Top Banner
Please cite this article in press as: Rizzo, L.B., et al., The theory of bipolar disorder as an illness of accelerated aging: Implications for clinical care and research. Neurosci. Biobehav. Rev. (2014), http://dx.doi.org/10.1016/j.neubiorev.2014.02.004 ARTICLE IN PRESS G Model NBR 1894 1–13 Neuroscience and Biobehavioral Reviews xxx (2014) xxx–xxx Contents lists available at ScienceDirect Neuroscience and Biobehavioral Reviews jou rn al h om epage: www.elsevier.com/locate/neubiorev Review The theory of bipolar disorder as an illness of accelerated aging: Implications for clinical care and research Lucas Bortolotto Rizzo a,b , Leonardo Gazzi Costa a,b , Rodrigo B. Mansur a,b,c , Q1 Walter Swardfager d , Síntia Iole Belangero b,e , Rodrigo Grassi-Oliveira f , Roger S. McIntyre c,d , Moisés E. Bauer f , Elisa Brietzke a,b,a Program for Recognition and Intervention in Individuals in At-Risk Mental States (PRISMA), Department of Psychiatry, Federal University of São Paulo, São Paulo, Brazil b Interdisciplinary Laboratory of Clinical Neuroscience (LINC), Department of Psychiatry, Federal University of São Paulo, São Paulo, Brazil c Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada d Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada e Department of Morphology and Genetics, Federal University of São Paulo, São Paulo, Brazil f Institute of Biomedical Research and Faculty of Biosciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil a r t i c l e i n f o Article history: Received 29 August 2013 Received in revised form 17 December 2013 Accepted 5 February 2014 Keywords: Bipolar disorder Aging Biomarkers Neuroprogression Telomeres Inflammation Immunosenescence BDNF Oxidative stress Amyloid Cognition Molecular imaging a b s t r a c t Bipolar Disorder (BD) has been conceptualized as both a cyclic and a progressive disorder. Mechanisms involved in neuroprogression in BD remain largely unknown although several non-mutually exclusive models have been proposed as explanatory frameworks. In the present paper, we propose that the pathophysiological Q2 changes observed in BD (e.g. brain structural alterations, cognitive deficits, oxidative stress imbalance, amyloid metabolism, immunological deregulation, immunosenescence, neurotrophic deficiencies and telomere shortening) converge on a model of accelerated aging (AA). Aging can be under- stood as a multidimensional process involving physical, neuropsychological, and social changes, which can be highly variable between individuals. Determinants of successful aging (e.g environmental and genetic factors), may also confer differential vulnerability to components of BD pathophysiology and contribute to the clinical presentation of BD. Herein we discuss how the understanding of aging and senes- cence can contribute to the search for new and promising molecular targets to explain and ameliorate neuroprogression in BD. We also present the strengths and limitations of this concept. © 2014 Published by Elsevier Ltd. Contents 1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 2. Neurobiological similarities between neuroprogression in BD and aging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 2.1. Changes at the structural level . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 2.2. Changes at the functional levels . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 2.3. Changes at the molecular level . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 2.3.1. Increased oxidative stress . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 2.3.2. Disturbances in amyloid metabolism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 2.4. Changes at the cellular level . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 2.4.1. Immunosenescence . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 2.4.2. Reduction in neurotrophins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 2.4.3. Short telomeres . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 Corresponding author at: R. Pedro de Toledo, 669, 3 andar Vila Clementino, CEP: 04039-032 São Paulo, Brazil. E-mail address: [email protected] (E. Brietzke). http://dx.doi.org/10.1016/j.neubiorev.2014.02.004 0149-7634/© 2014 Published by Elsevier Ltd. 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48
13

The theory of bipolar disorder as an illness of accelerated aging: Implications for clinical care and research

Jan 17, 2023

Download

Documents

Norman Madarasz
Welcome message from author
This document is posted to help you gain knowledge. Please leave a comment to let me know what you think about it! Share it to your friends and learn new things together.
Transcript
Page 1: The theory of bipolar disorder as an illness of accelerated aging: Implications for clinical care and research

N

R

TI

LQ1

WRa

Sb

c

d

e

f

a

ARR1A

KBABNTIIBOACM

Q2

C

h0

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18

19

20

21

22

23

24

25

26

27

28

29

30

31

32

33

34

35

36

37

38

39

40

41

42

43

44

45

46

47

48

ARTICLE IN PRESSG ModelBR 1894 1–13

Neuroscience and Biobehavioral Reviews xxx (2014) xxx–xxx

Contents lists available at ScienceDirect

Neuroscience and Biobehavioral Reviews

jou rn al h om epage: www.elsev ier .com/ locate /neubiorev

eview

he theory of bipolar disorder as an illness of accelerated aging:mplications for clinical care and research

ucas Bortolotto Rizzoa,b, Leonardo Gazzi Costaa,b, Rodrigo B. Mansura,b,c,alter Swardfagerd, Síntia Iole Belangerob,e, Rodrigo Grassi-Oliveira f,

oger S. McIntyrec,d, Moisés E. Bauer f, Elisa Brietzkea,b,∗

Program for Recognition and Intervention in Individuals in At-Risk Mental States (PRISMA), Department of Psychiatry, Federal University of São Paulo,ão Paulo, BrazilInterdisciplinary Laboratory of Clinical Neuroscience (LINC), Department of Psychiatry, Federal University of São Paulo, São Paulo, BrazilMood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, CanadaSunnybrook Research Institute, University of Toronto, Toronto, Ontario, CanadaDepartment of Morphology and Genetics, Federal University of São Paulo, São Paulo, BrazilInstitute of Biomedical Research and Faculty of Biosciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil

r t i c l e i n f o

rticle history:eceived 29 August 2013eceived in revised form7 December 2013ccepted 5 February 2014

eywords:ipolar disordergingiomarkerseuroprogressionelomeres

a b s t r a c t

Bipolar Disorder (BD) has been conceptualized as both a cyclic and a progressive disorder. Mechanismsinvolved in neuroprogression in BD remain largely unknown although several non-mutually exclusivemodels have been proposed as explanatory frameworks. In the present paper, we propose that thepathophysiological changes observed in BD (e.g. brain structural alterations, cognitive deficits, oxidativestress imbalance, amyloid metabolism, immunological deregulation, immunosenescence, neurotrophicdeficiencies and telomere shortening) converge on a model of accelerated aging (AA). Aging can be under-stood as a multidimensional process involving physical, neuropsychological, and social changes, whichcan be highly variable between individuals. Determinants of successful aging (e.g environmental andgenetic factors), may also confer differential vulnerability to components of BD pathophysiology andcontribute to the clinical presentation of BD. Herein we discuss how the understanding of aging and senes-cence can contribute to the search for new and promising molecular targets to explain and ameliorate

nflammationmmunosenescenceDNFxidative stressmyloid

neuroprogression in BD. We also present the strengths and limitations of this concept.© 2014 Published by Elsevier Ltd.

ognitionolecular imaging

ontents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 002. Neurobiological similarities between neuroprogression in BD and aging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00

2.1. Changes at the structural level . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 002.2. Changes at the functional levels . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 002.3. Changes at the molecular level . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00

2.3.1. Increased oxidative stress . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 002.3.2. Disturbances in amyloid metabolism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00

2.4. Changes at the cellular level . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00

Please cite this article in press as: Rizzo, L.B., et al., The theory of bipclinical care and research. Neurosci. Biobehav. Rev. (2014), http://dx.d

2.4.1. Immunosenescence . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .2.4.2. Reduction in neurotrophins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .2.4.3. Short telomeres . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

∗ Corresponding author at: R. Pedro de Toledo, 669, 3◦ andar Vila Clementino, CEP: 040E-mail address: [email protected] (E. Brietzke).

ttp://dx.doi.org/10.1016/j.neubiorev.2014.02.004149-7634/© 2014 Published by Elsevier Ltd.

olar disorder as an illness of accelerated aging: Implications foroi.org/10.1016/j.neubiorev.2014.02.004

. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00

. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00

39-032 São Paulo, Brazil.

Page 2: The theory of bipolar disorder as an illness of accelerated aging: Implications for clinical care and research

ARTICLE IN PRESSG ModelNBR 1894 1–13

2 L.B. Rizzo et al. / Neuroscience and Biobehavioral Reviews xxx (2014) xxx–xxx

3. Implications for research directed to clinical care . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 003.1. Pharmacological strategies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00

3.1.1. Current pharmacology modulating aging related mechanisms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 003.1.2. Sirtuins as possible new target . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00

3.2. Non-pharmacological strategies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 003.2.1. Physical activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00

4. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00Conflict of interest statement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00Acknowledgements. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00

. . . . . .

1

ooretGcnSm(

is2dtdtaavilesepss

uaastoGHoa2

aSdiAota

Normal aging is associated with alterations in brain structures.

49

50

51

52

53

54

55

56

57

58

59

60

61

62

63

64

65

66

67

68

69

70

71

72

73

74

75

76

77

78

79

80

81

82

83

84

85

86

87

88

89

90

91

92

93

94

95

96

97

98

99

100

101

102

103

104

105

106

107

108

109

110

111

112

113

114

115

116

117

118

119

120

121

122

123

124

125

126

127

128

129

130

131

132

133

134

135

136

137

138

139

140

141

142

143

144

145

146

147

References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

. Introduction

Bipolar Disorder (BD) is a prevalent and often severe mood dis-rder where individuals experience disruptive episodes of maniar hypomania and depression (Anderson et al., 2012). Causes of BDemain largely unknown but probably involve a set of genetic andnvironmental factors, which interact during neurodevelopmento determine vulnerability to the disease (Brietzke et al., 2012a).enetic studies have implicated many chromosomal regions andandidate genes, but results have been inconsistent and oftenot replicable (Sullivan et al., 2012). Genome-wide Associantiontudies (GWAS) studies indicate that genetic susceptibility is deter-ined by a high number of genes, each with a small effect size

Sullivan et al., 2012).Although BD has been understood classically as a cyclic disease,

n the last 10 years evidence has accumulated supporting progres-ive features of BD (Berk et al., 2010; Fries et al., 2012; Mansur et al.,013), reconceptualizing BD as both a cyclic and progressive disor-er. The starting point for this hypothesis was the clinical evidencehat individuals in early and late stages of BD present substantialifferences in the severity of clinical presentation and response toreatment (Berk et al., 2011). There are robust data suggesting that

greater number of episodes, especially those of manic polarity,re associated with a decrease in the length of the euthymic inter-al between episodes, worsening of neurocognitive performance,ncreasing risk of suicide, and poorer response to both pharmaco-ogical and psychosocial treatments (Berk et al., 2010; Magalhaest al., 2012). Nonetheless, evidence has been mixed and progres-ive models of mental illness are not universally accepted (Zipurskyt al., 2012). At present, operationalization of the concept of neuro-rogression in clinical practice manifests mainly in the “staging” ofevere mental disorders, including BD (for a detailed review, pleaseee Kapczinski et al., 2009).

Mechanisms involved in neuroprogression remain largelynknown and only few explanatory models exist that could justify

neuroprogressive disease course. Among these, one of the mostccepted is the concept of Allostatic Load (AL). AL implicates chronictress in overactivating homeostatic mechanisms that are collec-ively beyond the capability of the organism, leading to progressionf clinical and neurobiological parameters (Brietzke et al., 2011;oldstein et al., 2009b; Grande et al., 2012; Kapczinski et al., 2008).owever, it has been difficult to quantify the impact of stress biol-gy on neuroprogression, in part due to mechanisms of resistancend resilience that can modulate the impact of stress (Brietzke et al.,012b).

Another nascent theoretical framework is the concept of BDs a disorder of accelerated aging (AA) (Simon et al., 2006;odhi et al., 2012). Aging in humans refers to a persistentecline in the age-specific fitness components of an organ-

sm due to internal physiological degeneration (Rose, 1991).ging can also be understood as a multidimensional process

Please cite this article in press as: Rizzo, L.B., et al., The theory of bipclinical care and research. Neurosci. Biobehav. Rev. (2014), http://dx.d

f physical, neuropsychological, and social changes. With respecto neuropsychological changes, the effect of aging is non-uniformcross domains. For instance, psychomotor processing speed and

. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00

verbal memory performance decline with age, while knowledgeand wisdom can continue to expand. The rates of change in thesedomains differ between individuals and they can be modified bymany intervening genetic and environmental factors. A comple-mentary concept is the idea of senescence. The phenomenon ofcellular senescence was first described by Leonard Hayflick in 1961,referring to the limited capacity of isolated cells to proliferate inculture (the Hayflick Limit) (Hayflick and Moorhead, 1961), but theconcept can also be applied to whole organisms. For example, aftera period of near perfect renewal (in humans, between 20 and 35years of age), organismal senescence is characterized by the declin-ing ability to respond to stress, increasing homeostatic imbalanceand risk of disease.

Preliminary data suggest that individuals with BD present earlysenescent features consistent with AA. One of the most clinicallyconspicuous corollaries is the high prevalence and earlier age ononset of age related medical conditions e.g. cardiovascular con-ditions, hypertension, metabolic imbalances, autoimmunity andcancer (Crump et al., 2013; Czepielewski et al., 2013; Fagioliniet al., 2008; Goldstein et al., 2009a; McIntyre et al., 2005, 2006;Osby et al., 2001; Padmos et al., 2004; Rege and Hodgkinson, 2013;Soreca et al., 2008). In addition, the association between mooddisorders and dementia has been well recognized. Most studiesinclude individuals with Major Depressive Disorder (MDD), but arecent meta-analysis suggested that the association between BDand dementia might be stronger than that of MDD (da Silva et al.,2013). Although the degree of neurobiological overlap betweenthe two conditions remains a matter of debate, there is evidencethat inflammation, neurotrophic and amyloid cascades are alteredin both conditions (Aprahamian et al., 2013; Modabbernia et al.,2013).

Here we discuss how aging and senescence may contributeto the search for new and promising molecular targets to bet-ter understand neuroprogressive features of BD. Although somefindings are preliminary, we postulate they can be integrated ina new theoretical framework to better explain some elements ofBD pathophysiology.

2. Neurobiological similarities between neuroprogressionin BD and aging

Although there are few studies exploring aging in BD, there is asurprisingly high overlap between neurobiological mechanisms inthe two conditions, including progressive changes at the molecularand cellular levels, and in the structure and function of the centralnervous system (CNS).

2.1. Changes at the structural level

olar disorder as an illness of accelerated aging: Implications foroi.org/10.1016/j.neubiorev.2014.02.004

Post mortem and structural neuroimaging studies indicate thatthe human brain shrinks with age, with selective and differentialchanges that are not uniform or random. CNS volume atrophy is

148

149

150

Page 3: The theory of bipolar disorder as an illness of accelerated aging: Implications for clinical care and research

ING ModelN

iobeha

ialtcti(n0pfdi2J2

dettt2rwFit2dneo2

2

blspshifbahv

ofMibstfovfmfc

151

152

153

154

155

156

157

158

159

160

161

162

163

164

165

166

167

168

169

170

171

172

173

174

175

176

177

178

179

180

181

182

183

184

185

186

187

188

189

190

191

192

193

194

195

196

197

198

199

200

201

202

203

204

205

206

207

208

209

210

211

212

213

214

215

216

217

218

219

220

221

222

223

224

225

226

227

228

229

230

231

232

233

234

235

236

237

238

239

240

241

242

243

244

245

246

247

248

249

250

251

252

253

254

255

256

257

258

259

260

261

262

263

264

265

266

267

268

269

270

271

272

ARTICLEBR 1894 1–13

L.B. Rizzo et al. / Neuroscience and B

ndicated by reduced brain weight and volume, ventriculomegalynd sulcal expansion. Microscopic studies documented myelin pal-or, loss of neuronal bodies in the neocortex, the hippocampus andhe cerebellum, loss of myelinated fibers across the subcorticalerebrum, shrinkage and dysmorphology of neurons, accumula-ion of lipofuscin, rarefication of cerebral vasculature, reductionn synaptic density, deafferentation and loss of dendritic spinesRaz and Rodrigue, 2006). The main findings of magnetic reso-ance imaging studies include brain atrophy (total brain volume:.4–0.5% brain tissue loss per year); hippocampal volume loss (1.6%er year in normal individuals); white matter lesions as puncti-orm or early confluent lesions in periventricular or subcorticalistribution and cerebral microbleeds (prevalence more than 20%

n persons older than 60 years) (Allen et al., 2005; DeCarli et al.,005; Enzinger et al., 2005; Fotenos et al., 2005; Ikram et al., 2008;ernigan et al., 2001; van der Lijn et al., 2008; Vernooij and Smits,012).

BD is also associated with alterations in brain structures evi-enced by previous imaging studies that reported findings ofnlargement of the third and lateral ventricles; a reduction inhe gray matter volumes of the orbital and medial prefrontal cor-ex, ventral striatum and mesotemporal cortex and an increase inhe size of the amygdala (Hallahan et al., 2011; Konarski et al.,008; Lisy et al., 2011; Martinez-Aran et al., 2002). These neu-oanatomical changes tend to be more pronounced in patientsith repeated episodes (Lisy et al., 2011; Strakowski et al., 2002).

urthermore, one of the most consistently reported abnormalitiesn BD is an increased number and/or severity of white mat-er hyperintensities (WMH) (Mahon et al., 2010; Mahon et al.,009). The main findings of neuropathogical studies suggesteficits in neuroplasticity, particularly in cell resilience and con-ectivity (Connor et al., 2009; Harrison, 2002; Martinez-Arant al., 2002; Rajkowska, 2002) as well as data showing fewerligodendrocytes in prefrontal white matter (Vostrikov et al.,007).

.2. Changes at the functional levels

The tertiary association cortices, the neostriatum, and the cere-ellum are profoundly affected by aging throughout the adult

ifespan. Regional volumetric decline has been linked with domain-pecific declines in cognitive performance. For example, poorererformance on executive function tasks has been associated withmaller volume of the prefrontal cortex and increased white matteryperintensity burden. Skill acquisition performance is enhanced

n those individuals who show larger volumes of the striatum, pre-rontal cortex and cerebellum. Spatial memory performance haseen linked to hippocampal volume and the concentration of N-cetyl aspartate therein. In addition, entorhinal cortex shrinkageas been found to predict memory decline, even in healthy indi-iduals (Raz and Rodrigue, 2006).

BD is associated with cognitive impairment during acute phasesf illness and euthymia, with greater abnormalities reported asunction of more frequent episodes (Martinez-Aran et al., 2005;

artinez-Aran et al., 2004; Robinson et al., 2006). The most severempairments are seen in verbal working memory, response inhi-ition, sustained attention, psychomotor speed, abstraction andet-shifting (Martinez-Aran et al., 2007). These deficits in execu-ive function and verbal memory have been associated with poorerunctional outcomes (Torrent et al., 2006; Ustun, 1999) and burdenf illness associated with BD (Rosa et al., 2008). It is similar of indi-iduals with mild cognitive impairment and early dementia who

Please cite this article in press as: Rizzo, L.B., et al., The theory of bipclinical care and research. Neurosci. Biobehav. Rev. (2014), http://dx.d

requently report impairments in functioning such as poor perfor-ance in household management activities and in more advanced

unctions (Albert et al., 1999). Furthermore, impairments psychoso-ial functioning can take a progressive course (Rosa et al., 2012;

PRESSvioral Reviews xxx (2014) xxx–xxx 3

Rosa et al., 2011) and can be present during euthymia (Swannet al., 1999). Some authors have proposed that individuals with BDpresent an acceleration in age-related cognitive decline, when com-pared with healthy controls (Gualtieri and Johnson, 2008), althoughthere are some contrary data (Samame et al., 2013). Individualswith MDD, especially those who present the disorder in late life,carry a well-documented risk of dementia (Aprahamian et al., 2013;Osorio et al., 2013). The increased risk of dementia associated withmood disorders in epidemiological studies may be due to commonneuropathological changes shared by these disorders. The investi-gation of aging and senescence processes might lead to an improvedunderstanding of these associations (Gualtieri and Johnson, 2008).

2.3. Changes at the molecular level

2.3.1. Increased oxidative stressIncreased generation of reactive oxygen species (ROS) or dys-

regulation of anti-oxidant mechanisms can lead to the damage ofcellular components such as DNA, proteins and lipids (Andreazzaet al., 2009). Possible sources of ROS include changes in energygeneration and ATP production, for instance free radicals can begenerated during mitochondrial electron transport (Steckert et al.,2010). Other cell process can also generate ROS, including exces-sive dopaminergic transmission (due reductive potential of thisneurotransmitter and its metabolites) and increases in levels of glu-tamate leading to increases in levels of intracellular calcium andcytotoxicity (Berk et al., 2011). Under physiological conditions, theharmful potential of ROS is kept under control by enzymatic andnon-enzymatic antioxidant defense systems (Steckert et al., 2010).

As oxidative stress has been considered a major source of molec-ular and cellular damage, and aging can be understood as a lifelongprocess of cell damage, these two phenomena are presumablylinked. Key pathophysiological elements of several diseases relatedto aging cause end-organ damage through oxidative mechanisms,including chronic obstructive pulmonary disease (Ito et al., 2012),cancer (Ito et al., 2012), type II diabetes (Stadler, 2012), cardio-vascular disease (Penna et al., 2013), Parkinson’s disease (Torraoet al., 2012), and Alzheimer’s disease (Torrao et al., 2012). Thesedata initially lead to “the free radical theory of aging” supportedextensively by numerous in vivo and in vitro studies (Harman,2009). Cumulative oxidative stress causes DNA damage, reducesmechanisms of DNA repair, stimulates inflammatory activation andmodulates redox-sensitive transcriptional factors, such as activatorprotein (AP)-1 and nuclear factor-kappa B (NF-�B), which alter thetranscription of numerous genes, both adaptive and maladaptive,consequently disrupting homeostasis.

Increased oxidative stress and its cellular and molecular con-sequences have been found extensively in BD. In animal modelsof mania, increases in oxidative mediators such as thiobarbituricacid reactive substances (TBARS) are well documented (Ghedimet al., 2012). Studies that focused on individuals in manic episodesobserved increases in nitric oxide (NO) and TBARS, as well asdecreases in antioxidant enzymes, including superoxide dismutase(SOD) and catalase (CAT) (Gergerlioglu et al., 2007; Machado-Vieiraet al., 2007; Savas et al., 2002). Acute treatment with lithiumappears to have an antioxidant effect, as it was shown to affectTBARS, SOD and CAT levels (Machado-Vieira et al., 2007). The onlystudy that evaluated BD individuals in depressive episodes reportedhigher levels of NO and lower levels of SOD, which were alsoresponsive to pharmacological treatment (Selek et al., 2008).

olar disorder as an illness of accelerated aging: Implications foroi.org/10.1016/j.neubiorev.2014.02.004

While imbalances in oxidative stress can be detected in earlystages of disease, they tend to be more pronounced with increasingduration of illness or number of episodes, suggesting that oxidativestress might be a mechanism of neuroprogression and a source of

273

274

275

276

Page 4: The theory of bipolar disorder as an illness of accelerated aging: Implications for clinical care and research

ING ModelN

4 iobeha

pe

2

smadbrapddptsomnAc

paaettJec2fntde

ediSBse(l(t2dVrborJTtoiibt

277

278

279

280

281

282

283

284

285

286

287

288

289

290

291

292

293

294

295

296

297

298

299

300

301

302

303

304

305

306

307

308

309

310

311

312

313

314

315

316

317

318

319

320

321

322

323

324

325

326

327

328

329

330

331

332

333

334

335

336

337

338

339

340

341

342

343

344

345

346

347

348

349

350

351

352

353

354

355

356

357

358

359

360

361

362

363

364

365

366

367

368

369

370

371

372

373

374

375

376

377

378

379

380

381

382

383

384

385

386

387

388

389

390

391

392

393

394

395

396

397

398

399

400

401

ARTICLEBR 1894 1–13

L.B. Rizzo et al. / Neuroscience and B

lasma biomarkers to monitor the course of the disease (Andreazzat al., 2009; Berk et al., 2011; Magalhaes et al., 2012).

.3.2. Disturbances in amyloid metabolismAmyloid � (A�) peptides of 40 or 42 amino acids are formed after

equential cleavage of the amyloid precursor protein (APP), a transembrane glycoprotein of undetermined function, by successive

ction of the � and � secretases (Zhang et al., 2011). Alzheimer’sisease (AD) is characterized, from a neuropathological standpoint,y the extracellular deposition of the A� and by the intraneu-onal generation of neurofibrillary tangles, neuropil threads, andbnormal material in dystrophic nerve cell processes of neuriticlaques. These changes are also found in a large number of non-emented elderly people post-mortem. One difference betweenemented AD patients and nondemented elderly with AD-relatedathology is reflected by the distribution pattern of neurofibrillaryangles and A� deposits. The AD patients who manifest clinicalymptoms of dementia present a pattern of widespread lesionsccurring in many areas of the brain (all areas affected in nonde-ented patients plus in the brain stem and cerebellum) whereas

ondemented elderly people showed neurofibrillary tangles and� deposits restricted to distinct predilection sites (neocortex, allo-ortex, basal ganglia, and diencephalic nuclei) (Thal et al., 2004).

Alterations of A� peptides concentration in plasma from ADatients (i.e. reduced A� 42 and an increased A� 40/A� 42 ratio)re common findings in recent studies, and this alteration waslso found in patients suffering from MDD (Kita et al., 2009; Qiut al., 2007; Sun et al., 2007). The risk for dementia and for cogni-ive decline seems to be greater in patients with mood disordershan in the general population (Geerlings et al., 2008; Gualtieri andohnson, 2008). In BD, the number of affective episodes (Geerlingst al., 2008; Kessing and Andersen, 2004) and the presence of psy-hotic symptoms (Kessing and Andersen, 2004; Robinson et al.,006; Torres et al., 2007) have been implicated as additional riskactor. Another study on bipolar depressed patients, found a sig-ificant negative correlation between A� 42 plasma levels andhe duration of the illness, whereas a positive correlation wasetected between the A� 40/A� 42 ratio and the number of affectivepisodes (Piccinni et al., 2012).

Direct cytotoxic effects of A�, negative effects on monoamin-rgic transmission, and functional antagonism between brain-erived neurotrophic factor (BDNF) and A�, suggest the potential

nvolvement of A� in the pathophysiology of BD and cellular aging.tudies suggest that A� may exhibit functional interference withDNF actions, because BDNF stimulates glutamatergic transmis-ion and long-term potentiation (LTP) (Korte et al., 1995; Levinet al., 1998; Lue et al., 1999) whereas A� inhibits these phenomenaSnyder et al., 2005). Furthermore, A� can block the phosphory-ation of the transcription factor cAMP response element-bindingCREB) (Tong et al., 2001) and its nuclear translocation, inhibitinghe synthesis of BDNF (Arancio and Chao, 2007; Arvanitis et al.,007). The hypothesized increase in glutamatergic transmissionuring mood episodes (Kugaya and Sanacora, 2005; Machado-ieira et al., 2009; Yildiz-Yesiloglu and Ankerst, 2006) may play aole in the A�-mediated neurotoxicity, explaining the relationshipetween cognitive decline and the severity of the clinical coursef mood disorders, and the greater risk of developing dementiaeported in mood disorders (Geerlings et al., 2008; Gualtieri andohnson, 2008; Kessing and Andersen, 2004; Robinson et al., 2006;orres et al., 2007). It has been suggested that some presenta-ions may represent a prodromal manifestation of AD, or a subtypef amyloid-associate mood disorder characterized by cognitive

Please cite this article in press as: Rizzo, L.B., et al., The theory of bipclinical care and research. Neurosci. Biobehav. Rev. (2014), http://dx.d

mpairment and risk of dementia (Sun et al., 2007). More generally,ndividuals who present with considerable Alzheimer’s pathologyut who do not develop clinical dementia may be less suscep-ible to dementia due to greater “cognitive reserve” (i.e. greater

PRESSvioral Reviews xxx (2014) xxx–xxx

resilience to neuropathological processes by virtue of greater neu-ral substrate, higher premorbid IQ, extensive education, exercise,other lifestyle factors, etc.), which may be eroded by BD pathologyresulting in greater susceptibility to dementia in the presence ofthe same neuropathological burden.

2.4. Changes at the cellular level

2.4.1. ImmunosenescenceImmunosenescence refers to the decline of immunological func-

tion that occurs with aging. During aging, sustained low-gradeinflammatory activity gradually evolves and this is sometimesreferred to as “inflammaging” (Franceschi et al., 2007). In this con-text, healthy aging and longevity are likely result not only froma lower propensity to mount inflammatory responses but alsomaintenance of efficient anti-inflammatory networks (Franceschiet al., 2007). During aging, regulatory processes may fail to coun-teract the inflammatory responses consequent to exposure todamaging agents or to the lifelong acquired antigenic burden.Intriguing data from a large multi-ethnic cohort study suggeststhat cumulative pathogen exposure, particularly viral pathogenssuch as cytomegalovirus (CMV) and other herpes virus, can lead toincreased risks of vascular aging, and cognitive decline in later-life(Elkind et al.; Katan et al.; Swardfager and Black). Further investi-gation is needed, but it is possible that these infections, overwhelmthe diminishing capacity for immunoregulation/immunotoleracethat occurs with age, resulting in increased tissue damage. In ani-mal models, the inflammatory response to experimental infectionresolves more slowly in aged mice than in adult mice, which isassociated with prolonged depressive-like behavior in aged mice(Kelley et al.). According to this perspective, one key to successfulphysical, psychological and cognitive aging is decreased inflamma-tory activity without compromising an effective acute response tonew pathogens (Franceschi et al., 2007).

The paradigm of inflammaging has been largely accepted toexplain why age is an important risk factor for diseases such astype-2 diabetes, cardiovascular diseases and some types of can-cer (Macaulay et al., 2013). In addition, inflammaging has beenpostulated as an explanation for some neurobiological characteris-tics of Alzheimer’s disease. Patients with Alzheimer’s disease showincreased inflammatory activity compared to controls (Swardfageret al., 2010b) as do patients in the clinical mild cognitive impair-ment prodromal phase of the illness (Fuchs et al., 2013). There isevidence that IFN-� and other pro-inflammatory cytokines inter-act with processing and production of A� peptide, suggesting thatthe increased inflammatory activity that accompanies aging couldprecipitate, accelerate or exacerbate AD pathology (Giunta, 2008).Some treatments that counteract inflammaging have been pro-posed to prevent or treat AD, including celecoxib, naproxen, trifusaland indomethacin although limitations in trial design or uncer-tainty in these results limit their readiness for clinical application(de Jong et al., 2008; Gomez-Isla et al., 2008).

There is a consistent body of evidence suggesting that BD isassociated with a persistent and low intensity pro-inflammatorystates, which are more intense during mood episodes, especiallymanic episodes, and less intense in depressive episodes (Brietzkeet al., 2009b; Modabbernia et al., 2013). Even euthymia has beenassociated with detectable peripheral pro-inflammatory activity(Brietzke et al., 2009a). In the context of aging, blood cytokineconcentrations have been correlated with immune-stimulatedcatabolism of tryptophan and tyrosine, necessary precursors in thesynthesis of serotonin and dopamine, and this has been associ-

olar disorder as an illness of accelerated aging: Implications foroi.org/10.1016/j.neubiorev.2014.02.004

ated with affective and neurovegetative symptoms, respectively(Capuron et al., 2011). This mechanism may apply to BD patientsearlier in life. For instance, the enzymes and final products of thekynurenine pathway, which catabolizes tryptophan, are increased

402

403

404

405

Page 5: The theory of bipolar disorder as an illness of accelerated aging: Implications for clinical care and research

ING ModelN

iobeha

iaaS

iC2eccsstpqrw2iiltci2incir2

2

ctora12Pnd

mthiaBeBtBa2

alaip6e

406

407

408

409

410

411

412

413

414

415

416

417

418

419

420

421

422

423

424

425

426

427

428

429

430

431

432

433

434

435

436

437

438

439

440

441

442

443

444

445

446

447

448

449

450

451

452

453

454

455

456

457

458

459

460

461

462

463

464

465

466

467

468

469

470

471

472

473

474

475

476

477

478

479

480

481

482

483

484

485

486

487

488

489

490

491

492

493

494

495

496

497

498

499

500

501

502

503

504

505

506

507

508

509

510

511

512

513

514

515

516

517

518

519

520

521

522

523

524

525

526

527

528

529

530

531

ARTICLEBR 1894 1–13

L.B. Rizzo et al. / Neuroscience and B

n the cingulate cortex in BD (Miller et al., 2006, 2008). This has beenssociated with cognitive symptoms in mood disorders patientsnd proposed as a modifiable target for therapy (Wonodi andchwarcz, 2010).

BD patients exhibit other immunological alterations commonn elderly people; for instance higher proportions of circulatingD8+CD28− cells (do Prado et al., 2013; Wieck and Grassi-Oliveira,013), and lower proportions of regulatory T cells (do Pradot al., 2013; Wieck and Grassi-Oliveira, 2013) and increasedytomegalovirus (CMV) infection (Rizzo et al., 2013). CD8+CD28−

ells have lost the expression of the CD28 protein, which is neces-ary for their complete stimulation and clonal expansion. They havehort telomeres, impaired cytotoxic function, and they are resistanto apoptosis (Derhovanessian et al., 2009; Weng et al., 2009). Theroportion of CD8+CD28− cells increases with aging and a high fre-uency of CD28− cells in the elderly correlates with a less effectiveesponse to influenza virus vaccination (Goronzy et al., 2001) andith a higher risk of mortality (Olsson et al., 2000; Wikby et al.,

002). The main cause for the expansion of these senescent cellss related to infectious burden, particularly CMV infection. Afternfection with CMV, the body is not able to promote virus clearance,eading to chronic albeit asymptomatic infection. The long course ofhe infection promotes chronic immunological stimulation of spe-ific cells for CMV epitopes, and consequently restriction of themmunological repertoire (Almanzar et al., 2005; Ouyang et al.,003). Intriguingly, this effect is less pronounced other chronic

nfections (Pawelec and Derhovanessian, 2011). The motive is untilow not completely understood, but it is possible that CMV has aloser relation with the immunological system compared to othernfections, since dendritic and endothelial cells function as viraleservoirs (Benedict et al., 2008; Bentz et al., 2006; Tabata et al.,008).

.4.2. Reduction in neurotrophinsImpaired capacity for neuroplasticity has been proposed as a

ore-underlying feature of mood disorders, which may be relatedo both affective and cognitive symptoms as they affect the integrityf nodal brain structures and connectivity between them. Neu-oplasticity can be impaired by pro-inflammatory cytokines (Dasnd Basu, 2007; Goshen et al., 2007), normal aging (Kuhn et al.,996) and activation of the stress-response systems (Joca et al.,007; Karten et al., 2005). On the other hand, physical activity (vanraag et al., 1999) and mood stabilizers such as lithium can promoteeurogenesis (Chen et al.). Many treatment benefits are thought toepend on the release of neurotrophic factors.

Neurotrophins, notably BDNF, have been considered one of theost important mediators of synaptic plasticity and apoptosis con-

rol in mood disorders. Individuals with BD, when compared withealthy controls, have lower peripheral levels of BDNF in mania and

n depressive episodes, as described by a systematic review, with meta-regression analysis (Fernandes et al., 2011). In that study,DNF levels in euthymia were unaltered, although they were influ-nced by age and length of illness. A separate study reported thatDNF levels were decreased in euthymic individuals, but only inhe latter stages of illness (Kauer-Sant’Anna et al., 2009). Therefore,DNF has been considered both a marker of clinical symptoms and

mechanism underlying neuroprogression (Berk, 2009; Berk et al.,010).

Interestingly, reduced BDNF levels have also been found inging (Perovic et al., 2012). In non-demented elderly individuals,ower BDNF plasma levels have been correlated with white-mattertrophy (Driscoll et al., 2012). The age-related decline in BDNF

Please cite this article in press as: Rizzo, L.B., et al., The theory of bipclinical care and research. Neurosci. Biobehav. Rev. (2014), http://dx.d

s probably due to epigenetic changes. In one rodent study, hip-ocampal BDNF mRNA was lower in 12-month-old compared with

months old animals. In addition, there was a decrease in thexpression of TrkB, the BDNF receptor mediating neuroprotection

PRESSvioral Reviews xxx (2014) xxx–xxx 5

and structural plasticity, which was accompanied by lower levelsof the two main downstream effector kinases, pAkt and proteinkinase C (Perovic et al., 2012). Recently, a post-mortem studyshowed an age-related decrease in BDNF transcripts in amygdalaof both healthy and MDD individuals as well as in BDNF-dependentgene expression. Interestingly, most genes that are age-dependentin control subjects display greater age effects in MDD subjects(Douillard-Guilloux et al., 2013).

A second neurotrophin likely to be of importance to mood dis-orders, and particularly in senescence processes, is insulin-likegrowth factor 1 (IGF-1). In mouse behavioral studies, experimen-tal IGF-1 deficiency induces depressive-like symptoms (Mitschelenet al.), and in an inflammatory depression model, IGF-1 reduceddepression-like behavior (Park et al.). Polymorphisms in the IGF-1gene have been associated with BD, suggesting that genomic vari-ants may increase susceptibility (Pereira et al.). Insulin-like growthfactor-1 (IGF-1) can increase neural stem cell proliferation in thedentate gyrus of the hippocampus and increase CA1 pyramidalneuron dendritic spine density (Glasper et al.). IGF-1 activates anti-apoptotic signaling, protecting neurons against oxidative stress(Floratou et al.; Wang et al.). Moreover, a recent study found that BDpatients who respond to lithium express greater amounts of IGF-1(Squassina et al.) compared to non-responders. The IGF-1 systemis a well-known regulator of aging; however, the relationship maybe complex and the overall effect on lifespan may depend on theperiod of reduced IGF-1 signaling (Kenyon). In animals, the IGF-1receptor determines lifespan based on a neuroendocrine mecha-nism during normal development that increases metabolic functionand reduces longevity (Holzenberger et al.). In humans, mutationsthat decrease the function of the IGF-1 receptor or of signalingmolecules downstream of the IGF-1 receptor are associated withlongevity (Kenyon). However, plasma IGF-1 concentrations declinewith age and they are negatively associated with cardiometabolicrisk factors such as BMI, insulin resistance and blood pressure, sug-gesting that concentrations of IGF-1 may be a marker of metabolicdisease rendering patients with BD more susceptible to neuralinsult and impaired resilience (Sesti et al.). In later-life and in mooddisorders, increases in oxidative stress, apoptosis, inflammation,and declining endothelial function may be ameliorated by IGF-1(Higashi et al.). Further investigation into the relationships betweenBD, aging and the IGF-1 system are warranted.

2.4.3. Short telomeresIn humans and other animals, cellular senescence has been

attributed mainly to the shortening of telomeres. Telomeres arespecialized structures present at the end of chromosomes com-posed by DNA tandem repeats of (TTAGGG/CCCTAA)n ranging5–15 kbp (Aubert and Lansdorp, 2008) and proteins such as TRFs,POT, TIN2, which stabilize the region (Amiard et al., 2007; Teixeiraet al., 2004). Telomeres are believed to have evolved to “cap” chro-mosomal termini and prevent end-to-end recombination and thusserve a critical role in the maintenance of chromosomal integrity(Blackburn, 2001; Blackburn et al., 2000; Simon et al., 2006). Sincetelomerase levels are insufficient in normal human cells, in each celldivision telomeres get shorter due to the “end replication problem”.When telomeres become too short, the cell ceases to proliferate andenters a state known as “replicative senescence”. Thus, meteredloss of telomeres can serve as a cellular “mitotic clock” that ulti-mately limits the number of cell divisions and cellular life span.For instance, telomere length declines in stem cells, affecting theirfunctionality and capability to generate new cells (Vaziri et al.,1994). Interestingly, shorter telomeres have been associated with

olar disorder as an illness of accelerated aging: Implications foroi.org/10.1016/j.neubiorev.2014.02.004

higher risk of mortality due to cardiovascular and infection-relateddiseases (Cawthon et al., 2003). Although the replicative problemplay a role in telomere shortening, other mechanisms are involvedlike oxidative stress (von Zglinicki, 2000).

532

533

534

535

Page 6: The theory of bipolar disorder as an illness of accelerated aging: Implications for clinical care and research

ARTICLE IN PRESSG ModelNBR 1894 1–13

6 L.B. Rizzo et al. / Neuroscience and Biobehavioral Reviews xxx (2014) xxx–xxx

Fig. 1. Biological findings linking bipolar disorder and aging. White matter hyperintensities (WMH), T regulatory cells (Treg), cytomegalovirus (CMV), nitric Oxide (NO),t ase (C

fihsMnlotavtpe

gcspg(opItwl(

gm

536

537

538

539

540

541

542

543

544

545

546

547

548

549

550

551

552

553

554

555

556

557

558

559

560

561

562

563

564

565

566

567

568

569

570

571

572

573

574

575

576

577

578

579

580

581

582

583

584

585

586

587

588

589

590

591

592

593

594

595

hiobarbituric acid reactive substances, (TBARS), superoxide dismutase (SOD), catal

A preliminary study conducted by Simon and collaboratorsound reduction of telomere length from peripheral leukocytes inndividuals with mood disorders (MDD and BD) compared withealthy controls (Simon et al., 2006). Hartman et al. also foundhorter mean telomere length in leukocytes of individuals withDD compared to healthy controls, although the duration of ill-

ess and depression severity were not associated with telomereength (Hartmann et al., 2010). On the other hand, the numberf previous depressive episodes was an important determinant ofelomere shortening in another study (Wolkowitz et al., 2011). Inddition, pre-treatment telomerase activity was significantly ele-ated in depressed individuals compared with healthy controls andelomerase activity was correlated with depression severity andredicticting response to antidepressant treatment (Wolkowitzt al., 2012).

Telomere shortening is less investigated in BD than in MDD. In aroup of individuals with BD type 2, short telomeres were moreommon among the affected group that among healthy controlubjects (15.04% vs. 13.48%; p = 0.04). Mean telomere length in theatient group was 552 base pairs (bp) shorter than in the controlroup; however, the difference did not reach statistical significanceElvsashagen et al., 2011). The effect of lithium in telomere lengthf individuals with BD was also investigated. Lithium-treated BDatients had 35% longer telomeres compared with healthy controls.

n this study, telomere length correlated positively with lithiumreatment duration of >30 months and was negatively associatedith increasing number of depressive episodes. Thus, response to

ithium in BD patients may also be associated with longer telomeres

Please cite this article in press as: Rizzo, L.B., et al., The theory of bipclinical care and research. Neurosci. Biobehav. Rev. (2014), http://dx.d

Martinsson et al., 2013).Possible causes for shortening of telomeres have been investi-

ated. One study assessing telomere length in the cerebellar grayatter of patients diagnosed with MDD, BD and schizophrenia

AT).

found no difference in mean telomere length between the threegroups and controls. Since mean telomere length has been reportedto be a heritable quantitative trait, the authors also carried outgenome-wide mapping of genetic factors for telomere length andno association survived correction of multiple comparisons for thenumber of SNPs studied. This suggests that genetic predispositionto shorter telomere length could be determined by multiple lociwith small effect sizes (Zhang et al., 2010).

Although a cause-effect relationship cannot be definitivelyestablished, stress and its impact in the organism have been postu-lated as an important factor. In subjects with MDD, telomere lengthwas inversely correlated with oxidative stress and with inflamma-tion in depressed individuals (Wolkowitz et al., 2011). In addition,stress is implicated as a crucial causal mechanism for mood disor-ders, as well as for aging. In one study of individuals with MDD,short telomere length was associated with Perceived Stress Ques-tionnaire scores, and with a hypocortisolemic state, which wasespecially prevalent among patients with a high familial loadingof affective disorders and high levels of C-reactive protein levels(Wikgren et al., 2012).

A possible limitation of current research investigating telom-eres in mood disorders is the possibility of tissue-specific changes.It is possible that premature senescence may occur particularly inrapidly dividing tissues such as leukocytes, sparing others such asthe cerebellar gray matter, as suggested by Zhang and collaborators.Analyses that include other brain regions, particularly the sub-granular zone of the dentate gyrus or the subventrical zone whereneurogenesis continues into adulthood may be more appropriate.

olar disorder as an illness of accelerated aging: Implications foroi.org/10.1016/j.neubiorev.2014.02.004

Additionally, analyses that seek to identify correlations betweenother peripheral senescence-related biomarkers with telomerelength and clinical symptoms or staging in BD are also warranted Q3(Fig. 1).

596

597

598

599

Page 7: The theory of bipolar disorder as an illness of accelerated aging: Implications for clinical care and research

ARTICLE ING ModelNBR 1894 1–13

L.B. Rizzo et al. / Neuroscience and Biobeha

Fig. 2. The relation between Allostatic Load (AL) and Accelerated Aging (AA)hypotheses. Both hypotheses are complementary, the higher the allostatic loadexperienced by BD patients, the higher will the demand of mechanisms to recoverhomeostasis. This leads to an exhaustion of the entire system and promotes theaccelerated aging. The AA paradigm expands the AL hypothesis to its long termconsequences, but also offers new insights into the etiopathology of BD, putting inperspective some of the current knowledge and indicating new areas for furtherrg

3

pwtaadt

Afcmtcbpstrhp

rstOoa(astEsssto2

and interaction with GSK-3 and Bcl-2 (Lai et al., 2006).

600

601

602

603

604

605

606

607

608

609

610

611

612

613

614

615

616

617

618

619

620

621

622

623

624

625

626

627

628

629

630

631

632

633

634

635

636

637

638

639

640

641

642

643

644

645

646

647

648

649

650

651

652

653

654

655

656

657

658

659

660

661

662

663

664

665

666

667

668

669

670

671

672

673

674

675

676

677

678

679

680

681

682

683

684

685

686

687

688

689

690

691

692

693

694

695

696

697

698

699

700

701

esearch. AA hypothesis also opens the field on thinking BD as not just a neuropro-ressive, but a somatoprogressive disorder.

. Implications for research directed to clinical care

Bipolar disorder is a complex disease, accumulating evidenceoints to the involvement of a myriad of pathophysiological path-ays that are dynamic and interact with each other and with

he environment. Notwithstanding these foraging observations, unitary disease model is still unavailable, necessitating furtherdvancement of our understanding of this multifaceted mentalisorder, and the development and refinement of comprehensiveheoretical frameworks.

One integrative hypothesis for neuroprogression in BD is thellostatic Load (AL) which says that BD patients are under stress-

ul conditions (cyclic mood episodes, drug addiction and otheromorbidities) and struggle to restore homeostasis by allostaticechanisms. In the same way, aging can be thought as a loss in

he capability to reattain homeostasis (O’Neill, 1997). In this con-ept, the AA and AL hypotheses are complementary (Fig. 2), theurden of BD promotes exhaustion of allostatic mechanisms thatromote homeostasis. The AA paradigm expands the AL hypothe-is to its long term consequences, but also offer new insights intohe etiopathology of BD, putting in perspective some of the cur-ent knowledge and indicating new areas for further research. AAypothesis also opens the field on thinking BD as not just a neuro-rogressive, but a somatoprogressive disorder.

Although the most explored models for progression of BD areelated to stress, it is intuitive that, even organisms that are notubmitted to chronic stress will age. In addition to environmen-al agents, there is also a strong genetic component in aging.ne interesting insight about genetic component in aging can bebtained from diseases marked by a highly accelerated aging, suchs Hutchinson-Gilford Progeria (HGP) and Werner syndrome (WS)Hegele, 2003; Heyn et al., 2013). As naturally aged individuals, HGPnd WS patients present hair graying, skin thinning, atherosclero-is and osteopenia. Genetically, the disease could be traced backo mutations in lamin A (LMNA) (De Sandre-Giovannoli et al., 2003;riksson et al., 2003; Worman and Bonne, 2007) and the Werneryndrome RecQ helicase like (WRN) (Yu et al., 1996) genes. Althoughimilarities between these conditions and BD were never explored,chizophrenia was conceptualized by some authors as a segmen-

Please cite this article in press as: Rizzo, L.B., et al., The theory of bipclinical care and research. Neurosci. Biobehav. Rev. (2014), http://dx.d

al Progeria, due to similarities in genetic and epigenetic controlf CNS senescence between the disorders (Papanastasiou et al.,011).

PRESSvioral Reviews xxx (2014) xxx–xxx 7

From a clinical point of view, based on the evidence of the pro-gressive nature of BD, some authors propose that the notion ofclinical staging can be applied to BD (Kapczinski et al., 2009; Scottet al., 2013). The main benefits are allowing better prognosis andmore personalized treatments. The use of biomarkers, includingneurotrophins, inflammatory markers and oxidative stress mark-ers have been proposed as auxiliary tools to refine the descriptionof the stages (Kapczinski et al., 2009). In this sense, age-relatedfeatures, such as the presence of medical comorbidities, cognitiveimpairment and shortened telomere length could be useful in thecharacterization of clinical stages. Also, investigation of the pro-gression of these features along the course of the disease may allowearly interventions aimed at better management and prognoses forpatients.

Finally, the recognition of aging processes as part of the BD puz-zle leads to the possibility of treatments targeting specifically thesepathways the so-called “anti-aging” strategies, including nutri-tional approaches and pharmacological and non-pharmacologicalinterventions

3.1. Pharmacological strategies

3.1.1. Current pharmacology modulating aging relatedmechanisms

The mechanisms of action of drugs currently used to treat BD isnot completely understood, but there are some data showing theircapability to modulate pathways related to aging. Patients in treat-ment with lithium show reduced neuronal loss and higher level ofN-acetyl-aspartate (NAA) (marker of neuronal viability) (Beardenet al., 2007; Moore et al., 2000), suggesting a neuroprotectionaleffect by lithium which is already known to be partially mediatedby several mechanisms, its capability to reduce glutamate-inducedexcitotoxicity by NMDA receptors (Hashimoto et al., 2002), directand indirect inhibittion of the pro-apoptotic protein Glycogen syn-thase kinase 3 (GSK-3) (Chiu and Chuang, 2011; Stambolic et al.,1996), induction the antiapoptotic protein B-cell lymphoma 2 (Bcl-2) (Chen and Chuang, 1999) and induction of the growth factorsBDNF (Fukumoto et al., 2001) and vascular endothelial growth fac-tor (VEGF) (Guo et al., 2009).

Over the last years, GSK-3 was also identified as a regulator ofmany components of the immune system. GSK-3 plays an impor-tant role in the signal transmission to promote production of thepro-inflammatory cytokines IL-6, IL-1�, IL-12p40, IFN� and TNF-�and inhibition of the anti-inflammatory cytokine IL-10 and IL1-RA(Beurel et al., 2010). The inhibition of GSK-3 by lithium and itsconsequent impact in the immune system may be one of the ther-apeutic mechanisms of lithium. Recently, the recognition that BDpatients that were lithium intakers had longer telomeres raised thequestion by which means lithium could promote telomere exten-sion. It seems to be also related to the inhibition of GSK-3 andconsequently promoting the transcription hTERT the catalytic sub-unit bearing the enzymatic activity of telomerase (Martinsson et al.,2013).

Lithium also seems to plays a role in the reduction of oxida-tive stress, since lithium treated patients present lower peripherallevels of oxidative stress makers like TBARS, SOD and CAT com-pared to unmedicated patients (Machado-Vieira et al., 2007). Also,lithium pré-treatment was able to protect human neuroblastoma(SH-SY5Y) cell line from rotenone and H2O2-induced cytotoxicity(Lai et al., 2006). The mechanism by which the medication medi-ates oxidative protection is not yet completely understood, but itis partly due to induction of antioxidant proteins like CAT and SOD,

olar disorder as an illness of accelerated aging: Implications foroi.org/10.1016/j.neubiorev.2014.02.004

Valproic acid (VPA) also inhibits GSK-3 activity, although itis now clear yet if it shares the same direct effect as lithiumor other mechanism are involved (Chen et al., 1999; Rosenberg,

702

703

704

Page 8: The theory of bipolar disorder as an illness of accelerated aging: Implications for clinical care and research

ING ModelN

8 iobeha

2iecebc3eias(u

3

eir1rlc2iliohn2

fae(cmtBrt2tB

lcaVad2aptbpsagdg

705

706

707

708

709

710

711

712

713

714

715

716

717

718

719

720

721

722

723

724

725

726

727

728

729

730

731

732

733

734

735

736

737

738

739

740

741

742

743

744

745

746

747

748

749

750

751

752

753

754

755

756

757

758

759

760

761

762

763

764

765

766

767

768

769

770

771

772

773

774

775

776

777

778

779

780

781

782

783

784

785

786

787

788

789

790

791

792

793

794

795

796

797

798

799

800

801

802

803

804

805

806

807

808

809

810

811

812

813

814

815

816

817

818

819

820

821

822

823

824

825

ARTICLEBR 1894 1–13

L.B. Rizzo et al. / Neuroscience and B

007). VPA also impacts in the immunological system promot-ng anti-inflammatory effects in different models as ischemia andxperimental auto-immune neuritis reducing pro-inflammatoryytokines IFN-�, TNF-�, IL-1, IL-6 and IL-17, and also elevating lev-ls of Treg cell (Zhang et al., 2008; Zhang et al., 2012). Mechanismsy which VPA promotes the anti-inflammatory effect needs to belarified, but it seems to be partly mediated by inhibition of GSK-

and histone deacetylase inhibition (Beurel et al., 2010; Grabiect al., 2011). The relation between GSK-3 inhibition by VPA andnduction of hTERT transcription still needs to be investigated. VPAnd atypical antipsychotics also seems to promote effect on cellurvival by elevating levels BDNF and fibroblast growth factor 1FGF1)(Bai et al., 2003; Kao et al., 2013; Yasuda et al., 2009) andpregulating Bcl-2 (Bai et al., 2004).

.1.2. Sirtuins as possible new targetThe sirtuins are a family of ribosyltransferase or deacylase

nzymes that influence a plethora of processes involved in aging,ncluding apoptosis, DNA repair, the inflammatory response andesistance to stress. The mammalian sirtuin homologues, SIRTs–7, are also involved in energy metabolism, with a particularole in metabolic efficiency and maintenance of function duringow energy situations. The sirtuin genes can be upregulated byalorie restriction, with longevity promoting effects (Kanfi et al.,012). For example, SIRT6 is expressed in the nucleus where it

s involved in DNA repair, regulating cell metabolism, and regu-ating the secretion of TNF. In male mice, over-expression of Sirt6ncreases lifespan and alters IGF-1 signaling, consistent with a rolef IGF-1 in determining longevity (Kanfi et al., 2012). On the otherand, SIRT6 deficiency leads to enhanced NF-�B dependent sig-aling, apoptosis and accelerated cell senescence (Kawahara et al.,009).

Recently, decreased expression of SIRT1, SIRT2 and SIRT6 wasound in white blood cells collected from BD patients in states ofcute depression compared to controls. In contrast, SIRT mRNA lev-ls in euthymic BD patients were different from those of controlsAbe et al., 2011), suggesting that sirtuin genes may be defi-iently expressed in BD, and that sirtuin expression may be a statearker of illness. Previously, differences have been described in

he response of white blood cells to glucose deprivation betweenD subjects and controls – whereas healthy controls showed up-egulated transcripts related to mitochondrial energy metabolism,hese transcripts were down-regulated in BD (Naydenov et al.,007). Sirtuin expression would seem to be a compensatory pro-ective measure, and this resilience pathway may be defective inD.

These observations suggest the possibility for novel pharmaco-ogical interventions. For instance, in animal models, resveratrolan activate SIRT1, prolonging the lifespan in obese animalsnd reversing age-related cardiovascular dysfunction (Alcain andillalba, 2009; Barger et al., 2008). More potent pharmacologicalctivators of SIRT1 are under development. These agents can moreramatically increase mitochondrial activity (Alcain and Villalba,009). Interestingly, sirtuin activity is inhibited by nictotinamide,

final product of the kynurenine pathway and an important com-onent of NAD and NADPH involved in the mitochondrial electronransport chain. It has been suggested that blocking nicotinamideinding to its cognate receptors could enhance sirtuin activity andrevent tissue damage caused by age-related degenerative diseasesuch as diabetes, Alzheimers and atherosclerosis (David Adams

Please cite this article in press as: Rizzo, L.B., et al., The theory of bipclinical care and research. Neurosci. Biobehav. Rev. (2014), http://dx.d

nd Lori, 2007; Milne et al., 2007). These pharmacological strate-ies might be useful to mitigate the cellular damage that occursuring depressive episodes, and prevent the associated neuropro-ression.

PRESSvioral Reviews xxx (2014) xxx–xxx

3.2. Non-pharmacological strategies

3.2.1. Physical activityPhysical activity and cardiopulmonary fitness are well-

documented neuroprotective strategies that can increase brainvolumes (Colcombe et al., 2006; Colcombe et al., 2006) and pre-serve cognitive function later in life (Barnes et al., 2003) evenin groups of patients with concomitant cardiovascular morbidity(Swardfager et al., 2010a) or clinical mild cognitive impairment(Baker et al., 2010). It has been suggested that these effects may bemediated, in part by augmentation of neurotrophin concentrations(e.g. BDNF and IGF-1) (Erickson et al., 2011; Llorens-Martin et al.,2010; Swardfager et al., 2011) but improvements in other aspects ofphysiological function may also ameliorate neurocognitive decline,including improvements in vascular endothelial function (Banket al., 1998; Fuchsjager-Mayrl et al., 2002; Xiang et al., 2009). Vas-cular endothelial dysfunction is related to increased generationof oxidative species, which can irreversibly damage intracellularcomponents of the cerebral neurovascular unit and compromiseregulation of cerebral blood flow and thus brain function. Vascu-lar endothelial cell senescence and changes in vascular endothelialfunction occur with aging, favoring pro-inflammatory and pro-oxidant processes that may contribute substantially to age-relatedneurological decline (Yildiz, 2007), particularly since these pro-cesses are exacerbated by age-related cognitive risk factors suchas diabetes and hypertension.

Physical activity is also known to increase telomerase activityboth in human and animal models (Ludlow et al., 2012; Werneret al., 2008; Wolf et al., 2011). Telomerase has not yet been eval-uated in BD, but there are reports of decreased activity in majordepression and schizophrenia (Porton et al., 2008; Wolkowitz et al.,2012). These data suggest that BD may follow the same direction,and other mechanism by which patients could be benefited by exer-cise.

The extant literature suggests that BD, and manic or hypo-manic symptoms in particular, may be associated with prematureincreases in vascular endothelial dysfunction (Fiedorowicz et al.,2012; Rybakowski et al., 2006). Physical activity as a means to pro-mote vascular endothelial health or telomere length in BD has notyet been assessed; however, poorer physical fitness can confer anincreased risk of affective disorders including BD (Aberg et al., 2012)and exercise has been suggested as a strategy to improve cognitionin BD (Ng et al., 2007). Prospective controlled studies are needed,and related mechanisms require further investigation (Kucyi et al.,2010).

4. Conclusions

There is considerable overlap between neurobiological charac-teristics of BD and processes of both normal and pathological aging,which is evident in clinical, physiological, neurostructural, cellular,and molecular studies. The strengths of the AA integrative view ofBD progression are that the hypothesis comprises and also expendsthe neuroprogression and allostatic load view, also expands thefield of possible biological mechanisms involved in the pathophys-iology of the disease. The AA hypothesis also creates a theoreticalframework that encompasses not only neurological but also sys-temic alterations involved in the pathophysiology of BD.

Although AA hypothesis integrates the current knowledgeregarding progression of BD, it does not provide direct answersabout the mechanisms that generate mood cyclicity. Yet, in

olar disorder as an illness of accelerated aging: Implications foroi.org/10.1016/j.neubiorev.2014.02.004

our concept, mood cyclicity generates stressful conditions thatdemand the action of biological mechanisms to reattain homeo-stasis, the chronicity of this conditions could promote acceleratedaging (Fig. 2). Other limitation is that albeit there are compelling

826

827

828

829

Page 9: The theory of bipolar disorder as an illness of accelerated aging: Implications for clinical care and research

ING ModelN

iobeha

dm

fstsoae

aptii

C

e

UQ4

eePe

A

dN(

HT

R

A

A

AA

A

A

A

AA

A

830

831

832

833

834

835

836

837

838

839

840

841

842

843

844

845

846

847

848

849

850

851

852

853

854

855

856

857

858

859

860

861

862

863

864

865

866

867

868

869

870

871

872

873

874

875

876

877

878

879

880

881

882

883

884

885

886

887

888

889

890

891

892

893

894

895

896

897

898

899

900

901

902

903

904

905

906

907

908

909

910

911

912

913

914

915

916

917

918

919

920

921

922

923

924

925

926

927

928

929

930

931

932

933

934

935

936

937

938

939

940

941

942

943

944

945

946

947

948

949

950

951

952

953

954

955

956

957

958

959

960

961

962

963

964

965

966

967

968

969

970

971

972

973

974

975

ARTICLEBR 1894 1–13

L.B. Rizzo et al. / Neuroscience and B

ata supporting AA hypothesis, most studies use cross-sectionalethodology which are more susceptible to bias.The present work, creates the basis for new prospective studies

ocusing on AA framework and consequently bring more under-tanding about BD etiology and physiology. Although the scope ofhe present work made an in deep analysis of AA in BD, there areome data suggesting that this concept could be extrapolated forther psychiatric disorders like major depression, schizophreniand posttraumatic stress disorder (Simon et al., 2006; Wolkowitzt al., 2010).

Finally, the exploration of physiological processes involved inging provides useful insights into the biology of BD, and newotential to suggest new avenues of treatment. Notwithstanding,he associated of controversy and practical challenges, anti-agingnterventions could be beneficial in subpopulations of BD warrant-ng further investigation.

onflict of interest statement

The authors declare that they have no competing financial inter-sts.

ncited references

Chen et al., 2000, Elkind et al., 2010, Floratou et al., 2012, Glaspert al., 2010, Higashi et al., 2012, Holzenberger et al., 2003, Katant al., 2013, Kelley et al., 2013, Kenyon, 2010, Mitschelen et al., 2011,ark et al., 2011, Pereira et al., 2011, Sesti et al., 2005, Squassinat al., 2013, Swardfager and Black, 2013 and Wang et al., 2010.

cknowledgements

The authors acknowledge support by fellowships from Coor-ination of Improvement of Higher Education Personnel (CAPES),ational Counsel of Technological and Scientific Development

CNPq) and São Paulo Research Foundation (Fapesp).Dr. Swardfager acknowledges support by fellowships from the

eart & Stroke Foundation Centre for Stroke Recovery and theoronto Rehabilitation Institute.

eferences

be, N., Uchida, S., Otsuki, K., Hobara, T., Yamagata, H., Higuchi, F., Shibata, T., Watan-abe, Y., 2011. Altered sirtuin deacetylase gene expression in patients with amood disorder. J. Psychiatr. Res. 45, 1106–1112.

berg, M.A., Waern, M., Nyberg, J., Pedersen, N.L., Bergh, Y., Aberg, N.D., Nilsson, M.,Kuhn, H.G., Toren, K., 2012. Cardiovascular fitness in males at age 18 and risk ofserious depression in adulthood: Swedish prospective population-based study.Br. J. Psychiatry 201, 352–359.

lcain, F.J., Villalba, J.M., 2009. Sirtuin activators. Expert Opin. Ther. Pat. 19, 403–414.lbert, S.M., Michaels, K., Padilla, M., Pelton, G., Bell, K., Marder, K., Stern, Y.,

Devanand, D.P., 1999. Functional significance of mild cognitive impairmentin elderly patients without a dementia diagnosis. Am. J. Geriatr. Psychiatry 7,213–220.

llen, J.S., Bruss, J., Brown, C.K., Damasio, H., 2005. Normal neuroanatomical variationdue to age: the major lobes and a parcellation of the temporal region. Neurobiol.Aging 26, 1245–1260, discussion 1279-1282.

lmanzar, G., Schwaiger, S., Jenewein, B., Keller, M., Herndler-Brandstetter,D., Wurzner, R., Schonitzer, D., Grubeck-Loebenstein, B., 2005. Long-termcytomegalovirus infection leads to significant changes in the composition of theCD8+ T-cell repertoire, which may be the basis for an imbalance in the cytokineproduction profile in elderly persons. J. Virol. 79, 3675–3683.

miard, S., Doudeau, M., Pinte, S., Poulet, A., Lenain, C., Faivre-Moskalenko, C.,Angelov, D., Hug, N., Vindigni, A., Bouvet, P., Paoletti, J., Gilson, E., Giraud-Panis,M.J., 2007. A topological mechanism for TRF2-enhanced strand invasion. Nat.Struct. Mol. Biol. 14, 147–154.

nderson, I.M., Haddad, P.M., Scott, J., 2012. Bipolar disorder. BMJ 345, e8508.ndreazza, A.C., Kapczinski, F., Kauer-Sant’Anna, M., Walz, J.C., Bond, D.J., Goncalves,

Please cite this article in press as: Rizzo, L.B., et al., The theory of bipclinical care and research. Neurosci. Biobehav. Rev. (2014), http://dx.d

C.A., Young, L.T., Yatham, L.N., 2009. 3-Nitrotyrosine and glutathione antioxidantsystem in patients in the early and late stages of bipolar disorder. J. PsychiatryNeurosci. 34, 263–271.

prahamian, I., Nunes, P.V., Forlenza, O.V., 2013. Cognitive impairment and dementiain late-life bipolar disorder. Curr. Opin. Psychiatry 26, 120–123.

PRESSvioral Reviews xxx (2014) xxx–xxx 9

Arancio, O., Chao, M.V., 2007. Neurotrophins, synaptic plasticity and dementia. Curr.Opin. Neurobiol. 17, 325–330.

Arvanitis, D.N., Ducatenzeiler, A., Ou, J.N., Grodstein, E., Andrews, S.D., Tendulkar,S.R., Ribeiro-da-Silva, A., Szyf, M., Cuello, A.C., 2007. High intracellular concen-trations of amyloid-beta block nuclear translocation of phosphorylated CREB. J.Neurochem. 103, 216–228.

Aubert, G., Lansdorp, P.M., 2008. Telomeres and aging. Physiol. Rev. 88, 557–579.Bai, O., Chlan-Fourney, J., Bowen, R., Keegan, D., Li, X.M., 2003. Expression of brain-

derived neurotrophic factor mRNA in rat hippocampus after treatment withantipsychotic drugs. J. Neurosci. Res. 71, 127–131.

Bai, O., Zhang, H., Li, X.M., 2004. Antipsychotic drugs clozapine and olanzapine upreg-ulate bcl-2 mRNA and protein in rat frontal cortex and hippocampus. Brain Res.1010, 81–86.

Baker, L.D., Frank, L.L., Foster-Schubert, K., Green, P.S., Wilkinson, C.W., McTiernan,A., Plymate, S.R., Fishel, M.A., Watson, G.S., Cholerton, B.A., Duncan, G.E., Mehta,P.D., Craft, S., 2010. Effects of aerobic exercise on mild cognitive impairment: acontrolled trial. Arch. Neurol. 67, 71–79.

Bank, A.J., Shammas, R.A., Mullen, K., Chuang, P.P., 1998. Effects of short-termforearm exercise training on resistance vessel endothelial function in normalsubjects and patients with heart failure. J. Card. Fail. 4, 193–201.

Barger, J.L., Kayo, T., Vann, J.M., Arias, E.B., Wang, J., Hacker, T.A., Wang, Y., Raed-erstorff, D., Morrow, J.D., Leeuwenburgh, C., Allison, D.B., Saupe, K.W., Cartee,G.D., Weindruch, R., Prolla, T.A., 2008. A low dose of dietary resveratrol partiallymimics caloric restriction and retards aging parameters in mice. PLoS ONE 3,e2264.

Barnes, D.E., Yaffe, K., Satariano, W.A., Tager, I.B., 2003. A longitudinal study ofcardiorespiratory fitness and cognitive function in healthy older adults. J. Am.Geriatr. Soc. 51, 459–465.

Bearden, C.E., Thompson, P.M., Dalwani, M., Hayashi, K.M., Lee, A.D., Nicoletti, M.,Trakhtenbroit, M., Glahn, D.C., Brambilla, P., Sassi, R.B., Mallinger, A.G., Frank, E.,Kupfer, D.J., Soares, J.C., 2007. Greater cortical gray matter density in lithium-treated patients with bipolar disorder. Biol. Psychiatry 62, 7–16.

Benedict, C.A., Loewendorf, A., Garcia, Z., Blazar, B.R., Janssen, E.M., 2008. Dendriticcell programming by cytomegalovirus stunts naive T cell responses via the PD-L1/PD-1 pathway. J. Immunol. 180, 4836–4847.

Bentz, G.L., Jarquin-Pardo, M., Chan, G., Smith, M.S., Sinzger, C., Yurochko, A.D., 2006.Human cytomegalovirus (HCMV) infection of endothelial cells promotes naivemonocyte extravasation and transfer of productive virus to enhance hematoge-nous dissemination of HCMV. J. Virol. 80, 11539–11555.

Berk, M., 2009. Neuroprogression: pathways to progressive brain changes in bipolardisorder. Int. J. Neuropsychopharmacol. 12, 441–445.

Berk, M., Conus, P., Kapczinski, F., Andreazza, A.C., Yucel, M., Wood, S.J., Pantelis,C., Malhi, G.S., Dodd, S., Bechdolf, A., Amminger, G.P., Hickie, I.B., McGorry, P.D.,2010. From neuroprogression to neuroprotection: implications for clinical care.Med. J. Aust. 193, S36–S40.

Berk, M., Kapczinski, F., Andreazza, A.C., Dean, O.M., Giorlando, F., Maes, M., Yucel, M.,Gama, C.S., Dodd, S., Dean, B., Magalhaes, P.V., Amminger, P., McGorry, P., Malhi,G.S., 2011. Pathways underlying neuroprogression in bipolar disorder: focus oninflammation, oxidative stress and neurotrophic factors. Neurosci. Biobehav.Rev. 35, 804–817.

Beurel, E., Michalek, S.M., Jope, R.S., 2010. Innate and adaptive immune responsesregulated by glycogen synthase kinase-3 (GSK3). Trends Immunol. 31, 24–31.

Blackburn, E.H., 2001. Switching and signaling at the telomere. Cell 106, 661–673.Blackburn, E.H., Chan, S., Chang, J., Fulton, T.B., Krauskopf, A., McEachern, M., Prescott,

J., Roy, J., Smith, C., Wang, H., 2000. Molecular manifestations and moleculardeterminants of telomere capping. Cold Spring Harb. Symp. Quant. Biol. 65,253–263.

Brietzke, E., Kapczinski, F., Grassi-Oliveira, R., Grande, I., Vieta, E., McIntyre, R.S.,2011. Insulin dysfunction and allostatic load in bipolar disorder. Expert Rev.Neurother. 11, 1017–1028.

Brietzke, E., Kauer-Sant’Anna, M., Teixeira, A.L., Kapczinski, F., 2009a. Abnormalitiesin serum chemokine levels in euthymic patients with bipolar disorder. BrainBehav. Immun. 23, 1079–1082.

Brietzke, E., Kauer Sant’anna, M., Jackowski, A., Grassi-Oliveira, R., Bucker, J., Zug-man, A., Mansur, R.B., Bressan, R.A., 2012a. Impact of childhood stress onpsychopathology. Rev. Bras. Psiquiatr. 34, 480–488.

Brietzke, E., Mansur, R.B., Soczynska, J., Powell, A.M., McIntyre, R.S., 2012b. Atheoretical framework informing research about the role of stress in the patho-physiology of bipolar disorder. Prog. Neuro-Psychopharmacol. Biol. Psychiatry39, 1–8.

Brietzke, E., Stertz, L., Fernandes, B.S., Kauer-Sant’anna, M., Mascarenhas, M.,Escosteguy Vargas, A., Chies, J.A., Kapczinski, F., 2009b. Comparison of cytokinelevels in depressed, manic and euthymic patients with bipolar disorder. J. Affect.Disord. 116, 214–217.

Capuron, L., Schroecksnadel, S., Feart, C., Aubert, A., Higueret, D., Barberger-Gateau,P., Laye, S., Fuchs, D., 2011. Chronic low-grade inflammation in elderly per-sons is associated with altered tryptophan and tyrosine metabolism: role inneuropsychiatric symptoms. Biol. Psychiatry 70, 175–182.

Cawthon, R.M., Smith, K.R., O’Brien, E., Sivatchenko, A., Kerber, R.A., 2003. Associationbetween telomere length in blood and mortality in people aged 60 years or older.Lancet 361, 393–395.

olar disorder as an illness of accelerated aging: Implications foroi.org/10.1016/j.neubiorev.2014.02.004

Chen, G., Huang, L.D., Jiang, Y.M., Manji, H.K., 1999. The mood-stabilizing agent val-proate inhibits the activity of glycogen synthase kinase-3. J. Neurochem. 72,1327–1330.

Chen, G., Rajkowska, G., Du, F., Seraji-Bozorgzad, N., Manji, H.K., 2000. Enhancementof hippocampal neurogenesis by lithium. J. Neurochem. 75, 1729–1734.

976

977

978

979

980

Page 10: The theory of bipolar disorder as an illness of accelerated aging: Implications for clinical care and research

ING ModelN

1 iobeha

C

C

C

C

CQ5

C

d

D

D

d

D

D

D

d

D

D

E

E

E

E

E

F

F

F

F

F

F

F

981

982

983

984

985

986

987

988

989

990

991

992

993

994

995

996

997

998

999

1000

1001

1002

1003

1004

1005

1006

1007

1008

1009

1010

1011

1012

1013

1014

1015

1016

1017

1018

1019

1020

1021

1022

1023

1024

1025

1026

1027

1028

1029

1030

1031

1032

1033

1034

1035

1036

1037

1038

1039

1040

1041

1042

1043

1044

1045

1046

1047

1048

1049

1050

1051

1052

1053

1054

1055

1056

1057

1058

1059

1060

1061

1062

1063

1064

1065

1066

1067

1068

1069

1070

1071

1072

1073

1074

1075

1076

1077

1078

1079

1080

1081

1082

1083

1084

1085

1086

1087

1088

1089

1090

1091

1092

1093

1094

1095

1096

1097

1098

1099

1100

1101

1102

1103

1104

1105

1106

1107

1108

1109

1110

1111

1112

1113

1114

1115

1116

1117

1118

1119

1120

1121

1122

1123

1124

1125

1126

1127

1128

1129

1130

1131

1132

1133

1134

1135

1136

1137

1138

1139

1140

1141

1142

1143

1144

1145

1146

1147

ARTICLEBR 1894 1–13

0 L.B. Rizzo et al. / Neuroscience and B

hen, R.W., Chuang, D.M., 1999. Long term lithium treatment suppresses p53 andBax expression but increases Bcl-2 expression. A prominent role in neuropro-tection against excitotoxicity. J. Biol. Chem. 274, 6039–6042.

hiu, C.T., Chuang, D.M., 2011. Neuroprotective action of lithium in disorders of thecentral nervous system. Zhong Nan Da Xue Xue Bao Yi Xue Ban 36, 461–476.

olcombe, S.J., Erickson, K.I., Scalf, P.E., Kim, J.S., Prakash, R., McAuley, E., Elavsky,S., Marquez, D.X., Hu, L., Kramer, A.F., 2006. Aerobic exercise training increasesbrain volume in aging humans. J. Gerontol. A: Biol. Sci. Med. Sci. 61, 1166–1170.

onnor, C.M., Guo, Y., Akbarian, S., 2009. Cingulate white matter neurons inschizophrenia and bipolar disorder. Biol. Psychiatry 66, 486–493.

rump, C., Sundquist, K., Winkleby, M.A., Sundquist, J., 2013. Comorbidities andmortality in bipolar disorder: a Swedish National Cohort Study. JAMA Psychiatry.

zepielewski, L., Daruy Filho, L., Brietzke, E., Grassi-Oliveira, R., 2013. Bipolar dis-order and metabolic syndrome: a systematic review. Rev. Bras. Psiquiatr. 35,88–93.

a Silva, J., Goncalves-Pereira, M., Xavier, M., Mukaetova-Ladinska, E.B., 2013.Affective disorders and risk of developing dementia: systematic review. Br. J.Psychiatry 202, 177–186.

as, S., Basu, A., 2007. Inflammation: a new candidate in modulating adult neuro-genesis. J. Neurosci. Res.

avid Adams Jr., J.K., Lori, K., 2007. Sirtuins, nicotinamide and aging: a critical review.Lett. Drug Des. Discov. 4, 44–48.

e Jong, D., Jansen, R., Hoefnagels, W., Jellesma-Eggenkamp, M., Verbeek, M., Borm,G., Kremer, B., 2008. No effect of one-year treatment with indomethacin onAlzheimer’s disease progression: a randomized controlled trial. PLoS ONE 3,e1475.

e Sandre-Giovannoli, A., Bernard, R., Cau, P., Navarro, C., Amiel, J., Boccaccio, I.,Lyonnet, S., Stewart, C.L., Munnich, A., Le Merrer, M., Levy, N., 2003. Lamin atruncation in Hutchinson-Gilford progeria. Science 300, 2055.

eCarli, C., Massaro, J., Harvey, D., Hald, J., Tullberg, M., Au, R., Beiser, A., D’Agostino,R., Wolf, P.A., 2005. Measures of brain morphology and infarction in the fram-ingham heart study: establishing what is normal. Neurobiol. Aging 26, 491–510.

erhovanessian, E., Larbi, A., Pawelec, G., 2009. Biomarkers of human immunosenes-cence: impact of Cytomegalovirus infection. Curr. Opin. Immunol. 21, 440–445.

o Prado, C.H., Rizzo, L.B., Wieck, A., Lopes, R.P., Teixeira, A.L., Grassi-Oliveira, R.,Bauer, M.E., 2013. Reduced regulatory T cells are associated with higher levels ofTh1/TH17 cytokines and activated MAPK in type 1 bipolar disorder. Psychoneu-roendocrinology 38, 667–676.

ouillard-Guilloux, G., Guilloux, J.P., Lewis, D.A., Sibille, E., 2013. Anticipated brainmolecular aging in major depression. Am. J. Geriatr. Psychiatry 21, 450–460.

riscoll, I., Martin, B., An, Y., Maudsley, S., Ferrucci, L., Mattson, M.P., Resnick, S.M.,2012. Plasma BDNF is associated with age-related white matter atrophy but notwith cognitive function in older, non-demented adults. PLoS ONE 7, e35217.

lkind, M.S., Luna, J.M., Moon, Y.P., Boden-Albala, B., Liu, K.M., Spitalnik, S., Rundek,T., Sacco, R.L., Paik, M.C., 2010. Infectious burden and carotid plaque thickness:the northern Manhattan study. Stroke 41, e117–e122.

lvsashagen, T., Vera, E., Boen, E., Bratlie, J., Andreassen, O.A., Josefsen, D., Malt, U.F.,Blasco, M.A., Boye, B., 2011. The load of short telomeres is increased and associ-ated with lifetime number of depressive episodes in bipolar II disorder. J. Affect.Disord. 135, 43–50.

nzinger, C., Fazekas, F., Matthews, P.M., Ropele, S., Schmidt, H., Smith, S., Schmidt, R.,2005. Risk factors for progression of brain atrophy in aging: six-year follow-upof normal subjects. Neurology 64, 1704–1711.

rickson, K.I., Voss, M.W., Prakash, R.S., Basak, C., Szabo, A., Chaddock, L., Kim, J.S.,Heo, S., Alves, H., White, S.M., Wojcicki, T.R., Mailey, E., Vieira, V.J., Martin,S.A., Pence, B.D., Woods, J.A., McAuley, E., Kramer, A.F., 2011. Exercise train-ing increases size of hippocampus and improves memory. Proc. Natl. Acad. Sci.U.S.A. 108, 3017–3022.

riksson, M., Brown, W.T., Gordon, L.B., Glynn, M.W., Singer, J., Scott, L., Erdos, M.R.,Robbins, C.M., Moses, T.Y., Berglund, P., Dutra, A., Pak, E., Durkin, S., Csoka, A.B.,Boehnke, M., Glover, T.W., Collins, F.S., 2003. Recurrent de novo point mutationsin lamin A cause Hutchinson-Gilford progeria syndrome. Nature 423, 293–298.

agiolini, A., Chengappa, K.N., Soreca, I., Chang, J., 2008. Bipolar disorder and themetabolic syndrome: causal factors, psychiatric outcomes and economic bur-den. CNS Drugs 22, 655–669.

ernandes, B.S., Gama, C.S., Cereser, K.M., Yatham, L.N., Fries, G.R., Colpo, G., deLucena, D., Kunz, M., Gomes, F.A., Kapczinski, F., 2011. Brain-derived neu-rotrophic factor as a state-marker of mood episodes in bipolar disorders: asystematic review and meta-regression analysis. J. Psychiatr. Res. 45, 995–1004.

iedorowicz, J.G., Coryell, W.H., Rice, J.P., Warren, L.L., Haynes, W.G., 2012. Vas-culopathy related to manic/hypomanic symptom burden and first-generationantipsychotics in a sub-sample from the collaborative depression study. Psy-chother. Psychosom. 81, 235–243.

loratou, K., Giannopoulou, E., Antonacopoulou, A., Karakantza, M., Adonakis, G.,Kardamakis, D., Matsouka, P., 2012. Oxidative stress due to radiation in CD34(+)hematopoietic progenitor cells: protection by IGF-1. J. Radiat. Res. 53, 672–685.

otenos, A.F., Snyder, A.Z., Girton, L.E., Morris, J.C., Buckner, R.L., 2005. Normativeestimates of cross-sectional and longitudinal brain volume decline in aging andAD. Neurology 64, 1032–1039.

ranceschi, C., Capri, M., Monti, D., Giunta, S., Olivieri, F., Sevini, F., Panourgia, M.P.,Invidia, L., Celani, L., Scurti, M., Cevenini, E., Castellani, G.C., Salvioli, S., 2007.

Please cite this article in press as: Rizzo, L.B., et al., The theory of bipclinical care and research. Neurosci. Biobehav. Rev. (2014), http://dx.d

Inflammaging and anti-inflammaging: a systemic perspective on aging andlongevity emerged from studies in humans. Mech. Ageing Dev. 128, 92–105.

ries, G.R., Pfaffenseller, B., Stertz, L., Paz, A.V., Dargel, A.A., Kunz, M., Kapczinski, F.,2012. Staging and neuroprogression in bipolar disorder. Curr. Psychiatry Rep.14, 667–675.

PRESSvioral Reviews xxx (2014) xxx–xxx

Fuchs, T., Trollor, J.N., Crawford, J., Brown, D.A., Baune, B.T., Samaras, K., Campbell,L., Breit, S.N., Brodaty, H., Sachdev, P., Smith, E., 2013. Macrophage inhibitorycytokine-1 is associated with cognitive impairment and predicts cognitivedecline – the Sydney Memory and Ageing Study. Aging Cell.

Fuchsjager-Mayrl, G., Pleiner, J., Wiesinger, G.F., Sieder, A.E., Quittan, M., Nuhr, M.J.,Francesconi, C., Seit, H.P., Francesconi, M., Schmetterer, L., Wolzt, M., 2002. Exer-cise training improves vascular endothelial function in patients with type 1diabetes. Diabetes Care 25, 1795–1801.

Fukumoto, T., Morinobu, S., Okamoto, Y., Kagaya, A., Yamawaki, S., 2001. Chroniclithium treatment increases the expression of brain-derived neurotrophic factorin the rat brain. Psychopharmacology (Berl.) 158, 100–106.

Geerlings, M.I., den Heijer, T., Koudstaal, P.J., Hofman, A., Breteler, M.M., 2008. Historyof depression, depressive symptoms, and medial temporal lobe atrophy and therisk of Alzheimer disease. Neurology 70, 1258–1264.

Gergerlioglu, H.S., Savas, H.A., Bulbul, F., Selek, S., Uz, E., Yumru, M., 2007. Changes innitric oxide level and superoxide dismutase activity during antimanic treatment.Prog. Neuropsychopharmacol. Biol. Psychiatry 31, 697–702.

Ghedim, F.V., Fraga Dde, B., Deroza, P.F., Oliveira, M.B., Valvassori, S.S., Steckert, A.V.,Budni, J., Dal-Pizzol, F., Quevedo, J., Zugno, A.I., 2012. Evaluation of behavioraland neurochemical changes induced by ketamine in rats: implications as ananimal model of mania. J. Psychiatr. Res. 46, 1569–1575.

Giunta, S., 2008. Exploring the complex relations between inflammation andaging (inflamm-aging): anti-inflamm-aging remodelling of inflamm-aging, fromrobustness to frailty. Inflamm. Res. 57, 558–563.

Glasper, E.R., Llorens-Martin, M.V., Leuner, B., Gould, E., Trejo, J.L., 2010. Blockade ofinsulin-like growth factor-I has complex effects on structural plasticity in thehippocampus. Hippocampus 20, 706–712.

Goldstein, B.I., Fagiolini, A., Houck, P., Kupfer, D.J., 2009a. Cardiovascular disease andhypertension among adults with bipolar I disorder in the United States. BipolarDisord. 11, 657–662.

Goldstein, B.I., Kemp, D.E., Soczynska, J.K., McIntyre, R.S., 2009b. Inflammation andthe phenomenology, pathophysiology, comorbidity, and treatment of bipo-lar disorder: a systematic review of the literature. J. Clin. Psychiatry 70,1078–1090.

Gomez-Isla, T., Blesa, R., Boada, M., Clarimon, J., Del Ser, T., Domenech, G., Ferro,J.M., Gomez-Anson, B., Manubens, J.M., Martinez-Lage, J.M., Munoz, D., Pena-Casanova, J., Torres, F., 2008. A randomized, double-blind, placebo controlled-trial of triflusal in mild cognitive impairment: the TRIMCI study. Alzheimer Dis.Assoc. Disord. 22, 21–29.

Goronzy, J.J., Fulbright, J.W., Crowson, C.S., Poland, G.A., O’Fallon, W.M., Weyand,C.M., 2001. Value of immunological markers in predicting responsiveness toinfluenza vaccination in elderly individuals. J. Virol. 75, 12182–12187.

Goshen, I., Kreisel, T., Ben-Menachem-Zidon, O., Licht, T., Weidenfeld, J., Ben-Hur, T., Yirmiya, R., 2007. Brain interleukin-1 mediates chronic stress-induceddepression in mice via adrenocortical activation and hippocampal neurogenesissuppression. Mol. Psychiatry.

Grabiec, A.M., Tak, P.P., Reedquist, K.A., 2011. Function of histone deacetylaseinhibitors in inflammation. Crit. Rev. Immunol. 31, 233–263.

Grande, I., Magalhaes, P.V., Kunz, M., Vieta, E., Kapczinski, F., 2012. Mediators ofallostasis and systemic toxicity in bipolar disorder. Physiol. Behav. 106, 46–50.

Gualtieri, C.T., Johnson, L.G., 2008. Age-related cognitive decline in patients withmood disorders. Prog. Neuropsychopharmacol. Biol. Psychiatry 32, 962–967.

Guo, S., Arai, K., Stins, M.F., Chuang, D.M., Lo, E.H., 2009. Lithium upregulates vascularendothelial growth factor in brain endothelial cells and astrocytes. Stroke 40,652–655.

Hallahan, B., Newell, J., Soares, J.C., Brambilla, P., Strakowski, S.M., Fleck, D.E.,Kieseppa, T., Altshuler, L.L., Fornito, A., Malhi, G.S., McIntosh, A.M., Yurgelun-Todd, D.A., Labar, K.S., Sharma, V., MacQueen, G.M., Murray, R.M., McDonald,C., 2011. Structural magnetic resonance imaging in bipolar disorder: an inter-national collaborative mega-analysis of individual adult patient data. Biol.Psychiatry 69, 326–335.

Harman, D., 2009. Origin and evolution of the free radical theory of aging: a briefpersonal history, 1954–2009. Biogerontology 10, 773–781.

Harrison, P.J., 2002. The neuropathology of primary mood disorder. Brain 125,1428–1449.

Hartmann, N., Boehner, M., Groenen, F., Kalb, R., 2010. Telomere length of patientswith major depression is shortened but independent from therapy and severityof the disease. Depress Anxiety 27, 1111–1116.

Hashimoto, R., Hough, C., Nakazawa, T., Yamamoto, T., Chuang, D.M., 2002. Lithiumprotection against glutamate excitotoxicity in rat cerebral cortical neurons:involvement of NMDA receptor inhibition possibly by decreasing NR2B tyrosinephosphorylation. J. Neurochem. 80, 589–597.

Hayflick, L., Moorhead, P.S., 1961. The serial cultivation of human diploid cell strains.Exp. Cell Res. 25, 585–621.

Hegele, R.A., 2003. Drawing the line in progeria syndromes. Lancet 362, 416–417.Heyn, H., Moran, S., Esteller, M., 2013. Aberrant DNA methylation profiles in the

premature aging disorders Hutchinson-Gilford Progeria and Werner syndrome.Epigenetics 8, 28–33.

Higashi, Y., Sukhanov, S., Anwar, A., Shai, S.Y., Delafontaine, P., 2012. Aging,atherosclerosis, and IGF-1. J. Gerontol. A: Biol. Sci. Med. Sci. 67, 626–639.

Holzenberger, M., Dupont, J., Ducos, B., Leneuve, P., Geloen, A., Even, P.C., Cervera, P.,

olar disorder as an illness of accelerated aging: Implications foroi.org/10.1016/j.neubiorev.2014.02.004

Le Bouc, Y., 2003. IGF-1 receptor regulates lifespan and resistance to oxidativestress in mice. Nature 421, 182–187.

Ikram, M.A., Vrooman, H.A., Vernooij, M.W., van der Lijn, F., Hofman, A., van derLugt, A., Niessen, W.J., Breteler, M.M., 2008. Brain tissue volumes in the generalelderly population. The Rotterdam Scan Study. Neurobiol. Aging 29, 882–890.

1148

1149

1150

1151

1152

Page 11: The theory of bipolar disorder as an illness of accelerated aging: Implications for clinical care and research

ING ModelN

iobeha

I

J

J

K

K

K

K

K

K

K

K

K

KK

K

K

K

K

K

K

L

L

L

L

L

L

M

M

1153

1154

1155

1156

1157

1158

1159

1160

1161

1162

1163

1164

1165

1166

1167

1168

1169

1170

1171

1172

1173

1174

1175

1176

1177

1178

1179

1180

1181

1182

1183

1184

1185

1186

1187

1188

1189

1190

1191

1192

1193

1194

1195

1196

1197

1198

1199

1200

1201

1202

1203

1204

1205

1206

1207

1208

1209

1210

1211

1212

1213

1214

1215

1216

1217

1218

1219

1220

1221

1222

1223

1224

1225

1226

1227

1228

1229

1230

1231

1232

1233

1234

1235

1236

1237

1238

1239

1240

1241

1242

1243

1244

1245

1246

1247

1248

1249

1250

1251

1252

1253

1254

1255

1256

1257

1258

1259

1260

1261

1262

1263

1264

1265

1266

1267

1268

1269

1270

1271

1272

1273

1274

1275

1276

1277

1278

1279

1280

1281

1282

1283

1284

1285

1286

1287

1288

1289

1290

1291

1292

1293

1294

1295

1296

1297

1298

1299

1300

1301

1302

1303

1304

1305

1306

1307

1308

1309

1310

1311

1312

1313

1314

1315

1316

1317

1318

1319

ARTICLEBR 1894 1–13

L.B. Rizzo et al. / Neuroscience and B

to, K., Colley, T., Mercado, N., 2012. Geroprotectors as a novel therapeutic strategyfor COPD, an accelerating aging disease. Int. J. Chron. Obstruct. Pulmon. Dis. 7,641–652.

ernigan, T.L., Archibald, S.L., Fennema-Notestine, C., Gamst, A.C., Stout, J.C., Bonner,J., Hesselink, J.R., 2001. Effects of age on tissues and regions of the cerebrum andcerebellum. Neurobiol. Aging 22, 581–594.

oca, S.R., Ferreira, F.R., Guimaraes, F.S., 2007. Modulation of stress consequencesby hippocampal monoaminergic, glutamatergic and nitrergic neurotransmittersystems. Stress 10, 227–249.

anfi, Y., Naiman, S., Amir, G., Peshti, V., Zinman, G., Nahum, L., Bar-Joseph, Z.,Cohen, H.Y., 2012. The sirtuin SIRT6 regulates lifespan in male mice. Nature483, 218–221.

ao, C.Y., Hsu, Y.C., Liu, J.W., Lee, D.C., Chung, Y.F., Chiu, I.M., 2013. The mood stabi-lizer valproate activates human FGF1 gene promoter through inhibiting HDACand GSK-3 activities. J. Neurochem. 126, 4–18.

apczinski, F., Dias, V.V., Kauer-Sant’Anna, M., Brietzke, E., Vazquez, G.H., Vieta, E.,Berk, M., 2009. The potential use of biomarkers as an adjunctive tool for stagingbipolar disorder. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 33, 1366–1371.

apczinski, F., Vieta, E., Andreazza, A.C., Frey, B.N., Gomes, F.A., Tramontina, J., Kauer-Sant’anna, M., Grassi-Oliveira, R., Post, R.M., 2008. Allostatic load in bipolardisorder: implications for pathophysiology and treatment. Neurosci. Biobehav.Rev. 32, 675–692.

arten, Y.J., Olariu, A., Cameron, H.A., 2005. Stress in early life inhibits neurogenesisin adulthood. Trends Neurosci. 28, 171–172.

atan, M., Moon, Y.P., Paik, M.C., Sacco, R.L., Wright, C.B., Elkind, M.S., 2013. Infectiousburden and cognitive function: The Northern Manhattan Study. Neurology 80,1209–1215.

auer-Sant’Anna, M., Kapczinski, F., Andreazza, A.C., Bond, D.J., Lam, R.W., Young,L.T., Yatham, L.N., 2009. Brain-derived neurotrophic factor and inflammatorymarkers in patients with early- vs. late-stage bipolar disorder. Int. J. Neuropsy-chopharmacol. 12, 447–458.

awahara, T.L., Michishita, E., Adler, A.S., Damian, M., Berber, E., Lin, M., McCord, R.A.,Ongaigui, K.C., Boxer, L.D., Chang, H.Y., Chua, K.F., 2009. SIRT6 links histone H3lysine 9 deacetylation to NF-kappaB-dependent gene expression and organismallife span. Cell 136, 62–74.

elley, K.W., O’Connor, J.C., Lawson, M.A., Dantzer, R., Rodriguez-Zas, S.L., McCusker,R.H., 2013. Aging leads to prolonged duration of inflammation-induceddepression-like behavior caused by Bacillus Calmette-Guerin. Brain Behav.Immun. 32, 63–69.

enyon, C.J., 2010. The genetics of ageing. Nature 464, 504–512.essing, L.V., Andersen, P.K., 2004. Does the risk of developing dementia increase

with the number of episodes in patients with depressive disorder and in patientswith bipolar disorder. J. Neurol. Neurosurg. Psychiatry 75, 1662–1666.

ita, Y., Baba, H., Maeshima, H., Nakano, Y., Suzuki, T., Arai, H., 2009. Serum amyloidbeta protein in young and elderly depression: a pilot study. Psychogeriatrics 9,180–185.

onarski, J.Z., McIntyre, R.S., Kennedy, S.H., Rafi-Tari, S., Soczynska, J.K., Ketter, T.A.,2008. Volumetric neuroimaging investigations in mood disorders: bipolar dis-order versus major depressive disorder. Bipolar Disord. 10, 1–37.

orte, M., Carroll, P., Wolf, E., Brem, G., Thoenen, H., Bonhoeffer, T., 1995. Hip-pocampal long-term potentiation is impaired in mice lacking brain-derivedneurotrophic factor. Proc. Natl. Acad. Sci. U.S.A. 92, 8856–8860.

ucyi, A., Alsuwaidan, M.T., Liauw, S.S., McIntyre, R.S., 2010. Aerobic physical exer-cise as a possible treatment for neurocognitive dysfunction in bipolar disorder.Postgrad. Med. 122, 107–116.

ugaya, A., Sanacora, G., 2005. Beyond monoamines: glutamatergic function in mooddisorders. CNS Spectr. 10, 808–819.

uhn, H.G., Dickinson-Anson, H., Gage, F.H., 1996. Neurogenesis in the dentate gyrusof the adult rat: age-related decrease of neuronal progenitor proliferation. J.Neurosci. 16, 2027–2033.

ai, J.S., Zhao, C., Warsh, J.J., Li, P.P., 2006. Cytoprotection by lithium and valproatevaries between cell types and cellular stresses. Eur. J. Pharmacol. 539, 18–26.

evine, E.S., Crozier, R.A., Black, I.B., Plummer, M.R., 1998. Brain-derived neu-rotrophic factor modulates hippocampal synaptic transmission by increasingN-methyl-d-aspartic acid receptor activity. Proc. Natl. Acad. Sci. U.S.A. 95,10235–10239.

isy, M.E., Jarvis, K.B., DelBello, M.P., Mills, N.P., Weber, W.A., Fleck, D., Strakowski,S.M., Adler, C.M., 2011. Progressive neurostructural changes in adolescent andadult patients with bipolar disorder. Bipolar Disord. 13, 396–405.

lorens-Martin, M., Torres-Aleman, I., Trejo, J.L., 2010. Exercise modulates insulin-like growth factor 1-dependent and -independent effects on adult hippocampalneurogenesis and behaviour. Mol. Cell. Neurosci. 44, 109–117.

udlow, A.T., Witkowski, S., Marshall, M.R., Wang, J., Lima, L.C., Guth, L.M., Span-genburg, E.E., Roth, S.M., 2012. Chronic exercise modifies age-related telomeredynamics in a tissue-specific fashion. J. Gerontol. A: Biol. Sci. Med. Sci. 67,911–926.

ue, L.F., Kuo, Y.M., Roher, A.E., Brachova, L., Shen, Y., Sue, L., Beach, T., Kurth, J.H.,Rydel, R.E., Rogers, J., 1999. Soluble amyloid beta peptide concentration as apredictor of synaptic change in Alzheimer’s disease. Am. J. Pathol. 155, 853–862.

acaulay, R., Akbar, A.N., Henson, S.M., 2013. The role of the T cell in age-relatedinflammation. Age (Dordr.) 35, 563–572.

Please cite this article in press as: Rizzo, L.B., et al., The theory of bipclinical care and research. Neurosci. Biobehav. Rev. (2014), http://dx.d

achado-Vieira, R., Andreazza, A.C., Viale, C.I., Zanatto, V., Cereser Jr., V., da SilvaVargas, R., Kapczinski, F., Portela, L.V., Souza, D.O., Salvador, M., Gentil, V., 2007.Oxidative stress parameters in unmedicated and treated bipolar subjects duringinitial manic episode: a possible role for lithium antioxidant effects. Neurosci.Lett. 421, 33–36.

PRESSvioral Reviews xxx (2014) xxx–xxx 11

Machado-Vieira, R., Manji, H.K., Zarate, C.A., 2009. The role of the tripartite gluta-matergic synapse in the pathophysiology and therapeutics of mood disorders.Neuroscientist 15, 525–539.

Magalhaes, P.V., Jansen, K., Pinheiro, R.T., Colpo, G.D., da Motta, L.L., Klamt, F.,da Silva, R.A., Kapczinski, F., 2012. Peripheral oxidative damage in early-stagemood disorders: a nested population-based case-control study. Int. J. Neuropsy-chopharmacol. 15, 1043–1050.

Mahon, K., Burdick, K.E., Szeszko, P.R., 2010. A role for white matter abnormalities inthe pathophysiology of bipolar disorder. Neurosci. Biobehav. Rev. 34, 533–554.

Mahon, K., Wu, J., Malhotra, A.K., Burdick, K.E., DeRosse, P., Ardekani, B.A., Szeszko,P.R., 2009. A voxel-based diffusion tensor imaging study of white matter inbipolar disorder. Neuropsychopharmacology 34, 1590–1600.

Mansur, R.B., Cha, D.S., Asevedo, E., McIntyre, R.S., Brietzke, E., 2013. Selfish brainand neuroprogression in bipolar disorder. Prog. Neuropsychopharmacol. Biol.Psychiatry 43, 66–71.

Martinez-Aran, A., Vieta, E., Colom, F., Reinares, M., Benabarre, A., Torrent, C.,Goikolea, J.M., Corbella, B., Sanchez-Moreno, J., Salamero, M., 2002. Neu-ropsychological performance in depressed and euthymic bipolar patients.Neuropsychobiology 46 (Suppl. 1), 16–21.

Martinez-Aran, A., Vieta, E., Colom, F., Torrent, C., Reinares, M., Goikolea, J.M., Ben-abarre, A., Comes, M., Sanchez-Moreno, J., 2005. Do cognitive complaints ineuthymic bipolar patients reflect objective cognitive impairment? Psychother.Psychosom. 74, 295–302.

Martinez-Aran, A., Vieta, E., Reinares, M., Colom, F., Torrent, C., Sanchez-Moreno, J.,Benabarre, A., Goikolea, J.M., Comes, M., Salamero, M., 2004. Cognitive functionacross manic or hypomanic, depressed, and euthymic states in bipolar disorder.Am. J. Psychiatry 161, 262–270.

Martinez-Aran, A., Vieta, E., Torrent, C., Sanchez-Moreno, J., Goikolea, J.M., Salamero,M., Malhi, G.S., Gonzalez-Pinto, A., Daban, C., Alvarez-Grandi, S., Fountoulakis, K.,Kaprinis, G., Tabares-Seisdedos, R., Ayuso-Mateos, J.L., 2007. Functional outcomein bipolar disorder: the role of clinical and cognitive factors. Bipolar Disord. 9,103–113.

Martinsson, L., Wei, Y., Xu, D., Melas, P.A., Mathe, A.A., Schalling, M., Lavebratt, C.,Backlund, L., 2013. Long-term lithium treatment in bipolar disorder is associatedwith longer leukocyte telomeres. Translational psychiatry 3, e261.

McIntyre, R.S., Konarski, J.Z., Misener, V.L., Kennedy, S.H., 2005. Bipolar disorder anddiabetes mellitus: epidemiology, etiology, and treatment implications. Ann. Clin.Psychiatry 17, 83–93.

McIntyre, R.S., Konarski, J.Z., Soczynska, J.K., Wilkins, K., Panjwani, G., Bouffard, B.,Bottas, A., Kennedy, S.H., 2006. Medical comorbidity in bipolar disorder: impli-cations for functional outcomes and health service utilization. Psychiatr. Serv.57, 1140–1144.

Miller, C.L., Llenos, I.C., Cwik, M., Walkup, J., Weis, S., 2008. Alterations in kynurenineprecursor and product levels in schizophrenia and bipolar disorder. Neurochem.Int. 52, 1297–1303.

Miller, C.L., Llenos, I.C., Dulay, J.R., Weis, S., 2006. Upregulation of the initiating stepof the kynurenine pathway in postmortem anterior cingulate cortex from indi-viduals with schizophrenia and bipolar disorder. Brain Res. 1073–1074, 25–37.

Milne, J.C., Lambert, P.D., Schenk, S., Carney, D.P., Smith, J.J., Gagne, D.J., Jin, L., Boss,O., Perni, R.B., Vu, C.B., Bemis, J.E., Xie, R., Disch, J.S., Ng, P.Y., Nunes, J.J., Lynch,A.V., Yang, H., Galonek, H., Israelian, K., Choy, W., Iffland, A., Lavu, S., Medvedik,O., Sinclair, D.A., Olefsky, J.M., Jirousek, M.R., Elliott, P.J., Westphal, C.H., 2007.Small molecule activators of SIRT1 as therapeutics for the treatment of type 2diabetes. Nature 450, 712–716.

Mitschelen, M., Yan, H., Farley, J.A., Warrington, J.P., Han, S., Herenu, C.B., Csiszar,A., Ungvari, Z., Bailey-Downs, L.C., Bass, C.E., Sonntag, W.E., 2011. Long-termdeficiency of circulating and hippocampal insulin-like growth factor I inducesdepressive behavior in adult mice: a potential model of geriatric depression.Neuroscience 185, 50–60.

Modabbernia, A., Taslimi, S., Brietzke, E., Ashrafi, M., 2013. Cytokine alterations inbipolar disorder: a meta-analysis of 30 studies. Biol. Psychiatry 74, 15–25.

Moore, G.J., Bebchuk, J.M., Hasanat, K., Chen, G., Seraji-Bozorgzad, N., Wilds, I.B.,Faulk, M.W., Koch, S., Glitz, D.A., Jolkovsky, L., Manji, H.K., 2000. Lithiumincreases N-acetyl-aspartate in the human brain: in vivo evidence in supportof bcl-2’s neurotrophic effects? Biol. Psychiatry 48, 1–8.

Naydenov, A.V., MacDonald, M.L., Ongur, D., Konradi, C., 2007. Differences in lym-phocyte electron transport gene expression levels between subjects with bipolardisorder and normal controls in response to glucose deprivation stress. Arch.Gen. Psychiatry 64, 555–564.

Ng, F., Dodd, S., Berk, M., 2007. The effects of physical activity in the acute treatmentof bipolar disorder: a pilot study. J. Affect. Disord. 101, 259–262.

O’Neill, P., 1997. Aging homeostasis. Rev. Clin. Gerontol. 7, 199–211.Olsson, J., Wikby, A., Johansson, B., Lofgren, S., Nilsson, B.O., Ferguson, F.G., 2000.

Age-related change in peripheral blood T-lymphocyte subpopulations andcytomegalovirus infection in the very old: the Swedish longitudinal OCTOimmune study. Mech. Ageing Dev. 121, 187–201.

Osby, U., Brandt, L., Correia, N., Ekbom, A., Sparen, P., 2001. Excess mortality in bipolarand unipolar disorder in Sweden. Archiv. Gen. Psychiatry 58, 844–850.

Osorio, R.S., Gumb, T., Pomara, N., 2013. Soluble amyloid-beta levels and late-lifedepression. Curr Pharm Des.

Ouyang, Q., Wagner, W.M., Wikby, A., Walter, S., Aubert, G., Dodi, A.I., Travers, P.,

olar disorder as an illness of accelerated aging: Implications foroi.org/10.1016/j.neubiorev.2014.02.004

Pawelec, G., 2003. Large numbers of dysfunctional CD8+ T lymphocytes bearingreceptors for a single dominant CMV epitope in the very old. J. Clin. Immunol.23, 247–257.

Padmos, R.C., Bekris, L., Knijff, E.M., Tiemeier, H., Kupka, R.W., Cohen, D.,Nolen, W.A., Lernmark, A., Drexhage, H.A., 2004. A high prevalence of

1320

1321

1322

1323

1324

Page 12: The theory of bipolar disorder as an illness of accelerated aging: Implications for clinical care and research

ING ModelN

1 iobeha

P

P

P

P

P

P

P

P

Q

RR

R

R

R

R

R

R

RR

R

S

S

S

S

S

S

S

S

1325

1326

1327

1328

1329

1330

1331

1332

1333

1334

1335

1336

1337

1338

1339

1340

1341

1342

1343

1344

1345

1346

1347

1348

1349

1350

1351

1352

1353

1354

1355

1356

1357

1358

1359

1360

1361

1362

1363

1364

1365

1366

1367

1368

1369

1370

1371

1372

1373

1374

1375

1376

1377

1378

1379

1380

1381

1382

1383

1384

1385

1386

1387

1388

1389

1390

1391

1392

1393

1394

1395

1396

1397

1398

1399

1400

1401

1402

1403

1404

1405

1406

1407

1408

1409

1410

1411

1412

1413

1414

1415

1416

1417

1418

1419

1420

1421

1422

1423

1424

1425

1426

1427

1428

1429

1430

1431

1432

1433

1434

1435

1436

1437

1438

1439

1440

1441

1442

1443

1444

1445

1446

1447

1448

1449

1450

1451

1452

1453

1454

1455

1456

1457

1458

1459

1460

1461

1462

1463

1464

1465

1466

1467

1468

1469

1470

1471

1472

1473

1474

1475

1476

1477

1478

1479

1480

1481

1482

1483

1484

1485

1486

1487

1488

1489

1490

1491

ARTICLEBR 1894 1–13

2 L.B. Rizzo et al. / Neuroscience and B

organ-specific autoimmunity in patients with bipolar disorder. Biol. Psychiatry56, 476–482.

apanastasiou, E., Gaughran, F., Smith, S., 2011. Schizophrenia as segmental progeria.J. R. Soc. Med. 104, 475–484.

ark, S.E., Lawson, M., Dantzer, R., Kelley, K.W., McCusker, R.H., 2011. Insulin-likegrowth factor-I peptides act centrally to decrease depression-like behavior ofmice treated intraperitoneally with lipopolysaccharide. J. Neuroinflamm. 8, 179.

awelec, G., Derhovanessian, E., 2011. Role of CMV in immune senescence. VirusRes. 157, 175–179.

enna, C., Perrelli, M.G., Pagliaro, P., 2013. Mitochondrial pathways, permeabilitytransition pore, and redox signaling in cardioprotection: therapeutic implica-tions. Antioxid. Redox Signal. 18, 556–599.

ereira, A.C., McQuillin, A., Puri, V., Anjorin, A., Bass, N., Kandaswamy, R., Lawrence,J., Curtis, D., Sklar, P., Purcell, S.M., Gurling, H.M., 2011. Genetic association andsequencing of the insulin-like growth factor 1 gene in bipolar affective disorder.Am. J. Med. Genet. B: Neuropsychiatr. Genet. 156, 177–187.

erovic, M., Tesic, V., Mladenovic Djordjevic, A., Smiljanic, K., Loncarevic-Vasiljkovic,N., Ruzdijic, S., Kanazir, S., 2012. BDNF transcripts, proBDNF and proNGF, in thecortex and hippocampus throughout the life span of the rat. Age (Dordr).

iccinni, A., Origlia, N., Veltri, A., Vizzaccaro, C., Marazziti, D., Catena-Dell’osso, M.,Conversano, C., Moroni, I., Domenici, L., Dell’osso, L., 2012. Plasma beta-amyloidpeptides levels: a pilot study in bipolar depressed patients. J. Affect. Disord. 138,160–164.

orton, B., Delisi, L.E., Bertisch, H.C., Ji, F., Gordon, D., Li, P., Benedict, M.M., Greenberg,W.M., Kao, H.T., 2008. Telomerase levels in schizophrenia: a preliminary study.Schizophr. Res. 106, 242–247.

iu, W.Q., Sun, X., Selkoe, D.J., Mwamburi, D.M., Huang, T., Bhadela, R., Bergethon, P.,Scott, T.M., Summergrad, P., Wang, L., Rosenberg, I., Folstein, M., 2007. Depres-sion is associated with low plasma Abeta42 independently of cardiovasculardisease in the homebound elderly. Int. J. Geriatr. Psychiatry 22, 536–542.

ajkowska, G., 2002. Cell pathology in bipolar disorder. Bipolar Disord. 4, 105–116.az, N., Rodrigue, K.M., 2006. Differential aging of the brain: patterns, cognitive

correlates and modifiers. Neurosci. Biobehav. Rev. 30, 730–748.ege, S., Hodgkinson, S.J., 2013. Immune dysregulation and autoimmunity in bipo-

lar disorder: synthesis of the evidence and its clinical application. Aust. N. Z. J.Psychiatry.

izzo, L.B., Do Prado, C.H., Grassi-Oliveira, R., Wieck, A., Correa, B.L., Teix-eira, A.L., Bauer, M.E., 2013. Immunosenescence is associated with humancytomegalovirus and shortened telomeres in type I bipolar disorder. BipolarDisord..

obinson, L.J., Thompson, J.M., Gallagher, P., Goswami, U., Young, A.H., Ferrier, I.N.,Moore, P.B., 2006. A meta-analysis of cognitive deficits in euthymic patients withbipolar disorder. J. Affect. Disord. 93, 105–115.

osa, A.R., Franco, C., Martinez-Aran, A., Sanchez-Moreno, J., Reinares, M., Salamero,M., Arango, C., Ayuso-Mateos, J.L., Kapczinski, F., Vieta, E., 2008. Functionalimpairment in patients with remitted bipolar disorder. Psychother. Psychosom.77, 390–392.

osa, A.R., Gonzalez-Ortega, I., Gonzalez-Pinto, A., Echeburua, E., Comes, M.,Martinez-Aran, A., Ugarte, A., Fernandez, M., Vieta, E., 2012. One-year psychoso-cial functioning in patients in the early vs. late stage of bipolar disorder. ActaPsychiatr. Scand. 125, 335–341.

osa, A.R., Reinares, M., Amann, B., Popovic, D., Franco, C., Comes, M., Torrent, C.,Bonnin, C.M., Sole, B., Valenti, M., Salamero, M., Kapczinski, F., Vieta, E., 2011.Six-month functional outcome of a bipolar disorder cohort in the context of aspecialized-care program. Bipolar Disord. 13, 679–686.

ose, M.R., 1991. Evolutionary Biology of Aging. Oxford University Press, New York.osenberg, G., 2007. The mechanisms of action of valproate in neuropsychiatric

disorders: can we see the forest for the trees? Cell Mol Life Sci 64, 2090–2103.ybakowski, J.K., Wykretowicz, A., Heymann-Szlachcinska, A., Wysocki, H., 2006.

Impairment of endothelial function in unipolar and bipolar depression. Biol.Psychiatry 60, 889–891.

amame, C., Martino, D.J., Strejilevich, S.A., 2013. A quantitative review of neurocog-nition in euthymic late-life bipolar disorder. Bipolar Disord. 15, 633–644.

avas, H.A., Herken, H., Yurekli, M., Uz, E., Tutkun, H., Zoroglu, S.S., Ozen, M.E., Cengiz,B., Akyol, O., 2002. Possible role of nitric oxide and adrenomedullin in bipolaraffective disorder. Neuropsychobiology 45, 57–61.

cott, J., Leboyer, M., Hickie, I., Berk, M., Kapczinski, F., Frank, E., Kupfer, D., McGorry,P., 2013. Clinical staging in psychiatry: a cross-cutting model of diagnosis withheuristic and practical value. Br. J. Psychiatry 202, 243–245.

elek, S., Savas, H.A., Gergerlioglu, H.S., Bulbul, F., Uz, E., Yumru, M., 2008. The courseof nitric oxide and superoxide dismutase during treatment of bipolar depressiveepisode. J. Affect. Disord. 107, 89–94.

esti, G., Sciacqua, A., Cardellini, M., Marini, M.A., Maio, R., Vatrano, M., Succurro, E.,Lauro, R., Federici, M., Perticone, F., 2005. Plasma concentration of IGF-I is inde-pendently associated with insulin sensitivity in subjects with different degreesof glucose tolerance. Diabetes Care 28, 120–125.

imon, N.M., Smoller, J.W., McNamara, K.L., Maser, R.S., Zalta, A.K., Pollack, M.H.,Nierenberg, A.A., Fava, M., Wong, K.K., 2006. Telomere shortening and mooddisorders: preliminary support for a chronic stress model of accelerated aging.Biol. Psychiatry 60, 432–435.

nyder, E.M., Nong, Y., Almeida, C.G., Paul, S., Moran, T., Choi, E.Y., Nairn, A.C., Salter,

Please cite this article in press as: Rizzo, L.B., et al., The theory of bipclinical care and research. Neurosci. Biobehav. Rev. (2014), http://dx.d

M.W., Lombroso, P.J., Gouras, G.K., Greengard, P., 2005. Regulation of NMDAreceptor trafficking by amyloid-beta. Nat. Neurosci. 8, 1051–1058.

odhi, S.K., Linder, J., Chenard, C.A., Miller del, D., Haynes, W.G., Fiedorowicz, J.G.,2012. Evidence for accelerated vascular aging in bipolar disorder. J. Psychosom.Res. 73, 175–179.

PRESSvioral Reviews xxx (2014) xxx–xxx

Soreca, I., Fagiolini, A., Frank, E., Houck, P.R., Thompson, W.K., Kupfer, D.J., 2008.Relationship of general medical burden, duration of illness and age in patientswith bipolar I disorder. J. Psychiatr. Res. 42, 956–961.

Squassina, A., Costa, M., Congiu, D., Manchia, M., Angius, A., Deiana, V., Ardau, R.,Chillotti, C., Severino, G., Calza, S., Del Zompo, M., 2013. Insulin-like growth fac-tor 1 (IGF-1) expression is up-regulated in lymphoblastoid cell lines of lithiumresponsive bipolar disorder patients. Pharmacol. Res. 73, 1–7.

Stadler, K., 2012. Oxidative stress in diabetes. Adv. Exp. Med. Biol. 771, 272–287.Stambolic, V., Ruel, L., Woodgett, J.R., 1996. Lithium inhibits glycogen synthase

kinase-3 activity and mimics wingless signalling in intact cells. Curr. Biol. 6,1664–1668.

Steckert, A.V., Valvassori, S.S., Moretti, M., Dal-Pizzol, F., Quevedo, J., 2010. Role ofoxidative stress in the pathophysiology of bipolar disorder. Neurochem. Res. 35,1295–1301.

Strakowski, S.M., DelBello, M.P., Zimmerman, M.E., Getz, G.E., Mills, N.P., Ret, J., Shear,P., Adler, C.M., 2002. Ventricular and periventricular structural volumes in first-versus multiple-episode bipolar disorder. Am. J. Psychiatry 159, 1841–1847.

Sullivan, P.F., Daly, M.J., O’Donovan, M., 2012. Genetic architectures of psychi-atric disorders: the emerging picture and its implications. Nat. Rev. Genet. 13,537–551.

Sun, X., Mwamburi, D.M., Bungay, K., Prasad, J., Yee, J., Lin, Y.M., Liu, T.C., Summer-grad, P., Folstein, M., Qiu, W.Q., 2007. Depression, antidepressants, and plasmaamyloid beta (Beta) peptides in those elderly who do not have cardiovasculardisease. Biol. Psychiatry 62, 1413–1417.

Swann, A.C., Bowden, C.L., Calabrese, J.R., Dilsaver, S.C., Morris, D.D., 1999. Differen-tial effect of number of previous episodes of affective disorder on response tolithium or divalproex in acute mania. Am. J. Psychiatry 156, 1264–1266.

Swardfager, W., Black, S.E., 2013. Dementia: a link between microbial infection andcognition? Nature reviews. Neurology 9, 301–302.

Swardfager, W., Herrmann, N., Marzolini, S., Saleem, M., Kiss, A., Shammi, P., Oh,P.I., Lanctot, K.L., 2010a. Cardiopulmonary fitness is associated with cognitiveperformance in patients with coronary artery disease. J. Am. Geriatr. Soc. 58,1519–1525.

Swardfager, W., Herrmann, N., Marzolini, S., Saleem, M., Shammi, P., Oh, P.I., Albert,P.R., Daigle, M., Kiss, A., Lanctot, K.L., 2011. Brain derived neurotrophic factor,cardiopulmonary fitness and cognition in patients with coronary artery disease.Brain Behav. Immun. 25, 1264–1271.

Swardfager, W., Lanctot, K., Rothenburg, L., Wong, A., Cappell, J., Herrmann, N.,2010b. A meta-analysis of cytokines in Alzheimer’s disease. Biol. Psychiatry 68,930–941.

Tabata, T., Kawakatsu, H., Maidji, E., Sakai, T., Sakai, K., Fang-Hoover, J., Aiba, M.,Sheppard, D., Pereira, L., 2008. Induction of an epithelial integrin alphavbeta6 inhuman cytomegalovirus-infected endothelial cells leads to activation of trans-forming growth factor-beta1 and increased collagen production. Am. J. Pathol.172, 1127–1140.

Teixeira, M.T., Arneric, M., Sperisen, P., Lingner, J., 2004. Telomere length homeosta-sis is achieved via a switch between telomerase-extendible and -nonextendiblestates. Cell 117, 323–335.

Thal, D.R., Del Tredici, K., Braak, H., 2004. Neurodegeneration in normal brain agingand disease. Sci. Aging Knowledge Environ. 2004, pe26.

Tong, L., Thornton, P.L., Balazs, R., Cotman, C.W., 2001. Beta-amyloid-(1–42) impairsactivity-dependent cAMP-response element-binding protein signaling in neu-rons at concentrations in which cell survival is not compromised. J. Biol. Chem.276, 17301–17306.

Torrao, A.S., Cafe-Mendes, C.C., Real, C.C., Hernandes, M.S., Ferreira, A.F., Santos, T.O.,Chaves-Kirsten, G.P., Mazucanti, C.H., Ferro, E.S., Scavone, C., Britto, L.R., 2012.Different approaches, one target: understanding cellular mechanisms of Parkin-son’s and Alzheimer’s diseases. Rev. Bras. Psiquiatr. 34 (Suppl. 2), S194–S205.

Torrent, C., Martinez-Aran, A., Daban, C., Sanchez-Moreno, J., Comes, M., Goikolea,J.M., Salamero, M., Vieta, E., 2006. Cognitive impairment in bipolar II disorder.Br. J. Psychiatry 189, 254–259.

Torres, I.J., Boudreau, V.G., Yatham, L.N., 2007. Neuropsychological functioning ineuthymic bipolar disorder: a meta-analysis. Acta Psychiatr. Scand. Suppl., 17–26.

Ustun, T.B., 1999. The global burden of mental disorders. Am. J. Public Health 89,1315–1318.

van der Lijn, F., den Heijer, T., Breteler, M.M., Niessen, W.J., 2008. Hippocampussegmentation in MR images using atlas registration, voxel classification, andgraph cuts. Neuroimage 43, 708–720.

van Praag, H., Christie, B.R., Sejnowski, T.J., Gage, F.H., 1999. Running enhances neu-rogenesis, learning, and long-term potentiation in mice. Proc. Natl. Acad. Sci.U.S.A. 96, 13427–13431.

Vaziri, H., Dragowska, W., Allsopp, R.C., Thomas, T.E., Harley, C.B., Lansdorp, P.M.,1994. Evidence for a mitotic clock in human hematopoietic stem cells: loss oftelomeric DNA with age. Proc. Natl. Acad. Sci. U.S.A. 91, 9857–9860.

Vernooij, M.W., Smits, M., 2012. Structural neuroimaging in aging and Alzheimer’sdisease. Neuroimaging Clin. N. Am. 22, 33–55, vii–viii.

von Zglinicki, T., 2000. Role of oxidative stress in telomere length regulation andreplicative senescence. Ann. N. Y. Acad. Sci. 908, 99–110.

Vostrikov, V.M., Uranova, N.A., Orlovskaya, D.D., 2007. Deficit of perineuronal oligo-dendrocytes in the prefrontal cortex in schizophrenia and mood disorders.Schizophr. Res. 94, 273–280.

olar disorder as an illness of accelerated aging: Implications foroi.org/10.1016/j.neubiorev.2014.02.004

Wang, L., Yang, H.J., Xia, Y.Y., Feng, Z.W., 2010. Insulin-like growth factor 1 pro-tects human neuroblastoma cells SH-EP1 against MPP+-induced apoptosis byAKT/GSK-3beta/JNK signaling. Apoptosis 15, 1470–1479.

Weng, N.P., Akbar, A.N., Goronzy, J., 2009. CD28(−) T cells: their role in the age-associated decline of immune function. Trends Immunol. 30, 306–312.

1492

1493

1494

1495

1496

Page 13: The theory of bipolar disorder as an illness of accelerated aging: Implications for clinical care and research

ING ModelN

iobeha

W

W

W

W

W

W

W

W

1497

1498

1499

1500

1501

1502

1503

1504

1505

1506

1507

1508

1509

1510

1511

1512

1513

1514

1515

1516

1517

1518

1519

1520

1521

1522

1523

1524

1525

1526

1527

1528

1529

1530

1531

1532

1533

1534

1535

1536

1537

1538

1539

1540

1541

1542

1543

1544

1545

1546

1547

1548

1549

1550

1551

1552

ARTICLEBR 1894 1–13

L.B. Rizzo et al. / Neuroscience and B

erner, C., Hanhoun, M., Widmann, T., Kazakov, A., Semenov, A., Poss, J., Bauersachs,J., Thum, T., Pfreundschuh, M., Muller, P., Haendeler, J., Bohm, M., Laufs, U., 2008.Effects of physical exercise on myocardial telomere-regulating proteins, survivalpathways, and apoptosis. J. Am. Coll. Cardiol. 52, 470–482.

ieck, A., Grassi-Oliveira, R., do Prado, C.H., Rizzo, L.B., de Oliveira, A.S.,Kommers-Molina, J., Viola, T.W., Teixeira, A.L., Bauer, M.E., 2013. Differentialneuroendocrine and immune responses to acute psychosocial stress in womenwith type 1 bipolar disorder. Brain Behav. Immun.

ikby, A., Johansson, B., Olsson, J., Lofgren, S., Nilsson, B.O., Ferguson, F., 2002. Expan-sions of peripheral blood CD8 T-lymphocyte subpopulations and an associationwith cytomegalovirus seropositivity in the elderly: the Swedish NONA immunestudy. Exp. Gerontol. 37, 445–453.

ikgren, M., Maripuu, M., Karlsson, T., Nordfjall, K., Bergdahl, J., Hultdin, J., Del-Favero, J., Roos, G., Nilsson, L.G., Adolfsson, R., Norrback, K.F., 2012. Shorttelomeres in depression and the general population are associated with ahypocortisolemic state. Biol. Psychiatry 71, 294–300.

olf, S.A., Melnik, A., Kempermann, G., 2011. Physical exercise increases adult neu-rogenesis and telomerase activity, and improves behavioral deficits in a mousemodel of schizophrenia. Brain Behav. Immun. 25, 971–980.

olkowitz, O.M., Epel, E.S., Reus, V.I., Mellon, S.H., 2010. Depression gets oldfast: do stress and depression accelerate cell aging? Depress. Anxiety 27,327–338.

olkowitz, O.M., Mellon, S.H., Epel, E.S., Lin, J., Dhabhar, F.S., Su, Y., Reus, V.I.,Rosser, R., Burke, H.M., Kupferman, E., Compagnone, M., Nelson, J.C., Blackburn,E.H., 2011. Leukocyte telomere length in major depression: correlations withchronicity, inflammation and oxidative stress—preliminary findings. PLoS ONE

Please cite this article in press as: Rizzo, L.B., et al., The theory of bipclinical care and research. Neurosci. Biobehav. Rev. (2014), http://dx.d

6, e17837.olkowitz, O.M., Mellon, S.H., Epel, E.S., Lin, J., Reus, V.I., Rosser, R., Burke, H.,

Compagnone, M., Nelson, J.C., Dhabhar, F.S., Blackburn, E.H., 2012. Resting leuko-cyte telomerase activity is elevated in major depression and predicts treatmentresponse. Mol. Psychiatry 17, 164–172.

PRESSvioral Reviews xxx (2014) xxx–xxx 13

Wonodi, I., Schwarcz, R., 2010. Cortical kynurenine pathway metabolism: a noveltarget for cognitive enhancement in Schizophrenia. Schizophr. Bull. 36, 211–218.

Worman, H.J., Bonne, G., 2007. Laminopathies: a wide spectrum of human diseases.Exp. Cell Res. 313, 2121–2133.

Xiang, G.D., Pu, J., Sun, H., Zhao, L., Yue, L., Hou, J., 2009. Regular aerobic exer-cise training improves endothelium-dependent arterial dilation in patients withsubclinical hypothyroidism. Eur. J. Endocrinol. 161, 755–761.

Yasuda, S., Liang, M.H., Marinova, Z., Yahyavi, A., Chuang, D.M., 2009. The moodstabilizers lithium and valproate selectively activate the promoter IV of brain-derived neurotrophic factor in neurons. Mol. Psychiatry 14, 51–59.

Yildiz-Yesiloglu, A., Ankerst, D.P., 2006. Neurochemical alterations of the brain inbipolar disorder and their implications for pathophysiology: a systematic reviewof the in vivo proton magnetic resonance spectroscopy findings. Prog. Neuropsy-chopharmacol. Biol. Psychiatry 30, 969–995.

Yildiz, O., 2007. Vascular smooth muscle and endothelial functions in aging. Ann. N.Y. Acad. Sci. 1100, 353–360.

Yu, C.E., Oshima, J., Fu, Y.H., Wijsman, E.M., Hisama, F., Alisch, R., Matthews, S.,Nakura, J., Miki, T., Ouais, S., Martin, G.M., Mulligan, J., Schellenberg, G.D., 1996.Positional cloning of the Werner’s syndrome gene. Science 272, 258–262.

Zhang, D., Cheng, L., Craig, D.W., Redman, M., Liu, C., 2010. Cerebellar telomere lengthand psychiatric disorders. Behav. Genet. 40, 250–254.

Zhang, Y.W., Thompson, R., Zhang, H., Xu, H., 2011. APP processing in Alzheimer’sdisease. Mol. Brain 4, 3.

Zhang, Z., Zhang, Z.Y., Fauser, U., Schluesener, H.J., 2008. Valproic acid attenu-ates inflammation in experimental autoimmune neuritis. Cell Mol. Life Sci. 65,4055–4065.

olar disorder as an illness of accelerated aging: Implications foroi.org/10.1016/j.neubiorev.2014.02.004

Zhang, Z., Zhang, Z.Y., Wu, Y., Schluesener, H.J., 2012. Valproic acid amelioratesinflammation in experimental autoimmune encephalomyelitis rats. Neuro-science 221, 140–150.

Zipursky, R.B., Reilly, T.J., Murray, R.M., 2012. The myth of schizophrenia as a pro-gressive brain disease. Schizophr. Bull..

1553

1554

1555

1556

1557