for mood dis-orders? Expert Opinion on Investigational Drugs 17: 827–843.

Sohmiya M, Tanaka M, Tak NW, et al. (2004) Redox status of plasma coen-zyme Q10 indicates elevated systemic oxidative stress in Parkinson’s disease. Journal of the Neurological Sciences 223: 161–166.

Sorbi S, Forleo P, Fani C, et al. (2000) Double-blind, crossover, placebo-controlled clinical trial with L-acetylcarnitine in patients with degen-erative cerebellar ataxia. Clinical Neuropharmacology 23: 114–118.

Spagnoli A, Lucca U, Menasce G, et al. (1991) Long-term acetyl-L-car-nitine treatment in Alzheimer’s disease. Neurology 41: 1726–1732.

Storch A, Jost WH, Vieregge P, et al. (2007) Randomized, double-blind, placebo-controlled trial on symptomatic effects of coenzyme Q10 in Parkinson disease. Archives of Neurology 64: 938–944.

Stork C and Renshaw PF (2005) Mitochondrial dysfunction in bipolar disorder: Evidence from magnetic resonance spectroscopy research. Molecular Psychiatry 10: 900–919.

Strijks E, Kremer HPH and Horstink M (1997) Q(10) therapy in patients with idiopathic Parkinson’s disease. Molecular Aspects of Medicine 1997; 18 Suppl: S237–S240.

Sullivan PG, Geiger JD, Mattson MP, et al. (2000) Dietary supplement creatine protects against traumatic brain injury. Annals of Neurology 48: 723–729.

Sun X, Wang J-F, Tseng M, et al. (2006) Downregulation in components of the mitochondrial electron transport chain in the postmortem fron-tal cortex of subjects with bipolar disorder. Journal of Psychiatry & Neuroscience 31: 189–196.

Tabrizi SJ, Blamire AM, Manners DN, et al. (2005) High-dose creatine therapy for Huntington disease: A 2-year clinical and MRS study. Neurology 64: 1655–1656.

Tan DX, Manchester LC, Reiter RJ, et al. (1998) A novel melatonin metabolite, cyclic 3-hydroxymelatonin: A biomarker of in vivo hydroxyl radical generation. Biochemical and Biophysical Research Communications 253: 614–620.

Tankova T, Koev D and Dakovska L (2004) Alpha-lipoic acid in the treat-ment of autonomic diabetic neuropathy (controlled, randomized, open-label study). Romanian Journal of Internal Medicine 42: 457–464.

Tarnopolsky MA (2008) The mitochondrial cocktail: Rationale for com-bined nutraceutical therapy in mitochondrial cytopathies. Advanced Drug Delivery Reviews 60: 1561–1567.

Tarnopolsky MA and Beal MF (2001) Potential for creatine and other ther-apies targeting cellular energy dysfunction in neurological disorders. Annals of Neurology 49: 561–574.

Tarnopolsky MA, Mahoney DJ, Vajsar J, et al. (2004) Creatine monohy-drate enhances strength and body composition in Duchenne muscular dystrophy. Neurology 62: 1771–1777.

Tarnopolsky MA, Roy BD and MacDonald JR (1997) A randomized, con-trolled trial of creatine monohydrate in patients with mitochondrial cytopathies. Muscle & Nerve 20: 1502–1509.

Taylor RW and Turnbull DM (2005) Mitochondrial DNA mutations in human disease. Nature Reviews. Genetics 6: 389–402.

Tempesta E, Casella L, Pirrongelli C, et al. (1987) L-Acetylcarnitine in depressed elderly subjects. A cross-over study vs placebo. Drugs Under Experimental and Clinical Research 13: 417–423.

Terjung RL, Clarkson P, Eichner ER, et al. (2000) American College of Sports Medicine roundtable. The physiological and health effects of oral creatine supplementation. Medicine and Science in Sports and Exercise 32: 706–717.

Torrioli MG, Vernacotola S, Mariotti P, et al. (1999) Double-blind, pla-cebo-controlled study of L-acetylcarnitine for the treatment of hyper-active behavior in fragile X syndrome. American Journal of Medical Genetics 87: 366–368.

Torrioli MG, Vernacotola S, Peruzzi L, et al. (2008) A double-blind, paral-lel, multicenter comparison of L-acetylcarnitine with placebo on the attention deficit hyperactivity disorder in fragile X syndrome boys. American Journal of Medical Genetics. Part A 146A: 803–812.

Verbessem P, Lemiere J, Eijnde BO, et al. (2003) Creatine supplemen-tation in Huntington’s disease: A placebo-controlled pilot trial. Neurology 61: 925–930.

Villardita C, Smirni P and Vecchio I (1983) Acetyl-L-carnitine in depressed geriatric patients. European Review for Medical and Pharmacological Sciences 6: 1–12.

Vincent HK, Bourguignon CM, Vincent KR, et al. (2007) Effects of alpha-lipoic acid supplementation in peripheral arterial disease: A

at Fund Diag.Est Imstico PARENT on August 7, 2013anp.sagepub.comDownloaded from

Page 18: Aust N Z J Psychiatry 2013 Nierenberg 26 42

42 ANZJP Articles

Australian & New Zealand Journal of Psychiatry, 47(1)

pilot study. Journal of Alternative and Complementary Medicine 13: 577–584.

Virmani A, Gaetani F and Biniend Z (2005) Effects of metabolic modifiers such as carnitines, coenzyme Q10, and PUFAs against different forms of neurotoxic insults: Metabolic inhibitors, MPTP, and methampheta-mine. Annals of the New York Academy of Sciences 1053: 183–191.

Walter MC, Lochmuller H, Reilich P, et al. (2000) Creatine monohydrate in muscular dystrophies: A double-blind, placebo-controlled clinical study. Neurology 54: 1848–1850.

Wang J-F. (2007) Defects of mitochondrial electron transport chain in bipolar disorder: Implications for mood-stabilizing treatment. Canadian Journal of Psychiatry-Revue Canadienne De Psychiatrie 52: 753–762.

Weeber E, Levy M, Sampson M, et al. (2002) The role of mitochon-drial porins and the permeability transition pore in learning and synaptic plasticity. Journal of Biological Chemistry 277: 18,891–18,897.

Weissman MM, Bland RC, Canino GJ, et al. (1996) Cross-national epi-demiology of major depression and bipolar disorder. Journal of the American Medical Association 276: 293–299.

Winiarska K, Fraczyk T, Malinska D, et al. (2006) Melatonin attenu-ates diabetes-induced oxidative stress in rabbits. Journal of Pineal Research 40: 168–176.

Yadav V, Marracci G, Lovera J, et al. (2005) Lipoic acid in multiple sclerosis: A pilot study. Multiple Sclerosis 11: 159–165.

Yang F, He X, Russell J, et al. (2003) Ca2+ influx-independent synaptic potentiation mediated by mitochondrial Na(+)-Ca2+ exchanger and protein kinase C. Journal of Cell Biology 163: 511–523.

Yildiz A, Sachs GS, Dorer DJ, et al. (2001) P-31 Nuclear magnetic resonance spectroscopy findings in bipolar illness: A meta-analysis. Psychiatry Research Neuroimaging 106: 181–191.

Youle M and Osio M (2007) A double-blind, parallel-group, placebo-controlled, multicentre study of acetyl L-carnitine in the sympto-matic treatment of antiretroviral toxic neuropathy in patients with HIV-1 infection. HIV Medicine 8: 241–250.

Zanardi R and Smeraldi E (2006) A double-blind, randomised, con-trolled clinical trial of acetyl-L-carnitine vs. amisulpride in the treatment of dysthymia. European Neuropsychopharmacology 16: 281–287.

Ziegler D, Ametov A, Barinov A, et al. (2006) Oral treatment with alpha-lipoic acid improves symptomatic diabetic polyneuropathy: The SYDNEY 2 trial. Diabetes Care 29: 2365–2370.

Ziegler D, Hanefeld M, Ruhnau KJ, et al. (1995) Treatment of symptomatic diabetic peripheral neuropathy with the anti-oxidant alpha-lipoic acid. A 3-week multicentre randomized controlled trial (ALADIN Study). Diabetologia 38: 1425–1433.

Ziegler D, Hanefeld M, Ruhnau KJ, et al. (1999) Treatment of symp-tomatic diabetic polyneuropathy with the antioxidant alpha-lipoic acid: A 7-month multicenter randomized controlled trial (ALADIN III Study). ALADIN III Study Group. Alpha-Lipoic Acid in Diabetic Neuropathy. Diabetes Care 22: 1296–1301.

Ziegler D, Schatz H, Conrad F, et al. (1997) Effects of treatment with the antioxidant alpha-lipoic acid on cardiac autonomic neuropathy in NIDDM patients. A 4-month randomized controlled multicenter trial (DEKAN Study). Deutsche Kardiale Autonome Neuropathie. Diabetes Care 20: 369–373.

at Fund Diag.Est Imstico PARENT on August 7, 2013anp.sagepub.comDownloaded from


Recommended