Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2018 Jan 1.
Published in final edited form as: J Pain Symptom Manage. 2016 Oct 5;53(1):67–84.e7. doi: 10.1016/j.jpainsymman.2016.08.004

Associations Between Neurotransmitter Genes and Fatigue and Energy Levels in Women Following Breast Cancer Surgery

Jasmine Eshragh 1, Anand Dhruva 2, Steven M Paul 1, Bruce A Cooper 1, Judy Mastick 1, Deborah Hamolsky 1, Jon D Levine 2, Christine Miaskowski 1, Kord M Kober 1
PMCID: PMC5191954  NIHMSID: NIHMS821824  PMID: 27720787

Abstract

Context

Fatigue is a common problem in oncology patients. Less is known about decrements in energy levels and the mechanisms that underlie both fatigue and energy.

Objectives

In patients with breast cancer, variations in neurotransmitter genes between Lower and Higher Fatigue latent classes and between the Higher and Lower Energy latent classes were evaluated.

Methods

Patients completed assessments prior to and monthly for 6 months following surgery. Growth mixture modeling was used to identify distinct latent classes for fatigue severity and energy levels. Thirty candidate genes involved in various aspects of neurotransmission were evaluated.

Results

Eleven single nucleotide polymorphisms (SNPs) or haplotypes (i.e., ADRB2 rs1042718, BDNF rs6265, COMT rs9332377, CYP3A4 rs4646437, GALR1 rs949060, GCH1 rs3783642, NOS1 rs9658498, NOS1 rs2293052, NPY1R Haplotype A04, SLC6A2 rs17841327 and 5HTTLPR + rs25531 in SLC6A4) were associated with latent class membership for fatigue. Seven SNPs or haplotypes (i.e., NOS1 rs471871, SLC6A1 rs2675163, SLC6A1 Haplotype D01, SLC6A2 rs36027, SLC6A3 rs37022, SLC6A4 rs2020942, and TAC1 rs2072100) were associated with latent class membership for energy. Three of thirteen genes (i.e., NOS1, SLC6A2, SLC6A4) were associated with latent class membership for both fatigue and energy.

Conclusions

Molecular findings support the hypothesis that fatigue and energy are distinct, yet related symptoms. Results suggest that a large number of neurotransmitters play a role in the development and maintenance of fatigue and energy levels in breast cancer patients.

Keywords: fatigue, energy, neurotransmitter genes, growth mixture modeling, breast cancer, candidate genes

INTRODUCTION

Fatigue is the most common symptom associated with cancer and its treatments.1 While several studies have examined fatigue in breast cancer patients receiving chemotherapy (CTX)2 and radiation (RT),3 studies on the occurrence of and predictors for fatigue following surgery are scarce. In a recent study,4 patients with breast cancer reported relatively high levels of fatigue in the first two months after surgery followed by mild to moderate levels of fatigue that persisted for 12 months after surgery.

The measurement of a patient’s level of energy has received little or no attention in the cancer literature. While energy level is commonly thought of as the opposite of fatigue, evidence suggests that fatigue and energy are distinct, but related concepts.5,6 In oncology, fatigue is defined as a distressing and persistent sense of physical, emotional, and/or cognitive tiredness or exhaustion related to cancer or its treatment that is not proportional to recent activity and interferes with usual functioning.7 In contrast, energy is defined as an individual’s potential to perform physical and mental activity.6 In the only study that evaluated energy levels in patients with breast cancer prior to surgery,8 while 32% of the women reported clinically meaningful levels of fatigue, nearly 50% of these women reported clinically meaningful decrements in energy levels. Findings from this study,8 as well as a study of patients who underwent RT and their family caregivers,9 and a study of patients with HIV disease,10 support the hypothesis that energy is a distinct concept from fatigue.

Factors that contribute to fatigue severity are multidimensional and include numerous biopsychosocial characteristics.11 Some of the predictors of fatigue following breast cancer surgery include higher levels of anxiety; the personality characteristic of extraversion;12 increased fatigue prior to surgery;13 higher levels of emotional distress, mental fatigue, and pain;14 as well as depressive symptoms and receipt of CTX.4

Recent evidence suggests that genetic mechanisms contribute to the severity of fatigue experienced by breast cancer patients. For example, in one study,15 a number of pro-inflammatory cytokine genes were associated with fatigue. In another study,16 polymorphisms in IL1B and IL10 were associated with fatigue in women who underwent breast cancer surgery. Polymorphisms in cytokine genes may contribute to fatigue severity through the modulation of pro- and anti- inflammatory pathways.17,18

While the majority of studies on genetic associations with fatigue has focused on cytokine dysregulation, a number of additional pathways may influence fatigue and energy levels (for review see 19). Neurotransmitter dysregulation may play an important role in the severity of fatigue and/or changes in energy levels. The most commonly cited neurotransmitter associated with fatigue is serotonin. For example, increased serum levels of serotonin were linked to fatigue following prolonged exercise.20 However, it is unlikely that a single neurotransmitter is responsible for the development of/or changes in fatigue and/or energy levels. Rather, it is more likely that several neurotransmitters contribute to inter-individual variability in fatigue and energy.21 Some neurotransmitter genes that were associated with fatigue and energy in a variety of populations include alterations in the dopaminergic system, specifically polymorphyisms in catechol-o-methyl-transferase (COMT), dopamine-2 receptor (DRD2), and dopamine-1 transporter (DAT1).22 For example, in a recent study that evaluated for differences in gene expression in breast cancer patients who reported lower versus higher levels of evening fatigue during CTX,23 between group differences were identified in a number of neurotransmitter pathways. However, no studies were identified that evaluated for associations between neurotransmitter genes and fatigue and energy levels in patients with breast cancer.

The current study is based on our recent work that used growth mixture modeling (GMM) to identify distinct latent classes for fatigue severity and decrements in energy levels in women (n=398) prior to and for six months following breast cancer surgery.16 Fatigue and energy levels were evaluated using the Lee Fatigue Scale (LFS).24 In the GMM analysis for fatigue, two distinct latent classes were identified (i.e., Lower Fatigue (38.5%) and Higher Fatigue (61.5%)). At enrollment, mean fatigue scores were 1.60 and 3.90 for the Lower and Higher Fatigue classes, respectively. In both fatigue classes, fatigue scores remained relatively constant from the preoperative assessment to 6 months after breast cancer surgery. In the GMM analysis for energy, two distinct latent classes were identified (i.e., Higher Energy (32.0%) and Lower Energy (68.0%)). At enrollment, mean energy scores were 5.82 and 4.35 for the Higher and Lower Energy classes, respectively. In both energy groups, energy levels remained relatively constant from the preoperative assessment to 6 months after breast cancer surgery.

Given the paucity of research on the role of various neurotransmitters in the mechanism that underlie fatigue severity or decrements in energy levels in patients with breast cancer, the purpose of this study was to evaluate for associations between variations in a number of genes involved in neurotransmission, drug metabolism, and transport of molecules across cell membranes between the Lower and Higher Fatigue latent classes as well as between the Higher and Lower Energy latent classes.

METHODS

Patients and Settings

The study methods are described in detail elsewhere.2527 In brief, patients were recruited from 2004 to 2008, from Breast Care Centers located in a Comprehensive Cancer Center, two public hospitals, and four community practices. Patients were eligible to participate if they: were adult women (≥18 years) who were scheduled to undergo unilateral breast cancer surgery; were able to read, write, and understand English; agreed to participate; and gave written informed consent. Patients were excluded if they were having breast cancer surgery on both breasts and/or had distant metastasis at the time of diagnosis.

Instruments

The demographic questionnaire obtained information on age, marital status, education, ethnicity, employment status, and living situation. Functional status was evaluated using the Karnofsky Performance Status (KPS) scale.28 The number and impact of comorbid conditions was evaluated using the Self-Administered Comorbidity Questionnaire (SCQ).29

The LFS consists of 18 items designed to assess physical fatigue and energy.24 Each item was rated on a 0 to 10 numeric rating scale (NRS). Total fatigue and energy scores were calculated as the mean of the 13 fatigue items and the 5 energy items, with higher scores indicating greater fatigue severity and higher levels of energy. Patients were asked to rate each item based on how they felt “right now”. Cutoff scores of ≥4.4 and ≤4.8 indicate clinically meaningful levels of fatigue severity and low levels of energy.3 The LFS has well established validity and reliability.24,30 Cronbach’s alphas for fatigue and energy scales were .96 and .93, respectively.

Study Procedures

The study was approved by the Committee on Human Research at the University of California, San Francisco and by the Institutional Review Boards at each of the study sites. During the patients’ preoperative visit, a clinician explained the study and determined patients’ willingness to participate. The research nurse met with interested women, determined eligibility, and obtained written informed consent prior to surgery. After obtaining consent, patients completed the enrollment questionnaires an average of 4 days prior to surgery. Patients completed the LFS at enrollment and monthly for 6 months (i.e., 7 assessments). Medical records were reviewed for disease and treatment information.

Genomic analyses

Gene selection

Thirty candidate genes involved in various aspects of neurotransmission, drug metabolism, or transport of molecules across cell membranes were evaluated (Supplementary Table 1). Genes involved in catecholaminergic neurotransmission included alpha-1D-adrenergic receptor (ADRA1D); alpha-2A-adrenergic receptor (ADRA2A); beta-2-adrenergic receptor (ADRB2); beta-3-adrenergic receptor (ADRB3); beta adrenergic receptor kinase 2 (ADRBK2); solute-like carrier (SLC) family 6 member 2 – noradrenaline transporter (SLC6A2); SLC family 6 member 3 – dopamine transporter (SLC6A3); tyrosine hydroxylase (TH); and catecho-o-methyl transferase (COMT). The gene involved in gabaergic neurotransmission was the SLC family 6 member 1 – GABA transporter (SLC6A1). Genes involved in serotonergic neurotransmission included: 5-hydroxytryptamine receptor (HTR) 1A (HTR1A); HTR 1B (HTR1B); HTR 2A (HTR2A); HTR 3A (HTR3A); SLC family 6 member 4 – serotonin transporter (SLC6A4); and tryptophan hydroxylase 2 (TPH2). Genes involved in molecular transport and drug metabolism were: ATP-binding cassette, subfamily B member 1 (ABCB1) and cytochrome P450, family 3, subfamily A, polypeptide 4 (CYP3A4). A number of genes involved in various aspects of neurotransmission that were evaluated included: brain-derived neurotrophic factor (BDNF); galanin (GAL); galanin receptor 1 (GALR1); galanin receptor 2 (GALR2); GTP cyclohydrolase 1 (GCH1); nitric oxide synthase 1 (NOS1); nitric oxide synthase 2 (NOS2); neuropeptide Y (NPY); neuropeptide Y receptor 1 (NPY1R); prodynorphin (PDYN); tachykinin precursor 1 (TAC1); and tachykinin receptor 1 (TACR1). All genes were identified according to the approved symbol stored in the Human Genome Organization (HUGO) Gene Nomenclature Committee (HGNC) database (http://www.genenames.org).

Blood collection and genotyping

Of the 398 patients who completed the enrollment assessment, 310 provided a blood sample from which deoxyribonucleic acid (DNA) could be isolated from peripheral blood mononuclear cells (PBMCs). Genomic DNA was extracted using the PUREGene DNA Isolation System (Invitrogen, Carlsbad, CA). DNA was quantitated with a Nanodrop Spectrophotometer (ND-1000) and normalized to a concentration of 50 ng/μL (diluted in 10 mM Tris/1 mM EDTA). Genotyping was performed using a custom array on the Golden Gate genotyping platform (Illumina, San Diego, CA) and processed according to the standard protocol using GenomeStudio (Illumina, San Diego, CA).

SNP selection

Tagging SNPs and literature driven SNPs were selected for analysis. Tagging SNPs were required to be common (defined as having a minor allele frequency of ≥0.05) in public databases. To ensure robust genetic association analyses, quality control filtering of SNPs was performed. SNPs with call rates of <95% or Hardy-Weinberg p-values of <.001 were excluded. A total of 249 SNPs among the 30 candidate genes passed all of the quality control filters and were included in the genetic association analyses (Supplementary Table 1).

Localization of SNPs on the human genome was performed using the GRCh38 human reference assembly. Regional annotations were identified using the University of California Santa Cruz (UCSC) Human Genome Browser GRCh37/hg19 (http://genome.ucsc.edu/cgibin/hgTracks?db=hg38). Potential regulatory involvement of SNPs was investigated using ENCODE.31

Genotyping the serotonin-linked polymorphic region of SLC6A4

The serotonin transporter-linked polymorphic region (5-HTTLPR) occurs in the promoter region of the SLC6A4 gene. 5-HTTLPR occurs primarily as either a shorter (S, i.e., 14 repeats of 23 nucleotides) or longer (L, i.e., 16 repeats) sequence.32 5-HTT rs25531 is a SNP, which is present in either a common (A) or rare (G) variant, and is located immediately upstream of the 5-HTTLPR.33,34 The 5-HTTLPR/rs25531 haplotype (i.e., 5-HTTLPR triallelic polymorphism) is known to influence SLC6A4 expression levels. In vitro studies of the L allele suggest that the LA allele exhibits higher 5-HTT transcription and that the LG allele is more similar in function to the S allele.33,35 In this study the LA allele was used as the reference allele. The 5-HTTLPR polymorphism was measured using polymerase chain reaction (PCR) followed by resolution of PCR products by gel electrophoresis.36 The rs25531 genotype was obtained by DNA cycle sequencing.

Statistical Analyses for the Phenotypic Data

Data were analyzed using SPSS version 2237 and STATA Version 13.38 As described previously,16 unconditional GMM with robust maximum likelihood estimation was carried out to identify latent classes with distinct fatigue and energy trajectories using Mplus Version 5.21.3941 Descriptive statistics and frequency distributions were generated for sample characteristics. Independent sample t-tests, Mann-Whitney U tests, and Chi square analyses were used to evaluate for differences in demographic and clinical characteristics between the two latent classes for fatigue and energy. A p-value of <0.05 was considered statistically significant.

Statistical Analyses for the Genetic Data

The genomic analyses are described in detail elsewhere.16 In brief, allele and genotype frequencies were determined by gene counting. Hardy-Weinberg equilibrium was assessed by the Chi-square or Fisher Exact tests. Measures of linkage disequilibrium ((LD), i.e., D′ and r2) were computed using Haploview 4.2. Haplotypes were constructed using PHASE version 2.1.42 Only inferred haplotypes that occurred with a frequency estimate of ≥15% were included in the association analyses, assuming a dosage model.

Ancestry informative markers (AIMs) were used to minimize confounding due to population stratification.4345 Homogeneity in ancestry among patients was verified by principal component analysis,46 using HelixTree (GoldenHelix, Bozeman, MT). The first three PCs were used in all of the logistic regression models.

For association tests, three genetic models were assessed for each SNP: additive, dominant, and recessive. The genetic model that best fit the data, by maximizing the significance of the p-value was selected for each SNP. Logistic regression analysis, that controlled for significant covariates, as well as genomic estimates of (i.e., AIMs) and self-reported race/ethnicity, was used to evaluate the associations between genotype and Higher Fatigue and Lower Energy class memberships. Only those genetic associations identified as significant from the bivariate analyses were evaluated in the multivariate analyses. A backwards stepwise approach was used to create a parsimonious model. Except for race/ethnicity, only predictors with a p-value of <.05 were retained in the final model.

As was done in our previous studies,9,4751 based on the recommendations in the literature,52,53 as well as the implementation of rigorous quality controls for genomic data, the non-independence of SNPs/haplotypes in LD, and the exploratory nature of the analyses, adjustments were not made for multiple testing. In addition, significant SNPs identified in the bivariate analyses were evaluated further using logistic regression analyses that controlled for differences in phenotypic characteristics, potential confounding due to population stratification, and variations in other SNPs/haplotypes within the same gene. Only those SNPs that remained significant were included in the final presentation of the results. Therefore, the significant independent associations reported are unlikely to be due solely to chance. Unadjusted associations, for all of the SNPs evaluated, are found in Supplementary Table 1, to allow for subsequent comparisons and meta-analyses.

RESULTS

Differences in Demographic and Clinical Characteristics between the Fatigue Classes

Differences between the two fatigue classes are listed in Table 1. Patients in the Higher Fatigue class were significantly younger, had more years of education, a lower KPS score, a higher SCQ score, a higher number of lymph nodes removed and a higher fatigue severity score at enrollment. A larger percentage of patients in the Higher Fatigue class had received neoadjuvant CTX and had received CTX during the first 6 months after breast cancer surgery.

Table 1.

Differences in Demographic and Clinical Characteristics Between the Lower Fatigue (n= 153) and Higher Fatigue (n= 244) Classes at Enrollment

Characteristic Lower Fatigue Class

n=153
(38.4%)

Mean (SD)
Higher Fatigue Class

n= 244
(61.3%)

Mean (SD)
Statistic and p-value
Age (years) 57.8 (11.9) 53.1 (11.0) t=4.09, p=<.0001
Education (years) 15.3 (2.5) 15.9 (2.8) t=−2.02, p=.04
Karnofsky Performance Status score 96.6 (7.0) 91.1 (11.4) t=5.86, p=<.0001
Self-administered Comorbidity Questionnaire score 3.8 (2.6) 4.6 (3.0) t=−2.64, p=.009
Fatigue severity score at enrollment 1.6 (1.6) 4.1 (2.2) t=−12.55, p=<.0001
Number of breast biopsies in past year 1.5 (0.8) 1.5 (0.8) U, p=.47
Number of positive lymph nodes 0.8 (1.9) 1.0 (2.4) t=−0.88, p=.38
Number of lymph nodes removed 4.8 (5.1) 6.4 (7.5) t=−2.43, p=.016
n (%) n (%)
Ethnicity
 White
 Black
 Asian/Pacific Islander
 Hispanic/Mixed ethnic background/Other

100 (65.8)
19 (12.5)
17 (11.2)
16 (10.5)

155 (63.8)
21 (8.6)
32 (13.2)
35 (14.4)
χ2=2.82, p=.42
Married/partnered (% yes) 64 (42.1) 100 (41.5) FE, p=.92
Work for pay (% yes) 71 (46.4) 118 (49.0) FE, p=.68
Lives alone (% yes) 40 (26.5) 54 (22.4) FE, p=.40
Gone through menopause (% yes) 96 (63.6) 151 (64.3) FE, p=.91
Stage of disease
 0
 I
 IIA and IIB
 IIIA, IIIB, IIIC, and IV

29 (19.0)
66 (43.1)
48 (31.4)
10 (6.5)

44 (18.0)
85 (34.8)
92 (37.7)
23 (9.4)
U, p=.13
Surgical treatment
 Breast conservation
 Mastectomy

123 (80.4)
30 (19.6)

195 (79.9)
49 (20.1)
FE, p=1.00
Sentinel node biopsy (% yes) 130 (85.0) 197 (80.7) FE, p=.34
Axillary lymph node dissection (% yes) 50 (32.7) 98 (40.3) FE, p=.14
Breast reconstruction at the time of surgery (% yes) 33 (21.7) 53 (21.7) FE, p=1.00
Neoadjuvant chemotherapy (% yes) 21 (13.7) 58 (23.9) FE, p=.014
Radiation therapy during the first 6 months (% yes) 87 (56.9) 137 (56.1) FE, p=.92
Chemotherapy during the first 6 months (% yes) 36 (23.5) 97 (39.8) FE, p=.001

Abbreviations: FE=Fisher Exact test, SD = standard deviation, U=Mann Whitney U test

Candidate Gene Analyses for Fatigue

Genotype distributions differed between the Lower and Higher Fatigue classes for: 2 SNPs and one haplotype in ADRB2; 3 SNPs in BDNF; 1 SNP in COMT; 1 SNP in CYP3A4; 1 SNP in GALR1; 1 SNP in GCH1; 5 SNPs and 2 haplotypes in NOS1; 1 SNP and 1 haplotype in NPY1R; 1 SNP and 1 haplotype in SLC6A1; 2 SNPs and 1 haplotype in SLC6A2; 1 SNP in SLC6A3; and 2 SNPs and 1 haplotype in TAC1, as well as in the 5HTTLPR + rs25521 haplotype in the SLC6A4 gene.

Regression Analyses for Fatigue

In these regression analyses that included genomic estimates of and self-reported race/ethnicity, the only phenotypic characteristics that remained significant in the multivariate model were: age, KPS score, SCQ score, and receipt of CTX within six months after breast cancer surgery. Eleven gene loci remained significantly associated with fatigue class membership in the regression analyses (Table 2).

Table 2.

Multiple Logistic Regression Analyses for Neurotransmitter Genes and Lower Fatigue Versus Higher Fatigue Classes

Predictor Odds Ratio Standard Error 95% CI Z p-value
ADRB2 rs1042718 0.13 0.100 0.030, 0.582 −2.67 .008
Age 0.80 0.052 0.707, 0.912 −3.39 .001
KPS score 0.56 0.097 0.396, 0.783 −3.36 .001
SCQ score 1.11 0.062 0.998, 1.243 1.92 .054
Any chemotherapy 2.31 0.669 1.307, 4.072 2.88 .004
Overall model fit: χ2 = 59.87, p <.0001 R2 = 0.1479
BDNF rs6265 0.50 0.149 0.278, 0.897 −2.32 .020
Age 0.80 0.052 0.707, 0.910 −3.43 .001
KPS score 0.57 0.101 0.406, 0.810 −3.16 .002
SCQ score 1.13 0.063 1.010, 1.256 2.14 .032
Any chemotherapy 2.50 0.727 1.414, 4.420 3.15 .002
Overall model fit: χ2 = 56.84, p <.0001 R2 = 0.1404
COMT rs9332377 0.48 0.158 0.256, 0.919 −2.22 .026
Age 0.82 0.052 0.723, 0.928 −3.13 .002
KPS score 0.55 0.095 0.389, 0.767 −3.49 <.0001
SCQ score 1.13 0.063 1.011, 1.260 2.15 .031
Any chemotherapy 2.41 0.697 1.370, 4.251 3.05 .002
Overall model fit: χ2 = 56.34, p <.0001 R2 = 0.1392
CYP3A4 rs4646437 0.48 0.157 0.253, 0.914 −2.24 .025
Age 0.81 0.052 0.710, 0.914 −3.36 .001
KPS score 0.55 0.098 0.392, 0.783 −3.34 .001
SCQ score 1.12 0.063 1.005, 1.251 2.04 .041
Any chemotherapy 2.40 0.691 1.365, 4.221 3.04 .002
Overall model fit: χ2 = 56.43, p <.0001 R2 = 0.1394
GALR1 rs949060 2.46 0.950 1.150, 5.244 2.32 .020
Age 0.81 0.053 0.713, 0.920 −3.25 .001
KPS score 0.58 0.100 0.413, 0.814 −3.15 .002
SCQ score 1.12 0.063 1.007, 1.253 2.09 .037
Any chemotherapy 2.55 0.738 1.444, 4.496 3.23 .001
Overall model fit: χ2 = 56.98, p <.0001 R2 = 0.1411
GCH1 rs3783642 0.47 0.144 0.260, 0.859 −2.46 .014
Age 0.81 0.052 0.713, 0.917 −3.31 .001
KPS score 0.58 0.102 0.411, 0.818 −3.10 .002
SCQ score 1.12 0.064 1.006, 1.256 2.07 .039
Any chemotherapy 2.40 0.690 1.364, 4.216 3.04 .002
Overall model fit: χ2 = 57.66, p <.0001 R2 = 0.1424
NOS1 rs9658498 0.45 0.164 0.223, 0.920 −2.19 .029
NOS1 rs2293052 4.58 2.429 1.621, 12.953 2.87 .004
Age 0.80 0.053 0.705, 0.913 −3.33 .001
KPS score 0.54 0.095 0.383, 0.762 −3.51 <.0001
SCQ score 1.11 0.063 0.991, 1.240 1.80 .072
Any chemotherapy 2.45 0.721 1.373, 4.361 3.04 .002
Overall model fit: χ2 = 69.13, p <.0001 R2 = 0.1708
NPY1R Haplotype A04 1.77 0.346 1.207, 2.595 2.92 .003
Age 0.81 0.052 0.711, 0.917 −3.31 .001
KPS score 0.55 0.099 0.388, 0.784 −3.32 .001
SCQ score 1.11 0.063 0.994, 1.241 1.85 .064
Any chemotherapy 2.58 0.756 1.454, 4.584 3.24 .001
Overall model fit: χ2 = 60.22, p <.0001 R2 = 0.1487
SLC6A2 rs17841327 10.31 8.139 2.195, 48.439 2.96 .003
Age 0.81 0.053 0.717, 0.924 −3.18 .001
KPS score 0.56 0.101 0.395, 0.797 −3.23 .001
SCQ score 1.13 0.064 1.007, 1.257 2.08 .037
Any chemotherapy 2.68 0.784 1.514, 4.756 3.38 .001
Overall model fit: χ2 = 65.01, p <.0001 R2 = 0.1606
5HTTLPR + rs25531 in SLC6A4 0.53 0.148 0.305, 0.914 −2.28 .023
Age 0.81 0.505 0.720, 0.919 −3.33 .001
KPS score 0.59 0.101 0.422, 0.825 −3.09 .002
SCQ score 1.12 0.060 1.012, 1.246 2.19 .029
Any chemotherapy 2.23 0.623 1.292, 3.858 2.88 .004
Overall model fit: χ2 = 52.30, p <.0001 R2 = 0.1280

Multiple logistic regression analyses of candidate gene associations with lower fatigue versus higher fatigue classes (n=301). For each model, the first three principal components identified from the analysis of ancestry informative markers, as well as self-reported race/ethnicity, were retained in all models to adjust for potential confounding due to race/ethnicity (data not shown). For the regression analyses, predictors evaluated in each model included genotype (ADRB2 rs1042718: CC+CA versus AA; BDNF rs6265: GG versus GA+AA; COMT rs9332377: TT versus TC+CC; CYP3A4 rs4646437: CC versus CT+TT; GALR1 rs949060: GG+GC versus CC; GCH1 rs3783642: TT versus TC+CC; NOS1 rs9658498: TT+TC versus CC; NOS1 rs2293052: CC+CT versus TT; NPY1R HapA04: haplotype composed of the rs9764 common T allele and the rs7687423 common G allele; SLC6A2 rs17841327: CC+CA versus AA; and 5HTTLPR + rs25531 (the 5-HTTLPR triallelic polymorphism): zero doses of LA allele versus one or two doses of LA allele), age (5 years increments), functional status (KPS score in 10 unit increments), number of comorbid conditions, and receipt of chemotherapy within six months after surgery.

Abbreviations: ADRB2 = adrenergic, beta-2 receptor, surface; any chemotherapy = receipt of chemotherapy within six months after surgery; BDNF = brain derived neurotrophic factor; CI = confidence interval; COMT = catechol-O-methyltransferase; CYP3A4 = cytochrome P450, family 3, subfamily A, polypeptide 4; GALR1 = galanin receptor 1; GCH1 = GTP cyclohydrolase 1; Hap = haplotype; 5-HTTLPR = serotonin-linked polymorphic region; KPS = Karnofsky Performance Status; NOS1 = nitric oxide synthase 1; NPY1R = neuropeptide Y receptor Y1; SCQ = Self-administered Comorbidity Questionnaire; SLC6A2 = solute carrier family 6 (neurotransmitter transporter, noradrenaline) member 2

For ADRB2 rs1042718, carrying two doses of the rare A allele was associated with a 87% lower odds of belonging to the Higher Fatigue class (Figure 1A). For BDNF rs6265, carrying one or two doses of the rare A allele was associated with a 50% lower odds of belonging to the Higher Fatigue class (Figure 1B). For COMT rs9332377, carrying one or two doses of the rare C allele was associated with a 52% lower odds of belonging to the Higher Fatigue class (Figure 1C). For CYP3A4 rs4646437, carrying one or two doses of the rare T allele was associated with a 52% lower odds of belonging to the Higher Fatigue class (Figure 1D). For GALR1 rs949060, carrying two doses of the rare C allele was associated with a 2.46-fold higher odds of belonging to the Higher Fatigue class (Figure 1E). For GCH1 rs3783642, carrying one or two doses of the rare C allele was associated with a 53% lower odds of belonging to the Higher Fatigue class (Figure 1F).

Figure 1.

Figure 1

A–F – Differences between the fatigue latent classes in the percentages of patients who were homozygous for the common allele or heterozygous or homozygous for the rare allele in for each of the polymorphism identified. Values are plotted as unadjusted proportions with corresponding p-value.

For NOS1, two SNPs were associated with membership in the Higher Fatigue class. In the regression analysis, including both SNPs, for NOS1 rs9658498, carrying two doses of the rare C allele was associated with a 55% lower odds of belonging to the Higher Fatigue class (Figure 2A). In the same regression analysis, for NOS1 rs2293052, carrying two doses of the rare T allele was associated with a 4.58-fold higher odds of belonging to the Higher Fatigue class (Figure 2B). For NPY1R HapA04, that is composed of alleles at two SNPs (i.e., rs9764 [common T allele], and rs7687423 [common G allele]), each additional dose of NPY1R HapA04 was associated with a 1.77-fold higher odds of belonging to the Higher Fatigue class (Figure 3). For SLC6A2 rs17841327, carrying two doses of the rare A allele was associated with a 10.31-fold higher odds of belonging to the Higher Fatigue class (Figure 2C). For the 5-HTTLPR + rs25531 polymorphism in SLC6A4, carrying one or two doses of the LA allele was associated with a 47% lower odds of belonging to the Higher Fatigue class (Figure 2D).

Figure 2.

Figure 2

A–D – Differences between the fatigue latent classes in the percentages of patients who were homozygous for the common allele or heterozygous or homozygous for the rare allele in for each of the polymorphism identified. Values are plotted as unadjusted proportions with corresponding p-value.

Figure 3.

Figure 3

NPYR1 linkage disequibrium (LD)-based heatmap and haplotype analysis. The top white bar represents the physical distance along the human chromosome. Reference sequence identifiers (rsIDs) for each single-nucleotide polymorphism (SNP) are plotted on the white bar and equidistantly to render the pairwise LD estimates. The correlation statistics (r2 and D′) are provided in the heatmap. The haplotype is indicated in a bolded triangle and its component SNPs are rendered in bold font. Pairwise D′ value (range: 0–1, inclusive) was rendered in color, with the darker red diamond representing D′ value approaching 1.0. When the r2 (range 0–100 inclusive) is not equal to 0 or 100, it is provided in a given diamond. The 2-SNP haplotype associated with fatigue is composed of rs9764 and rs7687423.

Differences in Demographic and Clinical Characteristics between the Energy Classes

Differences between the two energy classes are listed in Table 3. Patients in the Lower Energy class had a lower KPS score, a higher SCQ score, and a lower mean energy score at enrollment. In addition, a higher percentage of patients with more advanced disease were in the Lower Energy class.

Table 3.

Differences in Demographic and Clinical Characteristics Between the Higher Energy (n=127) and Lower Energy (n=270) Classes at Enrollment

Characteristic Higher Energy Class

n=127
(31.9%)

Mean (SD)
Lower Energy Class

n= 270
(67.8%)

Mean (SD)
Statistic and p-value
Age (years) 56.5 (10.8) 54.2 (11.8) t=1.88, p=.061
Education (years) 15.7 (2.2) 15.7 (2.8) t=0.01, p=.994
Karnofsky Performance Status score 95.4 (9.4) 92.2 (10.6) t=3.06, p=.002
Self-administered Comorbidity Questionnaire score 3.6 (2.3) 4.6 (3.0) t=−3.47, p=.001
Mean energy score at enrollment 6.1 (2.7) 4.4 (2.2) t=−6.26, p=<.0001
Number of breast biopsies in past year 1.5 (0.8) 1.5 (0.8) U, p=.604
Number of positive lymph nodes 0.8 (2.0) 1.0 (2.3) t=0.76, p=.450
Number of lymph nodes removed 5.0 (6.3) 6.1 (6.9) t=−1.51, p=.132
n (%) n (%)
Ethnicity
 White
 Black
 Asian/Pacific Islander
 Hispanic/Mixed ethnic background/Other

86 (68.3)
10 (7.9)
16 (12.7)
14 (11.1)

169 (62.8)
30 (11.2)
33 (12.3)
37 (13.8)
χ2=1.75, p=.627
Married/partnered (% yes) 50 (39.7) 114 (42.7) FE, p=.586
Work for pay (% yes) 66 (52.4) 123 (45.9) FE, p=.236
Lives alone (% yes) 29 (23.0) 65 (24.4) FE, p=.801
Gone through menopause (% yes) 84 (68.3) 163 (62.0) FE, p=.256
Stage of disease
 0
 I
 IIA and IIB
 IIIA, IIIB, IIIC, and IV

29 (22.8)
51 (40.2)
39 (30.7)
8 (6.3)

44 (16.3)
100 (37.0)
101 (37.4)
25 (9.3)
U, p=.040
Surgical treatment
 Breast conservation
 Mastectomy

100 (78.7)
27 (21.3)

218 (80.7)
52 (19.3)
FE, p=.686
Sentinel node biopsy (% yes) 103 (81.1) 224 (83.0) FE, p=.673
Axillary lymph node dissection (% yes) 40 (31.7) 108 (40.0) FE, p=.120
Breast reconstruction at the time of surgery (% yes) 28 (22.2) 58 (21.5) FE, p=.896
Neoadjuvant chemotherapy (% yes) 22 (17.5) 57 (21.1) FE, p=.421
Radiation therapy during the first 6 months (% yes) 75 (59.1) 149 (55.2) FE, p=.515
Chemotherapy during the first 6 months (% yes) 34 (26.8) 99 (36.7) FE, p=.054

Abbreviations: FE=Fisher Exact test, SD = standard deviation, U=Mann Whitney U test

Candidate Gene Analyses for Energy

Genotype distributions differed between the Higher Energy and Lower Energy classes for: 1 SNP in COMT; 2 SNPs in HTR2A; 1 SNP in NOS1; 1 SNP in NOS2A; 4 SNPs and 3 haplotypes in SLC6A1; 4 SNPs in SLC6A2; 1 SNP in SLC6A3; 3 SNPs and 1 haplotype in SLC6A4; 1 SNP in TAC1; and 1 SNP in TACR1.

Regression Analyses for Energy

In these regression analyses that included genomic estimates of and self-reported race/ethnicity, the phenotypic characteristics that remained significant in the multivariate model were: KPS score and receipt of CTX within six months after breast cancer surgery. Seven gene loci remained significantly associated with energy class membership in the multivariate logistic regression analyses (Table 4).

Table 4.

Multiple Logistic Regression Analyses for Neurotransmitter Genes and Higher Energy Versus Lower Energy Classes

Predictor Odds Ratio Standard Error 95% CI Z p-value
NOS1 rs471871 0.28 0.138 0.103,0.736 −2.57 .010
KPS score 0.65 0.101 0.483, 0.884 −2.75 .006
Any chemotherapy 1.73 0.479 1.002, 2.972 1.97 .049
Overall model fit: χ2 = 24.43, p =.0037 R2 = 0.0638
SLC6A1 rs2675163 1.85 0.507 1.082, 3.166 2.25 .025
SLC6A1 Haplotype D01 0.60 0.116 0.413, 0.880 −2.62 .009
KPS score 0.68 0.105 0.503, 0.921 −2.49 .013
Any chemotherapy 1.56 0.440 0.898, 2.714 1.58 .114
Overall model fit: χ2 =30.86, p =.0006 R2 = 0.0810
SLC6A2 rs36027 0.59 0.107 0.415, 0.844 −2.90 .004
KPS score 0.66 0.102 0.484, 0.889 −2.72 .007
Any chemotherapy 1.75 0.485 1.014, 3.010 2.01 .044
Overall model fit: χ2 =26.25, p =.0019 R2 = 0.0686
SLC6A3 rs37022 9.75 10.612 1.155, 82.302 2.09 .036
KPS score 0.66 0.103 0.484, 0.895 −2.67 .008
Any chemotherapy 1.75 0.487 1.017, 3.022 2.02 .043
Overall model fit: χ2 = 24.77, p =.0032 R2 = 0.0647
SLC6A4 rs2020942 0.36 0.144 0.161, 0.787 −2.55 .011
KPS score 0.66 0.103 0.488, 0.898 −2.65 .008
Any chemotherapy 1.73 0.482 1.006, 2.991 1.98 .047
Overall model fit: χ2 = 24.16, p =.0041 R2 = 0.0631
TAC1 rs2072100 2.11 0.718 1.083, 4.113 2.19 .028
KPS score 0.67 0.102 0.498, 0.905 −2.61 .009
Any chemotherapy 1.73 0.480 1.007, 2.983 1.98 .047
Overall model fit: χ2 = 22.78, p =.0067 R2 = 0.0595

Multiple logistic regression analyses of candidate gene associations with higher energy versus lower energy classes (n=301). For each model, the first three principal components identified from the analysis of ancestry informative markers, as well as self-reported race/ethnicity, were retained in all models to adjust for potential confounding due to race/ethnicity. For the regression analyses, predictors evaluated in each model included genotype (NOS1 rs471871 genotype: AA +AT versus TT; SLC6A1 rs2675163 genotype: TT versus TC+CC; SLC6A1 HapD01 haplotype: composed of the rs10514669 common C allele, the rs2697138 common C allele, and the rs1062246 common A allele; SLC6A2 rs36027 genotype: AA versus AG versus GG; SLC6A3 rs37022 genotype: TT+TA versus AA; SLC6A4 rs2020942 genotype: GG+GA versus AA; TAC1 rs2072100 genotype: AA+AG versus GG), functional status (KPS score in 10 unit increments), and receipt of chemotherapy within six months after surgery.

Abbreviations: Any chemotherapy = receipt of chemotherapy within six months after surgery; CI = confidence interval; Hap = haplotype; KPS = Karnofsky Performance Status; NOS1 = nitric oxide synthase 1; SCQ = Self-administered Comorbidity Questionnaire; SLC6A1 = solute carrier family 6 (neurotransmitter transporter, GABA) member 1; SLC6A2 = solute carrier family 6 (neurotransmitter transporter, noradrenaline) member 2; SLC6A3 = solute carrier family 6 (neurotransmitter transporter, dopamine) member 3; SLC6A4 = solute carrier family 6 (neurotransmitter transporter, serotonin) member 4; TAC1 = tachykinin, precursor 1

For NOS1 rs471871, carrying two doses of the rare T allele was associated with a 72% lower odds of belonging to the Lower Energy class (Figure 4A). For SLC6A1, one SNP and one haplotype were associated with membership in the Lower Energy class. For SLC6A1 rs2675163, carrying one or two doses of the rare C allele was associated with a 1.85-fold higher odds of belonging to the Lower Energy class (Figure 4B). In the same regression analysis, for SLC6A1 HapD01, that is composed of alleles at three SNPs (i.e., rs10514669 [common C allele], rs2697138 [common C allele], and rs1062246 [common A allele]), each additional dose of SLC6A1 HapD01 was associated with a 40% lower odds of belonging to the Lower Energy class (Figure 5). For SLC6A2 rs36027, each additional dose of the rare G allele was associated with a 41% lower odds of belonging to the Lower Energy class (Figure 4C). For SLC6A3 rs37022, carrying two doses of the rare A allele was associated with a 9.75-fold higher odds of belonging to the Lower Energy class (Figure 4D). For SLC6A4 rs2020942, carrying two doses of the rare A allele was associated with a 64% lower odds of belonging to the Lower Energy class (Figure 4E). For TAC1 rs2072100, carrying two doses of the rare G allele was associated with a 2.11-fold higher odds of belonging to the Lower Energy class (Figure 4F).

Figure 4.

Figure 4

A–F – Differences between the energy latent classes in the percentages of patients who were homozygous for the common allele or heterozygous or homozygous for the rare allele in for each of the polymorphism identified. Values are plotted as unadjusted proportions with corresponding p-value.

Figure 5.

Figure 5

SLC6A1 linkage disequibrium (LD)-based heatmap and haplotype analysis. The top white bar represents the physical distance along the human chromosome. Reference sequence identifiers (rsIDs) for each single-nucleotide polymorphism (SNP) are plotted on the white bar and equidistantly to render the pairwise LD estimates. The correlation statistics (r2 and D′) are provided in the heatmap. The haplotype is indicated in a bolded triangle and its component SNPs are rendered in bold font. Pairwise D′ value (range: 0–1, inclusive) was rendered in color, with the darker red diamond representing D′ value approaching 1.0. When the r2 (range 0–100 inclusive) is not equal to 0 or 100, it is provided in a given diamond. The 3-SNP haplotype associated with energy is composed rs10514669, rs2697138, and rs1062246.

DISCUSSION

A discussion of the differences in phenotypic characteristics between the fatigue latent classes, as well as between the energy latent classes, is found in our recent paper.16 This discussion is focused on the genotypic findings.

Fatigue Polymorphisms

The ADRB2 receptor is part of the G-protein-coupled receptor family that influences sympathetic nervous system responses, as well as plays a role in the regulation of lipid metabolism. Polymorphisms in ADRB2 are associated with bronchodilation; insulin secretion; gluconeogenesis and glycogenolysis in skeletal muscle; as well as increased cardiac output; arterial dilation; and lipolysis.54 Sarpeshkar and Bentley hypothesized that alterations in this gene may be responsible for enhanced aerobic capacity and delayed exercise-induced fatigue.54 In addition, ADRB2 receptor stimulation inhibits production of type 1 pro-inflammatory cytokines55 and under-expression of ADRB2 receptors is associated with chronic fatigue syndrome.56

In our study, patients who were homozygous for the rare A allele for ADRB2 rs1042718 had a 87% lower odds of belonging to the Higher Fatigue class. Polymorphisms in ADRB2 rs1042718 produce a silent mutation. While no studies were identified that evaluated associations between this SNP and fatigue, significant associations were found between rs1042718 and enhanced longevity57 and negative emotions.58 In the negative emotions study,58 individuals who were heterozygous or homozygous for the rare allele in rs1042718 were less likely to report feelings of uselessness, loneliness, and anxiety. Given that previous studies of oncology patients found associations between higher levels of psychological distress and fatigue,5961 our findings are consistent with those reported by Zheng and colleagues.58

COMT is a key enzyme responsible for the metabolism and inactivation of dopamine, norepinephrine, and epinephrine.62 Alterations in the COMT gene were associated with fatigue and pain in breast cancer patients through interactions with two stress pathways (i.e., hypothalamic-pituitary-adrenal (HPA) axis, the sympathetic nervous system (SNS)).6264 In our study, patients who were heterozygous or homozygous for the rare C allele for COMT rs9332377 had a 52% lower odds of belonging to the Higher Fatigue class. This intronic SNP is located near the 3′ UTR of the COMT gene. While this location suggests that this polymorphism has a regulatory function and may affect COMT expression,65 we found no support for nearby regulatory regions when the ENCODE data were reviewed. Only three studies have reported significant associations between COMT rs9332377 and clinical phenotypes (i.e., hearing loss,66 suicidal ideation,67 nicotine dependence65). No studies have evaluated for associations between COMT rs9332377 and fatigue. However,, in the study of suicidal ideation,67 individuals who were homozygous for the rare C allele of COMT rs9332377 reported lower irritability scores on the Questionnaire for Measuring Factors of Aggression. This finding supports our association between rs9332377 and decreased fatigue when one considers COMT’s role in the manifestation of emotions, a possible marker for chronic fatigue syndrome.68

BDNF is a neural growth factor found throughout the central nervous system (CNS). BDNF is associated with overall brain health because it plays a role in the promotion of neurogenesis, neuroprotection, mental performance, and cognitive function.69 Altered BDNF levels are associated with fibromyalgia syndrome,70 chronic fatigue syndrome,71 and depression.72

BDNF rs6265 is a missense mutation that results in a non-synonymous conservative change in the amino acid sequence from valine (Val) to methionine (Met). In two studies,72,73 decreases in serum BDNF levels were associated with the Met allele. In our study, being heterozygous or homozygous for the rare allele was associated with a reduction in the odds of belonging to the Higher Fatigue class. One might hypothesize that lower levels of BDNF would be associated with membership in the Higher Fatigue class given that lower levels of BDNF were associated with depression72 and chronic fatigue syndrome.71 However, findings regarding changes in serum levels of BDNF associated with the Met allele are inconsistent.74 In addition, the effect of the Met allele on BDNF levels in the brain, where it may play a greater role in the perception of fatigue, remains unknown.

The CYP3A4 gene is a part of the cytochrome P450 superfamily. Cytochrome P450 enzymes are responsible for catalyzing multiple reactions involved in lipid synthesis and drug metabolism. These enzymes are responsible for the metabolism of approximately one-third of anticancer drugs.75 The rs4646437 SNP is located in intron 7 of CYP3A4. While no studies evaluated for associations between CYP3A4 rs4646437 and fatigue, in one study,76 an association was found between CYP3A4 rs4646437 and in vitro CYP3A expression and activity. In this study, women who carried the rare T allele of rs4646437 had higher expression and activity of the CYP3A4 enzyme. Considering CYP3A4’s role in metabolizing anti-cancer drugs, one can hypothesize that women who are able to more effectively metabolize CTX would be less likely to experience higher levels of fatigue. This hypothesis is supported by our findings that being heterozygous or homozygous for rare T allele for rs4646437 was associated with a 52% lower odds of belonging to the Higher Fatigue class.

Galanin, a neuropeptide found throughout the CNS, has an inhibitory effect on multiple neurotransmitters.77 Polymorphisms in the galanin gene are associated with eating disorders,78 cancer,79 Alzheimer’s disease,80,81 depression, and anxiety.77 Within the CNS, the functional effects of galanin are mediated by three G-protein-coupled receptor subtypes, including GALR1. The GAL1 receptor has an inhibitory effect on adenylate cyclase through coupling with the G proteins Gi/Go. This inhibition affects ATP metabolism and plays an important role in cellular energy pathways.81 Of note, Staines82 hypothesized that dysfunctions in G protein-coupled receptors (e.g., GALR1) contribute to the development of fatigue. In our study, patients who homozygous for the rare C allele for GALR1 rs949060 had a 2.46-fold higher odds of belonging to the Higher Fatigue class. GALR1 rs949060 is located on chromosome 18 approximately 3 kilobases upstream of the GALR1 gene in the promoter region. However, no nearby regulatory element was identified in the ENCODE data.

GCH1 is the rate-limiting enzyme involved in the synthesis of tetrahydrobiopterin (BH4). BH4 plays a role in nitric oxide (NO) production and hydroxylation of aromatic amino acids. Polymorphisms in GCH1 are associated with pain,83 altered cognitive performance,84 and dopa-responsive dystonia.85 In our study, being heterozygous or homozygous for the rare C allele for GCH1 rs3783642 was associated with a 53% lower odds of belonging to the Higher Fatigue class. While no studies reported on GCH1 rs3783642, in one study,86 a protective association between other polymorphisms in GCH1 and fibromyalgia syndrome. This locus resides in putative CCCTC-binding factor (CTCF) and RAD22 transcription factor binding sites which suggests a possible role in the regulation of GCH1.

NPY1R is part of a family of G protein-coupled receptors that binds NPY. NPY acts in both the CNS and peripheral nervous system (PNS). Peripherally, NPY is a neurotransmitter that is released from sympathetic nerve endings. Centrally, NPY acts on receptors present in those areas of the brain that are involved with emotion.87 NPY is involved in sleep regulation, anxiety, memory, pain, and energy homeostasis.88,89 Alterations in NPY are implicated in chronic fatigue syndrome87 and depression.90 Alterations in NPY signaling through variations in NPY1R may have an effect on any of the aforementioned processes, including fatigue.

In our study, each additional dose of NPY1R HapA04 was associated with a 1.77-fold higher odds of belonging to the Higher Fatigue class. HapA04 is comprised of a 3-prime UTR SNP (rs9764) and one intronic SNP (rs7687423). Although no studies were identified that reported on NPYR1 HapA04, polymorphisms in rs9764 and rs7687423 were associated with nicotine91 and methamphetamine92 dependence, respectively. No studies were identified that reported on associations with either SNP and fatigue.

Energy Polymorphisms

The SLC6A1 gene encodes for one of the four GABA transporters found in the brain. The role of this transporter is to remove GABA from the synaptic cleft which decreases extracellular levels of GABA. The inhibitory neurotransmitter GABA is important for normal brain function. Based on studies of knockout mice,93 deficiencies in SLC6A1 were associated with depression, reduced aggression, and reduced anxiety. Furthermore, an association was found between polymorphisms in SLC6A1 and anxiety disorders.94 In a genome-wide association study,94 a SNP in SLC6A1 was associated with symptoms of inattention and hyperactivity in attention-deficit/hyperactivity disorder (ADHD).

In our study, one SNP and one haplotype in the SLC6A1 gene were associated with membership in the Lower Energy class. Being heterozygous or homozygous for the rare C allele of SLC6A1 rs2675163 was associated with a 1.85-fold higher odds of belonging to the Lower Energy class, while each additional dose of SLC6A1 HapD01, that is composed of alleles at three SNPs (i.e., rs10514669, rs2697138, and rs1062246), was associated with a 40% lower odds of belonging to the Lower Energy class. No studies were identified that reported on polymorphisms in any of the SLC6A1 SNPs and energy.

The SLC6A3 gene encodes for a dopamine transporter. The dopamine transporter protein is responsible for re-uptake of dopamine from the synaptic cleft which results in decreased extracellular levels of dopamine.96 Decreased levels of dopamine are hypothesized to play a role in the development of central fatigue because of dopamine’s known effects on initiation of movement.97 Therefore, alterations in dopaminergic circuits, including its transport receptors, may affect an individual’s energy level and fatigue.

While the majority of the literature on polymorphisms in the SLC6A3 gene has focused on ADHD,98,99 associations were found between dopaminergic polymorphisms and fatigue,22 as well as decreases in mental energy and sustained attention.100 In our study, being homozygous for the rare A allele of SLC6A3 rs37022 was associated with a 9.75-fold higher odds of belonging to the Lower Energy class. No studies were identified that reported on polymorphisms in this SNP.

The TAC1 gene encodes for a group of tachykinin peptide hormones (e.g., Substance P) that function as neurotransmitters. Substance P is implicated in fibromyalgia syndrome101 and with fatigue and other negative mood states.102 Therefore, polymorphisms in tachykinin pathway genes may have an effect on fatigue and energy levels. In our study, being homozygous for the rare G allele of TAC1 rs2072100 was associated with a 2.11-fold higher odds of belonging to the Lower Energy class. While the rs2072100 polymorphism was linked with increased risk for colorectal cancer103 and susceptibility to multiple sclerosis,104 no studies were identified that reported on associations with energy.

Fatigue and Energy Polymorphisms

Three genes (i.e., NOS1, SLC6A2, SL6A4) were associated with latent class membership for both fatigue and energy. NOS1 is part of a group of nitric acid synthases (NOS) responsible for the synthesis of NO. NO mediates vasodilation, neural regulation of skeletal muscle, and neurotransmission.105 Elevated NO levels are implicated in chronic fatigue syndrome,106 fatigue in muscular dystrophies,107,108 and fatigue in post-radiation syndrome.109 While no studies were identified on associations between NOS polymorphisms and fatigue, other studies found associations between polymorphisms in the NOS1 gene and depression110 and anxiety.111

In our study, two SNPs in the NOS1 gene were associated with membership in the Higher Fatigue class. Being homozygous for the rare C allele of rs9658498 was associated with a 55% lower odds of belonging to the Higher Fatigue class, while carrying two doses of the rare T allele of rs2293052 was associated with a 4.58-fold higher odds of belonging to the Higher Fatigue class. No studies were identified that reported on NOS1 rs9658498. However, an association was found between rs2293052 and Parkinson’s disease (PD).112 These results support our findings of an association between this SNP and increased fatigue because similar to the aforementioned fatigue-syndromes, PD is associated with increased NO levels.113 In addition, fatigue is a common symptom associated with PD114 and may share similar susceptibility gene polymorphisms. A different SNP in the NOS1 gene was associated with energy levels. Being homozygous for the rare T allele of rs471871 was associated with a 72% lower odds of belonging to the Lower Energy class. No studies were identified that reported on NOS1 rs471871.

The SLCA2 gene encodes for the norepinephrine transporter (NET) protein. The NET found at noradrenergic synapses, is responsible for the removal of NE from the synaptic cleft and plays a major role in NE homeostasis.115 Impairments in the NET protein may contribute to the development of fatigue.116 Mutations in the SLCA2 gene are associated with orthostatic intolerance, a syndrome that includes fatigue as a significant symptom.115,117 In addition, polymorphisms in the SLCA2 gene were associated with major depression, a condition that includes fatigue as a major symptom.118 In our study, being homozygous for the rare A allele of SLC6A2 rs17841327 was associated with a 10.31-fold higher odds of belonging to the Higher Fatigue group. In addition, a different SNP in the SLC6A2 gene was associated with energy levels. Each additional dose of the rare G allele of SLC6A2 rs36027 was associated with a 41% higher odds of belonging to the Lower Energy class. No studies were identified that reported on either SLC6A2 SNP.

The SLC6A4 gene encodes for a membrane protein that is responsible for re-uptake of serotonin from the synaptic cleft. The serotonergic neurotransmitter system is hypothesized to play a role in cancer-related fatigue.119,120 Serotonin is involved in various human behaviors including sleep, mood, appetite, memory, and learning. Increased levels of serotonin in the brain are hypothesized to contribute to fatigue through its interaction with the HPA axis leading to a sensation of reduce potential to perform physical activity.119 Yamamoto et al.121 demonstrated a reduced density of serotonin transporters in the rostral subdivision of the anterior cingulate of patients with chronic fatigue syndrome. In addition, an association was found between polymorphisms in the promoter of the SLC6A4 gene and chronic fatigue syndrome.122

In our study, being homozygous for the rare A allele of SLC6A4 rs2020942 was associated with a 64% lower odds of belonging to the Lower Energy class. In addition, carrying one or two doses of the LA allele for the 5-HTTLPR + rs25531 polymorphism in SLC6A4 was associated with a 47% decrease in the odds of belonging to the Higher Fatigue class. While rs2020942 was linked with obsessive-compulsive symptoms123 and risk for nonsyndromic cleft lip with or without cleft palate,124 no studies have reported on associations between SLC6A4 rs2020942 and energy level.

While the functional consequences of SLC6A4 rs2020942 are not known, a considerable amount of research has evaluated the functional consequences of the A>G polymorphism (rs25531) within the L allele of 5-HTTLPR. In this triallelic polymorphism (i.e., S, LA, and LG), nearly equivalent expression of the serotonin transporter is observed with the S and LG alleles and increased transcriptional activity is observed with the LA allele.33,35 In addition, other studies found that being homozygous for the LA allele was associated with increased serotonin transporter binding.125,126 Since the serotonin transporter modulates the concentration of serotonin at the synaptic cleft, an increase in the transcription of the transporter would decrease the amount of serotonin at the synaptic cleft. In a number of studies, individuals who possessed one or two doses of the low activity alleles (S or LG) were more likely to develop behavioral disorders (e.g., depression) when they experienced stressful life events.127,128 Our findings are consistent with these observations in that patients who carried one or two doses of the LA allele were less likely to be in the Higher Fatigue class. One can hypothesize that patients with the LA allele have increased transcription of the serotonin transporter which would decrease the concentration of serotonin at the synaptic cleft. The decreased concentration of serotonin would result in decreased levels of fatigue.

Conclusions

Several limitations must be acknowledged. While our sample size was sufficient, additional studies with independent samples are needed to confirm the latent classes as well as the genetic associations. In order to increase the generalizability of these results, women were recruited from 7 different centers and approximately 30% of the patients were ethnic minorities. However, the single diagnosis of breast cancer limits the generalizability of the findings to other cancer diagnoses. Finally, recent studies suggest that diurnal variations in fatigue and energy levels constitute distinct symptom phenotypes.9,129,130 Future studies need to evaluate for differences in the levels of morning and evening fatigue and decrements in energy levels in women following breast cancer surgery.

The molecular findings from this study support the hypothesis that fatigue and energy are distinct, yet related symptoms. Only three of the 13 genes identified in this study were associated with membership in both the fatigue and energy latent classes. Additional support for this hypothesis comes from recent studies that explored the concepts of fatigue and energy in patients with cancer and their family caregivers9,47 and in women with HIV.131 Lerdal et al.131 concluded that fatigue and energy are distinct constructs and should not be used interchangeably, neither clinically nor in research. Additional studies are needed to determine which common and distinct phenotypic and molecular characteristics are associated with increased fatigue severity and decrements in energy. Findings from these types of studies will provide insights into the mechanisms that underlie one or both of these symptoms and facilitate the development and testing of interventions to decrease fatigue and/or increase energy levels of patients undergoing cancer treatment.

Supplementary Material

Acknowledgments

This study was funded by grants from the National Cancer Institute (NCI, CA107091 and CA118658). Dr. Christine Miaskowski is an American Cancer Society Clinical Research Professor and is supported by a K05 award from the NCI (CA168960). This project is supported by NIH/NCRR UCSF-CTSI Grant Number UL1 RR024131. Its contents are solely the responsibility of the authors and do not necessarily represent the official views of the NIH.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Prue G, Rankin J, Allen J, Gracey J, Cramp F. Cancer-related fatigue: A critical appraisal. Eur J Cancer. 2006;42:846–863. doi: 10.1016/j.ejca.2005.11.026. [DOI] [PubMed] [Google Scholar]
  • 2.Siefert ML. Fatigue, pain, and functional status during outpatient chemotherapy. Oncol Nurs Forum. 2010;37:E114–123. doi: 10.1188/10.ONF.114-123. [DOI] [PubMed] [Google Scholar]
  • 3.Dhruva A, Dodd M, Paul SM, et al. Trajectories of fatigue in patients with breast cancer before, during, and after radiation therapy. Cancer Nurs. 2010;33:201–212. doi: 10.1097/NCC.0b013e3181c75f2a. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Huang HP, Chen ML, Liang J, Miaskowski C. Changes in and predictors of severity of fatigue in women with breast cancer: A longitudinal study. Int J Nurs Stud. 2014;51:582–592. doi: 10.1016/j.ijnurstu.2013.09.003. [DOI] [PubMed] [Google Scholar]
  • 5.Lerdal A. A theoretical extension of the concept of energy through an empirical study. Scand J Caring Sci. 2002;16:197–206. doi: 10.1046/j.1471-6712.2002.00079.x. [DOI] [PubMed] [Google Scholar]
  • 6.Lerdal A. A concept analysis of energy. Its meaning in the lives of three individuals with chronic illness. Scand J Caring Sci. 1998;12:3–10. doi: 10.1080/02839319850163075. [DOI] [PubMed] [Google Scholar]
  • 7.Berger AM, Mooney K, Alvarez-Perez A, et al. Cancer-Related Fatigue, Version 2.2015. J Natl Compr Canc Netw. 2015;13:1012–1039. doi: 10.6004/jnccn.2015.0122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Van Onselen C, Aouizerat BE, Dunn LB, et al. Differences in sleep disturbance, fatigue and energy levels between women with and without breast pain prior to breast cancer surgery. Breast. 2013;22:273–276. doi: 10.1016/j.breast.2012.07.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Aouizerat BE, Dhruva A, Paul SM, et al. Phenotypic and molecular evidence suggests that decrements in morning and evening energy are distinct but related symptoms. J Pain Symptom Manage. 2015;50:599–614. doi: 10.1016/j.jpainsymman.2015.05.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Aouizerat BE, Gay CL, Lerdal A, Portillo CJ, Lee KA. Lack of energy: an important and distinct component of HIV-related fatigue and daytime function. J Pain Symptom Manage. 2013;45:191–201. doi: 10.1016/j.jpainsymman.2012.01.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Berger AM, Gerber LH, Mayer DK. Cancer-related fatigue: implications for breast cancer survivors. Cancer. 2012;118:2261–2269. doi: 10.1002/cncr.27475. [DOI] [PubMed] [Google Scholar]
  • 12.De Vries J, Van der Steeg AF, Roukema JA. Determinants of fatigue 6 and 12 months after surgery in women with early-stage breast cancer: a comparison with women with benign breast problems. J Psychosom Res. 2009;66:495–502. doi: 10.1016/j.jpsychores.2008.11.008. [DOI] [PubMed] [Google Scholar]
  • 13.Lockefeer JP, De Vries J. What is the relationship between trait anxiety and depressive symptoms, fatigue, and low sleep quality following breast cancer surgery? Psychooncology. 2013;22:1127–1133. doi: 10.1002/pon.3115. [DOI] [PubMed] [Google Scholar]
  • 14.Bortolon C, Krikorian A, Carayol M, et al. Cancer-related fatigue in breast cancer patients after surgery: a multicomponent model using partial least squares-path modeling. Psychooncology. 2014;23:444–451. doi: 10.1002/pon.3438. [DOI] [PubMed] [Google Scholar]
  • 15.Bower JE, Ganz PA, Irwin MR, et al. Cytokine genetic variations and fatigue among patients with breast cancer. J Clin Oncol. 2013;31:1656–1661. doi: 10.1200/JCO.2012.46.2143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Kober KM, Smoot B, Paul SM, et al. Polymorphisms in cytokine genes are associated with higher levels of fatigue and lower levels of energy in women following breast cancer surgery. J Pain Symptom Manage. doi: 10.1016/j.jpainsymman.2016.04.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Illi J, Miaskowski C, Cooper B, et al. Association between pro- and anti-inflammatory cytokine genes and a symptom cluster of pain, fatigue, sleep disturbance, and depression. Cytokine. 2012;58:437–447. doi: 10.1016/j.cyto.2012.02.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Collado-Hidalgo A, Bower JE, Ganz PA, Irwin MR, Cole SW. Cytokine gene polymorphisms and fatigue in breast cancer survivors: early findings. Brain Behav Immun. 2008;22:1197–1200. doi: 10.1016/j.bbi.2008.05.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Saligan LN, Olson K, Filler K, et al. The biology of cancer-related fatigue: a review of the literature. Support Care Cancer. 2015;23:2461–2478. doi: 10.1007/s00520-015-2763-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Meeusen R, Roelands B. Central fatigue and neurotransmitters, can thermoregulation be manipulated? Scand J Med Sci Sports. 2010;20(Suppl 3):19–28. doi: 10.1111/j.1600-0838.2010.01205.x. [DOI] [PubMed] [Google Scholar]
  • 21.Meeusen R, Watson P, Hasegawa H, Roelands B, Piacentini MF. Brain neurotransmitters in fatigue and overtraining. Appl Physiol Nutr Metab. 2007;32:857–864. doi: 10.1139/H07-080. [DOI] [PubMed] [Google Scholar]
  • 22.Malyuchenko NV, Schegolkova JV, Kulikova MA, et al. Effects of genetic variations in the dopaminergic system on fatigue in humans: gender aspects. Bull Exp Biol Med. 2010;149:226–232. doi: 10.1007/s10517-010-0913-4. [DOI] [PubMed] [Google Scholar]
  • 23.Kober KM, Dunn L, Mastick J, et al. Gene expression profiling of evening fatigue in women undergoing chemotherapy for breast cancer. Biol Res Nurs. doi: 10.1177/1099800416629209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Lee KA, Hicks G, Nino-Murcia G. Validity and reliability of a scale to assess fatigue. Psychiatry Res. 1991;36:291–298. doi: 10.1016/0165-1781(91)90027-m. [DOI] [PubMed] [Google Scholar]
  • 25.McCann B, Miaskowski C, Koetters T, et al. Associations between pro- and anti-inflammatory cytokine genes and breast pain in women prior to breast cancer surgery. J Pain. 2012;13:425–437. doi: 10.1016/j.jpain.2011.02.358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Miaskowski C, Cooper B, Paul SM, et al. Identification of patient subgroups and risk factors for persistent breast pain following breast cancer surgery. J Pain. 2012;13:1172–1187. doi: 10.1016/j.jpain.2012.09.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Van Onselen C, Aouizerat BE, Dunn LB, et al. Differences in sleep disturbance, fatigue and energy levels between women with and without breast pain prior to breast cancer surgery. Breast. 2013;22:273–276. doi: 10.1016/j.breast.2012.07.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Karnofsky D, Abelmann WH, Craver LV, Burchenal JH. The use of nitrogen mustards in the palliative treatment of carcinoma. Cancer. 1948;1:634–656. [Google Scholar]
  • 29.Sangha O, Stucki G, Liang MH, Fossel AH, Katz JN. The Self-Administered Comorbidity Questionnaire: a new method to assess comorbidity for clinical and health services research. Arthritis Rheum. 2003;49:156–163. doi: 10.1002/art.10993. [DOI] [PubMed] [Google Scholar]
  • 30.Gay CL, Lee KA, Lee SY. Sleep patterns and fatigue in new mothers and fathers. Biol Res Nurs. 2004;5:311–318. doi: 10.1177/1099800403262142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Encode Project Consortium. Dunham I, Kundaje A, et al. An integrated encyclopedia of DNA elements in the human genome. Nature. 2012;489:57–74. doi: 10.1038/nature11247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Nakamura M, Ueno S, Sano A, Tanabe H. The human serotonin transporter gene linked polymorphism (5-HTTLPR) shows ten novel allelic variants. Mol Psychiatry. 2000;5:32–38. doi: 10.1038/sj.mp.4000698. [DOI] [PubMed] [Google Scholar]
  • 33.Hu XZ, Lipsky RH, Zhu G, et al. Serotonin transporter promoter gain-of-function genotypes are linked to obsessive-compulsive disorder. Am J Hum Genet. 2006;78:815–826. doi: 10.1086/503850. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Wendland JR, Martin BJ, Kruse MR, Lesch KP, Murphy DL. Simultaneous genotyping of four functional loci of human SLC6A4, with a reappraisal of 5-HTTLPR and rs25531. Mol Psychiatry. 2006;11:224–226. doi: 10.1038/sj.mp.4001789. [DOI] [PubMed] [Google Scholar]
  • 35.Praschak-Rieder N, Kennedy J, Wilson AA, et al. Novel 5-HTTLPR allele associates with higher serotonin transporter binding in putamen: a [(11)C] DASB positron emission tomography study. Biol Psychiatry. 2007;62:327–331. doi: 10.1016/j.biopsych.2006.09.022. [DOI] [PubMed] [Google Scholar]
  • 36.Kaiser R, Tremblay PB, Schmider J, et al. Serotonin transporter polymorphisms: no association with response to antipsychotic treatment, but associations with the schizoparanoid and residual subtypes of schizophrenia. Mol Psychiatry. 2001;6:179–185. doi: 10.1038/sj.mp.4000821. [DOI] [PubMed] [Google Scholar]
  • 37.SPSS. IBM SPSS for Windows (Version 23) Chicago, Illinois: SPSS, Inc; 2015. [Google Scholar]
  • 38.StataCorp. Stata Statistical Software: Release 14. College Station, Texas: Stata Corporation; 2015. [Google Scholar]
  • 39.Jung T, Wickrama KAS. An introduction to latent class growth analysis and growth mixture modeling. Social and Personality Psychology Compass. 2008;2:302–317. [Google Scholar]
  • 40.Nylund KL, Asparouhov T, Muthen BO. Deciding on the number of classes in latent class analysis and growth mixture modeling: A Monte Carlo simulation study. Struct Equ Modeling. 2007;14:535–569. [Google Scholar]
  • 41.Tofighi D, Enders CK. Identifying the correct number of classes in growth mixture models. Charlotte, NC: Information Age Publishing; 2008. [Google Scholar]
  • 42.Stephens M, Smith NJ, Donnelly P. A new statistical method for haplotype reconstruction from population data. Am J Hum Genet. 2001;68:978–989. doi: 10.1086/319501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Halder I, Shriver M, Thomas M, Fernandez JR, Frudakis T. A panel of ancestry informative markers for estimating individual biogeographical ancestry and admixture from four continents: utility and applications. Human Mutation. 2008;29:648–658. doi: 10.1002/humu.20695. [DOI] [PubMed] [Google Scholar]
  • 44.Hoggart CJ, Parra EJ, Shriver MD, et al. Control of confounding of genetic associations in stratified populations. Am J Hum Genet. 2003;72:1492–1504. doi: 10.1086/375613. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Tian C, Gregersen PK, Seldin MF. Accounting for ancestry: population substructure and genome-wide association studies. Hum Mol Genet. 2008;17:R143–150. doi: 10.1093/hmg/ddn268. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Price AL, Patterson NJ, Plenge RM, et al. Principal components analysis corrects for stratification in genome-wide association studies. Nat Genet. 2006;38:904–909. doi: 10.1038/ng1847. [DOI] [PubMed] [Google Scholar]
  • 47.Dhruva A, Aouizerat BE, Cooper B, et al. Cytokine gene associations with self-report ratings of morning and evening fatigue in oncology patients and their family caregivers. Biol Res Nurs. 2015;17:175–184. doi: 10.1177/1099800414534313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Miaskowski C, Cataldo JK, Baggott CR, et al. Cytokine gene variations associated with trait and state anxiety in oncology patients and their family caregivers. Support Care Cancer. 2015;23:953–965. doi: 10.1007/s00520-014-2443-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Dunn LB, Aouizerat BE, Langford DJ, et al. Cytokine gene variation is associated with depressive symptom trajectories in oncology patients and family caregivers. Eur J Oncol Nurs. 2013;17:346–353. doi: 10.1016/j.ejon.2012.10.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Miaskowski C, Dodd M, Paul SM, et al. Lymphatic and angiogenic candidate genes predict the development of secondary lymphedema following breast cancer surgery. PLoS One. 2013;8:e60164. doi: 10.1371/journal.pone.0060164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Miaskowski C, Cooper BA, Dhruva A, et al. Evidence of associations between cytokine genes and subjective reports of sleep disturbance in oncology patients and their family caregivers. PLoS One. 2012;7:e40560. doi: 10.1371/journal.pone.0040560. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Rothman KJ. No adjustments are needed for multiple comparisons. Epidemiology. 1990;1:43–46. [PubMed] [Google Scholar]
  • 53.Bakan D. The test of significance in psychological research. Psychol Bull. 1966;66:423–437. doi: 10.1037/h0020412. [DOI] [PubMed] [Google Scholar]
  • 54.Sarpeshkar V, Bentley DJ. Adrenergic-beta(2) receptor polymorphism and athletic performance. J Hum Genet. 2010;55:479–485. doi: 10.1038/jhg.2010.42. [DOI] [PubMed] [Google Scholar]
  • 55.Elenkov IJ, Wilder RL, Chrousos GP, Vizi ES. The sympathetic nerve–an integrative interface between two supersystems: the brain and the immune system. Pharmacol Rev. 2000;52:595–638. [PubMed] [Google Scholar]
  • 56.Light AR, White AT, Hughen RW, Light KC. Moderate exercise increases expression for sensory, adrenergic, and immune genes in chronic fatigue syndrome patients but not in normal subjects. J Pain. 2009;10:1099–1112. doi: 10.1016/j.jpain.2009.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Zhao L, Yang F, Xu K, et al. Common genetic variants of the beta2-adrenergic receptor affect its translational efficiency and are associated with human longevity. Aging Cell. 2012;11:1094–1101. doi: 10.1111/acel.12011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Zeng Y, Cheng L, Zhao L, et al. Interactions between Social/behavioral factors and ADRB2 genotypes may be associated with health at advanced ages in China. BMC Geriatr. 2013;13:91. doi: 10.1186/1471-2318-13-91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.So WK, Marsh G, Ling WM, et al. The symptom cluster of fatigue, pain, anxiety, and depression and the effect on the quality of life of women receiving treatment for breast cancer: A multicenter study. Oncol Nurs Forum. 2009;36:E205–214. doi: 10.1188/09.ONF.E205-E214. [DOI] [PubMed] [Google Scholar]
  • 60.Fox SW, Lyon D, Farace E. Symptom clusters in patients with high-grade glioma. J Nurs Scholarsh. 2007;39:61–67. doi: 10.1111/j.1547-5069.2007.00144.x. [DOI] [PubMed] [Google Scholar]
  • 61.Hwang SS, Chang VT, Rue M, Kasimis B. Multidimensional independent predictors of cancer-related fatigue. J Pain Symptom Manage. 2003;26:604–614. doi: 10.1016/s0885-3924(03)00218-5. [DOI] [PubMed] [Google Scholar]
  • 62.Light KC, White AT, Tadler S, Iacob E, Light AR. Genetics and gene expression involving stress and distress pathways in fibromyalgia with and without comorbid chronic fatigue syndrome. Pain Res Treat. 2012;2012:427869. doi: 10.1155/2012/427869. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Fernandez-de-Las-Penas C, Cantarero-Villanueva I, Fernandez-Lao C, et al. Influence of catechol-o-methyltransferase genotype (Val158Met) on endocrine, sympathetic nervous and mucosal immune systems in breast cancer survivors. Breast. 2012;21:199–203. doi: 10.1016/j.breast.2011.09.012. [DOI] [PubMed] [Google Scholar]
  • 64.Fernandez-de-las-Penas C, Fernandez-Lao C, Cantarero-Villanueva I, et al. Catechol-O-methyltransferase genotype (Val158met) modulates cancer-related fatigue and pain sensitivity in breast cancer survivors. Breast Cancer Res Treat. 2012;133:405–412. doi: 10.1007/s10549-011-1757-y. [DOI] [PubMed] [Google Scholar]
  • 65.Greenbaum L, Kanyas KS, Rigbi A, et al. Why do young women smoke? VII COMT as a risk modifying gene for Nicotine dependence – role of gene-gene interaction, personality, and environmental factors. Hum Psychopharmacol. 2010;25:536–542. doi: 10.1002/hup.1149. [DOI] [PubMed] [Google Scholar]
  • 66.Ross CJ, Katzov-Eckert H, Dube MP, et al. Genetic variants in TPMT and COMT are associated with hearing loss in children receiving cisplatin chemotherapy. Nat Genet. 2009;41:1345–1349. doi: 10.1038/ng.478. [DOI] [PubMed] [Google Scholar]
  • 67.Calati R, Porcelli S, Giegling I, et al. Catechol-o-methyltransferase gene modulation on suicidal behavior and personality traits: review, meta-analysis and association study. J Psychiatr Res. 2011;45:309–321. doi: 10.1016/j.jpsychires.2010.07.004. [DOI] [PubMed] [Google Scholar]
  • 68.Goertzel BN, Pennachin C, de Souza Coelho L, et al. Combinations of single nucleotide polymorphisms in neuroendocrine effector and receptor genes predict chronic fatigue syndrome. Pharmacogenomics. 2006;7:475–483. doi: 10.2217/14622416.7.3.475. [DOI] [PubMed] [Google Scholar]
  • 69.Seifert T, Secher NH. Sympathetic influence on cerebral blood flow and metabolism during exercise in humans. Prog Neurobiol. 2011;95:406–426. doi: 10.1016/j.pneurobio.2011.09.008. [DOI] [PubMed] [Google Scholar]
  • 70.Nugraha B, Karst M, Engeli S, Gutenbrunner C. Brain-derived neurotrophic factor and exercise in fibromyalgia syndrome patients: a mini review. Rheumatol Int. 2012;32:2593–2599. doi: 10.1007/s00296-011-2348-2. [DOI] [PubMed] [Google Scholar]
  • 71.Chen R, Liang FX, Moriya J, et al. Chronic fatigue syndrome and the central nervous system. J Int Med Res. 2008;36:867–874. doi: 10.1177/147323000803600501. [DOI] [PubMed] [Google Scholar]
  • 72.Lotrich FE, Albusaysi S, Ferrell RE. Brain-derived neurotrophic factor serum levels and genotype: association with depression during interferon-alpha treatment. Neuropsychopharmacology. 2013;38:985–995. doi: 10.1038/npp.2012.263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Ozan E, Okur H, Eker C, et al. The effect of depression, BDNF gene val66met polymorphism and gender on serum BDNF levels. Brain Res Bull. 2010;81:61–65. doi: 10.1016/j.brainresbull.2009.06.022. [DOI] [PubMed] [Google Scholar]
  • 74.Terracciano A, Martin B, Ansari D, et al. Plasma BDNF concentration, Val66Met genetic variant and depression-related personality traits. Genes Brain Behav. 2010;9:512–518. doi: 10.1111/j.1601-183X.2010.00579.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Oneda B, Crettol S, Jaquenoud Sirot E, et al. The P450 oxidoreductase genotype is associated with CYP3A activity in vivo as measured by the midazolam phenotyping test. Pharmacogenet Genomics. 2009;19:877–883. doi: 10.1097/FPC.0b013e32833225e7. [DOI] [PubMed] [Google Scholar]
  • 76.Schirmer M, Rosenberger A, Klein K, et al. Sex-dependent genetic markers of CYP3A4 expression and activity in human liver microsomes. Pharmacogenomics. 2007;8:443–453. doi: 10.2217/14622416.8.5.443. [DOI] [PubMed] [Google Scholar]
  • 77.Holmes A, Heilig M, Rupniak NM, Steckler T, Griebel G. Neuropeptide systems as novel therapeutic targets for depression and anxiety disorders. Trends Pharmacol Sci. 2003;24:580–588. doi: 10.1016/j.tips.2003.09.011. [DOI] [PubMed] [Google Scholar]
  • 78.Leibowitz SF. Regulation and effects of hypothalamic galanin: relation to dietary fat, alcohol ingestion, circulating lipids and energy homeostasis. Neuropeptides. 2005;39:327–332. doi: 10.1016/j.npep.2004.12.022. [DOI] [PubMed] [Google Scholar]
  • 79.Misawa K, Ueda Y, Kanazawa T, et al. Epigenetic inactivation of galanin receptor 1 in head and neck cancer. Clin Cancer Res. 2008;14:7604–7613. doi: 10.1158/1078-0432.CCR-07-4673. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Wrenn CC, Crawley JN. Pharmacological evidence supporting a role for galanin in cognition and affect. Prog Neuropsychopharmacol Biol Psychiatry. 2001;25:283–299. doi: 10.1016/s0278-5846(00)00156-1. [DOI] [PubMed] [Google Scholar]
  • 81.Ogren SO, Kuteeva E, Hokfelt T, Kehr J. Galanin receptor antagonists: a potential novel pharmacological treatment for mood disorders. CNS Drugs. 2006;20:633–654. doi: 10.2165/00023210-200620080-00003. [DOI] [PubMed] [Google Scholar]
  • 82.Staines D. Are vasoactive neuropeptide autoimmune fatigue-related disorders mediated via G protein-coupled receptors? Med Hypotheses. 2005;65:29–31. doi: 10.1016/j.mehy.2005.02.013. [DOI] [PubMed] [Google Scholar]
  • 83.Lotsch J, Klepstad P, Doehring A, Dale O. A GTP cyclohydrolase 1 genetic variant delays cancer pain. Pain. 2010;148:103–106. doi: 10.1016/j.pain.2009.10.021. [DOI] [PubMed] [Google Scholar]
  • 84.Yasuda Y, Hashimoto R, Ohi K, et al. A functional polymorphism of the GTP cyclohydrolase 1 gene predicts attention performance. Neurosci Lett. 2014;566:46–49. doi: 10.1016/j.neulet.2014.02.019. [DOI] [PubMed] [Google Scholar]
  • 85.Theuns J, Crosiers D, Debaene L, et al. Guanosine triphosphate cyclohydrolase 1 promoter deletion causes dopa-responsive dystonia. Mov Disord. 2012;27:1451–1456. doi: 10.1002/mds.25147. [DOI] [PubMed] [Google Scholar]
  • 86.Kim SK, Kim SH, Nah SS, et al. Association of guanosine triphosphate cyclohydrolase 1 gene polymorphisms with fibromyalgia syndrome in a Korean population. J Rheumatol. 2013;40:316–322. doi: 10.3899/jrheum.120929. [DOI] [PubMed] [Google Scholar]
  • 87.Fletcher MA, Rosenthal M, Antoni M, et al. Plasma neuropeptide Y: a biomarker for symptom severity in chronic fatigue syndrome. Behav Brain Funct. 2010;6:76. doi: 10.1186/1744-9081-6-76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Dyzma M, Boudjeltia KZ, Faraut B, Kerkhofs M. Neuropeptide Y and sleep. Sleep Med Rev. 2010;14:161–165. doi: 10.1016/j.smrv.2009.09.001. [DOI] [PubMed] [Google Scholar]
  • 89.Chambers AP, Woods SC. The role of neuropeptide Y in energy homeostasis. Handb Exp Pharmacol. 2012:23–45. doi: 10.1007/978-3-642-24716-3_2. [DOI] [PubMed] [Google Scholar]
  • 90.Morales-Medina JC, Dumont Y, Quirion R. A possible role of neuropeptide Y in depression and stress. Brain Res. 2010;1314:194–205. doi: 10.1016/j.brainres.2009.09.077. [DOI] [PubMed] [Google Scholar]
  • 91.Wei J, Chu C, Wang Y, et al. Association study of 45 candidate genes in nicotine dependence in Han Chinese. Addict Behav. 2012;37:622–626. doi: 10.1016/j.addbeh.2012.01.009. [DOI] [PubMed] [Google Scholar]
  • 92.Okahisa Y, Ujike H, Kotaka T, et al. Association between neuropeptide Y gene and its receptor Y1 gene and methamphetamine dependence. Psychiatry Clin Neurosci. 2009;63:417–422. doi: 10.1111/j.1440-1819.2009.01961.x. [DOI] [PubMed] [Google Scholar]
  • 93.Zhou Y, Danbolt NC. GABA and glutamate transporters in brain. Front Endocrinol. 2013;4:165. doi: 10.3389/fendo.2013.00165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Thoeringer CK, Ripke S, Unschuld PG, et al. The GABA transporter 1 (SLC6A1): a novel candidate gene for anxiety disorders. J Neural Transm. 2009;116:649–657. doi: 10.1007/s00702-008-0075-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Ebejer JL, Duffy DL, van der Werf J, et al. Genome-wide association study of inattention and hyperactivity-impulsivity measured as quantitative traits. Twin Res Hum Genet. 2013;16:560–574. doi: 10.1017/thg.2013.12. [DOI] [PubMed] [Google Scholar]
  • 96.Sikora M, Gese A, Czypicki R, et al. Correlations between polymorphisms in genes coding elements of dopaminergic pathways and body mass index in overweight and obese women. Endokrynol Pol. 2013;64:101–107. [PubMed] [Google Scholar]
  • 97.Foley TE, Fleshner M. Neuroplasticity of dopamine circuits after exercise: implications for central fatigue. Neuromolecular Med. 2008;10:67–80. doi: 10.1007/s12017-008-8032-3. [DOI] [PubMed] [Google Scholar]
  • 98.Althaus M, Groen Y, Wijers AA, et al. Variants of the SLC6A3 (DAT1) polymorphism affect performance monitoring-related cortical evoked potentials that are associated with ADHD. Biol Psychol. 2010;85:19–32. doi: 10.1016/j.biopsycho.2010.04.007. [DOI] [PubMed] [Google Scholar]
  • 99.Yang B, Chan RC, Jing J, et al. A meta-analysis of association studies between the 10-repeat allele of a VNTR polymorphism in the 3′-UTR of dopamine transporter gene and attention deficit hyperactivity disorder. Am J Med Genet B Neuropsychiatr Genet. 2007;144B:541–550. doi: 10.1002/ajmg.b.30453. [DOI] [PubMed] [Google Scholar]
  • 100.Lim J, Ebstein R, Tse CY, et al. Dopaminergic polymorphisms associated with time-on-task declines and fatigue in the Psychomotor Vigilance Test. PLoS One. 2012;7:e33767. doi: 10.1371/journal.pone.0033767. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Iqbal R, Mughal MS, Arshad N, Arshad M. Pathophysiology and antioxidant status of patients with fibromyalgia. Rheumatol Int. 2011;31:149–152. doi: 10.1007/s00296-010-1470-x. [DOI] [PubMed] [Google Scholar]
  • 102.Lieberman HR, Kellogg MD, Kramer FM, Bathalon GP, Lesher LL. Lipid and other plasma markers are associated with anxiety, depression, and fatigue. Health Psychol. 2012;31:210–216. doi: 10.1037/a0026499. [DOI] [PubMed] [Google Scholar]
  • 103.Yu Y, Pan Y, Jin M, et al. Association of genetic variants in tachykinins pathway genes with colorectal cancer risk. Int J Colorectal Dis. 2012;27:1429–1436. doi: 10.1007/s00384-012-1478-7. [DOI] [PubMed] [Google Scholar]
  • 104.Cunningham S, O’Doherty C, Patterson C, et al. The neuropeptide genes TAC1, TAC3, TAC4, VIP and PACAP(ADCYAP1), and susceptibility to multiple sclerosis. J Neuroimmunol. 2007;183:208–213. doi: 10.1016/j.jneuroim.2006.11.002. [DOI] [PubMed] [Google Scholar]
  • 105.Colasanti M, Suzuki H. The dual personality of NO. Trends Pharmacol Sci. 2000;21:249–252. doi: 10.1016/s0165-6147(00)01499-1. [DOI] [PubMed] [Google Scholar]
  • 106.Pall ML. Elevated, sustained peroxynitrite levels as the cause of chronic fatigue syndrome. Med Hypotheses. 2000;54:115–125. doi: 10.1054/mehy.1998.0825. [DOI] [PubMed] [Google Scholar]
  • 107.Angelini C, Tasca E. Fatigue in muscular dystrophies. Neuromuscul Disord. 2012;22(Suppl 3):S214–220. doi: 10.1016/j.nmd.2012.10.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Heydemann A, McNally E. NO more muscle fatigue. J Clin Invest. 2009;119:448–450. doi: 10.1172/JCI38618. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Pall ML. Post-radiation syndrome as a NO/ONOO- cycle, chronic fatigue syndrome-like disease. Med Hypotheses. 2008;71:537–541. doi: 10.1016/j.mehy.2008.05.023. [DOI] [PubMed] [Google Scholar]
  • 110.Galecki P, Maes M, Florkowski A, et al. Association between inducible and neuronal nitric oxide synthase polymorphisms and recurrent depressive disorder. J Affect Disord. 2011;129:175–182. doi: 10.1016/j.jad.2010.09.005. [DOI] [PubMed] [Google Scholar]
  • 111.Kurrikoff T, Lesch KP, Kiive E, et al. Association of a functional variant of the nitric oxide synthase 1 gene with personality, anxiety, and depressiveness. Dev Psychopathol. 2012;24:1225–1235. doi: 10.1017/S0954579412000661. [DOI] [PubMed] [Google Scholar]
  • 112.Hancock DB, Martin ER, Vance JM, Scott WK. Nitric oxide synthase genes and their interactions with environmental factors in Parkinson’s disease. Neurogenetics. 2008;9:249–262. doi: 10.1007/s10048-008-0137-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Beal MF. Excitotoxicity and nitric oxide in Parkinson’s disease pathogenesis. Ann Neurol. 1998;44:S110–114. doi: 10.1002/ana.410440716. [DOI] [PubMed] [Google Scholar]
  • 114.Wen HB, Zhang ZX, Wang H, et al. Epidemiology and clinical phenomenology for Parkinson’s disease with pain and fatigue. Parkinsonism Relat Disord. 2012;18(Suppl 1):S222–225. doi: 10.1016/S1353-8020(11)70068-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Robertson D, Flattem N, Tellioglu T, et al. Familial orthostatic tachycardia due to norepinephrine transporter deficiency. Ann N Y Acad Sci. 2001;940:527–543. doi: 10.1111/j.1749-6632.2001.tb03703.x. [DOI] [PubMed] [Google Scholar]
  • 116.Morriss RK, Robson MJ, Deakin JF. Neuropsychological performance and noradrenaline function in chronic fatigue syndrome under conditions of high arousal. Psychopharmacology. 2002;163:166–173. doi: 10.1007/s00213-002-1129-8. [DOI] [PubMed] [Google Scholar]
  • 117.Shannon JR, Flattem NL, Jordan J, et al. Orthostatic intolerance and tachycardia associated with norepinephrine-transporter deficiency. N Engl J Med. 2000;342:541–549. doi: 10.1056/NEJM200002243420803. [DOI] [PubMed] [Google Scholar]
  • 118.Ryu SH, Lee SH, Lee HJ, et al. Association between norepinephrine transporter gene polymorphism and major depression. Neuropsychobiology. 2004;49:174–177. doi: 10.1159/000077361. [DOI] [PubMed] [Google Scholar]
  • 119.Ryan JL, Carroll JK, Ryan EP, et al. Mechanisms of cancer-related fatigue. Oncologist. 2007;12(Suppl 1):22–34. doi: 10.1634/theoncologist.12-S1-22. [DOI] [PubMed] [Google Scholar]
  • 120.Alexander S, Stone P, White S, et al. Evaluation of central serotonin sensitivity in breast cancer survivors with cancer-related fatigue syndrome. J Pain Symptom Manage. 2010;40:892–898. doi: 10.1016/j.jpainsymman.2010.03.023. [DOI] [PubMed] [Google Scholar]
  • 121.Yamamoto S, Ouchi Y, Onoe H, et al. Reduction of serotonin transporters of patients with chronic fatigue syndrome. Neuroreport. 2004;15:2571–2574. doi: 10.1097/00001756-200412030-00002. [DOI] [PubMed] [Google Scholar]
  • 122.Narita M, Nishigami N, Narita N, et al. Association between serotonin transporter gene polymorphism and chronic fatigue syndrome. Biochem Biophys Res Commun. 2003;311:264–266. doi: 10.1016/j.bbrc.2003.09.207. [DOI] [PubMed] [Google Scholar]
  • 123.Lei X, Chen C, He Q, et al. Sex determines which section of the SLC6A4 gene is linked to obsessive-compulsive symptoms in normal Chinese college students. J Psychiatr Res. 2012;46:1153–1560. doi: 10.1016/j.jpsychires.2012.05.002. [DOI] [PubMed] [Google Scholar]
  • 124.Mostowska A, Hozyasz KK, Wojcicka K, Lianeri M, Jagodzinski PP. Polymorphisms of stress-related genes and the risk of nonsyndromic cleft lip with or without cleft palate. Birth Defects Res A Clin Mol Teratol. 2011;91:948–955. doi: 10.1002/bdra.20843. [DOI] [PubMed] [Google Scholar]
  • 125.Baldinger P, Kraus C, Rami-Mark C, et al. Interaction between 5-HTTLPR and 5-HT1B genotype status enhances cerebral 5-HT1A receptor binding. Neuroimage. 2015;111:505–512. doi: 10.1016/j.neuroimage.2015.01.049. [DOI] [PubMed] [Google Scholar]
  • 126.Lothe A, Boni C, Costes N, et al. 5-HT1A gene promoter polymorphism and [18F]MPPF binding potential in healthy subjects: a PET study. Behav Brain Funct. 2010;6:37. doi: 10.1186/1744-9081-6-37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Caspi A, Hariri AR, Holmes A, Uher R, Moffitt TE. Genetic sensitivity to the environment: the case of the serotonin transporter gene and its implications for studying complex diseases and traits. Am J Psychiatry. 2010;167:509–527. doi: 10.1176/appi.ajp.2010.09101452. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Caspi A, Sugden K, Moffitt TE, et al. Influence of life stress on depression: moderation by a polymorphism in the 5-HTT gene. Science. 2003;301:386–389. doi: 10.1126/science.1083968. [DOI] [PubMed] [Google Scholar]
  • 129.Wright F, D’Eramo Melkus G, Hammer M, et al. Trajectories of evening fatigue in oncology outpatients receiving chemotherapy. J Pain Symptom Manage. 2015;50:163–175. doi: 10.1016/j.jpainsymman.2015.02.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Wright F, D’Eramo Melkus G, Hammer M, et al. Predictors and trajectories of morning fatigue are distinct from evening fatigue. J Pain Symptom Manage. 2015;50:176–189. doi: 10.1016/j.jpainsymman.2015.02.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Lerdal A, Kottorp A, Gay CL, Lee KA. Lee fatigue and energy scales: Exploring aspects of validity in a sample of women with HIV using an application of a Rasch model. Psychiatry Res. 2013;205:241–246. doi: 10.1016/j.psychres.2012.08.031. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

RESOURCES