Impact of obesity and overweight on DNA stability: Few facts and many hypotheses

Impact of obesity and overweight on DNA stability: Few facts and many hypotheses

Accepted Manuscript Title: Impact of obesity and overweight on DNA stability: Few facts and many hypotheses Authors: Tahereh Setayesh, Armen Nersesyan...

3MB Sizes 0 Downloads 20 Views

Accepted Manuscript Title: Impact of obesity and overweight on DNA stability: Few facts and many hypotheses Authors: Tahereh Setayesh, Armen Nersesyan, Miroslav Miˇs´ık, Franziska Ferk, Sabine Langie, Vanessa M. Andrade, Alexander Haslberger, Siegfried Knasmuller ¨ PII: DOI: Reference:

S1383-5742(17)30096-0 https://doi.org/10.1016/j.mrrev.2018.07.001 MUTREV 8246

To appear in:

Mutation Research

Received date: Revised date: Accepted date:

27-10-2017 3-5-2018 6-7-2018

Please cite this article as: Setayesh T, Nersesyan A, Miˇs´ık M, Ferk F, Langie S, Andrade VM, Haslberger A, Knasmuller ¨ S, Impact of obesity and overweight on DNA stability: Few facts and many hypotheses, Mutation Research-Reviews in Mutation Research (2018), https://doi.org/10.1016/j.mrrev.2018.07.001 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 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.

Impact of obesity and overweight on DNA stability:

IP T

few facts and many hypotheses

Tahereh Setayesh1, Armen Nersesyan1, Miroslav Mišík1, Franziska Ferk1, Sabine Langie2,

1.

SC R

Vanessa M. Andrade3, Alexander Haslberger4, Siegfried Knasmüller1*

Institute of Cancer Research, Department of Medicine I, Medical University of Vienna,

Environmental Risk and Health Unit, Flemish Institute for Technological Research

N

2.

U

Vienna, Austria

Laboratório de Biologia Celulare Molecular, Programa de Pós-Graduação em Ciências da

M

3.

A

(VITO), Mol, Belgium

Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul

Department of Nutritional Sciences, University Vienna, Vienna, Austria

CC E

PT

4.

ED

Catarinense (UNESC), Brazil

* Corresponding author: Siegfried Knasmüller, Institute of Cancer Research, Department

A

of Medicine I, Medical University of Vienna, Vienna, Austria, Tel: +43 1 4016057562; Fax: +43 1 40160957500; E-mail: [email protected]

1

Abstract: Health authorities are alarmed worldwide about the increase of obesity and overweight in the last decades which lead to adverse health effects including inflammation, cancer, accelerated aging and infertility. We evaluated the state of knowledge concerning the impact of elevated body mass on genomic instability. Results of investigations with humans (39 studies) in which

IP T

DNA damage was monitored in lymphocytes and sperm cells, are conflicting and probably as

a consequence of heterogeneous study designs and confounding factors (e.g. uncontrolled

SC R

intake of vitamins and minerals and consumption of different food types). Results of animal studies with defined diets (23 studies) are more consistent and show that excess body fat

U

causes DNA damage in multiple organs including brain, liver, colon and testes. Different

N

molecular mechanisms may cause genetic instability in overweight/obese individuals. ROS

A

formation and lipid peroxidation were found in several investigations and may be caused by

M

increased insulin, fatty acid and glucose levels or indirectly via inflammation. Also reduced DNA repair and formation of advanced glycation end products may play a role but more data

ED

are required to draw firm conclusions. Reduction of telomere lengths and hormonal imbalances are characteristic for overweight/obesity but the former effects are delayed and

PT

moderate and hormonal effects were not investigated in regard to genomic instability in obese

CC E

individuals. Increased BMI values affect also the activities of drug metabolizing enzymes which activate/detoxify genotoxic carcinogens, but no studies concerning the impact of these alterations of DNA damage in obese individuals are available. Overall, the knowledge

A

concerning the impact of increased body weight and DNA damage is poor and further research is warranted to shed light on this important issue. Abbreviations: AGEs, advanced glycation end products; AP-1, activating protein -1; APE1, apurinic/apyrimidinic endonuclease 1; BER, base excision repair; BMI, body mass index; CAF, cafeteria diet; CAT, Catalase; CBMN-Cyt, cytokinesis block micronucleus cytome assay; CEL, carboxyl ester lipase; CML, Nε-carboxymethyllysine; CRP-1, C-reactive protein; DDR, DNA damage 2

response; DFI, DNA fragmentation index; DMBA, 7,12-dimethylbenz(α)anthracene; DNA, deoxyribonucleic acid; DNMTs, DNA methyltransferases; DSBs, double strand breaks; EGCG, Epigallocatechin gallate; ELISA, enzyme linked immunosorbent assay; ENDO III, endonuclease III; ER, estrogen receptor; FA, fatty acid; FACS, fluorescence-activated cell sorting; FPG, formamidopyrimidine DNA glycosylase; GPx, glutathione peroxidase; GSH/GSSG, glutathione/ oxidised glutathione disulphide; GSK-3β, glycogen synthase kinase-3β; GST, glutathione-Stransferase; HFD, high fat diet; Hras, harvey rat sarcoma virus oncogene 1; HPLC, high-performance

IP T

liquid chromatography; 5hmU, 5-hydroxymethyluracil;4-HNE, 4-hydroxy-2-nonenal; HO-1, haem oxygenase-1; HUVEC, human umbilical vein endothelial cells; IGF, insulin-like growth factor; IL-6,

interleukin-6; IR, insulin receptor; IRAC, international agency for research cancer; JNK, Jun N-

SC R

terminal kinase; LC-MC, liquid chromatography-mass spectrometry; LP, lipid peroxidation; MAPK,

mitogen activated protein kinase; MDA, malondialdehyde; MetS, metabolic syndrome; MLH-1, MutL homolog 1; MMR, mismatch repair; MN, micronuclei; NAT, N-acetyltransferase; NBs, nuclear bridges; Nbuds, nuclear buds; NER, nucleotide excision repair; NF-kB, nuclear factor kappa-light-

U

chain-enhancer of activated B cells; NOX1, NADPH oxidase 1; 8-OHdG, 8-hydroxy-2'– deoxyguanosine; 8-oxodG, 8-oxo-7,8-dihydro-2' –deoxyguanosine; 8-OH-Gua, 8-OH-guanosine; 8-

N

oxoGuo, 8-oxo-7,8-dihydroguanosine; 16α-OHE1, 16α-hydroxyestrone; PAHs, polycyclic aromatic

A

hydrocarbons; PCNA, proliferating cell nuclear antigen; PDK1, phosphatidylinositol-4,5-

M

bisphosphate 3-kinase; PI3K, phosphatidylinositol-3-kinase; PKC, proteinkinase C; PTEN, phosphatase and tensin homolog; RAGE, receptor advanced glycation end products; ROS, reactive oxygen species; SAM, S-adenosyl-L-methionine; SCGE, single cell gel electrophoresis assay; SDH,

ED

sorbitol dehydrogenase; SOD, superoxide dismutases; SSBs, single strand breaks; SCSA, sperm chromatin structure assay; TBARS, thiobarbituric reactive substances; T2D, type 2 diabetes; TdT,

PT

terminal deoxynucleotidyl transferase; TGF-β, transforming growth factor-β; TNF-α, tumor necrosis factor; TP53, tumor suppressor gene p53; TUNEL, terminal deoxynucleotidyl transferase dUTP

CC E

nick end labelling UGT, uridine diphosphate glucuronosyltransferase; YAP, yes-associated protein

A

Keywords: overweight; obesity; DNA damage; genomic instability; somatic cells; germ cells

Contents 1.

Introduction ................................................................................................................... 4

2.

Data collection .............................................................................................................. 7 3

3.

Methods used in investigations concerning associations between genetic instability and overweight/obesity ................................................................................................ 8

4.

Human studies ............................................................................................................... 9 4.1. Studies with overweight/obese individuals ............................................................... 9 4.2. Studies with MetS patients ...................................................................................... 11 4.3. Impact of body weight on DNA damage in human sperm cells .............................. 12 4.4. Comprehensive evaluation of human studies .......................................................... 13 Results of animal studies ............................................................................................ 15

IP T

5.

5.1. DNA damage in somatic cells ................................................................................. 15

SC R

5.2. DNA damage in germ cells...................................................................................... 17 5.3. Comprehensive evaluation of animal studies .......................................................... 18 Impact of obesity/overweight on mutations in oncogenes .......................................... 18

7.

Results of in vitro studies............................................................................................ 19

8.

Molecular mechanisms ............................................................................................... 21

U

6.

N

8.1. Direct formation of ROS ......................................................................................... 22 8.2. Obesity induced lipid peroxidation and DNA damage ............................................ 25

A

8.3. Impact of overweight on sex hormones ................................................................... 26

M

8.4. Glycation end products ............................................................................................ 29 8.5. Impact of obesity on DNA repair ............................................................................ 30

ED

8.6. Obesity and telomere lengths................................................................................... 33 8.7. Impact of obesity/overweight on the activities of enzymes that activate and detoxify mutagens .................................................................................................... 35

9.

PT

8.8. Inflammation and DNA damage .............................................................................. 37 Consequences of obesity induced DNA damage ........................................................ 38

CC E

10. Conclusions and knowledge gaps ............................................................................... 40 Conflict of interest statement ............................................................................................. 43

Acknowledgments.................................................................................................................. 43

A

References .............................................................................................................................. 44

1.

Introduction Obesity and overweight are characterized by abnormal accumulation of body mass and

are associated with increased health risks, in particular with diseases such as type 2 diabetes 4

(T2D), cardiovascular diseases, polycystic ovary syndrome, infertility and cancer [1]. Scientists and health authorities are alarmed by the fact that the increase of overweight/obesity is a worldwide phenomenon. A global estimate indicates that about 640 million adults were obese in 2014, this number corresponds to a 6-fold increase since 1975 [2]. Obesity has a prevalence of 10.8 % among men and 14.9 % among women [3]. The worldwide incidence

IP T

of childhood overweight and obesity increased from 4.2% in 1990 to 6.7% in 2010. This trend is expected to reach 9.1% or 60 million, in 2020 [4].

SC R

A typical feature of excess body weight is the formation of ROS and of cytokines which are characteristic for inflammation. These processes lead to damage of the genetic material

U

which in turn plays a key role in the induction of cancer and infertility; these disorders are

N

increased in overweight/obese humans [5, 6]. Aim of this article is to describe the current

A

state of knowledge of the relation between excess body weight and genomic instability.

M

Recently, several studies have been published which concern the association between MetS and DNA damage [7-9], however they are based on mechanistic considerations but not on a

ED

systematic evaluation of experimental data. The first part of the present paper focuses on the description of results of animal and human studies in which different parameters of DNA

PT

instability and chromosomal damage were measured in overweight/obese individuals in somatic and germ cells. Furthermore we describe also measurements which were performed

CC E

in these studies concerning biological processes that lead to DNA stability such as inhibition of DNA repair processes, decrease of the telomere lengths, changes of the redox status and of

A

the activities of antioxidant enzymes, lipid peroxidation and of the immune status. The association between overweight/obesity and cancer is well documented; it is, apart

from cardiovascular diseases, the most important health consequence of excess body weight. In 2016, the IARC published a report which is based on the evaluation of more than 1000 individual epidemiological studies. The Agency comes to the conclusion that the absence of 5

excess body fatness lower the risks of most cancer [2]. A comprehensive study from the US states that 14% of all cancer cases in man and 20% in women are due to obesity [10]. Numerous investigations showed that gene mutations as well as chromosomal aberrations play a key role in malignant transformation [11, 12]. Nevertheless, it is apparent

IP T

that most studies which concerned the mechanisms by which excess body weight leads to cancer focused on other modes of action such as alterations of signaling pathways, increased

production of growth factors and hormones and activation of transcription factors (for reviews

SC R

see [13-15]).

In the last decades, a variety of methods was developed which enable the investigation

U

of DNA stability in human and animal models [16]. These approaches were frequently used

N

to study occupational, lifestyle and dietary factors [16-20]. Furthermore, in vitro assays [21]

A

and rodent mutagenicity tests [22, 23] are conducted worldwide for routine monitoring of

M

chemicals. These models can be also used to identify drugs and dietary components which protect humans against DNA damage and its consequences [16, 24, 25] .

ED

This review describes human and animal studies which concern the impact of overweight/obesity on DNA integrity. The results of these investigations may contribute to a

PT

better understanding of the mechanisms by which excess body weight causes adverse health

CC E

consequences such as cancer and infertility and can also contribute to the development of strategies to prevent these effects. We included also investigations concerning the MetS, which is the most important consequence of abdominal obesity. Studies concerning T2D and

A

the polycystic ovary syndrome were excluded; these disorders are related to obesity but are characterized by severe additional pathological changes, which may have an impact on DNA stability but are not attributable to excess body weight [26, 27]. The following chapter defines the search strategy (chapter 2) and the methods which were used (chapter 3). The subsequent paragraphs describe investigations with humans 6

(chapter 4) and animals (chapter 5), the impact of obesity on mutations in oncogenes (chapter 6) and in vitro studies (chapter 7). Furthermore we describe the molecular mechanisms by which overweight/obesity may cause DNA damage (chapter 8), the consequence of obesity induced DNA damage and the links between DNA damage and cancer (chapter 9) and define

IP T

knowledge gaps and future research strategies (chapter 10).

2. Data collection

Library

of

Medicine,

SC R

The literature search was performed by using the MedLine/PubMed data-base (National http://www.ncbi.nlm.nih.gov/PubMed),

SCOPUS

(Elsevier,

U

http://www.scopus.com), Thomson ISI’s Web of Science (Thomson Reuters Corporation,

N

http://apps.webofknowl-edge.com), Google Scholar (http://www.google.scholar.-com) and

A

covered the period between December 1961 and August 2017.

M

Additionally, a manual search of the reference list of studies and of reviews was conducted. References of retrieved articles were analysed to identify further publications

ED

which may have been missed. Only articles in English were considered. The keywords and combinations of search terms were: body mass index, chromosomal aberrations,

PT

chromosomal damage, comet assay, DNA damage, DNA adducts, DNA repair, DNA

CC E

instability, γ-H2AX, genomic instability, metabolic syndrome, micronuclei, mutation, obesity, 8-OHdG, 8-oxodG; overweight, single cell gel electrophoresis (SCGE), sister chromatid exchange, sperm DNA integrity, telomere length and TUNEL assay.

A

The quality of the individual studies was assessed by a quality score (QS) system, which

is similar to a model which was used for MN- studies with occupationally exposed humans [28]. A detailed description of this system is described in the section “supplementary information”. According to the criteria, the studies were classified and grouped in different

7

categories (Low-, Medium- and High- quality). The score and the categories are indicated in the supplementary tables S1 and S2. Exclusion criteria for human studies were as follows (i) an insufficient number of participants (less than 20 individuals), (ii) lack of information concerning the number of

IP T

individuals per group, (iii) lack of information concerning the number of scored cell, (iv) an insufficient number of evaluated cells (<1000 per experimental point in MN studies, in comet

assays <100 cells per experimental point, in TUNEL tests <200 and in SCSA experiments

SC R

<5000 cells per experimental point respectively and (v) small differences in the BMI-values of different study group (≤0.5%). Exclusion criteria for animal studies were (i) an insufficient

U

number of animals per group (less than 3) and (ii) an inadequate number of scored cells

N

(<1000 per experimental point in MN studies with bone marrow and lymphocytes, in comet

A

assays <100 cells per experimental point, in TUNEL tests <200 and in SCSA experiments

Methods used in investigations concerning associations between genetic instability

PT

3.

ED

addition of the individual scores).

M

<5000 cells per experimental point respectively The overall QS values were determined by

CC E

and overweight/obesity

Figure 1 gives an overview of the methods which are used in studies with overweight/

A

obese humans and laboratory animals.

Figure 1

Table. 1 describes different methods which were used to study impact of overweight/obesity on DNA stability. Most of these approaches are also used to investigate the effect of 8

chemicals, lifestyle factors and chemical exposure in somatic cells, for studies with sperm cells additional test systems such as TUNEL assay and SCS assays were employed.

IP T

The most frequently method in human studies are comet assay experiments with and without restriction enzymes (in total 12 studies were published) followed by measurement of

SC R

8-OHdG (7 studies), MN (8 studies), only two publications concern γ-H2X measurements.

Also in animal studies, comet assays are dominating (8 investigation), followed by 8-

Human studies

A

4.

N

U

OHdG or 8-oxodG (4 studies) and by MN and γ-H2X (3 studies per endpoints).

M

The following chapters describe results which were obtained with somatic cells of healthy individuals (chapter 4.1) and MetS patients (chapter 4.2), chapter 4.3 summarizes the

PT

human studies.

ED

results with sperm cells and the last paragraph contains a comprehensive evaluation of the

4.1. Studies with overweight/obese individuals

CC E

Only relatively few studies (in total 22) were realized which focused on the impact of

A

body weight on genomic instability in somatic cells (Table. 2 to 5).

In five out of six comet assay trials with parallel design, increased DNA migration was

seen in lymphocytes of overweight/obese individuals [41-45]. In MN experiments lymphocytes, positive effects were found only in two of five studies with parallel design [47, 50]. Other markers of genomic instability (Nbuds and NBs) which were additionally scored

9

in a MN study by Donmez-Altuntas et al. [47] were also increased while no association was found between MN induction and formation of oxidised guanine (8-OHdG) levels in plasma [47]. Another endpoint which was elevated in the study by Scarpato et al. was γ-H2AX [50] Two studies were conducted with buccal cells. Unexpectedly the authors found in Malaysian farmers, which were exposed to pesticides and fertilizers, higher MN-rates in

IP T

individuals with normal BMI compared to participants with higher body weights [51]. A second trial was realized with school children in Mexico and no differences were seen

SC R

between normal, obese and overweight participants [52]. In the latter study a Giemsa based

stain was used which may leads to false positive results [60] and in both trials [51, 52] only

U

1000 instead of 2000 cells were scored per participants which is not in agreement with the

N

standard protocol [61].

A

Table 2. describes also the results of several intervention trials. Clear reduction of DNA

M

damage was seen in a study with calorie restriction [38]. No clear effect was seen in an intervention trial in which comparisons were made between consumption of red meat and a

ED

high carbohydrate diet [46]. A clear difference of the rate of Nbuds was observed at the end of this study but no differences between meat and carbohydrate consumption were detected.

PT

It is mentioned that the weight loss at the end of intervention was significant but the extent is not specified [46]. The most recent study was conducted with bariatric surgery patients

CC E

(n=56); the authors found no alteration of the comet formation in lymphocytes after 6 months after the surgery, but reduced DNA damage was observed after 1year [39].

A

Table 5. concerns findings from experiments in which phosphorylation of H2X (which

is indicative for DSBs) was monitored. Seven studies concerned formation of oxidised guanosin in plasma, whole blood or urine. In three trials a positive association between base oxidation and overweight was found [54, 56, 57], three studies reported an inverse relation [47, 53, 55]. No effect was seen in an Italian study; but clear evidence for formation of FPG

10

sensitive sites was detected in the same trial [41]. Two γ-H2AX studies were performed with lymphocytes and significant correlations were found in both trials [50, 58]. The last line of Table 5. summarizes the results of a study concerning associations between telomere lengths and damage of DNA bases before and after calorie restriction. The authors found increased telomere lengths and in parallel reduced formation of abasic sites

IP T

which were monitored with a commercial test kit in rectal biopsy samples. This assay was rarely used and not standardized [59].

SC R

It is notable, that changes which were seen in intervention trials are not necessarily due to reduction of the BMI but may be caused by other factors, i.e. by alterations of the intake of

N

U

vitamins and macronutrients which have an impact on genomic instability [62].

A

4.2. Studies with MetS patients

M

Table 6. summarizes the results of currently available studies. Five investigations concerned comparisons between overweight MetS patients and healthy individuals. Karaman

ED

et al. [63] and Demiberg et al. [64] found evidence for increased comet formation in lymphocytes with higher BMI values. Furthermore, also evidence for increased MN

PT

frequencies and higher levels of malondialdehyde (MDA) and decreased activities of

CC E

antioxidant enzymes was found in one of the studies [63]. On the contrary, no elevated comet formation was seen in a trial which was realized in Croatia [65]; the authors hypothesized that the lack of an effect may be due to the fact that the study was performed with young

A

participants who had a high repair capacity. The most recent study concerned DNA damage in morbidly obese women [45]. The

authors found no evidence for increased DNA damage in lymphocytes, also oxidised purines (FPG-sensitive lesions) were not increased. Interestingly, the number of individuals with gene

11

polymorphism in the fat mass and obesity associated gene (AA-FTO) was higher in the obese group, while another polymorphism in the same gene (TT-FTO) was found less frequently. Results of 8-OHdG measurements were performed in two MetS studies. Abdilla and co-workers [66] monitored several endpoints in different MetS groups and in healthy controls and detected no differences while Milić et al. [65] found even lower levels of 8-OHdG in the

IP T

urine of patients compared to the controls; they hypothesized that this unexpected result may

SC R

be due an adaptive response.

4.3. Impact of body weight on DNA damage in human sperm cells

The findings of different studies are listed in Table 7. The results of comet experiments

U

are controversial. No effects were seen in a trial in which the neutral version (which detects

N

mainly DSBs and cross-links) was used [67], while clear differences were found in a study

A

from Brazil with the standard protocol i.e. under alkaline conditions which detect DSBs, SSBs

M

and apurinic sites [68].

ED

The results of TUNEL assays indicate that differences exist between obese and normal weight individuals while no effects were found between overweight and lean participants [69,

PT

70]. Vignera et al. [69] analysed additional parameters and found a higher rate of sperm cells

CC E

with increased chromatin compactness in the former group. The authors postulated that this is a sign of early uncontrolled apoptosis. This assumption was supported by the observation of increased externalization of phosphatidylserine and reduced mitochondrial membrane

A

potential as a consequence of elevated body weight. These processes lead to programmed cell death and DNA fragmentation [78]. Interestingly, an increase of the DFI by 50% was observed in obese but not in overweight individuals in a large study (n=330) by Dupont et al. [70].

12

The results of 7 studies in which the effects of body weight were analysed in SCSA assays are controversial. The findings of Bandel et al. [79] are unexpected as the authors found an inverse association between BMI values and sperm DNA integrity, also the motility of the cells increased with the body weight. No such effects were found in several other trials [71, 73-76].

IP T

Two further studies are notable; one is an intervention trial in which the DFI of the

participants was compared after a weight loss period of 14 weeks in three groups. Several

SC R

sperm parameters were improved after weight reduction, also the DFI values were lower at

the end of the trial. The strongest reduction of this parameter (from 18 to 10) was found in the

U

group with the highest BMI values (46.1-60.9), however the effect did not reach significance

N

[71].

A

An interesting study from Denmark concerned the impact of the BMI of mothers on the

M

DFI in sperm of the sons. Higher values were found in the offspring of overweight mothers compared to mothers with normal weight [72].

ED

4.4. Comprehensive evaluation of human studies

PT

The main methods which were used in human studies are MN assays and the comet experiments. It is notable that MN reflect persisting chromosomal aberration, while “comets”

CC E

are caused by SSBs and DSBs and apurinic sites and may disappear as a consequence of repair processes. For MN studies a standard protocol is available and MN can be easily identified [80]. As described above, MN experiments yielded conflicting findings, only 3 out of 8

A

investigations found positive correlations with the BMI. In comet experiments different parameters (tail intensity (synonymous with % DNA in tail), tail moment and olive tail moment) can be measured by automated scoring systems while other authors use “manual scoring” and determine DNA migration by definition of arbitrary units. All these parameters were found acceptable, however it was emphasised by various expert groups that tail intensity 13

(%DNA in tail) is the most reliable one [23, 81, 82]. While studies which used this marker yielded strongly controversial results (6 negative, 9 positive) consistently positive findings were obtained in all manual scoring experiments which may be indicative for a biased evaluation.

IP T

Also the results of studies which concerned the induction of DNA damage in sperm are conflicting. It is difficult to assess which endpoints are the most reliable ones (SCSA, TUNEL assay and comet assay). It was emphasized that the different techniques reflect different

SC R

aspects i.e. the TUNEL assay and the comet assay are indicative for “real” damage while the

SCSA reflects the susceptibility of the DNA to denaturation [83]. Only for the later assay a

U

standardized protocol is available [84].

N

In the case of experiments which concern the quantification of the oxidation of DNA

A

bases not only differences of antioxidants in the diet but also methodological aspects may

M

play a role. It was emphasize that HPLC measurements are more reliable as compared to

ED

ELISA based measurements [85]. Furthermore, 8-oxodG and oxoGuo were monitored either in urine, plasma or in different target organs which may add to heterogeneity of the results.

PT

The calculation of the QS shows that the quality of the most studies is in a relatively

CC E

narrow range, therefore it is not possible to explain the conflicting results by shortcomings of the study design of the individual studies. As described in more detail in chapter 10 none of the investigations which were published so far was controlled in regard to the mineral and

A

vitamin intake of the participants which may have a strong impact on the results. It is conceivable that the composition of the foods in lean and overweight human has a strong impact on the outcome of the studies, also weight loss programs are based on substantial changes of the dietary patents.

14

5. Results of animal studies 5.1. DNA damage in somatic cells Different models were used in experiments with laboratory rodents. In most trials, the

IP T

animals were fed with a high fat diet (HFD) or a cafeteria diet (CAF) for several weeks which

lead to an increase of the body fat and also to excess body weight. These studies were mainly

SC R

conducted with C57BL/6j mice; these animals develop symptoms which are characteristic for

obese humans namely hyperinsulinemia, hyperglycemia, and hypertension [86]. In some

U

studies Zucker rats were used; obesity is in these animals an autosomal recessive trait and

N

resembles the early onset of human obesity [87] i.e. the animals have hyperlipidaemia,

A

hyperinsulinemia and peripheral insulin resistance. In one study, WNIN/Ob rats were used

M

which are hyperglycaemic and insulin–resistant and reflect the metabolic syndrome in humans [88].

ED

The first paper which described chromosomal aberrations as a consequence of obesity was published by the Swedish scientist Karl Fredga [89] more than 50 years ago. He observed

PT

in a pilot study (2 animals per group) that a specific strain of hyperglycaemic mice had

CC E

aberrant numbers of chromosomes in epithelial cells from the cornea. Our literature search identified 18 further studies which concerned the effects of

overweight on DNA stability in somatic cells (Table 8. to 11.). Results of all comet trials

A

(n=8) indicate that excess body fat causes genomic instability in a variety of inner organs [88, 90-96]. Tenorio and co-workers [94] analysed DNA migration in different tissues of rats and found larger comets in liver and lymphocytes but not in kidneys, brain and heart. Also in a Brazilian study [96] clear evidence for increased DNA damage was seen in obese mice which were fed with a carbohydrate and fat rich cafeteria diet (consisting of various supermarket

15

products). The consumption of this chow induced glucose intolerance and DNA damage in lymphocytes, kidneys, liver and in the brain [96]. A positive result was also obtained in MN experiments with bone marrow cells in the same study [96]. Also in HFD fed mice induction of DNA–migration was detected in liver and colon in a number of studies [90-93]. Remely et al. [90, 91] found an effect in both organs; Setayesh et al. [92] detected additionally increased

IP T

damage in brain, liver and colons while no significant induction of DNA migration was

observed in blood cells and adipose tissue. Gutzkow et al. [93], analysed in a more recent

SC R

study the extent of damage in different cell types (liver, blood and testes) of HFD fed mice.

They found in standard comet assays only in the liver an effect; furthermore an increase of

U

formation of oxidised purines was detected in hepatocytes and also in white blood cells. Table

N

8. describes the results of a study with genetically modified rats, which are a model for the

A

MetS. Clear evidence for increased damage was observed in different regions of the brain.

M

The authors state that the effects which they observed in 3-months old obese rats are similar to those seen in 15-months old parental control animals and conclude that obesity leads to

ED

premature ageing [88].

PT

Also in γ-H2AX measurements which are indicative for DSBs, clear effects were seen in livers and colons of obese animals [101]. Furthermore, alterations of the urinary excretion

CC E

of 8-oxodG were monitored in the same trial. All these markers of DNA damage were decreased as a consequence of weight loss after gastric bypass surgery [101]. Another γ-

A

H2AX study was published with obese Zucker rats; the authors found clear induction of DSBs in the lungs of the animals when they conducted the measurements after intraperitoneal injection of glucose, which leads to hyperglycemia, but not in fasting animals [105]. A further γ-H2AX study concerned the relation between DSB induction in ovaries and alterations of DNA repair proteins [106]. Also in a study with obese rats in which 5-hydroxymethyluracil

16

was measured in the livers and in the mammary glands, a protective effect was detected after calorie restriction [107]. Most studies, which are available so far were carried out with nuclear DNA; only few investigations concerned on mitochondrial DNA [103, 104, 108]. In one of them, the authors measured damage in liver and skeletal muscles of mice and detected pronounced differences

in liver, heart, kidney and testes of HFD mice were detected [103].

IP T

between obese and normal weight animals [108]. In another study, higher levels of 8-oxodG

SC R

Table 10. describes also findings of a study in which obese and lean rats were treated

with the breast carcinogen 7,12-dimethylbenz(α)anthracene (DMBA). The authors found in

U

obese animals increased oxidative damage (8-OH-guanosine) in the liver and in parallel an

N

altered ratio of reduced to glutathione/oxidised glutathione disulphide (GSH/GSSG), which

A

is indicative for oxidative stress. This effect was was paralleled by changes of the overall

DNA damage in germ cells

ED

5.2.

M

DNA methylation status [104].

We found in total five animal studies which concerned the effects of obesity on sperm

PT

integrity. In four of them clear associations were observed, regardless if the damage was

CC E

quantified in comet assay, TUNEL assays or SCSA experiments [93, 109-112]. Only in one study, the authors did not find an increase of DNA damage in testes of HFD mice; it is notable that the differences of the body weights of the experimental groups were only marginal in this

A

study [93].

17

5.3.

Comprehensive evaluation of animal studies In contrast to the conflicting findings which were obtained in the human studies in

lymphocytes and sperm, consistent results were found in animal studies with rodents, i.e. most of the comet experiments which have been published so far with somatic and sperm cells yielded positive results. The reasons for the unequivocal results of experiments with rodents

IP T

are probably due the use of indicator organisms with a similar genetic background and feeding

SC R

of standardized diets.

In contrast to the findings of comet assay studies, several MN experiments with cells from the haematopoietic system failed to detect an association between DNA damage and

U

increased body weight. The reason for the discrepancy to the comet results may be differences

N

in the sensitivity of the tissues which were analyzed in comet assay and MN experiments and

A

not differences in the sensitivity of the endpoints. Sasaki et al. [114] stressed that 54 out of

M

165 proven rodent carcinogens were not detected in MN experiments with haematopoietic

ED

cells and that 49 of these chemicals (which gave false negative results) were positive in comet

Impact of obesity/overweight on mutations in oncogenes

CC E

6.

PT

assays in at least one or more organs in mice in comet experiments.

Only few studies concerned mutations in cancer associated genes of laboratory rodents

and humans. These investigations are important as they enable to establish direct causal links

A

between overweight induced genomic instability and malignant transformation. Shen et al. [115] found in whole genome sequencing experiments with tissue from

hepatocellular carcinomas of obese mice increased rates of mutations in the carboxyl ester lipase (Cel) gene and in the Harvey rat sarcoma virus oncogene 1 (Hras). In Cel, multiple mutations were detected which lead to functional loss, while in Hras single mutations were 18

found in a specific codon (61). Mutant Cel causes accumulation of cholesteryl ester in liver cells which leads to induction of endoplasmic reticulum stress and consequently activates IRE1 α/c-Jun N-terminal kinase (JNK)/c-Jun/activating protein -1 (AP-1) signalling cascade to promote liver cell growth. In the case of Hras, gain of function mutations promote liver cell growth through activation of mitogen-activated protein kinase (MAPK) and

IP T

phosphatidylinositol-4,5-bisphosphate 3-kinase(PI3K)/3-phosphoinositide-dependent protein kinase-1 (PDK1)/Akt pathways [116].

SC R

A study on Kras mutations in colon cancer found in colon tissue of obese and lean

patients differences in the mutation rates in several genes [117]. The most striking observation was a lower number of mutations in patients with high BMI. The authors hypothesised that

N

U

the reduced number found in “driver genes” of overweight individuals is due to the fact that mutations in genes which lead to malignant cells are not required in obese patients as in this

M

A

case “other alterations of signalling pathways” may substitute them [117]. The impact of overweight on tumor suppressor gene p53 mutations was analysed by

ED

Ecke et al. [118] in bladder cancer patients and no impact of the BMI on the mutation

Results of in vitro studies

CC E

7.

PT

frequencies was detected.

It is not possible to investigate biochemical changes which are caused by excessive

calorie intake and elevated body weight under in vitro conditions. Nevertheless, results of

A

studies were published with mammalian and human cells which reflect physiological conditions that are characteristic for obese individuals such as increased concentrations of glucose, fatty acids and insulin. These findings contribute to a better understanding of the mechanisms by which overweight/obesity lead to the damage of the genetic material.

19

Cavazos et al. [119] exposed human prostate cells to serum from obese and non-obese mice. They monitored induction of DSBs in γ-H2AX experiments and found increased damage in malignant and non-malignant cells with sera from overweight animals. This effect correlated with ROS formation; furthermore, several markers of aerobic glycolysis were induced. The authors found that a potent antioxidant (N-tert-butylhydroxylamine) prevents

involved in aerobic glycolysis) and formation of γ-H2AX foci [119].

IP T

induction of hypoxia-inducible factor 1-α (a transcriptional regulator of genes which is

SC R

The effects of insulin were investigated in several studies by Othman et al. [120-122]. They observed induction of DNA damage in colon derived cells (HT29) and also in

U

lymphocytes in MN and comet experiments and explained these effects by intracellular ROS

N

formation [120, 122]. Also with primary kidney cells, positive results were obtained.

A

Subsequent in vivo studies with rats confirmed the assumption that insulin causes ROS

M

formation [120, 121]. Results of a further experiment in which the authors investigated the mechanisms of insulin induced DNA damage are described in chapter 7.

ED

Several investigations concerned the impact of glucose, which is increased in the blood of diabetic patients, but also as a consequence of consumption of carbohydrate and sugar rich

PT

diets. Lorenzi et al. [123] studied the effects of glucose on induction of DNA damage with a fluorometric technique which detects the unwinding of DNA in endothelial cells from

CC E

umbilical veins and human fibroblasts already 30 years ago. They found positive results in the former cell type; furthermore they detected also increased hydroxyurea incorporation of

A

radio-labelled thymidine which is indicative for DNA repair. Quagliaro et al. [124] used human umbilical vein endothelial cells (HUVEC) and found evidence for formation of 8OHdG as well as increased concentrations of nitrotyrosine (an indicator of NO· formation) and apoptosis as a result of high glucose levels [124]. The authors postulated that the oxidative stress is caused by protein kinase C (PKC) dependent activation of NAD(P)H oxidase. This

20

mechanism was also proposed by a Japanese group which worked with aortic smooth muscle and endothelial cells [125]. The findings of the latter studies are in contrast to results of Shimoi et al. [126], they found in HUVEC induction of oxidised purines but failed to explain these effects by formation of ROS formation and postulated that this phenomenon is caused by glyoxal.

IP T

Increased levels of fatty acids (FA) are typical for obese and diabetic individuals.It was reported by Beeharry et al. [127] that palmitic acid (a major saturated FA in plasma) causes

SC R

comet formation in an insulin producing cell line and also in fibroblasts. This effect was

inhibited by linoleic acid and the authors postulated that the protection is due to prevention of

U

oxidative stress. The assumption of ROS formation from palmitate was confirmed by

N

Inoguchi et al. [125] who found in electron spin resonance spectroscopy experiments evidence

8.

Molecular mechanisms

M

A

for the involvement of PKC.

ED

The following paragraphs concern mechanisms which may play a role in the association between excess body weight and genomic instability (Figure 2). Apart from enzymatic

PT

processes which lead to formation of ROS also other modes of action could be involved. Lipid

CC E

peroxidation (LP) processes are induced by ROS and lead to formation of DNA reactive products. Furthermore, inhibition of DNA repair and induction of inflammation have been found in a number of studies. Also formation of advanced glycation products, hormonal

A

effects and telomere shortening are phenomena which may lead to genomic instability.

Figure 2

21

8.1.

Direct formation of ROS The evaluation of human and animal studies (Tables 2-5 and 8-11) shows that in some

of them evidence for induction of oxidative DNA damage and/or decreased total antioxidant capacity and induction of antioxidant enzymes was observed [41, 53, 54, 56, 57, 63, 101, 103, 108]; also increased LP by ROS was found in a number of investigations (see chapter 8.2).

IP T

The results of Othman et al. [120] concerning the effects of insulin are of particular

interest as the authors measured in their experiments also genetic damage in comet assays. By

SC R

use of specific inhibitors they developed a model for the induction of DNA migration in colon and kidney cells (Figure. 4). Insulin binds either to its receptor (IR) or to the insulin-like growth

factor-1 (IGF-1) receptor. After internalization of the receptor-insulin-complex into the cells,

U

the hormone is digested by proteases. Binding to both types of receptors leads to activation

N

of PI3K. Results with a specific PI3K inhibitor showed that this enzyme plays a key role in

A

signalling and also in the induction of genomic damage by insulin. The subsequent step is the

M

activation of Akt by PI3K which translocates from the membrane to the nucleus where it

ED

phosphorylates a wide array of downstream proteins.

PT

Figure 3

CC E

The reduction of PI3K/Akt levels which was seen few hours after insulin exposure was

explained as a consequence of activation of p53 which controls the phosphatase and tensin

A

homolog (PTEN), a deactivator of Akt and PI3K. Two pathways lead subsequently to ROS formation: (i) insulin enhances the translocation of Akt into mitochondria where it causes ROS formation; (ii) another pathway is the activation of NADPH-oxidase. Different isoforms of this enzyme are involved in specific cell types. While NADPH oxidase 1 (NOX1) was found to be activated in colon cells, NOX4 is active in the kidney.

22

In this context it is notable that is known that cancer cells produce their energy by increasing the rate of anaerobic glycolysis (Warburg-Effect), therefore it is possible that elevated glucose levels may accelerate the proliferation of transformed cells [128]. This phenomenon has been demonstrated under in vitro conditions, and it is notable in this context that lower blood glucose levels in cancer patients correlated with better outcomes [129].

IP T

Increased glucose levels in blood lead to ROS formation via different pathways. (i) In

cultured vasoendothelial cells, radical formation was observed via PKC-dependent activation

SC R

of NADPH-oxidase (Figure 4) [124]. (ii) Glycoxidation products are formed as a consequence

of sequential glycation and oxidation reactions between proteins and reducing sugars [130]. (iii) Furthermore, an alteration of the ratio of NADH/NAD+ was found in hyperglycaemic

U

tissues. This condition is also termed “pseudohypoxia” as the tissue levels of oxygen are

N

normal and is characterized by increased production of O2· and NO·[131]. (iv) Finally,

M

A

intracellular oxidation processes may cause O2· formation in mitochondria. This process starts with glycolysis in the cytoplasm which generates NADH and pyruvate. Cytoplasmic NADH

ED

can donate reducing equivalents to the transport chain in the mitochondria. Pyruvate is reduced in the cytoplasm to lactate or migrates into the mitochondria. Two main sites in these

PT

organelles generate O2· namely the NADH-dehydrogenase complex I and the interface

A

CC E

between ubiquinone and complex III [132].

Figure 4

The assumption of a relation between insulin levels and DNA damage is supported by

the results of a calorie restriction study; the authors found lower hormone levels and in parallel less DNA damage at the end of the intervention phase. Notably, also the blood glucose levels were reduced at the end of the trial [38]. Also in a Korean study associations were detected

23

between insulin and glucose levels in blood and comet formation [42]. Furthermore, it is notable that correlations between DNA damage in sperm and insulin and glucose levels were found in the blood of mice [111]. It is well known that lipid levels are elevated as a consequence of excess consumption of fat and in metabolic diseases [133]. In this context, it is notable that palmitate, which is the

IP T

most abundant fatty acid in plasma was shown to increase the activity of PKC and to cause ROS production [125].

SC R

Only few studies measured ROS formation in obese and lean individuals and monitored its relation to DNA damage. Demirbag et al. [64] found reduced total antioxidant capacity in

U

a study with MetS patients and in parallel increased comet formation in lymphocytes. Higher

N

mitochondrial ROS levels in sperm cells were found in two studies (i.e. in a human

A

intervention trial and in an experiment with mice); in both, elevated DNA damage was

M

detected [110, 111].

Also investigations which concerned the reduction of the activities of antioxidant

ED

enzymes (superoxide dismutase (SOD), glutathione peroxidase (GPx) and glutathione-Stransferase (GST)) and of oxidised glutathione as a consequence of excess body weight

PT

provide evidence for oxidative stress. Such effects were seen in a number of studies with humans [55, 63, 88, 108]. Also in the brain activities of SOD and catalase (CAT) were

CC E

decreased in a rat model which reflects the MetS in humans; in parallel higher MDA levels were found. These effects correlated with induction of DNA damage in comet experiments

A

[88].

In a study with 7,12-dimethylbenzo anthracene (DMBA; a polycyclic aromatic

hydrocarbon which causes breast cancer) reduction of the ratio of oxidised/reduced glutathione (GSH/GSSG) which is indicative for oxidative stress was observed in obese rats. This effect was paralleled by increased 8-OHdG levels [104].

24

Another marker of oxidative stress are protein carbonyl concentrations in serum [134], no alterations in this marker were seen in a 6-months calorie restriction study with overweight individuals [38].

8.2. Obesity induced lipid peroxidation and DNA damage

IP T

Oxidative stress leads to oxidation of fatty acids in biological membranes; as a consequence of this chain reaction, lipid peroxides and alkoxyl radicals as well as aldehydes

SC R

and ketones are formed [135]. Some of these molecules attack DNA and were postulated to

play a key role in the aetiology of cancer and other diseases [136]. The formation of DNA adducts by LP products for example by MDA, acrolein, 4-hydroxy-2-nonenal (4-HNE) and

U

other compounds are described in recent reviews [137, 138]. Elevated levels of different LP

N

markers were repeatedly seen in blood and muscles and also in adipose tissue of obese

M

A

individuals [42, 56, 63, 88, 95, 101, 103, 139-143].

The most commonly monitored biomarkers which are used in LP studies are MDA,

ED

thiobarbituric reactive substances (TBARs), hydroperoxides, 4-HNE, isoprostanes and

PT

conjugated dienes [134]. We found only few publications in which LP markers and DNA stability were monitored in parallel in obese/overweight and in normal weight subjects.

CC E

Karaman et al. [63] conducted such a study in Turkey; they found increased MDA levels in individuals with MetS and also higher MN rates in peripheral lymphocytes. Also in experiments with obese rats evidence for an association between MDA formation and DNA

A

damage was observed [95]. In a study from Poland, the serum levels of MDA and 4hydroxyalkenals were measured as an index for LP, in parallel 8-oxodG concentrations were determined in blood [56]. The authors found that both parameters are associated with the BMI and with hip circumference. Furthermore, correlations between LP and 8-oxodG were observed. 25

Notably, also a clear relation between isoprostane (8-epi-prostaglandin F2α) concentrations in urine and MDA in plasma (which are both markers for LP) and comet formation in lymphocytes was seen in a study which focused on the effect of visceral body fat in non-obese Korean men [42]. Yu et al. [103] compared the association between MDA in different organs of rats which

IP T

received a HFD and formation of oxidised DNA (8-OHdG) in mitochondria. The LP product was increased in heart, kidney, testes and liver tissue after 10 weeks but in the hepatic tissue

SC R

even a decrease was observed after 8 weeks.

A large number of studies focused exclusively on the impact of obesity on LP markers

U

but not on genomic stability. In total, about twenty papers have been published and in most

N

of them evidence for increased LP as a consequence of excess body weight was found (for

A

review see [139]).

M

Also mitochondrial membranes are attacked by ROS in overweight individuals and in MetS patients. This process leads to alterations of proteins which are involved in the electron

ED

transport chain. As a consequence, increased release of ROS formation was postulated [144].

PT

8.3. Impact of overweight on sex hormones

CC E

It is well documented that excess body weight causes hormonal imbalance. One of the enzymes which plays a key role is aromatase which catalyses the conversion of androgens to estrogens [145]. Ovaries are the main site of estrogen production in pre-menopausal women,

A

but peripheral tissues including body fat become the primary source in overweight postmenopausal individuals [15, 146, 147]. Cauley et al. [148] showed that estriol and estradiol levels are increased by more than 40% in obese post-menopausal women. Also in obese prepubertal girls, elevated levels of these hormones were found [149]. This effect was

26

explained by enhanced production of aromatase as a consequence of secretion of TNF-alpha and IL-6 adipocytes. Apart from enhanced production of humans also decreased levels of serum hormonebinding globulin were found in obese individuals [150]. This glycoprotein binds estrogens

IP T

and testosterones and inhibits their activities. Alterations of the hormonal status were proposed to play a key role in the induction of breast and endometrial cancer which have an increased prevalence in post-menopausal obese

SC R

women [151-154]. Furthermore, it was also found that excess body weight in children increases the overall cancer death rates as well as breast cancer mortality [155].

U

Receptor-dependent as well as independent mechanisms are involved in estrogen

N

induced breast carcinogenesis [156]. The hormones enhance the proliferation of breast cells

A

by binding to specific receptors. As a consequence of increased mitotic activity, errors in

M

DNA-replication are induced, the repair of mutations is less efficient and DNA adducts,

ED

breaks and other lesions are more frequently converted to persisting mutations [157]; furthermore, cell divisions cause mitotic recombinations which lead to errors that are passed

PT

to the daughter cells [157].

It is well documented that certain estrogen metabolites cause direct DNA damage.

CC E

Estradiol (17-βE2) is converted to catechol metabolites which are hydroxylated at the 2- or 4positions to form 2-hydroxyestrogen (2-OHE) and 4-hydroxyestrogen (4-OHE). The latter

A

compound is mutagenic in rat and human cells; i.e. it is converted to E(2)-3,4-quinone which forms covalent estrogen-DNA-adducts [158]. Additionally, E(2)3-4 quinoline can undergo redox cycling with E(2)3-4 hydroquinone leading to release of ROS. Quinones are backconverted via quinone reductase to create a cycle which leads to continuous release of radicals and also to formation of unstable depurination products with adenine and guanine [159].

27

Estrone undergoes a similar metabolism as estradiol but can be also converted to 16αhydroxyestrone (16α-OHE1) which binds covalently to DNA and enhances the proliferation of cells via binding to the ER [155, 160]. The genotoxic effects of estrogens in human derived breast cancer cells are well studied.

IP T

For example, Mobley and co-workers [161-163] found evidence for induction of oxidative damage under conditions which are relevant for humans. Comparisons of effects in cells with

SC R

and without ERs indicate that their presence increases the extent of oxidative damage.

The associations between formation of genotoxic estrogen metabolites and breast cancer have been investigated in a number of studies. It was shown that E(2)3-4 quinoline

U

causes cancer in animals [160, 164-168]. However, it is not clear, if the concentrations of

N

DNA reactive estrogen metabolites are indeed increased in obese individuals. Mauras et al.

A

[149] found elevated concentrations of 16α-OHE1 in a small study in sera of obese

M

prepubertal girls (12 lean, 24 obese individuals) while no relation between the 4- and the 16α-

ED

OHE1 pathway and BMI values was found in a larger study (n=603) with post-menopausal women [169].

PT

Hormonal effects are also involved in the development of endometrial tumors [170]. Apart from enhanced cell proliferation, alterations of signalling pathways may play a role.

CC E

However, at present experimental evidence is lacking that genomic instability is enhanced in the endometrium of obese women and further studies are required to assess the impact of

A

overweight induced genomic instability on this form of cancer.

28

8.4. Glycation end products Advanced glycation end products (AGEs) are formed as a consequence of reactions between sugar moieties and proteins; their biological effects are described in a number of reviews [171-173].

IP T

The endogenous formation of these products in the human body takes place nonenzymatically via the Maillard reaction but also via the so called “polyol pathway”, a reaction

SC R

which is catalysed by sorbitol dehydrogenase (SDH) and phosphokinase. Reactive αoxoaldehydes which are formed during these reactions (glyoxal, methylglyoxal and 3deoxyglucosone) are converted irreversibly by further reactions to products such as Nε-

U

carboxyethyllysine, arginine pyrimidine, pentosidine, pyrralin, methylglyoxal, glyoxal lysine

N

and Nε-carboxymethyllysine (CML) [171].

A

AGEs bind in the human body to specific receptors (receptor advanced glycation end

M

products (RAGE)) which are involved in different signalling pathways; the most relevant ones

ED

are shown in Figure 5. These interactions cause formation of ROS via activation of NADH-

Figure 5

CC E

PT

oxidase which leads to activation of various transcription factors including NF-κB [174].

A

Consumption of carbohydrate and sugar rich Western diets causes increased formation

of AGEs; also excess body fat may lead to elevated concentrations in specific tissues. Some findings indicate that the serum levels of CML are inversely related to the BMI and also to body fat, but enhanced levels were found in adipose tissue [175-178]. A different observation was made with sera from the MetS patients, i.e. the concentrations of AGEs were elevated in

29

these individuals and the concentrations correlated with the levels of markers which are indicative for insulin resistance and inflammation [179]. Also in experiments with HFD fed mice it was found that the concentrations of AGEs are increased in adipose tissue and in the liver [180].

IP T

AGEs cause DNA damage directly and via interactions with signalling pathways. It is well documented that methylglyoxal causes gene mutations in bacterial test [181, 182] and also in yeast [182]. Furthermore, it was shown that CML accumulates in nuclear proteins and

SC R

causes DNA strand breaks in human skin cells [183].

Stopper and co-workers [184] studied the effects of methylglyoxal and CML in comet

U

assays with different cell types. They found induction of DNA damage and postulated on the

N

basis of results obtained with inhibitors that formation of ROS as a consequence of the

A

activation of the transcription factor NF-κB plays a key role (Figure 5). One of the proteins

M

which is regulated by NF-κB is angiotensin which is vasoconstrictive and causes down

ED

regulation of peroxisomes. The authors observed pronounced reduction of DNA damage when they added a specific receptor antagonist (candesartan) to the incubation mix [184, 185].

PT

Apart from formation of AGEs, sugars undergo also direct reactions with DNA-bases [186]. The products cause depurination [187] as well as SSBs and mutations in vitro [188]. In

CC E

recent years, improved techniques (for example LC-MS and HPLC measurements) as well as antibodies have been developed which enable to quantify these products in humans [189-191]

A

but it is not known if increased concentrations are present in obese individuals.

8.5. Impact of obesity on DNA repair Overweight has an impact on different DNA repair pathways (Figure 6), but the tissue specificity and the underlying molecular mechanisms are only partly understood.

30

Recently, results of a study with young adults were published in which the authors found an association between the BMI and NER in a modified comet experiments with lymphocytes [192]. Langie and co-workers [102] performed interesting experiments with mice in which the mothers received a low folate diet and the offspring a HFD. They found reduction of BER in several brain regions of the progeny. Furthermore, they analysed the impact of this feeding

SC R

is involved in repair of oxidised DNA bases in the hippocampus [102].

IP T

scheme on the expression of BER related repair genes and observed alterations of Ogg1 which

U

Figure 6

N

It is well documented that defects of BER and NER are associated with increased cancer

A

rates. Single nucleotide polymorphisms in genes that encode for proteins involved in these

M

pathways were found to cause reduced activities of repair enzymes and to increase the cancer

ED

risks in humans [193]. Furthermore, it is known that defective NER in Xeroderma pigmentosum patients leads to skin cancer and glioblastomas [194, 195].

PT

It is notable that also other repair pathways are affected by oxidation of the DNA [196198] and by LP products [199, 200]. A study with transgenic overweight mice concerned the

CC E

induction of DSBs, the synthesis of DNA repair proteins and transcription of repair associated genes after administration of phosphoramide mustard. The authors found clear evidence for

A

induction of DNA damage and activation of certain proteins (PRKDC and XRCC6) as well as increased levels of the ATM protein in ovaries of obese animals [106]. A number of investigation concerned the impact of obesity on the methylation status of genes which are directly or indirectly involved in DNA repair. The MutL homolog 1 (MLH1) is one of seven proteins which play a role in DNA mismatch repair (MMR) [201]. Remely 31

et al. [91, 98] found an inverse association between DNA damage in liver tissue of obese mice (which was monitored in comet experiments) and alterations of the methylation status of MLH1 in CpG1 in the same organ [91, 98]; on the contrary a positive association of the methylation of the same gene was seen in the liver in CpG4 [98]. In colon mucosa cells an inverse relation was seen with this gene in CpG2. These effects were reversed by

IP T

supplementation of the drinking water with the green tea constituent, EGCG and also with vitamin E. [91] On the contrary, no relation between BMI and MLH-1 methylation was

SC R

detected in a human study (n=128) with biopsies from endometrial tumors [202].

DNA methyltransferases (DNMTs) are responsible for the transfer of methyl groups

U

from the universal methyl donor S-adenosyl-L-methionine (SAM) to the 5-position of

N

cytosine residues in DNA. One of the members of this group, DNMT1 encodes for the

A

maintenance methyltransferase and plays a key role in MMR [201]. Remely et al. [91, 98],

M

found in the aforementioned studies no significant differences in the CpG methylation status in the promoter region of DNMT1 in colon and liver tissue of obese HFD mice.

ED

Hypomethylation of this gene leads to increased transcription of genes which are involved in

PT

MMR.

A recent study with lymphocytes of obese and normal weight adolescents indicated that

CC E

the capacity mitomycin C induced DNA repair is lower in the latter groups [58]. The authors measured in this investigation the time kinetics of disappearance of γ-H2AX foci in

A

lymphocytes as a parameter of the repair capacity. A further protein which may play an important role in obesity associated repair is p53.

The activation of this tumor suppressor protein as a consequence of excess body weight is well documented [7]. One of its functions is the inhibition of cell divisions which prolongs the repair phase [7]. It is known that p53 is activated by different types of DNA damage,

32

including oxidation of bases [7, 203]. Apart from beneficial effects of p53, newer findings indicate that its activation in adipocytes contributes to the development of insulin resistance [203, 204]. The impact of obesity on sex hormones is described in chapter 8.3. It was emphasized

IP T

that ER signalling leads to suppression of DNA repair and apoptosis in favour of cell proliferation [205]. The most important processes which are influenced by steroid hormones concern the regulation of DSB repair, non-homologous end-joining and homologous

SC R

recombination [206]. Most data come from in vitro experiments and it is not known at present

to which extent obesity induced alterations of the hormonal status have an impact on repair

U

function in humans.

N

Also telomeres have an impact on DNA repair; the impact of increased BMI on their

M

A

length is described in the following chapter.

ED

8.6. Obesity and telomere lengths

Telomeres are DNA sequences (tandem repeats of the sequence TTAGGG) which are

PT

localized at the end of chromosomes [207]. It was shown in vitro that shortening leads to

CC E

cellular senescence and that overexpression of the enzyme telomerase (which maintains the levels of telomeres) leads to immortalization [208]. Also results of animal experiments

A

support the assumption of the involvement of these structures in senescence and ageing [209]. The impact of obesity on these structures may be an important factor which affects DNA

stability. The mechanisms which cause this phenomenon are partly known [210]. One of the modes of action which may be involved is the dependency of DSB repair on telomeric regions. Chromosomal fusion in mammalian cells involves homologues end joining which is one of the major forms of DSB repair. Chromosome fusion initiates chromosome instability 33

involving breakage fusion cycles in which dicentric chromosomes form bridges and breaks when the cells attempt to divide [211]. One of the mechanisms which could lead to telomere shortening in overweight/obese individuals is oxidative stress as it was shown in in vitro that telomeres are highly sensitive to damage by ROS [212, 213].

IP T

Only one intervention study has been conducted which concerned the correlation between alterations of telomere lengths and DNA base damage in overweight humans [59]. The authors monitored alterations of these parameters after weight loss (for details see Table

SC R

5) and found an increase of telomere lengths and in parallel reduction of apurinic/apyrimidinic

sites. In another study with a parallel design, no connections were detected between obesity

U

and telomere length and DNA damage [40].

N

A large number of studies was published which concerned the effects of body weight

A

on telomeres. The results are reviewed in two papers, which contain meta-analyses [214, 215].

M

In the first, the authors evaluated 16 studies. Only in cross-sectional investigations (n=14), a

ED

inverse relation was observed [214]. A second analysis [215] comprised 63 studies (119.439 subjects); in 39 an inverse correlation between obesity and telomere lengths was observed.

PT

The authors found in a pooled analysis a trend for an inverse association. However, they emphasize the need for further longitudinal studies and stress that the assumption of a

CC E

relationship between obesity and telomere lengths remains open due to the heterogeneity of the results.

A

Several studies indicate that the relation between telomere lengths and adiposity is more

pronounced in younger subjects; this observation was also confirmed in a meta-analysis of Müezzinler [214]. Another aspect which was addressed in a study by Martens et al. [216] concerns the impact of the maternal BMI on telomere integrity in new-borns. The authors found evidence for shorter telomeres in babies of overweight/obese mothers.

34

The impact of weight loss on telomere lengths was investigated in several trials; overall the findings are controversial. Moreno-Navarrete et al. [217] concluded that shorter telomere length in obese subjects is an established irreversible feature which cannot be improved by weight loss while O’Callaghan et al. [59] state that weight loss as a consequence of consumption of a calorie restricted diet prevents telomere shortening. Also results of studies

IP T

with bariatric surgery patients are inconsistent. No effect was seen in a trial with 97 subjects

[218] one year after the operation, while an increase was found in a 10 year prospective study

SC R

[219].

N

U

8.7. Impact of obesity/overweight on the activities of enzymes that activate and detoxify mutagens

A

Many dietary and environmental carcinogens such as polycyclic aromatic

M

hydrocarbons, heterocyclic aromatic amines, aflatoxins and nitrosamines are converted in the body to DNA reactive metabolites by specific phase I enzymes and partly also by phase II

ED

enzymes [220]. The latter ones catalyse also the detoxification of DNA reactive intermediates and of directly active compounds [221]. A number of human and animal studies addressed

PT

the question if obesity affects the activities of these enzymes. In experiments with humans,

CC E

the clearance rates of enzyme specific drugs were studied with chemical analytical methods while in animal models mainly hepatic enzyme activities were monitored. A comprehensive review of human studies was published by Brill et al. [222]; the

A

authors state that there is evidence that the clearance of CYP3A4 substrates it reduced in individuals with high BMI while increased excretion was observed with substrates for CYP2E1 and CYP1A2. Also in children, increased activities of the later isozymes was found [223]. These observations are partly in agreement with animal data; increased i.e. CYP2E1 was observed for example in genetically modified obese female mice but not in males [224],

35

in a trial with monosodium glutamate treated overweight male mice an increase of this enzyme and also of CYP2A5 and 3A were detected in hepatic tissue and in colon mucosa [225]; furthermore, there is also evidence for CYP2E1 induction in obese rats [226]. This isozyme plays a key role in the activation of genotoxic nitrosamines. In regard to phase II enzymes, increased clearance of N-acetyltransferase (NAT) and

IP T

uridine diphosphate glucuronosyltransferase (UGT) substrates was found in humans [222]; the later effects were also seen in overweight children [223]. UGT activity was also increased

SC R

in livers of obese mice with steatosis [227].

Data concerning the impact of obesity on GST are controversial. In diabetic mouse

U

model, decreased activity was found in the liver [224]; no such effects were seen in genetically

N

modified (ob/ob) mice in females but a higher activity was detected in animals in the same

A

study [228].

M

It is conceivable that alterations of the activities of the different drug metabolizing enzymes have a strong impact on the genotoxic properties of chemicals; however we are not

ED

aware of studies in which the consequences of overweight on the genotoxic effects of dietary, occupationally and environmentally relevant groups of DNA reactive carcinogens were

PT

investigated and related to alterations of drug metabolizing enzymes. An interesting study was published by Johnson et al. [229], who quantified cyclophosphamide DNA crosslinks in blood

CC E

cells of overweight and lean patients who underwent hematologic cell transplantation and found no evidence for BMI associated differences despite the fact that the overweight patients

A

received substantially higher doses of the drug. This cytostatic is activated by CYP2B6 and the authors hypothesized that their finding may be due to altered metabolism of the drug in obese individuals but no enzyme measurements were conducted to confirm this assumption [229]. Data concerning the impact of polycyclic aromatic hydrocarbon (PAH) exposure on oxidative DNA damage are controversial [55]. In a large study with PAH exposed coke oven

36

workers, an inverse relation with the BMI was observed in lymphocytes, while elevated levels of 8-OHdG were detected in livers of obese mice which were treated with PAH in comparison to lean controls [104].

Inflammation and DNA damage

IP T

8.8.

Inflammations are defence mechanisms which protect the host against infections and

SC R

other insults (for reviews see [230, 231]). It was found in many investigations that overweight/obesity are associated low grade inflammation [232, 233].

Adipose tissue plays a central role in this reaction. Adipocytes release so-called

U

“adipokines” into the blood stream which exert systemic effects [234]. These inflammatory

N

mediators belong to several groups including hormones (e.g. leptin which is pro-inflammatory

A

and adiponectin which is anti-inflammatory), chemokines and pro-inflammatory cytokines

M

(e.g. interleukins 1β, 6, 10 and TNF-α), acute phase proteins and growth factors such as IGF-

ED

1 [235]. Apart from adipocytes also macrophages and other cell types accumulate in adipose tissue and are important contributors to the production of substances which trigger

PT

inflammation [236].

Pro-inflammatory proteins which are produced in adipose tissue including the enzyme

CC E

haem oxygenase-1 (HO-1) cause substantial alterations of several pathways which activate transcription factors [237]. Probably the most relevant one is NF-kB which is activated by ROS [174] and adipokines [238] and causes increased transcription of genes encoding for

A

inflammatory molecules including COX-2. This enzyme catalyzes the formation of proinflammatory eicosanoids from arachidonic acid [239]. Inflammations cause DNA damage which in turn induces further inflammatory reactions [240] and it is likely that genomic instability plays a key role in the aetiology of

37

diseases which are associated with inflammations, in particular with cancer [241, 242]. However, there is only limited evidence for formation of oxidised DNA bases in obese humans by ROS as a consequence of inflammation (see Tables 2-6). In a study by Scarpato et al. the levels of inflammation markers (IL-6 and C-reactive protein) were found to be

9.

IP T

significantly higher in individuals with increased body weight [50].

Consequences of obesity induced DNA damage

SC R

Overweight/obesity are associated with increased prevalence of different forms of cancer in humans [243, 244]. The assumption of an association between excess body weight and cancer is also supported by results of rodent experiments [2, 90-92]. We are not aware of

U

any targeted studies in which correlations between DNA damage and cancer rates were

N

investigated in obese humans and animals; however, direct links can be postulated on the

A

basis of mechanistic studies.

M

Another relevant issue is the impact of obesity on infertility which was seen in a number

ED

of studies. As described in chapter 4.3 there is some evidence for increased DNA damage in sperm cells of obese infertile men, but further investigations concerning this relation are

PT

warranted. It is also conceivable that obesity causes mutations in germ cells which lead to heritable diseases in the offspring but this issue has never been investigated according to our

CC E

knowledge.

The term “DNA damage response” (DDR) concerns the consequences of genomic

A

instability [7, 205, 245, 246]. It describes pathways by which cells react to damaged DNA to prevent adverse health effects such as initiation of mitotic arrest, senescence, repair and cell death [205]. It was postulated that overweight and the MetS cause processes which prevent malignant transformation by activation of processes which lead to senescence and programmed cell death. The induction of inflammation via DNA damage and the activation

38

of transcription factors as well as telomere dysfunctions play a key role in these response mechanisms. A detailed description of this concept can be found in the papers of Shimizu et al. [246] and Erol [7]. The results of comet experiments with rodents indicate that excess body weight causes

IP T

genomic instability in multiple organs. The extent of damage which we measured in experiments with overweight mice in different tissues was similar (e.g. in colon, brain and liver) and no pronounced gender specific differences was observed [92]. It is conceivable that

SC R

the formation of tumors involve different organs specific tumors promoting mechanisms

which cause clonal expansion of damaged cells. These mechanisms are described in several

U

reviews [14, 247]. In regard to breast and endometrial cancer hormonal effects play an

N

important role, whereas in the liver the key mechanisms include lipogenesis and insulin

A

resistance which cause inflammation. In regard to formation of pancreatic tumors,

M

dysfunctional autophagy and toxic effects are discussed while obesity associated colorectal cancer is probably a consequence of dysbiosis and thinning of the intestinal mucosa layer

ED

resulting in increased permeability of the intestinal epithelial to microbial products.

PT

Apart from these organ specific effects, several mechanisms have been identified in obese individuals which may favour the development of tumors at multiple sites from initiated

CC E

cells including sustained angiogenesis, tissue invasion and metastasis and specific metabolic disturbances. These effects are described in detail in a recent working report of the IRAC [2,

A

248].

39

10. Conclusions and knowledge gaps Obesity and metabolic diseases are among the most relevant health risk factors for humans worldwide. The evaluation of the literature show that the impact of overweight/obesity on DNA stability which plays a key role in the etiology of cancer, infertility and ageing is not adequately investigated. We found in total 27 human studies

IP T

concerning the effects of obesity/overweight individuals and MetS patients on genomic instability in somatic cells, 12 human and 5 animal investigations concerning on DNA damage

SC R

in sperm and 18 investigations with rodents in which DNA damage was studied in blood cells

and in inner organs. It is interesting that a substantially higher number of human studies has been published which concern the genotoxic effects of chemicals. On the basis of a computer

U

aided search we estimate that about 220 articles deal with the effects of selected heavy metals

N

(Cr, Ni, Pb, As and Hg), the impact of radioactivity was monitored in more than 700 trials.

A

The quality of studies concerning the impact of overweight/obesity on DNA stability is

M

in general low since potential confounding factors such as intake of vitamins, minerals and

ED

the effects of plant derived DNA protective components were not taken into consideration in the most trials. Only in two studies the intake of micronutrients were recorded [53, 63]. It is

PT

known from a number of human studies that vitamins such as folate, B6, B12 and minerals such as Se, Zn and Fe have a strong impact on genomic instability [249, 250]. Furthermore it

CC E

is notable that most animal trials were conducted with diets which differ in regard to their macronutrient levels, this leads to differences of the body weight and to accumulation of fat

A

but the differences of the composition of the chows may cause results which do not necessarily reflect the effects of excess body weight. The literature provides only limited evidence for an association between overweight/obesity and damage of the genetic material in men. The evaluation of human studies on the basis of the QS values shows that in most comet trials with medium quality (no 40

study reached a high score) evidence for an association was detected. Four of six investigations with medium quality found that increased BMI values are associated with comet formation while results of MN studies with medium quality are conflicting (50% positive, 50% no relation). Also 8-OHdG measurements in humans do not allow to draw firm conclusions (positive, as well as negative correlations were detected); notably the study with

IP T

the highest QS found even a significant inverse association [53]. Notable, significant positive

correlations between induction of DSBs and excess body weight were detected in all γ-H2AX

SC R

experiments. Overall, 24 human studies with somatic and sperm cells found evidence for an

increase of DNA damage with the BMI while 22 trials reported no or even inverse

U

associations. It is probable that the highly conflicting results are due to the differences in the

N

diets, i.e. it is likely that normal and slim individuals consume higher amounts of vegetables,

A

fruits and full grain products which contain a broad variety of DNA protective compounds

M

while obesity is associated with increased intake of fats and foods which contain sugars and purified flour.

ED

The results of animal studies in which standardized diets were fed under controlled conditions provide clearer answers; i.e. positive associations between excess body weight and

PT

comet formation were seen in all studies with a medium quality (no high quality trials were

CC E

published so far) in somatic cells. Also the findings of sperm experiments with medium quality are in good agreement (two TUNEL and two SCSA trials, found consistently positive associations). Results of MN studies with animals are conflicting (one positive and two

A

negative were reported) but all of them had only low quality scores. The assumption of a causal relation between excess body weight and DNA damage is

strongly supported by results of mechanistic studies which indicate that several modes of action may cause genetic instability in overweight/obese individuals. Many investigations indicate that oxidative stress may play an important role; the formation of ROS may be a 41

consequence of increased insulin and glucose levels. ROS may cause either direct oxidative DNA damage (which was seen not in all studies), or via formation of DNA reactive LP products. As mentioned above, pronounced LP was seen in many trials with animals and humans. Also inflammation may be casually related as it leads to formation of oxygen and nitrogen radicals. Increased levels of pro-inflammatory cytokines and activation of NF-kB are

IP T

characteristic features of increased excess body fat; however, further investigations are warranted to allow firm conclusion. As described in chapters 8.3 to 8.6 also hormonal effects,

SC R

formation of AGEs, inhibition of DNA repair process and reduction of telomere lengths may play a role in the association between increased BMI values and genomic instability.

U

However, further experimental data are required which address these issues. In the case of

N

hormonal effects it is conceivable that they are organ specific, but similar DNA damage was

A

seen in multiple organs in a number of studies and no gender specific effects were observed.

M

The impact of excess body weight on telomere lengths was quite moderate and was only observed after longer time periods while DNA protective effects were seen in obese humans

ED

and laboratory rodents after short intervention periods with dietary components indicating that the impact of the BMI on telomeres may be of minor relevance. The effects of AGEs are

PT

difficult to assess at present as no studies are available in which their formation was correlated

CC E

with DNA stability in humans and animals. Many important knowledge gaps exist at present. It is not known exactly which BMI

values lead to a significant increase of genomic instability and if the effects increase linear or

A

exponentially. Further research is also warranted to elucidate the consequences of overweight/obesity induced DNA damage in regards to malignant transformation of cells. So far, research concerning the induction of cancer by excess body weight focused mainly on inflammation, signaling pathways and hormonal effects [251-253]. Other relevant topics for further studies concern the consequences of DNA damage on cognitive dysfunctions and 42

investigations concerning induction of mutations in germ cells. As mentioned above, some animal studies indicate that increased BMI leads to DNA damage in different regions of the brain [88, 92, 96], furthermore some animal studies found elevated levels of DNA damage in sperm cells as a consequence of excess body weight.

IP T

Genotoxicity studies will also enable to assess if and to which extent weight loss strategies as well as therapeutic interventions with drugs and bioactive natural dietary components lead to an improvement of the stability of the genetic material. Weight loss diets

SC R

which are advocated worldwide are generally not designed on the basis of scientific data and

it is conceivable that drastic reduction of the caloric intake and imbalanced consumption of

U

vitamins and minerals (which are required to maintain the stability of genetic material) will

N

lead to adverse health effects including destabilization of the DNA. Also this important issue

ED

Conflict of interest statement

M

A

should be addressed in future investigations.

PT

There are no conflict of interest.

CC E

Acknowledgments This works was supported by COST Action CA15132 (hCOMET); the authors are

A

also thankful to participants of this research initiative for discussions.

43

References

8.

9. 10. 11. 12. 13. 14. 15.

IP T

CC E

16.

SC R

7.

U

6.

N

5.

A

4.

M

3.

ED

2.

Calle, E.E. and R. Kaaks, Overweight, obesity and cancer: epidemiological evidence and proposed mechanisms. Nat Rev Cancer, 2004. 4(8): p. 579-91. Lauby-Secretan, B., et al., Body fatness and cancer—Viewpoint of the IARC working group. N Engl J Med, 2016. 375(8): p. 794-798. Di Cesare, M., Bentham, J., Stevens, G A., Zhou, B., Danaei, G. et al., Trends in adult bodymass index in 200 countries from 1975 to 2014: a pooled analysis of 1698 population-based measurement studies with 19.2 million participants. The Lancet, 2016. 387(10026): p. 13771396. De Onis, M., M. Blössner, and E. Borghi, Global prevalence and trends of overweight and obesity among preschool children. The American journal of clinical nutrition, 2010. 92(5): p. 1257-1264. Lee, S.C. and J.C. Chan, Evidence for DNA damage as a biological link between diabetes and cancer. Chin Med J (Engl), 2015. 128(11): p. 1543. Katib, A., Mechanisms linking obesity to male infertility. Cent European J Urol, 2015. 68(1): p. 79-85. Erol, A., Systemic DNA damage response and metabolic syndrome as a premalignant state. Curr Mol Med, 2010. 10(3): p. 321-334. Stefani, G.P., et al., Metabolic Syndrome and DNA Damage: The Interplay of Environmental and Lifestyle Factors in the Development of Metabolic Dysfunction. Open J Endocr Metab Dis, 2015. 5(07): p. 65. Ross, S.A. and J.A. Milner, Epigenetic modulation and cancer: effect of metabolic syndrome? Am J Clin Nutr, 2007. 86(3): p. s872-7. Stein, C.J. and G.A. Colditz, Modifiable risk factors for cancer. Br J Cancer, 2004. 90(2): p. 299-303. Rivlin, N., et al., Mutations in the p53 tumor suppressor gene important milestones at the various steps of tumorigenesis. Genes Cancer, 2011. 2(4): p. 466-474. McCubrey, J.A., et al., Roles of the Raf/MEK/ERK pathway in cell growth, malignant transformation and drug resistance. Biochim Biophys Acta, 2007. 1773(8): p. 1263-84. Wolin, K.Y., K. Carson, and G.A. Colditz, Obesity and cancer. Oncologist, 2010. 15(6): p. 556565. Font-Burgada, J., B. Sun, and M. Karin, Obesity and cancer: the oil that feeds the flame. Cell Metab, 2016. 23(1): p. 48-62. Calle, E.E. and R. Kaaks, Overweight, obesity and cancer: epidemiological evidence and proposed mechanisms. Nat Rev Cancer, 2004. 4(8): p. 579-591. Nersesyan, A., M. Mišík, and S. Knasmüller, Methods used for the detection of antimutagens: an overview. Knasmueller, S., DeMarini, D.M., Johnson, I., Gerhauser, C. (Eds.), Chemoprevention of cancer and DNA damage by dietary factors, Wiley-VCH, Weheinim. 2009. 209-227. Nersesyan, A., et al., Use of Single-cell Gel Electrophoresis Assays in Dietary Intervention Trials. The Comet Assay in Toxicology. Vol. 30. 2016. 314. Anderson, D., A. Dhawan, and J. Laubenthal, The comet assay in human biomonitoring. Methods Mol Biol, 2013. 1044: p. 347-62. Vazquez Boucard, C., et al., A study of DNA damage in buccal cells of consumers of welland/or tap-water using the comet assay: Assessment of occupational exposure to genotoxicants. Environ Mol Mutagen, 2017. 58(8): p. 619-627. Wasson, G.R., V.J. McKelvey-Martin, and C.S. Downes, The use of the comet assay in the study of human nutrition and cancer. Mutagenesis, 2008. 23(3): p. 153-62. OECD, Guidelines for the Testing of Chemicals, No: 487; In vitro mammalian cell micronucleus test. 2010, OECD.

PT

1.

17.

A

18. 19.

20. 21.

44

31. 32. 33. 34. 35. 36.

37. 38.

CC E

39.

IP T

29. 30.

SC R

28.

U

27.

N

26.

A

25.

M

24.

ED

23.

OECD, Guidline for the testing of chemicals, No: 474; Mammalian erythrocyte micronuclei test. 1997, OECD. OECD, Guidelines for the Testing of Chemicals, Section 4, Test No. 489: In Vivo Mammalian Alkaline Comet Assay. 2016, OECD. p. 1-27. Williams, E.S., J. Panko, and D.J. Paustenbach, The European Union's REACH regulation: a review of its history and requirements. Crit Rev Toxicol, 2009. 39(7): p. 553-75. Corvi, R. and F. Madia, In vitro genotoxicity testing-Can the performance be enhanced? Food Chem Toxicol, 2017. 106(Pt B): p. 600-608. Barber, T., et al., Obesity and polycystic ovary syndrome. Clin Endocrinol, 2006. 65(2): p. 137-145. Olokoba, A.B., O.A. Obateru, and L.B. Olokoba, Type 2 diabetes mellitus: a review of current trends. Oman Med J, 2012. 27(4): p. 269-73. Nersesyan, A., et al., Use of the lymphocyte cytokinesis-block micronucleus assay in occupational biomonitoring of genome damage caused by in vivo exposure to chemical genotoxins: Past, present and future. Mutat Res, 2016. 770(Pt A): p. 1-11. Collins, A.R., et al., The comet assay: topical issues. Mutagenesis, 2008. 23(3): p. 143-151. Phillips, D., et al., Methods of DNA adduct determination and their application to testing compounds for genotoxicity. Environ Mol Mutagen, 2000. 35(3): p. 222-233. Azqueta, A., et al., Comet assay to measure DNA repair: approach and applications. Front Genet, 2014. 5: p. 288. Bolognesi, C., et al., Clinical application of micronucleus test in exfoliated buccal cells: A systematic review and metanalysis. Mutat Res Rev Mutat Res, 2015. 766: p. 20-31. Bonassi, S., et al., Micronuclei frequency in peripheral blood lymphocytes and cancer risk: evidence from human studies. Mutagenesis, 2011. 26(1): p. 93-100. Ivashkevich, A.N., et al., γH2AX foci as a measure of DNA damage: a computational approach to automatic analysis. Mutat Res, 2011. 711(1-2): p. 49-60. Loo, D.T., In situ detection of apoptosis by the TUNEL assay: an overview of techniques. Methods Mol Biol, 2011. 682: p. 3-13. Evenson, D.P., The Sperm Chromatin Structure Assay (SCSA®) and other sperm DNA fragmentation tests for evaluation of sperm nuclear DNA integrity as related to fertility. Anim Reprod Sci, 2016. 169: p. 56-75. Giovannelli, L., et al., Nutritional and lifestyle determinants of DNA oxidative damage: a study in a Mediterranean population. Carcinogenesis, 2002. 23(9): p. 1483-1489. Heilbronn, L.K., et al., Effect of 6-month calorie restriction on biomarkers of longevity, metabolic adaptation, and oxidative stress in overweight individuals: a randomized controlled trial. Jama, 2006. 295(13): p. 1539-48. Bankoglu, E.E., et al., Reduction of DNA damage in peripheral lymphocytes of obese patients after bariatric surgery-mediated weight loss. Mutagenesis, 2017. Jones, D., et al., Changes in markers of oxidative stress and DNA damage in human visceral adipose tissue from subjects with obesity and type 2 diabetes. Diabetes Res Clin Pract, 2014. 106(3): p. 627-633. Tomasello, B., et al., DNA damage in normal-weight obese syndrome measured by Comet assay. Med J Nutrition Metab, 2011. 4(2): p. 99-104. Jang, Y., et al., Visceral fat accumulation determines postprandial lipemic response, lipid peroxidation, DNA damage, and endothelial dysfunction in nonobese Korean men. J Lipid Res, 2003. 44(12): p. 2356-64. Gandhi, G. and G. Kaur, Assessment of DNA damage in obese individuals. Research Journal of Biology, 2012. 2(2): p. 37-44. Bukhari, S.A., et al., Plasma homocysteine and DNA damage profiles in normal and obese subjects in the Pakistani population. Mol Biol Rep, 2010. 37(1): p. 289-95.

PT

22.

40.

A

41. 42.

43. 44.

45

52. 53.

54. 55.

56. 57. 58. 59.

IP T

CC E

60.

SC R

51.

U

50.

N

49.

A

48.

M

47.

ED

46.

Luperini, B.C., et al., Gene polymorphisms and increased DNA damage in morbidly obese women. Mutat Res, 2015. 776: p. 111-7. Benassi-Evans, B., et al., High protein–high red meat versus high carbohydrate weight loss diets do not differ in effect on genome stability and cell death in lymphocytes of overweight men. Mutagenesis, 2009: p. gep006. Donmez-Altuntas, H., et al., Evaluation of chromosomal damage, cytostasis, cytotoxicity, oxidative DNA damage and their association with body-mass index in obese subjects. Mutat Res Genet Toxicol Environ Mutagen., 2014. 771: p. 30-36. Ros‐Llor, I., et al., Effect of mobile phones on micronucleus frequency in human exfoliated oral mucosal cells. Oral Dis, 2012. 18(8): p. 786-792. Rossnerova, A., et al., Mapping the factors affecting the frequency and types of micronuclei in an elderly population from Southern Bohemia. Mutat Res, 2016. 793-794: p. 32-40. Scarpato, R., et al., Nuclear damage in peripheral lymphocytes of obese and overweight Italian children as evaluated by the γ-H2AX focus assay and micronucleus test. FASEB J, 2011. 25(2): p. 685-693. Hamid, Z.A., et al., The association of nuclear abnormalities in exfoliated buccal epithelial cells with the health status of different agricultural activities farmers in Peninsular Malaysia. Genes Environ, 2016. 38(1): p. 1. Torres-Bugarin, O., et al., Genetic profile of overweight and obese school-age children. Toxicol Environ Chem, 2009. 91(4): p. 789-795. Loft, S., et al., Oxidative DNA damage estimated by 8-hydroxydeoxyguanosine excretion in humans: influence of smoking, gender and body mass index. Carcinogenesis, 1992. 13(12): p. 2241-2247. Kocael, A., et al., The effects on oxidative DNA damage of laparoscopic gastric band applications in morbidly obese patients. Can J Surg, 2014. 57(3): p. 183-7. Liu, A.-L., et al., Elevated levels of urinary 8-hydroxy-2'-deoxyguanosine, lymphocytic micronuclei, and serum glutathione S-transferase in workers exposed to coke oven emissions. Environ Health Perspect, 2006: p. 673-677. Karbownik-Lewinska, M., et al., Direct contribution of obesity to oxidative damage to macromolecules. Neuro Endocrinol Lett, 2012. 33(4): p. 453-61. Cejvanovic, V., et al., Markers of oxidative stress in obese men with and without hypertension. Scand J Clin Lab Invest, 2016. 76(8): p. 620-625. Azzara, A., et al., Different repair kinetic of DSBs induced by mitomycin C in peripheral lymphocytes of obese and normal weight adolescents. Mutat Res, 2016. 789: p. 9-14. O'Callaghan, N.J., et al., Weight loss in obese men is associated with increased telomere length and decreased abasic sites in rectal mucosa. Rejuvenation Res, 2009. 12(3): p. 169176. Nersesyan, A., et al., Effect of staining procedures on the results of micronucleus assays with exfoliated oral mucosa cells. Cancer Epidemiol Biomarkers Prev, 2006. 15(10): p. 183540. Thomas, P., et al., Buccal micronucleus cytome assay. Nat Protoc, 2009. 4(6): p. 825-37. Fenech, M., et al., Low intake of calcium, folate, nicotinic acid, vitamin E, retinol, β-carotene and high intake of pantothenic acid, biotin and riboflavin are significantly associated with increased genome instability—results from a dietary intake and micronucleus index survey in South Australia. Carcinogenesis, 2005. 26(5): p. 991-999. Karaman, A., et al., DNA damage is increased in lymphocytes of patients with metabolic syndrome. Mutat Res Genet Toxicol Environ Mutagen, 2015. 782: p. 30-35. Demirbag, R., et al., DNA damage in metabolic syndrome and its association with antioxidative and oxidative measurements. Int J Clin Pract, 2006. 60(10): p. 1187-1193. Milić, M., et al., Level of primary DNA damage in the early stage of metabolic syndrome. Mutat Res Genet Toxicol Environ Mutagen, 2013. 758(1): p. 1-5.

PT

45.

A

61. 62.

63. 64. 65.

46

73. 74. 75. 76. 77. 78.

79. 80. 81. 82.

IP T

CC E

83.

SC R

72.

U

71.

N

70.

A

69.

M

68.

ED

67.

Abdilla, N., et al., Impact of the components of metabolic syndrome on oxidative stress and enzymatic antioxidant activity in essential hypertension. J Hum Hypertens, 2007. 21(1): p. 68-75. Chavarro, J.E., et al., Body mass index in relation to semen quality, sperm DNA integrity, and serum reproductive hormone levels among men attending an infertility clinic. Fertil Steril, 2010. 93(7): p. 2222-2231. Fariello, R.M., et al., Association between obesity and alteration of sperm DNA integrity and mitochondrial activity. BJU Int, 2012. 110(6): p. 863-867. Vignera, S., et al., Negative effect of increased body weight on sperm conventional and nonconventional flow cytometric sperm parameters. J Androl, 2012. 33(1): p. 53-58. Dupont, C., et al., Obesity leads to higher risk of sperm DNA damage in infertile patients. Asian J Androl, 2013. 15(5): p. 622-625. Håkonsen, L.B., et al., Does weight loss improve semen quality and reproductive hormones? Results from a cohort of severely obese men. Reprod Health, 2011. 8(1): p. 1. Håkonsen, L., et al., Exposures that may affect sperm DNA integrity: two decades of followup in a pregnancy cohort. Reprod Toxicol, 2012. 33(3): p. 316-321. Rybar, R., et al., Male obesity and age in relationship to semen parameters and sperm chromatin integrity. Andrologia, 2011. 43(4): p. 286-291. Radwan, M., et al., Sperm DNA damage-the effect of stress and everyday life factors. Int J Impot Res, 2016. 28(4): p. 148-54. Eisenberg, M.L., et al., The relationship between male BMI and waist circumference on semen quality: data from the LIFE study. Hum Reprod, 2014. 29(2): p. 193-200. Hammiche, F., et al., Sperm quality decline among men below 60 years of age undergoing IVF or ICSI treatment. J Androl, 2011. 32(1): p. 70-76. Bandel, I., et al., No association between body mass index and sperm DNA integrity. Hum Reprod, 2015: p. dev111. Zhang, H.B., et al., Early apoptotic changes in human spermatozoa and their relationships with conventional semen parameters and sperm DNA fragmentation. Asian J Androl, 2008. 10(2): p. 227-235. Bandel, I., et al., No association between body mass index and sperm DNA integrity. Hum Reprod, 2015. 30(7): p. 1704-13. Fenech, M., Cytokinesis-block micronucleus cytome assay. Nature Protocols, 2007. 2(5): p. 1084-1104. Kumaravel, T.S. and A.N. Jha, Reliable Comet assay measurements for detecting DNA damage induced by ionising radiation and chemicals. Mutat Res, 2006. 605(1-2): p. 7-16. Burlinson, B., et al., Fourth International Workgroup on Genotoxicity testing: results of the in vivo Comet assay workgroup. Mutat Res, 2007. 627(1): p. 31-5. Henkel, R., et al., TUNEL assay and SCSA determine different aspects of sperm DNA damage. Andrologia, 2010. 42(5): p. 305-13. Evenson, D.P., The Sperm Chromatin Structure Assay (SCSA((R))) and other sperm DNA fragmentation tests for evaluation of sperm nuclear DNA integrity as related to fertility. Anim Reprod Sci, 2016. 169: p. 56-75. Gedik, C.M. and A. Collins, Establishing the background level of base oxidation in human lymphocyte DNA: results of an interlaboratory validation study. Faseb j, 2005. 19(1): p. 824. Collins, S., et al., Genetic vulnerability to diet-induced obesity in the C57BL/6J mouse: physiological and molecular characteristics. Physiol Behav, 2004. 81(2): p. 243-8. York, D.A., J. Steinke, and G.A. Bray, Hyperinsulinemia and insulin resistance in genetically obese rats. Metabolism, 1972. 21(4): p. 277-84.

PT

66.

84.

A

85.

86. 87.

47

95.

96.

97.

98.

99. 100. 101.

IP T

CC E

102.

SC R

94.

U

93.

N

92.

A

91.

M

90.

ED

89.

Sinha, J.K., et al., Increased macromolecular damage due to oxidative stress in the neocortex and hippocampus of WNIN/Ob, a novel rat model of premature aging. Neuroscience, 2014. 269: p. 256-64. Fredga, K., Studies of the chromosomes in american obese-hyperglycaemic mice. Hereditas, 1961. 47(3‐4): p. 615-618. Remely, M., et al., Vitamin E Modifies High-Fat Diet-Induced Increase of DNA Strand Breaks, and Changes in Expression and DNA Methylation of Dnmt1 and MLH1 in C57BL/6J Male Mice. Nutrients, 2017. 9(6): p. 607. Remely, M., et al., EGCG Prevents High Fat Diet-Induced Changes in Gut Microbiota, Decreases of DNA Strand Breaks, and Changes in Expression and DNA Methylation of Dnmt1 and MLH1 in C57BL/6J Male Mice. Oxid Med Cell Longev, 2017. 2017: p. 3079148. Setayesh T, F.F., Nersesyan A, Haslberger A, Huber W, Haslinger E, Knasmüller S, Impact of gallic acid on genotoxical and biochemical parameters in mice fed on a high fat diet, in global summit on obesity and diet managment. 2016: Los Angeles, USA. Gutzkow, K.B., et al., Enhanced susceptibility of obese mice to glycidamide-induced sperm chromatin damage without increased oxidative stress. Andrology, 2016. 4(6): p. 1102-1114. Tenorio, N.M., et al., The influence of sleep deprivation and obesity on DNA damage in female Zucker rats. Clinics, 2013. 68(3): p. 385-389. De Sibio, M.T., et al., A comparative genotoxicity study of a supraphysiological dose of triiodothyronine (T 3) in obese rats subjected to either calorie-restricted diet or hyperthyroidism. PloS one, 2013. 8(2): p. e56913. Leffa, D.D., et al., Corrective effects of acerola (Malpighia emarginata DC.) juice intake on biochemical and genotoxical parameters in mice fed on a high-fat diet. Mutat Res, 2014. 770: p. 144-152. Remely, M., et al., EGCG Prevents High Fat Diet-Induced Changes in Gut Microbiota, Decreases of DNA Strand Breaks, and Changes in Expression and DNA Methylation of Dnmt1 and MLH1 in C57BL/6J Male Mice. 2017. 2017: p. 3079148. Remely, M., et al., Vitamin E Modifies High-Fat Diet-Induced Increase of DNA Strand Breaks, and Changes in Expression and DNA Methylation of Dnmt1 and MLH1 in C57BL/6J Male Mice. Nutrients, 2017. 9(6). Vares, G., et al., Diet-induced obesity modulates epigenetic responses to ionizing radiation in mice. PLoS One, 2014. 9(8): p. e106277. Wickliffe, J.K., et al., Diet-induced obesity increases the frequency of Pig-a mutant erythrocytes in male C57BL/6J mice. Environ Mol Mutagen, 2016. 57(9): p. 668-677. Bankoglu, E.E., et al., Impact of weight loss induced by gastric bypass or caloric restriction on oxidative stress and genomic damage in obese Zucker rats. Free Radic Biol Med, 2016. 94: p. 208-217. Langie, S.A., et al., Maternal folate depletion and high-fat feeding from weaning affects DNA methylation and DNA repair in brain of adult offspring. FASEB J, 2013. 27(8): p. 33233334. Yu, H.T., et al., Oxidative damage of mitochondrial respiratory chain in different organs of a rat model of diet-induced obesity. Eur J Nutr, 2017: p. 1-11. Melnyk, S., et al., Effects of Obesity on Pro-Oxidative Conditions and DNA Damage in Liver of DMBA-Induced Mammary Carcinogenesis Models. Metabolites, 2017. 7(2). Del Ry, S., et al., Transcriptional alterations of ET-1 axis and DNA damage in lung tissue of a rat obesity model. DNA Cell Biol, 2015. 34(3): p. 170-7. Ganesan, S., et al., Obesity alters phosphoramide mustard-induced ovarian DNA repair in mice. Biol Reprod, 2017. 96(2): p. 491-501. Djuric, Z., et al., Oxidative DNA damage levels in rats fed low-fat, high-fat, or calorierestricted diets. Toxicol Appl Pharmacol, 1992. 115(2): p. 156-160.

PT

88.

103.

A

104. 105. 106. 107.

48

115. 116. 117. 118. 119. 120.

121. 122. 123.

IP T

CC E

124.

SC R

114.

U

113.

N

112.

A

111.

M

110.

ED

109.

Yuzefovych, L.V., et al., Mitochondrial DNA damage and dysfunction, and oxidative stress are associated with endoplasmic reticulum stress, protein degradation and apoptosis in high fat diet-induced insulin resistance mice. PloS one, 2013. 8(1): p. e54059. Vendramini, V., et al., Reproductive function of the male obese Zucker rats: alteration in sperm production and sperm DNA damage. Reprod Sci, 2014. 21(2): p. 221-9. Bakos, H.W., et al., The effect of paternal diet-induced obesity on sperm function and fertilization in a mouse model. Int J Androl, 2011. 34(5 Pt 1): p. 402-10. Palmer, N.O., et al., Diet and exercise in an obese mouse fed a high-fat diet improve metabolic health and reverse perturbed sperm function. Am J Physiol Endocrinol Metab, 2012. 302(7): p. E768-80. Duale, N., et al., Impaired sperm chromatin integrity in obese mice. Andrology, 2014. 2(2): p. 234-43. Vendramini, V., et al., Reproductive Function of the Male Obese Zucker Rats Alteration in Sperm Production and Sperm DNA Damage. Reprod Sci, 2013: p. 1933719113493511. Sasaki, Y.F., et al., The comet assay with multiple mouse organs: comparison of comet assay results and carcinogenicity with 208 chemicals selected from the IARC monographs and U.S. NTP Carcinogenicity Database. Crit Rev Toxicol, 2000. 30(6): p. 629-799. Shen, J., et al., Oncogenic mutations and dysregulated pathways in obesity-associated hepatocellular carcinoma. Oncogene, 2016. 35(49): p. 6271-6280. Shen, J., et al., Oncogenic mutations and dysregulated pathways in obesity-associated hepatocellular carcinoma. 2016. 35(49): p. 6271-6280. Bordonaro, M. and D. Lazarova, Hypothesis: Obesity Is Associated with a Lower Mutation Threshold in Colon Cancer. J Cancer, 2015. 6(9): p. 825-31. Ecke, T.H., et al., Body mass index (BMI) and mutations of tumor suppressor gene p53 (TP53) in patients with urinary bladder cancer. Urol Oncol, 2008. 26(5): p. 470-3. Cavazos, D.A., et al., Obesity promotes aerobic glycolysis in prostate cancer cells. Nutr Cancer, 2014. 66(7): p. 1179-1186. Othman, E.M., et al., Insulin-mediated oxidative stress and DNA damage in LLC-PK1 pig kidney cell line, female rat primary kidney cells, and male ZDF rat kidneys in vivo. Endocrinology, 2013. 154(4): p. 1434-1443. Othman, E.M., H. Hintzsche, and H. Stopper, Signaling steps in the induction of genomic damage by insulin in colon and kidney cells. Free Radic Biol Med, 2014. 68: p. 247-257. Othman, E.M., A. Leyh, and H. Stopper, Insulin mediated DNA damage in mammalian colon cells and human lymphocytes in vitro. Mutat Res, 2013. 745: p. 34-39. Lorenzi, M., et al., High glucose induces DNA damage in cultured human endothelial cells. J Clin Invest, 1986. 77(1): p. 322. Quagliaro, L., et al., Intermittent high glucose enhances apoptosis related to oxidative stress in human umbilical vein endothelial cells the role of protein kinase C and NAD (P) H-oxidase activation. Diabetes, 2003. 52(11): p. 2795-2804. Inoguchi, T., et al., High glucose level and free fatty acid stimulate reactive oxygen species production through protein kinase C--dependent activation of NAD (P) H oxidase in cultured vascular cells. Diabetes, 2000. 49(11): p. 1939-1945. Shimoi, K., et al., Oxidative DNA damage induced by high glucose and its suppression in human umbilical vein endothelial cells. Mutat Res, 2001. 480: p. 371-378. Beeharry, N., J. Chambers, and I. Green, Fatty acid protection from palmitic acid-induced apoptosis is lost following PI3-kinase inhibition. Apoptosis, 2004. 9(5): p. 599-607. Vander Heiden, M.G., L.C. Cantley, and C.B. Thompson, Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science, 2009. 324(5930): p. 1029-33. Klement, R.J. and U. Kämmerer, Is there a role for carbohydrate restriction in the treatment and prevention of cancer? Nutr Metab (Lond), 2011. 8: p. 75.

PT

108.

125.

A

126. 127. 128. 129.

49

138. 139.

140. 141.

142. 143.

144.

145.

CC E

146.

IP T

137.

SC R

136.

U

135.

N

134.

A

133.

M

132.

ED

131.

Baynes, J.W., Role of oxidative stress in development of complications in diabetes. Diabetes, 1991. 40(4): p. 405-412. Williamson, J.R., et al., Hyperglycemic pseudohypoxia and diabetic complications. Diabetes, 1993. 42(6): p. 801-813. Nishikawa, T., et al., Normalizing mitochondrial superoxide production blocks three pathways of hyperglycaemic damage. Nature, 2000. 404(6779): p. 787-790. Bays, H.E., et al., Obesity, adiposity, and dyslipidemia: a consensus statement from the National Lipid Association. J Clin Lipidol, 2013. 7(4): p. 304-383. Knasmüller, S., et al., Use of conventional and-omics based methods for health claims of dietary antioxidants: a critical overview. Br J Nutr, 2008. 99(E-S1): p. ES3-ES52. Lü, J.M., et al., Chemical and molecular mechanisms of antioxidants: experimental approaches and model systems. J Cell Mol Med, 2010. 14(4): p. 840-860. Lobo, V., et al., Free radicals, antioxidants and functional foods: Impact on human health. Pharmacogn Rev, 2010. 4(8): p. 118. Wei, X. and H. Yin, Covalent modification of DNA by α, β-unsaturated aldehydes derived from lipid peroxidation: Recent progress and challenges. Free Radic Res, 2015. 49(7): p. 905-917. Winczura, A., et al., Lipid peroxidation product 4-hydroxy-2-nonenal modulates base excision repair in human cells. DNA Repair (Amst), 2014. 22: p. 1-11. Vincent, H.K., K.E. Innes, and K.R. Vincent, Oxidative stress and potential interventions to reduce oxidative stress in overweight and obesity. Diabetes Obes Metab, 2007. 9(6): p. 813839. Vincent, H., et al., Mechanism for obesity-induced increase in myocardial lipid peroxidation. Int J Obes Relat Metab Disord, 2001. 25(3). Russell, A.P., et al., Lipid peroxidation in skeletal muscle of obese as compared to endurance‐trained humans: a case of good vs. bad lipids? FEBS Lett, 2003. 551(1-3): p. 104106. Cohen, G., Y. Riahi, and S. Sasson, Lipid peroxidation of poly-unsaturated fatty acids in normal and obese adipose tissues. Arch Physiol Biochem, 2011. 117(3): p. 131-139. Reddy, K.K., et al., Free radical and antioxidant status in urban and rural Tirupati men: interaction with nutrient intake, substance abuse, obesity and body fat distribution. Asia Pac J Clin Nutr, 1997. 6(4): p. 296-311. Nicolson, G.L., Metabolic syndrome and mitochondrial function: molecular replacement and antioxidant supplements to prevent membrane peroxidation and restore mitochondrial function. J Cell Biochem, 2007. 100(6): p. 1352-1369. Chen, J., et al., The correlation of aromatase activity and obesity in women with or without polycystic ovary syndrome. J Ovarian Res, 2015. 8(1): p. 1. van Kruijsdijk, R.C., E. van der Wall, and F.L. Visseren, Obesity and cancer: the role of dysfunctional adipose tissue. Cancer Epidemiol Biomarkers Prev, 2009. 18(10): p. 25692578. Boon, W.C., J.D. Chow, and E.R. Simpson, The multiple roles of estrogens and the enzyme aromatase. Prog Brain Res, 2010. 181: p. 209-232. Cauley, J.A., et al., The epidemiology of serum sex hormones in postmenopausal women. Am J Epidemiol, 1989. 129(6): p. 1120-1131. Mauras, N., et al., Estrogens and their genotoxic metabolites are increased in obese prepubertal girls. J Clin Endocrinol Metab, 2015. 100(6): p. 2322-2328. Morisset, A.-S., K. Blouin, and A. Tchernof, Impact of diet and adiposity on circulating levels of sex hormone-binding globulin and androgens. Nutr Rev, 2008. 66(9): p. 506-516. Cleary, M.P. and M.E. Grossmann, Obesity and breast cancer: the estrogen connection. Endocrinology, 2009. 150(6): p. 2537-2542.

PT

130.

147.

A

148. 149. 150. 151.

50

159. 160.

161. 162. 163. 164. 165. 166.

IP T

CC E

167.

SC R

158.

U

157.

N

156.

A

155.

M

154.

ED

153.

Cleary, M.P. and N.J. Maihle, The role of body mass index in the relative risk of developing premenopausal versus postmenopausal breast cancer. Exp Biol Med (Maywood), 1997. 216(1): p. 28-43. Morimoto, L.M., et al., Obesity, body size, and risk of postmenopausal breast cancer: the Women's Health Initiative (United States). Cancer Causes & Control, 2002. 13(8): p. 741751. Moley, K.H. and G.A. Colditz, Effects of obesity on hormonally driven cancer in women. Sci Transl Med, 2016. 8(323): p. 323ps3-323ps3. Weiderpass, E., et al., A prospective study of body size in different periods of life and risk of premenopausal breast cancer. Cancer Epidemiol Biomarkers Prev, 2004. 13(7): p. 11211127. Yue, W., et al., Estrogen receptor-dependent and independent mechanisms of breast cancer carcinogenesis. Steroids, 2013. 78(2): p. 161-170. Martino, S., et al., Continuing outcomes relevant to Evista: breast cancer incidence in postmenopausal osteoporotic women in a randomized trial of raloxifene. J Natl Cancer Inst., 2004. 96(23): p. 1751-1761. Dwivedy, I., et al., Synthesis and characterization of estrogen 2, 3-and 3, 4-quinones. Comparison of DNA adducts formed by the quinones versus horseradish peroxidaseactivated catechol estrogens. Chem Res Toxicol, 1992. 5(6): p. 828-833. Yue, W., et al., Estrogen receptor-dependent and independent mechanisms of breast cancer carcinogenesis. Steroids, 2013. 78(2): p. 161-70. Zhao, Z., et al., Mutagenic activity of 4-hydroxyestradiol, but not 2-hydroxyestradiol, in BB rat2 embryonic cells, and the mutational spectrum of 4-hydroxyestradiol. Chem Res Toxicol, 2006. 19(3): p. 475-479. Mobley, J.A. and R.W. Brueggemeier, Increasing the DNA damage threshold in breast cancer cells. Toxicol Appl Pharmacol, 2002. 180(3): p. 219-226. Mobley, J.A., A.S. Bhat, and R.W. Brueggemeier, Measurement of oxidative DNA damage by catechol estrogens and analogues in vitro. Chem Res Toxicol, 1999. 12(3): p. 270-277. Mobley, J.A. and R.W. Brueggemeier, Estrogen receptor-mediated regulation of oxidative stress and DNA damage in breast cancer. Carcinogenesis, 2004. 25(1): p. 3-9. Fuhrman, B.J., et al., Estrogen metabolism and risk of breast cancer in postmenopausal women. J Natl Cancer Inst, 2012. 104(4): p. 326-339. Im, A., et al., Urinary estrogen metabolites in women at high risk for breast cancer. Carcinogenesis, 2009. 30(9): p. 1532-1535. Eliassen, A.H., et al., Circulating 2-hydroxy-and 16α-hydroxy estrone levels and risk of breast cancer among postmenopausal women. Cancer Epidemiol Biomarkers Prev, 2008. 17(8): p. 2029-2035. Jernström, H., et al., Predictors of the plasma ratio of 2-hydroxyestrone to 16αhydroxyestrone among pre-menopausal, nulliparous women from four ethnic groups. Carcinogenesis, 2003. 24(5): p. 991-1005. Mauras, N., et al., Obesity without established comorbidities of the metabolic syndrome is associated with a proinflammatory and prothrombotic state, even before the onset of puberty in children. J Clin Endocrinol Metab, 2010. 95(3): p. 1060-1068. Xie, J., et al., Body size in relation to urinary estrogens and estrogen metabolites (EM) among premenopausal women during the luteal phase. Horm Cancer, 2012. 3(5-6): p. 249260. Brinton, L.A. and A.S. Felix, Menopausal Hormone Therapy and Risk of Endometrial Cancer. J Steroid Biochem Mol Biol, 2014. 142: p. 83-9. Ott, C., et al., Role of advanced glycation end products in cellular signaling. Redox Biol, 2014. 2: p. 411-429.

PT

152.

A

168.

169.

170. 171.

51

179.

180. 181.

182.

183. 184. 185.

IP T

CC E

186.

SC R

178.

U

177.

N

176.

A

175.

M

174.

ED

173.

Poulsen, M.W., et al., Advanced glycation endproducts in food and their effects on health. Food Chem Toxicol, 2013. 60: p. 10-37. Nedić, O., et al., Molecular effects of advanced glycation end products on cell signalling pathways, ageing and pathophysiology. Free Radic Res, 2013. 47(sup1): p. 28-38. Morgan, M.J. and Z.-g. Liu, Crosstalk of reactive oxygen species and NF-κB signaling. Cell Res, 2011. 21(1): p. 103-115. Semba, R.D., et al., Fat mass is inversely associated with serum carboxymethyl-lysine, an advanced glycation end product, in adults. J Nutr, 2011. 141(9): p. 1726-1730. Gaens, K.H., et al., Protein-Bound Plasma Nε-(Carboxymethyl) lysine Is Inversely Associated With Central Obesity and Inflammation and Significantly Explain a Part of the Central Obesity–Related Increase in Inflammation The Hoorn and CODAM Studies. Arterioscler Thromb Vasc Biol, 2015. 35(12): p. 2707-2713. Gaens, K.H., et al., Nε-(carboxymethyl) lysine-receptor for advanced glycation end product axis is a key modulator of obesity-induced dysregulation of adipokine expression and insulin resistance. Arterioscler Thromb Vasc Biol, 2014. 34(6): p. 1199-1208. Gaens, K.H., et al., Endogenous formation of N ε-(carboxymethyl) lysine is increased in fatty livers and induces inflammatory markers in an in vitro model of hepatic steatosis. J Hepatol, 2012. 56(3): p. 647-655. Uribarri, J., et al., Elevated serum advanced glycation endproducts in obese indicate risk for the metabolic syndrome: A link between healthy and unhealthy obesity? J Clin Endocrinol Metab, 2015. 100(5): p. 1957-1966. Song, F., et al., RAGE regulates the metabolic and inflammatory response to high-fat feeding in mice. Diabetes, 2014. 63(6): p. 1948-1965. Onoda, A., M. Asanoma, and H. Nukaya, Identification of methylglyoxal as a major mutagen in wood and bamboo pyroligneous acids. Biosci Biotechnol Biochem, 2016. 80(5): p. 833839. Murata, K., et al., Phenotype character of the methylglyoxal resistance gene in Saccharomyces cerevisiae: expression in Escherichia coli and application to breeding wildtype yeast strains. Appl Environ Microbiol, 1985. 50(5): p. 1200-1207. Roberts, M.J., et al., DNA damage by carbonyl stress in human skin cells. Mutat Res, 2003. 522(1): p. 45-56. Stopper, H., et al., Genotoxicity of advanced glycation end products in mammalian cells. Cancer Lett, 2003. 190(2): p. 151-156. Schupp, N., et al., Genotoxicity of advanced glycation end products: involvement of oxidative stress and of angiotensin II type 1 receptors. Ann N Y Acad Sci, 2005. 1043(1): p. 685-695. Gkogkolou, P. and M. Böhm, Advanced glycation end products: Key players in skin aging? Dermatoendocrinol, 2012. 4(3): p. 259-270. Seidel, W. and M. Pischetsrieder, DNA-glycation leads to depurination by the loss of N2carboxyethylguanine in vitro. Cell Mol Biol (Noisy-le-grand), 1998. 44(7): p. 1165-1170. Pischetsrieder, M., et al., N 2-(1-Carboxyethyl) deoxyguanosine, a nonenzymatic glycation adduct of DNA, induces single-strand breaks and increases mutation frequencies. Biochem Biophys Res Commun, 1999. 264(2): p. 544-549. Schneider, M., et al., Determination of Glycated Nucleobases in Human Urine by a New Monoclonal Antibody Specific for N 2-Carboxyethyl-2'-deoxyguanosine. Chem Res Toxicol, 2004. 17(10): p. 1385-1390. Bidmon, C., M. Frischmann, and M. Pischetsrieder, Analysis of DNA-bound advanced glycation end-products by LC and mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci, 2007. 855(1): p. 51-58. Matsumoto, K., et al., Immunohistochemical detection of advanced glycation end products in human bladder with specific monoclonal antibody. Int J Urol, 2009. 16(4): p. 402-405.

PT

172.

187.

A

188.

189.

190.

191.

52

199. 200.

201. 202. 203. 204. 205. 206. 207. 208.

IP T

CC E

209.

SC R

198.

U

197.

N

196.

A

195.

M

194.

ED

193.

Tyson, J., et al., Inter-individual variation in nucleotide excision repair in young adults: effects of age, adiposity, micronutrient supplementation and genotype. Br J Nutr, 2009. 101(09): p. 1316-1323. Langie, S.A., et al., Causes of genome instability: the effect of low dose chemical exposures in modern society. Carcinogenesis, 2015. 36(Suppl 1): p. S61-S88. Emmert, B., et al., [Xeroderma pigmentosum (XP) : A genetic disease sheds light on UVinduced skin cancer]. Hautarzt, 2011. 62(2): p. 91-7. Lai, J.-P., et al., The influence of DNA repair on neurological degeneration, cachexia, skin cancer and internal neoplasms: autopsy report of four xeroderma pigmentosum patients (XP-A, XP-C and XP-D). Acta Neuropathol Commun, 2013. 1(1): p. 4. Jaiswal, M., et al., Human Ogg1, a protein involved in the repair of 8-oxoguanine, is inhibited by nitric oxide. Cancer Res, 2001. 61(17): p. 6388-6393. Graziewicz, M., D.A. Wink, and F. Laval, Nitric oxide inhibits DNA ligase activity: potential mechanisms for NO-mediated DNA damage. Carcinogenesis, 1996. 17(11): p. 2501-2505. Laval, F. and D.A. Wink, Inhibition by nitric oxide of the repair protein, O6-DNAmethyltransferase. Carcinogenesis, 1994. 15(3): p. 443-447. Hiramatsu, K., et al., Monochloramine inhibits ultraviolet B-induced p53 activation and DNA repair response in human fibroblasts. Biochim Biophys Acta, 2006. 1763(2): p. 188-196. Feng, Z., et al., Malondialdehyde, a major endogenous lipid peroxidation product, sensitizes human cells to UV-and BPDE-induced killing and mutagenesis through inhibition of nucleotide excision repair. Mutat Res, 2006. 601(1): p. 125-136. Jin, B. and K.D. Robertson, DNA methyltransferases, DNA damage repair, and cancer. Adv Exp Med Biol, 2013. 754: p. 3-29. McCourt, C.K., et al., Body mass index: relationship to clinical, pathologic and features of microsatellite instability in endometrial cancer. Gynecol Oncol, 2007. 104(3): p. 535-9. Vergoni, B., et al., DNA damage and the activation of the p53 pathway mediate alterations in metabolic and secretory functions of adipocytes. Diabetes, 2016. 65(10): p. 3062-3074. Minamino, T., et al., A crucial role for adipose tissue p53 in the regulation of insulin resistance. Nat Med, 2009. 15(9): p. 1082-1087. Caldon, C.E., Estrogen signaling and the DNA damage response in hormone dependent breast cancers. Front Oncol, 2014. 4: p. 106. Schiewer, M.J. and K.E. Knudsen, Linking DNA damage and hormone signaling pathways in cancer. Trends Endocrinol Metab, 2016. 27(4): p. 216-225. Rosenberg, M., et al., Characterization of short tandem repeats from thirty-one human telomeres. Genome Res, 1997. 7(9): p. 917-923. Cong, Y.-S., W.E. Wright, and J.W. Shay, Human telomerase and its regulation. Microbiol Mol Biol Rev, 2002. 66(3): p. 407-425. Wong, K.-K., et al., Telomere dysfunction and Atm deficiency compromises organ homeostasis and accelerates ageing. Nature, 2003. 421(6923): p. 643-648. Mathieu, N., et al., Telomeres and chromosomal instability. Cell Mol Life Sci, 2004. 61(6): p. 641-656. Murnane, J.P., Telomere dysfunction and chromosome instability. Mutat Res, 2012. 730(1): p. 28-36. von Zglinicki, T., Oxidative stress shortens telomeres. Trends Biochem Sci, 2002. 27(7): p. 339-344. Coluzzi, E., et al., Oxidative stress induces persistent telomeric DNA damage responsible for nuclear morphology change in mammalian cells. PloS one, 2014. 9(10): p. e110963. Müezzinler, A., A. Zaineddin, and H. Brenner, Body mass index and leukocyte telomere length in adults: a systematic review and meta‐analysis. Obes Rev, 2014. 15(3): p. 192-201. Mundstock, E., et al., Effect of obesity on telomere length: Systematic review and meta‐ analysis. Obesity, 2015. 23(11): p. 2165-2174.

PT

192.

210.

A

211. 212. 213. 214. 215.

53

223. 224.

225.

226. 227. 228.

229.

230.

IP T

CC E

231.

SC R

222.

U

221.

N

220.

A

219.

M

218.

ED

217.

Martens, D.S., et al., Maternal pre-pregnancy body mass index and newborn telomere length. BMC Med, 2016. 14(1): p. 148. Moreno-Navarrete, J., et al., Telomere length of subcutaneous adipose tissue cells is shorter in obese and formerly obese subjects. Int J Obes (Lond), 2010. 34(8): p. 1345-1348. Formichi, C., et al., Weight loss associated with bariatric surgery does not restore short telomere length of severe obese patients after 1 year. Obes Surg, 2014. 24(12): p. 20892093. Laimer, M., et al., Telomere length increase after weight loss induced by bariatric surgery: results from a 10 year prospective study. nt J Obes (Lond), 2016. 40(5): p. 773-778. Glatt, H., Xennnobiotic metabolism: a target for nutritional chemoprevention of cancer?, in chemoprevention of cancer and DNA damage by dietary factors, S. Knasmüller, et al., Editors. 2009, Wiley-VCH Weinheim: Germany. p. 93-107. Knasmüller, S.E., et al., Chemoprevention of cancer and DNA damage by dietary factors. 2009: Wiley-VCH Weinheim, Germany. Brill, M.J., et al., Impact of obesity on drug metabolism and elimination in adults and children. Clin Pharmacokinet, 2012. 51(5): p. 277-304. Chiney, M.S., S.J. Schwarzenberg, and L.A. Johnson, Altered xanthine oxidase and Nacetyltransferase activity in obese children. Br J Clin Pharmacol, 2011. 72(1): p. 109-15. Roe, A.L., et al., Characterization of cytochrome P450 and glutathione S-transferase activity and expression in male and female ob/ob mice. Int J Obes Relat Metab Disord, 1999. 23(1): p. 48-53. Tomankova, V., et al., Altered cytochrome P450 activities and expression levels in the liver and intestines of the monosodium glutamate-induced mouse model of human obesity. Life Sci, 2015. 133: p. 15-20. Enriquez, A., et al., Altered expression of hepatic CYP2E1 and CYP4A in obese, diabetic ob/ob mice, and fa/fa Zucker rats. Biochem Biophys Res Commun, 1999. 255(2): p. 300-6. Xu, J., et al., UDP-glucuronosyltransferase expression in mouse liver is increased in obesityand fasting-induced steatosis. Drug Metab Dispos, 2012. 40(2): p. 259-66. Barnett, C.R., et al., Cytochrome P-450-dependent mixed-function oxidase and glutathione S-transferase activities in spontaneous obesity-diabetes. Biochem Pharmacol, 1992. 43(8): p. 1868-71. Johnson, L.A., N. Tretyakova, and P.A. Jacobson, Obesity effects on cyclophosphamideinduced DNA damage in hematopoietic cell transplant recipients. In Vivo, 2012. 26(5): p. 853-7. Chovatiya, R. and R. Medzhitov, Stress, inflammation, and defense of homeostasis. Mol Cell, 2014. 54(2): p. 281-288. Sirisinha, S., Insight into the mechanisms regulating immune homeostasis in health and disease. Asian Pac J Allergy Immunol, 2011. 29(1): p. 1. Calder, P.C., et al., Dietary factors and low-grade inflammation in relation to overweight and obesity. Br J Nutr, 2011. 106 Suppl 3: p. S5-78. Emanuela, F., et al., Inflammation as a link between obesity and metabolic syndrome. J Nutr Metab, 2012. 2012. Lumeng, C.N. and A.R. Saltiel, Inflammatory links between obesity and metabolic disease. J Clin Invest, 2011. 121(6): p. 2111-2117. Makki, K., P. Froguel, and I. Wolowczuk, Adipose tissue in obesity-related inflammation and insulin resistance: cells, cytokines, and chemokines. ISRN Inflamm, 2013. 2013. Calder, P.C., et al., Dietary factors and low-grade inflammation in relation to overweight and obesity. Br J Nutr, 2011. 106(S3): p. S1-S78. Araujo, J.A., M. Zhang, and F. Yin, Heme oxygenase-1, oxidation, inflammation, and atherosclerosis. Front Pharmacol, 2012. 3: p. 119.

PT

216.

232. 233.

A

234. 235. 236. 237.

54

246. 247. 248. 249.

250.

251. 252. 253.

CC E

254.

IP T

245.

SC R

244.

U

243.

N

242.

A

241.

M

240.

ED

239.

Lawrence, T., The nuclear factor NF-κB pathway in inflammation. Cold Spring Harb Perspect Biol, 2009. 1(6): p. a001651. Baker, R.G., M.S. Hayden, and S. Ghosh, NF-κB, inflammation, and metabolic disease. Cell Metab, 2011. 13(1): p. 11-22. Pálmai-Pallag, T. and C.Z. Bachrati, Inflammation-induced DNA damage and damageinduced inflammation: a vicious cycle. Microbes Infect, 2014. 16(10): p. 822-832. Jackson, A.L. and L.A. Loeb, The contribution of endogenous sources of DNA damage to the multiple mutations in cancer. Mutat Res, 2001. 477(1): p. 7-21. Reuter, S., et al., Oxidative stress, inflammation, and cancer: how are they linked? Free Radic Biol Med, 2010. 49(11): p. 1603-1616. Renehan, A.G., et al., Body-mass index and incidence of cancer: a systematic review and meta-analysis of prospective observational studies. The Lancet, 2008. 371(9612): p. 569578. Bracci, P.M., Obesity and pancreatic cancer: overview of epidemiologic evidence and biologic mechanisms. Mol Carcinog, 2012. 51(1): p. 53-63. Erol, A., Metabolic syndrome is a real disease and premalignant state induced by oncogenic stresses to block malignant transformation. Med Hypotheses, 2010. 74(6): p. 1038-1043. Shimizu, I., et al., DNA damage response and metabolic disease. Cell Metab, 2014. 20(6): p. 967-977. Simpson, E.R. and K.A. Brown, Obesity and breast cancer: role of inflammation and aromatase. J Mol Endocrinol, 2013. 51(3): p. T51-9. O'Flanagan C.H, B.L.W., Allott E.H,Hursting S. D., Molecular and metabolic mechanisms underlying the obesity-cancer link, I.w.g. No.10, Editor. 2017, IARC. p. 95-104. Fenech, M., The Genome Health Clinic and Genome Health Nutrigenomics concepts: diagnosis and nutritional treatment of genome and epigenome damage on an individual basis. Mutagenesis, 2005. 20(4): p. 255-69. Fenech, M.F., Dietary reference values of individual micronutrients and nutriomes for genome damage prevention: current status and a road map to the future. Am J Clin Nutr, 2010. 91(5): p. 1438s-1454s. Sun, B. and M. Karin, Obesity, inflammation, and liver cancer. J Hepatol, 2012. 56(3): p. 704-713. Ramos-Nino, M.E., The role of chronic inflammation in obesity-associated cancers. ISRN Oncol, 2013. 2013. Jung, U.J. and M.-S. Choi, Obesity and its metabolic complications: the role of adipokines and the relationship between obesity, inflammation, insulin resistance, dyslipidemia and nonalcoholic fatty liver disease. Int J Mol Sci, 2014. 15(4): p. 6184-6223. Fenech, M., Nutritional treatment of genome instability: a paradigm shift in disease prevention and in the setting of recommended dietary allowances. Nutr Res Rev, 2003. 16(01): p. 109-122. Azqueta, A. and A.R. Collins, The essential comet assay: a comprehensive guide to measuring DNA damage and repair. Arch Toxicol, 2013. 87(6): p. 949-68. Bakos, H.W., et al., Sperm DNA damage is associated with assisted reproductive technology pregnancy. Int J Androl, 2008. 31(5): p. 518-26. Evenson, D.P., K.L. Larson, and L.K. Jost, Sperm chromatin structure assay: its clinical use for detecting sperm DNA fragmentation in male infertility and comparisons with other techniques. J Androl, 2002. 23(1): p. 25-43.

PT

238.

255.

A

256.

257.

Legends for figures

55

Figure 1. Use of different methods for the detection of DNA damage in obesity studies. DNA damage was analysed in animal experiments mainly in comet assays (in different organs) which were also used in human studies with lymphocytes. Micronucleus (MN) experiments are conducted in bone marrow cells of rodents and in human studies with lymphocytes and with exfoliated epithelial cells. For the analysis of sperm cells, terminal deoxynucleotidyl-

IP T

transferase dUTP nick end labelling (TUNEL) and sperm chromatin structure assays (SCSA) are conducted. Oxidative damage of the DNA and alterations of DNA repair processes (BER,

SC R

NER) can be monitored with a modified protocol of the comet assay (not shown) and by

U

determination of 8-OHdG and/or 8-oxodG in urine, plasma and bloods cells.

N

Figure 2. Induction of DNA damage as a consequence of obesity/overweight. DNA damage

A

can be induced directly via estrogen metabolites, formation of advanced glycation end

and

alterations

of

the

activation

of

enzymes

which

catalyse

the

ED

shortening

M

products, LP products and radicals or indirectly via increased mitotic activity, telomere

activation/detoxification of mutagens and by inhibition of repair pathways. Phenomenona

PT

which were observed in overweight/obese humans (H) or animals (A) are indicated in circles. Solid lines indicate that experimental evidence shows that they lead to DNA stability or ROS

CC E

formation in rodents or men. Dotted lines are indicative for effects which were not demonstrated so far in overweight/obese subjects were observed in in vitro experiments.

A

Numbers indicate chapters in which the different effects are described.

Figure 3. Induction of ROS by insulin. The hormone binds to receptors (insulin receptor and insulin growth factor receptor) which leads to activation of phosphatidylinositol-3-kinase (PI3K) and Akt, the subsequent steps are activation of NADPH-oxidase and translocation of 56

Akt into mitochondria. Both processes lead to formation of ROS. Most findings come from in vitro experiments with mammalian cells but it was also shown that increased insulin levels cause DNA damage in rodents.

IP T

Figure 4. Induction of ROS by increased glucose levels. ROS can be formed by different molecular mechanisms, namely via activation of PKC or as a consequence of glycolysis in

SC R

the cytosol. AGEs are formed as a consequence of reactions of sugars with proteins, they activate transcription factors and cause also direct DNA damage. Another pathway of ROS

formation is caused by pseudohypoxia. All findings come from in vitro experiments with

A

N

U

mammalian cells but it was shown that increased insulin levels cause DNA damage in rodents.

M

Figure 5. Formation of AGEs and activation of NF-kB via RAGEs. Dotted line: intermediate processes which are not described in detail. AGEs are formed via Maillard reactions or via

ED

the polyol pathway and cause activation of transcription factors via interactions with signalling pathways. All findings come from in vitro experiments with mammalian cells but

CC E

PT

it was shown that increased insulin levels cause DNA damage in rodents.

Figure 6. Impact of obesity on DNA repair processes. Inflammation and estrogens increase

A

the mitotic activity of cells which results in errors of DNA replication and reduces the duration of the repair phase. Hormones have also a direct impact on different forms of repair. Telomere shortening was found in a number of investigations with overweight humans and leads to chromosomal aberrations as a consequence of inhibition of repair pathways. It is documented that ROS have an impact on repair functions (e.g. on NER); they also activate p53 which results in inhibition of cell divisions. Few studies are available indicating that 57

overweight/obesity have an impact on the methylation of DNA repair enzymes. Phenomenon which were observed in overweight/obese humans (H) or animals (A) are indicated in circles. Solid lines indicate that experimental evidence shows that they lead to DNA stability or ROS formation in rodents or men. Dotted lines are indicative for effects which were not

A

CC E

PT

ED

M

A

N

U

SC R

IP T

demonstrated so far in overweight/obese subjects but were observed in in vitro experiments.

58

59

A ED

PT

CC E

IP T

SC R

U

N

A

M

60

A ED

PT

CC E

IP T

SC R

U

N

A

M

A ED

PT

CC E

IP T

SC R

U

N

A

M Table. 1

61

I N U SC R

Table. 1. Overview of genotoxicity methods which are used in studies concerning the impact of overweight/ obesity on DNA stability Endpoints

Principle



BER/NER



cells are exposed to ROS generating chemicals e.g. (hydrogen peroxide or radiation) [30]  detection of alterations of DNA repair activities by induction of BER and NER specific DNA damage [31] MN are formed as a consequence of structural and numerical chromosomal aberrations, additional markers in MN cytome studies are nuclear buds and nuclear bridges which reflect genomic instability, furthermore acute toxicity markers are evaluated i.e. karyolysis, karyorrhexis, pyknotic nuclei, binucleated cells and condensed chromatin [32] determination of oxidised deoxyguanosine by use of HPLC, ELISA or calorimetric assays [30]

ED



FPG/Endo sensitive sites ROS sensivity

A



detection of DNA migration in an electric field. in humans: lymphocytes and whole  reflect formation of single and double strand breaks blood in rodents: different inner organs (SSBs and DSBs) and apurinic sites [29] *the methods can be also used to detect DNA damage in sperm cells III  detection of oxidised purines and pyrimidines

M

Comet formation  Standard conditions

CC E

PT

Micronuclei

A

8-OXOdG/ 8-OHdG/

γ-H2X

Target cells/ tissues and body fluids

in humans: lymphocytes and exfoliated cells from the mouth in rodents: bone marrow and erythrocytes

Remarks the predictive value of comet formation in regard to human cancer risks is not known.

MN in lymphocytes of humans are a reliable marker for cancer [33]

in humans: lymphocytes, plasma and relation to urine human cancer in rodents: plasma and urine risks not known. Induced by inflammatory diseases detection of double strand breaks by measurement of in humans: lymphocytes relation to human phosphorylation of a specific histone as a consequence of in rodents: different inner organs cancer risks not double strand DNA repair [34] known.

62

I N U SC R

Table. 1. continued Principle

Terminal deoxynucleotidyltransferase dUTP nick end labelling (TUNEL) assay

based on the ability of the enzyme terminal sperm cells in humans and animals deoxynucleotidyl- transferase (TdT) to incorporate labelled dUTP into free 3hydroxyl termini generated by fragmentation of genomic DNA into low molecular weight double stranded DNA and high molecular weight single stranded DNA [35] based on heat treatment or acidic denaturation sperm cells in humans and animals of sperm DNA and subsequent staining with acridine orange which enables to discriminate between DSBs and SSBs (double strands stain green, single strands red) [36]

M

ED

A

CC E

PT

Sperm chromatin structure assay (SCS)

Target cells/ tissues

A

Endpoints

63

Remarks in some animal and human studies results correlated with reduced fertility.

in some animal and human studies results correlated with reduced fertility.

I N U SC R

Table 2. Results of comet assay in overweight/obese subjects1 Study design

Parameter (indicator cells)

Participants and treatments

Intervention

tail intensity (lymphocytes)

48♂/23♀, BMI=18.0–23.9, n=21 BMI=24.0–26.9, n=26 BMI=27.0–39.9, n=24 age (all groups) 35-64

Intervention

arbitrary units (leukocytes)

Results

QS

Remarks

Ref

↔ DNA damage ↔ FPG in groups with different BMI

22 (M)

↑ oxidative DNA damage after consumption of sugar (1.8-fold) ↔ gender ↔ age

[37]

calorie restriction (CR): n=6♂/6♀, CR + exercise: n=6♂/6♀, very low- calorie diet: n=6♂/6♀, C: n=6♂/6♀, baseline BMI (all groups)=27.8, age (all groups) 26-49 9♂/47♀, BMI=51.1 n=56 before surgery 6 and 12 months after surgery age (all groups) 44.70 ± 1.4

↓ DNA damage after CR and after CR+exercise and very low-calorie diet groups at month 6 vs. baseline (1.03-fold)

18 (M)

weight loss after very low-caloric diet (12 kg) weight loss after CR + exercise and CR (8 kg) ↓ fasting insulin level after 3 and 6 months in CR + exercise and CR ↔ fasting glucose ↔ serum protein carbonyl

[38]

↓ DNA damage after 12 months (38%) ↔ DNA damage after 6 months

20 (M)

[39]

OB: n=20, BMI ≥30 OB+ type 2 diabetes (T2D): n=21, BMI ≥30 C: n=20, BMI ≤30 age (all groups) 44-55

↔ DNA damage OB vs. C

19 (M)

↓ ferric reducing antioxidant power assay after 6 and 12 months ↓ weight (22% after 6 months) and (29% after 12 months) ↑ total glutathione after 6 months (1.1-fold) ↑ total oxidised glutathione after 6 months (1.6-fold) ↔ total glutathione and oxidised glutathione after 12 months ↔MDA ↓ MDA in T2D vs. C (75%) ↓ telomere length in OB+T2D vs. OB

CC E

PT

ED

M

A

Comet assay2

tail intensity (whole blood)

A

Intervention

Parallel

tail intensity (visceral adipose tissue)

64

[40]

I N U SC R

Study design

Parameter (indicator cells)

Participants and treatments

Parallel

tail intensity (lymphocytes)

OB: n=20♀, BMI ≥25 C: n=20♀, BMI ≤25 normal weight obese syndrome3 (NWO): n=20♀, BMI ≤25 age (all groups) 25-35

Parallel

tail intensity (lymphocytes)

ED

M

A

Comet assay

QS

Remarks

Ref

↑ DNA damage OB vs. C (1.5-fold) ↑ DNA damage NWO vs. C (1.5-fold) ↑ FPG NWO vs. C (1.7fold) ↑ FPG OB vs. C (1.6-fold) ↑ DNA damage in high visceral fat vs. normal visceral fat (1.6-fold)

18 (L)

↔ 8-OHdG in whole blood

[41]

↑ MDA in high visceral fat vs. normal visceral fat (1.419 (M) fold)

[42]

↑ DNA damage in OB vs. C (5.0-fold)

↔ WC 21 (M) ↔ waist to hip ratio ↔ gender ↔ age

[43]

CC E

PT

healthy, non-obese Koreans, BMI=23.8, high visceral fat ≥100 cm2, n=27♂ normal visceral fat,<100 cm2, n=27♂ age (all groups) 36.5±0.8 OB: n=18♂/17♀, BMI= 31.9 (age <20: n=12, age 25-53: n=23) C: n= 8♂/7♀, BMI= 23.3 (age<20: n=9 and age 2553: n=6) OB: n=80♂/80♀, BMI=29 C: n=80♂/80♀, BMI =16.022.0 age (all groups) young and old (n.s)

Results

A

Parallel

Parallel

arbitrary units (leukocytes)

length of DNA migration (μm) (peripheral blood)

↑ DNA damage in ♀ young 17 OB vs. C (2.5-fold) (L) ↑ DNA damage in ♂ young OB vs. C (2.2-fold) ↑ DNA damage in ♀ old OB vs. C (2.6-fold) ↑ DNA damage in ♂ old OB vs. C (2.3-fold)

65

alterations of DNA damage and plasma homocysteine correlated with triglycerides, LDL-cholesterol, systolic blood pressure, cholesterol, MDA and total oxygen stress in OB. (-) association DNA damage and plasma homocysteine with total oxidant status and globulin.

[44]

I Study design

Parameter (indicator cells)

Participants and treatments

tail intensity (lymphocytes)

OB-MetS pts.: n= 30♀, BMI=45.7, age 32.4±6.8 OB-without MetS: n= 30♀, BMI=21.9 age 32.4±6.5

ED

M

A

Comet assay Parallel

N U SC R

Table 2. Continued

1

Results

QS

Remarks

Ref

↑ DNA damage in OB vs. C (1.7-fold) ↑ FPG in OB vs. C (1.4fold) ↑ endonuclease (ENDO) in OB vs. C (1.5-fold)

19 (M)

↑ AA-FTO genotype and allele A OB♀ (1.2-fold) ↓ TT-FTO genotype and allele T in morbidly OB♀ (1.2-fold)

[45]

3

Unless otherwise indicated comet assays were conducted under standard condition (SC), alkaline version which detects single/ double and apurinic sites.

CC E

2

PT

Abbreviations: BMI, body mass index; C, control; FPG, formamido pyrimidine glycosylase; LPO, lipid peroxidation; MN, micronuclei; MDA, malondialdehyde; NMO, normal weight obese; n, number of participants; n.s., not specified; OB, obese; OW, overweight; 8-OHdG, 8-hydroxy-2’-deoxyguanosine; SC, standard conditions; T2D, type 2 diabetes; wk, week; WC, waist circumference; high (H), medium (M) and low (L) refer to classification on the basis of quality scoring (QS). (For details see supplementary Table)

A

Normal weight obese (NWO): according to the article by Tomasello et al [41], NWO known as a metabolic condition which observed in women with normal index but fat mass (FM) >30%.

66

I N U SC R

Table 3. Results of cytokinesis – block micronucleus cytome (CBMN-Cyt) assay in overweight/obese subjects1 Parameter Participants and treatments (indicator cells) Cytokinesis – block micronucleus cytome (CBMN-Cyt) assay

MN (lymphocytes)

CC E

M

PT

Parallel

OB: n=33♂, 2 dietary interventions (both diets: 7000 kJ): either high protein-high red meat group (n=16), BMI=32.0 or high carbohydrate-low red meat diet group (n=17), BMI=31.0 age (all groups) 20-65 OB: n=36♂/47♀, BMI= 38.0, age 37.9±10.5 OW: n=10♂/11♀, BMI= 26.0, age 40.5±10.6 C: n=7♂/14♀, BMI= 22.0, age 34.8±11.5

ED

Intervention MN (randomized (lymphocytes) trial)

A

Study design

A

Parallel

Parallel

MN (lymphocytes)

OW/OB: n=14♀/♂, BMI>25 C: n=23, 20
MN (lymphocytes)

OB: n=33♂, BMI= 33.6 OW: n=55♂, BMI= 27.9, age elderly population (all groups) 68.0±6.8

Results

QS

Remarks

Ref

↔ MN (after weight loss) ↔ MN (by diet and time) ↑ nuclear buds during intervention (specific diet)

17 (M)

only 1000 lymphocytes scored for MN ↓ weight loss by both interventions after 12 wks (12%)

[46]

↑ MN in OB vs. C (1.7-fold) ↑ MN in OB vs. OW (1.5-fold) ↑ nucleoplasmic bridges in OB vs. C (1.7-fold) ↑ nucleoplasmic bridges in OB vs. OW (1.5-fold) ↑ nuclear buds in OB vs. C (2.2-fold) ↑ nuclear buds in OW vs. C (1.6fold) ↔ MN in OW/OB vs. C

16 (M)

only 1000 lymphocytes scored for MN ↓ 8OHdG in OB vs. C (7%) ↔ correlation waist to hip ratio and CBMN assay parameters and 8-OHdG (+) correlation between MN and BMI in OW and OB (+) correlation between BMI and nuclear buds in OW and OB

[47]

15 (M)

↔ gender ↔ age ↔ smoking ↔ effect of mobile phone

[48]

↔ MN in OB vs. OW

14 (M)

↔ smoking ↔ age ↔ BMI lower DNA damage in former uranium workers who received X-ray examination

[49]

67

I N U SC R

Table 3. Continued

Study Parameter Participants and treatments design (indicator cells) Cytokinesis – block micronucleus cytome (CBMN-Cyt) assay

Parallel

MN (buccal cells)

Crosssectional

MN (buccal cells)

OB: n= 31♂/30♀, BMI= 29, age: 10.8±3.4 OW: n= 9♂/11♀, BMI= 24.4 age: 12.2±2.6 C: n=16♂/22♀, BMI= 17.5 age:10.8±2.6 Malaysian farmers ♀/♂ OB: n=3, 35≤BMI OB: n=5, 30≤BMI<34.9 OW: n=35, 25≤BMI<29.9 normal: n=21, 18.5≤BMI<24.9 UW: n=4, BMI <18.5 age (all groups) 26-65 OB: n=20, BMI=22.4 OW: n=20, BMI=18.3 C: n=20, BMI=16.2 age (all groups) 7-11

A

MN (lymphocytes)

QS

↑ MN in OW vs. C (2.5fold) ↑ MN in OB vs. C (2.7fold)

17 (M)

↑ MN in normal vs. OW (1.25-fold) ↔ MN in normal vs. OB

17 (M)

↔ MN in different groups

18 (M)

A

CC E

PT

ED

M

Parallel

Results (fold)

1

Remarks

Ref

↑ γ-H2AX foci in OW vs. C (5.6-fold) ↑ γ-H2AX foci in OB vs. C (8-fold) ↔ tumor necrosis factor-alpha in all groups ↑ interleukin 6 in OW and OB vs. C (9- fold) ↑ C-reactive protein in OW and OB vs. C (17.7-fold) only 1000 cells scored for MN ↔ binucleated between different groups ↑ MN in pesticide exposed farmers vs. C ↔ age

[50]

only 1000 cells scored for MN ↔ binucleated cells ↔ pycnosis cells ↔ broken-eggs cells ↔ karyorrhexis cells ↔ condensed chromatins ↔ total nuclear abnormalities

[52]

[51]

Abbreviations: BMI, body mass index; C, control; CBMN-Cyt, cytokinesis block micronucleus cytome assay; MN, micronuclei; n, number of participants; n.s., not specified; OB, obese; OW, overweight; SC, standard conditions; UW, under-weight; wk, week; high (H), medium (M) and low (L) refer to classification on the basis of quality scoring (QS). (For details see supplementary Table)

68

I N U SC R

Table 4. Results of 8-hydroxy-2' -deoxyguanosine (8-OHdG) in overweight/obese subjects1 Parameter (indicator cells)

Participants and treatments

8-hydroxy-2' -deoxyguanosine (8-OHdG)

A

Study design

HPLC (urine)

n=31♂/52♀, BMI= 26.0, age (all groups) 40-64

Parallel

colorimetric assay (whole blood)

OB: n=20♀, BMI ≥25 C: n=20♀, BMI <25 NWO2: n=20♀, BMI ≤25 age (all groups) 25-35 OB: n=12♂/8♀, BMI= 46.8, age 44.2±12.0 and laparoscopic gastric banding C: n=10♂/10♀, BMI= 22.5, age 40.7±12.3

ED

ELISA (plasma & urine)

CC E

PT

Parallel

M

Intervention (randomizedtrial)

HPLC (plasma)

A

Parallel

Parallel

HPLC (plasma)

coke oven workers ♂ BMI ≤ 24: n=18 exposed, 12 non exposed BMI > 24: n=29 exposed, 19 non exposed, age according to BMI (n.s) OW and OB: n=6♂/52♀, BMI=31.9 C: n=3♂/17♀, BMI=22.3 age (all groups) 46-54

Results

QS

Remarks

↓ 8-OHdG in high BMI (4%) ↑ 8-OHdG in ♂ vs. ♀(1.3fold) ↔ 8-OHdG

18

↔ intake of vitamin C, E, A on DNA damage [53] ↑ 8-OHdG in smokers (2.0-fold)

(H) 14

(M) ↑ urinary 8-OHdG in OB vs. C (2.6-fold) ↔ plasma 8-OHdG ↓ urinary 8-OHdG after surgery (59%) ↓ plasma 8-OHdG after surgery (50%) ↓8-OHdG with BMI (not significant)

14

↑ 8-oxodG in OW and OB vs. C (2.0-fold)

17 (M)

69

(M)

15

(M)

Ref

↑ FPG NWO vs. C (1.7-fold) ↑ FPG OB vs. C (1.6-fold) ↑ DNA damage NWO vs. C (1.5-fold) ↑ DNA damage OB vs. C (1.5-fold) ↓ BMI after surgery (20%) ↓ body weight after surgery (19%)

[41]

↑ 8-OHdG and glutathione S-transferase in the exposed group (not significant)

[55]

↑ LPO in OW and OB vs. C (3.0-fold) (+) correlation 8-oxodG and BMI, WC, hip circumference, triglycerides (+) correlation LPO and BMI

[56]

[54]

I Study design

Parameter (indicator cells)

Participants and treatments

ELISA (plasma)

Parallel

ELISA (urine)

OB: n=36♂/47♀, BMI= 38.0, age 37.9±10.5 OW: n=10♂/11♀, BMI= 26.0, age 40.5±10.6 C: n=7♂/14♀, BMI= 22.0, age 34.8±11.5

↓ 8-OHdG in OB vs. C (7%)

QS

Remarks

Ref

17 (M)

↔ association waist to hip ratio and CBMN assay parameters and 8-OHdG ↑ MN in OB vs. C (1.7-fold) ↑ MN in OB vs. OW (1.5-fold) ↑ nucleoplasmic bridges in OB vs. C (1.7-fold) ↑ nucleoplasmic bridges in OB vs. OW (1.5-fold) ↑ nuclear buds in OB vs. C (2.2-fold) ↑ nuclear buds in OB vs. C (1.6-fold) (+) correlation between MN and BMI in OW and OB (+) correlation between BMI and nuclear buds in OW and OB association between 8-oxoGuo and waist-hip ratio association between 8-oxodG and adrenaline (↔) no association between 8-oxoGuo and 8-oxodG with leptin, adiponectin and high sensitive-Creactive protein

[47]

CC E

1

PT

ED

M

Parallel

Results

A

8-hydroxy-2' -deoxyguanosine (8-OHdG)

N U SC R

Table 4. Continued

OB hypertensive (OB-H): n=63♂, BMI≤30 OB normotensive (OB-N): n=40♂, BMI≤30 lean normotensive (L-N): n=27♂, 20< BMI≤25 age (all groups) 39-58

↔ 8-oxodG ↑ 8-oxoGuo in OB-H vs OB-N (1.1-fold) ↑ 8-oxoGuo in OB-N vs LN (1.2-fold) ↑ 8-oxoGuo in OB-H vs LN (1.3-fold)

14 (M)

[57]

A

Abbreviations: BMI, body mass index; C, control; ELISA, enzyme linked immunosorbent assay, FACS, fluorescence-activated cell sorting; FPG, formamido pyrimidine glycosylase; HPLC, high-performance liquid chromatography, LPO, lipid peroxidation; MN, micronuclei; MDA, malondialdehyde; NWO, normal weight obese; n, number of participants; n.s., not specified; OB, obese; OW, overweight; 8-OHdG, 8-hydroxy-2’-deoxyguanosine; SC, standard conditions; wk, week; WC, waist circumference; high (H), medium (M) and low (L) refer to classification on the basis of quality scoring (QS). (For details see supplementary Table) 2

Normal weight obese (NWO): according to the article by Tomasello et al [41], NWO known as a metabolic condition which observed in women with normal index but fat mass (FM) >30%.

70

I N U SC R

Table 5. Results of Gamma-H2AX (γ-H2AX) in overweight/obese subjects1 Participants and treatments

Parallel

FACS (lymphocytes)

OB: n= 11♂/12♀, BMIZscore2=2.8,age:11.0±0.8 C: n=7♂/8♀, BMIZ-score =0.7 age: 12.4±0.4

Parallel

FACS (lymphocytes)

M

A

Parameter (indicator cells) Gamma-H2AX (γ-H2AX)

ED

Study design

QS

Remarks

Ref

↑ γ-H2AX foci in OB vs. C after 2 hours inducing DNA damage by mitomycin C (2.0-fold) ↑ γ-H2AX foci in OB vs. C after 4 hours inducing DNA damage by mitomycin C (1.7-fold) ↑ γ-H2AX foci in OW vs. C (5.6-fold) ↑ γ-H2AX foci in OB vs. C (8.0-fold)

14 (M)

↑ expression of the high mobility group box-1 protein (HMGB1) involved in doublestrand breaks repair in OB

[58]

15 (M)

↑ MN in OW vs. C (2.5-fold) ↑ MN in OB vs. C (2.7-fold) ↔ TNF-α in all groups ↑ IL-6 in OW and OB vs. C (9.0- fold) ↑ CRP in OW and OB vs. C (17.7-fold)

[50]

↓abasic sites in DNA at 12 wks (30%) ↓abasic sites in DNA at 52 wks (65%)

-

↑ telomere lengths after weight loss (not significant)

CC E

PT

OB: n= 31♂/30♀, BMI= 29, age: 10.8±3.4 OW: n= 9♂/11♀, BMI= 24.4 age: 12.2±2.6 C: n=16♂/22♀, BMI= 17.5 age:10.8±2.6

Results

Miscellaneous

A

Intervention

test kit for abasic sites (apyrimidinic/ apurinic) (midrectal)

OB: after weight loss for 12wks (7000 kJ/day): n=45♂, BMI=31.9, age 51±1.0 and after 52 wks weight loss: n=12, BMI=31.6, age 56±1.1

1

[59]

Abbreviations:; BMI, body mass index; C, control; CRP, c-reactive protein; FACS, fluorescence-activated cell sorting; IL-6, interleukin 6; MN, micronuclei; n, number of participants; OB, obese; OW, overweight; TNF-α, tumor necrosis factor-alpha; wk, week; high (H), medium (M) and low (L) refer to classification on the basis of quality scoring (QS). (For details see supplementary Table) 2

BMI Z-score: Body mass index z-scores, also called BMI standard deviation (s.d.) scores, are measures of relative weight adjusted for child age and sex.

Table 6. Genomic instability in metabolic syndrome (MetS) patients1

71

I Parameter (indicator cells) Comet assay2

tail intensity early stage of MetS pts: (lymphocytes) n=65 ♂, BMI=33.0,age 47.4±5.5 C: n=56♂, BMI=24.5 age (all groups) 46.8±4.8 arbitrary units MetS pts: n= 38♂/27♀, (lymphocytes) BMI= 27.8 C: n=42♂/23♀, BMI= 26, age (all groups) 54-57 arbitrary units MetS pts: n=20♂/32♀, (whole blood) BMI=30.3, age 32-80 C: n=♂17/♀20, BMI=25, age (all groups) 27-70

Results

QS

Remarks

Ref

↔ DNA damage ↔ hOGG1 modified comet assay ↔ neutral comet assay

19 (M)

↓ 8-OHdG in urine MetS pts. (38%)

[65]

↑ DNA damage in MetS pts (1.7fold)

18 (M)

↓ total antioxidant capacity in MetS pts (50%)

[64]

↑ DNA damage in MetS pts (3.3fold)

20 (M)

↔ DNA damage in OB-MetS pts vs. OB-without MetS ↔ FPG in OB-MetS pts vs. OBwithout MetS ↔ endonuclease (ENDO) in OBMetS pts vs. OB-without MetS

19 (M)

↑ MN in MetS pts (2.2-fold) [63] ↑ malondialdehyde (MDA) in MetS pts (2.0-fold) ↓ superoxide dismutase in MetS pts (44%) ↓ glutathione peroxidase in MetS pts (27%) ↑ AA-FTO genotype and allele A OB♀ (1.2- [45] fold) ↓ TT-FTO genotype and allele T in morbidly OB♀ (1.2-fold)

tail intensity OB-MetS pts.: n= 30♀, (lymphocytes) BMI=45.7, age 32.4±6.8 OB-without MetS: n= 30♀, BMI=21.9 age 32.4±6.5

A

CC E

Parallel

ED

Parallel

PT

Parallel

M

A

Parallel

Participants and treatment

N U SC R

Study design

72

I N U SC R

Table 6. Continued

Study Parameter Participants and treatment design (indicator cells) Cytokinesis – block micronucleus cytome (CBMN-Cyt) assay MetS pts: n=20♂/32♀, BMI=30.3, age 32-80 C: n=17♂/20♀, BMI=25, age 27-70

A

MN (lymphocytes)

QS

Remarks

Ref

↑ MN in MetS pts (2.2-fold)

17 (M)

↑ DNA damage in MetS pts (3.3-fold) ↑ MDA in MetS pts (2.0-fold) ↓ superoxide dismutase in MetS pts (44%) ↓glutathione peroxidase in MetS pts (27%)

[63]

↔ 8-OHdG levels in different MetS groups

13 (M)

↔ oxidative stress in the different groups ↑ 8-OHdG in hypertensive pts vs. C (1.5-fold)

[66]

↓ 8-OHdG in MetS pts.(38%)

15 (M)

↔ DNA damage in lymphocytes ↔ hOGG1 modified comet assay ↔ neutral comet assay

[65]

M

Parallel

Results

HPLC (genomic and mitochondrial – mononuclear cells)

CC E

PT

Parallel

ED

8-hydroxy-2' –deoxyguanosine (8-OHdG)

A

Parallel

ELISA (urine)

MetS pts: n=108♂/77♀ number of groups of MetS: 1- n=10♂/11♀, BMI=23 2- n=28♂/12♀, BMI=28 3- n=40♂/26♀, BMI=29 4- n=24♂/20♀, BMI=32 5- n=6♂/8♀, BMI=32 C (no MetS): n=16♂/14♀, BMI= 24 age (all groups) 25-50 early stage of MetS pts: n=65 ♂, BMI=33.0,age 47.4±5.5 C: n=56♂, BMI=24.5 age (all groups) 46.8±4.8

1

Abbreviations: BMI, body mass index; C, controls; CBMN-Cyt, Cytokinesis block micronucleus cytome assay; ENDO, endonuclease; FPG, Formamidopyrimidine DNA glycosylase; HPLC, high-performance liquid chromatography; MN, micronuclei; MetS, metabolic syndrome; 8-OHdG, 8-hydroxy-2' –deoxyguanosine; n, number of participants; OB, obese; OW, overweight; 8-oxo-dG, 8-hydroxy-2' –deoxyguanosine; pts, patients; SC, standard conditions; wk, week; high (H), medium (M) and low (L) refer to classification on the basis of quality scoring (QS). (For details see supplementary Table) 2

Unless otherwise indicated comet assays were conducted under standard condition (SC), alkaline version which detects single/ double and apurinic sites

73

I Method/ Participants and treatments Results parameter Terminal deoxynucleotidyl transferase dUTP nick end labelling (TUNEL assay)

A

Study design

N U SC R

Table 7. Obesity related DNA damage in sperm

♂ in subfertile couples, n=330 OB: n=43, BMI≥30 OW: n=137, BMI=25-29.9 C: n=151, BMI=18.5-24.9 age (all groups) 37.6±6.2 Sperm chromatin structure assay (SCSA) TUNEL assay

Intervention

SCSA, DFI

Intervention

SCSA, DFI

ED

M

Parallel

PT

CC E

A

♂n=43, BMI=33-61, 14 wks residential weight loss program n=15, BMI=46.0-61.0 n=14, BMI=41.7-46.0 n=14, BMI=33.3-41.6 age (in all group) 20-59 weight of women before pregnancy: under weight (UW): n=33 BMI<18.5 normal weight (C): n=263 BMI=18.5-24.9 OW: n=22, BMI≥25.0 off spring: UW: n=11 BMI<18.5 C: n=243 BMI=18.5-24.9 OB: n=83, BMI≥25.0

QS

Remarks

Ref

↑ DFI in OB vs. C (1.5-fold) ↔ DFI in OW vs. C (1.2-fold)

16 (M)

↑ viable spermatozoa OB vs. C (30%)

[70]

↔ DFI after weight loss

16 (M)

(+) correlation DFI and BMI weight loss associated with increased testosterone levels

[71]

↑ DFI in sons of OW mothers vs. sons of C mothers (1.2-fold)

15 (M)

↑ DFI in OW vs. C (but not significant), after 20years of follow-up cohort study

[72]

Table 7. Continued

74

I N U SC R

Table 7. Continued Study design

Method/ parameter

Participants and treatments

Parallel

SCSA (flow cytometry) DFI

Parallel

SCSA DFI

Crosssectional

SCSA (flow cytometry), DFI

PT

CC E A

♂ in subfertile couples, n=153 OB: n=16, BMI≥30, age 32.5±4.0 OW: n=63, BMI=25.0-29.9, age 32.8±6.7 C: n=74, BMI<24.9, age 30.2±5.9 ♂n=287, C: n=91 OW: n=136 OB: n=59 age (all groups) 23-45 ♂n=468, n=72, BMI≥35 n=122, BMI=30-35 n=191, BMI=25-29 n=83, BMI<25 age (all groups) 31.8 ± 4.8 n=175♂, BMI=25.5 age (all groups) 25-60

QS

Remarks

Ref

↔ DFI

13 (M)

↔ levels of chromatin integrity impairment ↑ cells with defective chromatin condensationin OB vs. C ↔ BMI and basic semen parameters, chromatin integrity and chromatin condensation

[73]

↔ DFI in different BMI ↑ high DNA sustainability in OB vs OW (4-fold) ↔DFI in different BMI

15 (M)

↑ cell phone use (11-25 years) vs (6-10 years) ↑ occupational stress (work)

[74]

14 (M)

(-) association WC and sperm count no significant association body size and semen concentration , motility, vitality or morphology

[75]

↔ BMI and DFI

12 (L)

(-) significant between association DFI and age

[76]

M

SCSA (flow cytometry) DFI

ED

Parallel

A

Sperm chromatin structure assay (SCSA)

Results

75

I Study Method/ Participants and treatment design parameter Sperm chromatin structure assay (SCSA)

♂;cohort A: n=275, age 18 cohort B: n=304 Swedish military recruits, BMI=22.6, age 18 cohort C: n=724 fertile men and men without fertility problems, BMI=25.3, age 18-69 cohort D: n=200 of men without known fertility problems, BMI=24.6, age 19-40

A

SCSA DFI

ED

M

Crosssectional

N U SC R

Table 7. Continued

Results

QS

Remarks

Ref

↑ DFI in C vs. OW (when excluding cohort C, the significance was lost)

14 (M)

↑ progressive motile spermatozoa in OW vs. C (when excluding cohort C, the statistical significance was lost)

[77]

A

CC E

PT

Abbreviations: AU, arbitrary unit; BMI, body mass index; DFI, DNA fragmentation index; N, number; n.s. , not specified; OB, obese; OW, over weight; SCSA, sperm chromatin structure assay; TUNEL, terminal deoxynucleotidyl transferase dUTP nick end labelling; UW, underweight; WC, waist circumference; Y, year; high (H), medium (M) and low (L) refer to classification on the basis of quality scoring (QS) (For details see supplementary Table)

76

Participants and treatments

I

Method/ parameter

Results

N U SC R

Study design Comet assay

♂ in subfertile couples, n=413 ↔ DNA damage between high obese (OB): n=40, BMI=39.4, groups age 35.7±5.6 low OB: n=87, BMI=32.0. age 37.2±5.9 overweight (OW): n=233, BMI=27.0, age 36.3±5.2 control (C): n=123, BMI=23.0, age 35.7 ±5.2 Parallel standard ♂ in subfertile couples ↑ DNA damage in OB vs. C comet assay OB: n=36, BMI≥30, age 34.3±4.9 (1.8-fold) (arbitrary OW: n=187, BMI=25.0-29.9, age ↑ DNA damage in OW vs. C units) 34.7±7.9 (1.1-fold) C: n=82, BMI<24.9, age 33.5±6.1 Terminal deoxynucleotidyl transferase dUTP nick end labelling (TUNEL assay) neutral comet assay (tail intensity)

Remarks

Ref

19 (M)

↑ estradiol in both OB groups vs. C (1.2-fold) ↓ total testosterone in both OB groups vs. C (36%) ↑ sex hormone-binding globulin in both OB groups vs. C (40%)

[67]

18 (M)

↓ mitochondrial activity in OB vs. C (27%) ↓ sperm motility in OB vs. C (17%)

[68]

14 (M)

↓ viable spermatozoa in OB vs. C ↑ spermatozoa with decondensed chromatin in OB and OW vs. C (2.0-fold) ↑ spermatozoa with low mitochondrial membrane potential OB vs. C (14.0-fold) ↑ spermatozoa with low mitochondrial membrane potential OW vs. C (10.0-fold)

[69]

PT

TUNEL assay, flow cytometry

♂ healthy non-smokers, n=150 OB: n=50, BMI≥30, 31.6 ± 1.7 OW: n=50, BMI=25.0-29.9, age 31.2 ± 1.2 C: n=50, BMI=19.0-24.9, age 31.5 ± 1.1

↑ DFI in OB vs. C (3.0-fold) ↔ DFI in OW vs. C (1.5fold)

A

CC E

Parallel

ED

M

A

Parallel

QS

Table 8. Results of comet assay in animal studies concerning the impact of obesity on DNA stability1

77

I Zucker rats

Results

QS

Remarks

Ref

obese (OB) and control (C), n=5 p.g.♀ 3, 6, 12 months old weight after 12 months: OB: 747g; C: 259g OB: n=40♂, C: n=10♂ fed either with a hypercaloric (weight 620g) or a standard (STD) diet (weight 450g) for 30 wks after 20 wks , half of the OB rat were fed a 25%-caloric restriction diet (CRD) of STD for 10 wks n=6 p.g.♂ fed either STD or cafeteria diet (CAF) weight after 13 wks: STD: 44g, CAF:46g

↑ DNA damage in peripheral blood cell and liver OB vs. C (2.0-fold) ↔ DNA damage in kidney, brain and heart OB vs. C

13 (M)

↑ DNA damage in old rats vs. younger rats (3.0-fold in blood cells)

[94]

↑ DNA damage in OB vs. CRD (2.1-fold) ↑ DNA damage in OB vs. STD (10.0-fold)

13 (M)

↑ malondialdehyde (MDA) in OB vs. STD (2.2-fold) ↑ MDA in OB vs. CRD (1.8-fold) ↑ body weight in OB vs. STD animal lost weight after CRD vs. OB

[95]

↑ DNA damage in kidney CAF vs. STD (3.1-fold) ↑ DNA damage in peripheral blood CAF vs. STD (5.0-fold) ↑ DNA damage in liver CAF vs. STD (3.8-fold) ↑ DNA damage in brain CAF vs. STD (5.2-fold)

14 (M)

[96]

n=5p.g. ♂ and ♀ fed with either HFD (60% fat) or STD weight after 12 wks: ♂. OB: 43g; STD: 32g ♀. OB: 30g; STD:23g

↑ DNA damage in liver OB vs. STD in ♂ (1.6fold) and ♀ (2-fold) ↑ DNA damage in colon OB vs. STD in ♂ (1.5fold) and ♀ (1.5-fold) ↑ DNA damage in brain OB vs. STD in ♂ (1.9fold) and ♀ (1.7-fold) ↔ DNA damage in blood OB vs. STD in ♂ and ♀

15 (M)

↔ ratio polychromatic: normochromatic erythrocytes from bone marrow ↑ MN in CAF vs. STD (4.0-fold) ↑ oral glucose tolerance OB vs. STD (2.0-fold) ↑ TNF-α in plasma OB vs. STD in ♂ (1.9-fold) and ♀ (2.1-fold) ↑ MCP-1 in plasma OB vs. STD in ♂ (1.9-fold) and ♀ (3.3-fold) ↑ insulin in plasma OB vs. STD in ♂ (9.0-fold) and ♀ (9.9-fold) only abstract available

M

ED

tail moment (kidney, liver, heart, brain, blood cells) Wistar rats

N U SC R

Experimental design

A

Animal/parameter (indicator cells) comet assay2

CC E

PT

tail intensity (lymphocytes)

Swiss albino mice

A

arbitrary units (peripheral blood, kidney, liver, brain) C57BL/6J mice tail intensity (liver, colon, brain, blood)

78

[92]

I Experimental design

C57BL/6J mice

A

tail intensity (liver, colon)

Remarks

Ref

n=14 per group.♂ fed with HFD (45% fat) or STD weight after 4 months: OB: 46g STD: 27g

↑ DNA damage in liver OB vs. STD (2.1-fold) ↑ DNA damage in colon OB vs. STD (2.4-fold)

14 (M)

[97]

n=6 per group ♂ 5-6 wks fed either STD (10% fat) or HFD (45 % fat) weight after 6 wks.: HFD: 32g, STD: 28g

↑ DNA damage in in liver STD vs. HFD ↔ DNA damage in testes and blood ↑ FPG in white blood cells and liver in STD vs. HFD (1.1-fold) ↔ FPG in testes

14 (M)

↓ gene expression of MLH1 in liver OB vs. STD (50%) ↔ gene expression of MLH1 in colon ↓ gene expression of IL-6 in colon OB vs. STD (70%) ↓ gene expression of Dnmt1 in colon OB vs. STD (60%) ↓ gene expression of Dnmt1 in liver OB vs. STD (50%) ↓ bacterial abundance in OB vs. STD ↑ haemoglobin glycidamide adducts in HFD vs. STD ↑ weight gain HFD vs. STD (1.5-fold) glycidamide-induced sperm DNA fragmentation higher in HFD vs. STD ↔DFI in sperm HFD vs. STD

n=14 per group ♂ fed with STD or HFD (45% fat) weight after 4 months: OB: 46g, STD : 27g

↑ DNA damage in liver OB vs. STD (2.3-fold) ↑ DNA damage in colon OB vs. STD (2.6-fold)

14 (M)

↓ gene expression of MLH1 in liver OB vs. STD (49%) ↔ gene expression of MLH1 in colon OB vs. STD ↓ gene expression of Dnmt1 in colon OB vs. STD (61%) ↓ gene expression of Dnmt1 in liver OB vs. STD (61%) ↓ bacterial abundance in OB vs. STD ↔ gene expression Nrf2 and MGMT

[98]

ED PT

CC E

tail intensity (testes, blood, liver) and SCSA (sperm)

C57BL/6J mice

QS

M

tail intensity (liver, colon)

C57BL/6J mice

Results

A

Animal/method/parameter (indicator cells) Comet assay

N U SC R

Table 8. Continued

79

[93]

I N U SC R

Table 8. Continued

Experimental design

Results

QS

Remarks

Ref

WNIN/Ob rat (Met S- model)

n=6 per group ♂, OB: WNIN/Ob-obese (3-months) C: WNIN/Ob-lean (3months)

↑ DNA damage (SSB) in hippocampus in OB vs. C (1.2-fold) ↑ DNA damage (SSB) in neocortex in OB vs. C (1.4-fold) ↑ DNA damage (double strand breaks (DSBs) in hippocampus in OB vs. C (2.0-fold) ↑ DNA damage (DSBs) in neocortex in OB vs. C (1.9-fold)

13 (M)

↑ MDA in OB vs. C (1.5-fold) ↓ superoxide dismutase in OB vs. C (50%) ↓ activity of catalyse in OB vs. C (44%) weight (n.s)

[88]

M

ED

tail moment (neocortex & hippocampus)

A

Animal/method/parameter (indicator cells) Comet assay

1

Unless otherwise indicated comet assays were conducted under standard conditions (SC), alkaline version which detects single/ double and apurinic sites

A

2

CC E

PT

Abbreviations: C, controls; CAF, cafeteria diet; CBMN-Cyt, cytokinesis block micronucleus cytome assay; CRD, calorie restricted diet; DIO, diet induced obesity FPG, formamido pyrimidine glycosylase; HFD: high fat diet; IL-6, interlukin 6; MN, micronuclei; MDA, malondialdehyde; n, number of participants; OB, obese; OGG1, 8oxoguanine DNA glycosylase; OW, overweight; 8-oxodG, 8-oxo-7,8-dihydro-2' –deoxyguanosine; p.g.: per group; RSD, restricted diet; SC, standard conditions; STD, standard diet, TNF-α, tumor necrosis factor-alpha; wk, week; high (H), medium (M) and low (L) refer to classification on the basis of quality scoring (QS). (For details see supplementary Table)

80

I N U SC R

Table 9. Results of Cytokinesis – block micronucleus cytome (CBMN-Cyt) assay in animal studies concerning the impact of obesity on DNA stability Animal (indicator cells)

Experimental design

Results

QS

Remarks

Ref

↑ DNA damage in kidney CAF vs. STD (3.1[96] fold) ↑ DNA damage in peripheral blood CAF vs. STD (5.0-fold) ↑ DNA damage in liver CAF vs. STD (3.8-fold) ↑ DNA damage in brain CAF vs. STD (5.2fold) ↑ oral glucose tolerance test OB vs. STD the promoters of p16 and Dapk genes were [99] methylated in the livers of C57BL/6J mice fed a HFD (irradiated and non-irradiated) Mgmt promoter was methylated in irradiated and HFD mice.

11 (M)

n=5, C57BL/6J DIO: fed for 3 wks, age=14 wks C3H/HeJ: fed for 12 wks, age=5 wks both strains fed either HFD (60% fat) or STD (10 % fat) gender (n.s) n=36♂ fed either with HFD (60% fat) or with LFD (10% fat) for 18 wks, weight 28 g treated either with N-ethylN-nitrosourea (ENU) or with the vehicle control for 3 days

↔ MN in HFD in vs. STD (in both strains) ↔ P/N ratio in HFD vs. STD (in both strains)

10 (M)

↔ MN-NCEs HFD vs. LFD ↔ MN RETs HFD vs. LFD ↔ MN after ENU treatment in HFD vs. LFD ↑ MN-NCEs after ENU treatment in HFD and LFD vs. control (1.1-fold) ↑ MN RETs after ENU treatment in HFD and LFD vs. control (4-fold)

9 (M)

CC E

PT

C57BL/6J DIO mice (prediabetic T2D) and C3H/HeJ mice

C57BL/6J mice

flow cytometry (blood)

A

↑ MN in CAF vs. STD (4.0 -fold) ↔ polychromatic: normochromatic (P/N) ratio in bone marrow

ED

(bone marrow)

(bone marrow)

n=6 per group ♂ fed either STD or cafeteria diet (CAF) weight after 13 wks: STD: 44g, cafeteria diet (CAF): 46g

M

Swiss albino mice

A

Cytokinesis – block micronucleus cytome (CBMN-Cyt) assay

81

↑ Pig-α mutant RBCs in HFD vs. LFD (2.5fold)

[100]

I N U SC R

Table 10. Results of 8-oxo-7,8-dihydro-2' -deoxyguanosine (8-oxodG) in animal studies concerning the impact of obesity on DNA stability1

C57BL/6J mice

n=10 per group ♀ mother: either folate-depleted (2 mg folic acid/kg diet) or folate-adequate (0.4 mg folic acid/kg diet) 4 wks before mating off-spring: at weaning (age: 22-25 days), either HFD (20% anhydrous milk fat) or C (5% anhydrous milk fat) for 5 months n=40 per group ♂ fed either with HFD (45 %) or C (12%) for 10 wks.

PT

OB Zucker fa/fa rat and lean Zucker fa/+ rats

M

HPLC (urine)

♂, OB: weight=440 g, lean (C): weight=350 g, weight loss via roux-en-Y gastric bypass surgery (n=15), weight 410 g or sham surgery (n=17), weight 580 g

ED

A

Method/parameter Experimental design Results QS Remarks (indicator cells) 8-oxo-7,8-dihydro-2' -deoxyguanosine (8-oxodG) and 8-OH-Guanosine (8-OH-Gua) and 8-oxo-7,8-dihydroguanosine (8-oxoGuo)

CC E

HPLC (subcortical regions of brain)

A

Diet induced obesity (DIO) rat HPLC (in mitochondria of heart, liver, kidney and testes)

weight: HFD: 628 g, C: 570 g

↑ 8-oxodG after sham surgery vs. C (3.5-fold) ↔ 8-oxodG in OB vs. C ↑ 8-oxoGua in OB vs. C (2.5-fold) ↑ 8-oxoGuo in OB vs. C (3-fold)

11 (M)

↓ 8-oxodG in subcortical region of HFD vs. C (not significant)

11 (M)

↑ 8-oxodG in heart of HFD vs. C (1.1-fold) ↑ 8-oxodG in kidney of HFD vs. C (1.2-fold) ↑ 8-oxodG in testes of HFD vs. C (1.2-fold) ↑ 8-oxodG in liver of HFD vs. C in 10 wks.(1.1-fold) ↓ 8-oxodG in liver of HFD vs. C in 2 and 8 wks. (25%)

11 (M)

82

Ref

↑ MDA in OB vs. C (3.0-fold) ( + ) correlation between DNA damage and basal plasma insulin values ↑ DSBs in sham surgery vs. C (3.0fold) ↑ γ-H2AX in OB rats (7.0-fold) ↓ after intervention by bypass surgery and caloric restriction (3.2-fold) ↑8-OxodG levels correlated with lower BER (not significant) ↓ BER activity in off-spring HFD vs. C in cortex (40%), hippocampus (40%) and subcortical regions (45%)

[101]

↑ MDA in liver of HFD vs. C (1.3fold) ↑ MDA in heart of HFD vs. C (1.2fold) ↔ MDA in kidneys and testes of HFD vs. C

[103]

[102]

I N U SC R

Table 10. Continued

Method/parameter Experimental design Results QS Remarks (indicator cells) 8-oxo-7,8-dihydro-2' -deoxyguanosine (8-oxodG) and 8-OH-Guanosine (8-OH-Gua) and 8-oxo-7,8-dihydroguanosine (8-oxoGuo)

A

↓ 8-OH-Gua in OB vs. C (24%)

ED

HPLC with ultraviolet (liver)

n=46♀ obese fa/fa: n=20 lean (C): n=26 mice received 7,12 dimethylbenz(α)anthracene (DMBA) and after 155 days, liver removed.

M

Zucker rat

1

11 (M)

↑ ratio of glutathione/ oxidised glutathione disulphide in OB vs. C (1.3-fold) ↓ S-adenosylmethionine/Sadenosylhomocysteine levels (45%) ↑methionine in OB vs. C (1.3-fold) ↑5-methylcytosine in OB vs. C (1.1-fold)

Ref

[104]

A

CC E

PT

Abbreviations: C, controls; CAF, cafeteria diet; CBMN-Cyt, cytokinesis block micronucleus cytome assay; CRD, calorie restricted diet; DIO, diet induced obesity; DMBA, 7,12 dimethylbenz(α)anthracene, ELISA, enzyme linked immunosorbent assay; ENU, N-ethyl-N-nitrosourea; GSH/GSSG, glutathione/ oxidised glutathione disulphide; HFD: high fat diet; HPLC, high-performance liquid chromatography; MDA, malondialdehyde; n, number of participants; OB, obese; OGG1, 8-oxoguanine DNA glycosylase; OW, overweight; 8-oxodG, 8-oxo-7,8-dihydro-2' –deoxyguanosine; 8-OH-Gua, 8-OH-Guanosine; 8-oxoGuo, 8-oxo-7,8-dihydroguanosine; RSD, restricted diet; SC, standard conditions; STD, standard diet; wk, week; high (H), medium (M) and low (L) refer to classification on the basis of quality scoring (QS) (For details see supplementary Table).

83

I N U SC R

Table 11. Results of Gamma-H2AX (γ-H2AX) in animal studies concerning the impact of obesity on DNA stability

Method (indicator cells)

Experimental design

Results

QS

Remarks

Ref

OB: n=22♂, weight 360g C: n=18♂, weight 300g either fasting or acute hyperglycemia ♂, weight 440 g, lean Zucker fa/+ rats (12 wks), weight 350g weight loss either via Roux-en-Y gastric bypass (RYGB) surgery (n=15), weight 410g or sham surgery (n=17), weight 580g

↑ γ-H2AX in OB vs. C (4.5-fold) ↑ γ-H2AX in OB/acute hyperglycemia vs. C/acute hyperglycemia (4.5-fold)

9 (L)

↑ insulin in OB vs. C (2.6-fold)

[105]

↑ DSBs in sham surgery vs. C (3.0-fold) ↑ γ-H2AX in OB rats (7.0-fold) and decrease after intervention by RYGB surgery and caloric restriction (3.2-fold)

[101]

♀, OB: n=10, lean (c): n=10, 15 wks in both the lean and obese groups were ip dosed once with SO and phosphoramide mustard (PM)

↑ γ-H2AX in OB (PM) vs. lean (PM) (1.5-fold) ↑ γ-H2AX in OB(PM) vs. OB (1.5-fold)

↑ 8-OxodG after RYGB vs. lean 11 (M) (2.5-fold) ↔ 8-OxodG in OB vs. lean ↑ 8-oxoGua in OB vs. lean (2.5-fold) ↑ 8-oxoGuo in OB vs. lean (3.0-fold) ↑ malondialdehyde in OB vs. lean (3.0-fold) (+) correlation DNA damage and basal plasma insulin ↔ DNA repair genes Prkdc, Parp1 9 (M) and Rad51 mRNA by obesity ↓relative Xrcc6 mRNA in OB vs. C (45%) ↑ relative Atm mRNA in OB vs. C (1.8-fold) ↑relative Brca1 mRNA in OB vs. C (1.3-fold)

A

Gamma-H2AX (γ-H2AX)

M

Zucker rat

OB Zucker fa/fa rat

CC E

PT

(colon and kidney)

ED

(lung)

A

Normal nonagouti (a/a; designated lean), agouti lethal yellow (KK.Cg-Ay/J; designated obese) Western blot (ovaries)

84

[106]

I Method/parameter (indicator cells) Miscellaneous

Experimental design

n=344♀ groups: LFD (3% fat) hydroxymethyluracil, weight=136 g, gas spectrometry HFD (20 % fat) weight 146 (liver and mammary g, dietary restriction (CRD, gland) 40 %) weight 113 g, C (5% fat) weight 137 g, for 2 wks. C57BL/6J mice n=36♂ fed either with HFD (60% Pig-α mutation, flow fat) or LFD (10% fat) for 18 cytometry wks, weight 28 g (in normochromatic treated either with N-ethylerythrocytes (NCEs) and N-nitrosourea (ENU) or reticulocytes(RET)) vehicle control for 3 days C57BL/6J mice n=7-9♂ fed either with HFD (60% quantitative alkaline fat) or LFD (10% fat) for 16 Southern blot wks. (liver and skeletal weight: muscles) HFD: 38g LFD: 28g

Results (fold)

QS

Remarks

Ref

↓ 5-hydroxymethyluracil in liver in CRD vs. C (43.3%) ↓ 5-hydroxymethyluracil in mammary gland in HFD vs. LFD (31.8%)

-

↓ body weight in CRD vs. HFD (28%) ↑ body weight in HFD vs. STD (1.1-fold) ↑ body weight in HFD vs. LFD (1.1-fold) ↑ body weight in HFD vs. CRD (1.3-fold)

[107]

↑ Pig-α mutation in RBCs in HFD vs. LFD (2.5-fold)

-

[100]

↑ DNA damage (mitochondrial) in HFD in liver and muscle

-

↔ MN-NCEs HFD vs. LFD ↔ MN RETs HFD vs. LFD ↔ MN after ENU treatment in HFD vs. LFD ↑ MN-NCEs after ENU treatment in HFD and LFD vs. control (1.1-fold) ↑ MN RETs after ENU treatment in HFD and LFD vs. control (4-fold) ↓ ratio glutathione/glutathione disulfide ↔APE1 in skeletal muscles in mitochondria base excision DNA repair: ↑ apurinic/apyrymidinic Endonuclease 1 in liver (2.0-fold) and skeletal muscles (1,8-fold) ,in nucleus in HFD ↑ OGG1 (8-oxoguanine DNA glycosylase/AP lyase), in skeletal muscles in nucleus (1.6-fold) and mitochondrial (1.5fold) in HFD vs. LFD ↔ OGG1 in liver (nucleus and mitochondrial) ↓ GSH/GSSG HFD vs. LFD in liver (2.0-fold) ↓ GSH/GSSG HFD vs. LFD in muscle (2.8-fold) ↑ free fatty acid HFD vs. LFD (2.0-fold) ↑ insulin HFD vs. LFD (2.0-fold) ↑ triglyceride HFD vs. LFD (2.5-fold)

A

CC E

PT

ED

M

A

Fischer rats

N U SC R

Table 11. Continued

85

[108]

I N U SC R

A

Abbreviations: C, controls; CAF, cafeteria diet; CRD, calorie restricted diet; DIO, diet induced obesity; DMBA, 7,12 dimethylbenz(α)anthracene; ENU, N-ethyl-N-nitrosourea; GSH/GSSG, glutathione/ oxidised glutathione disulphide; HFD: high fat diet; MN, micronuclei; MDA, malondialdehyde; n, number of participants; OB, obese; OGG1, 8oxoguanine DNA glycosylase; OW, overweight; 8-oxodG, 8-oxo-7,8-dihydro-2' –deoxyguanosine; 8-OH-Gua, 8-oH-Guanosine; 8-oxoGuo, 8-oxo-7,8-dihydroguanosine; PM, Phosphoramide mustard; RET, reticulocytes; RSD, restricted diet; SC, standard conditions; STD, standard diet, TNF-α, tumor necrosis factor-alpha; wk, week; high (H), medium (M) and low (L) refer to classification on the basis of quality scoring (QS) (For details see supplementary Table).

A

CC E

PT

ED

M

Table 12. Results of animal studies concerning obesity related DNA damage in sperm

86

I Zucker rat

N U SC R

Experimental design

Results

QS

Remarks

Ref

n=6 per group ♂ after 64 and 100 days OB: weight in day 64=344g, day100=490g C: weight in day 64=233g, day 100=344g

↑ DNA damage (tail length) in OB vs. C (1.3-fold) ↑ DNA damage (tail extent moment) in OB vs. C (1.8-fold) ↔ DNA damage (tail DNA and olive tail moment)

12 (M)

↑ daily sperm production in C vs. OB (1.3-fold) ↑ testes weight in C vs. OB (1.5-fold)

[113]

n=40♂, 6 wks old, HFD: n=7, STD: n=8 fed either STD (6% fat) or HFD (21% fat) weight after 18wks: HFD=34.2g, STD=28.3g

↑ sperm DNA damage (%) in OB vs. STD (1.6-fold)

10 (M)

[111]

n=36♂, 6 wks old, HFD: n=18, STD: n=18 fed either with STD (6% fat) or HFD (22% fat, 0.15% cholesterol) weight after 9 wks: HFD=30.0g, STD=29.4 g

↑ sperm DNA damage (%) in OB vs. STD (9.4-fold)

10 (M)

↑ mitochondrial ROS in OB vs. STD (1.1-fold) ↓ sperm mobility in HFD vs. STD (11%) ↑ abnormal sperm tail in OB vs. STD (1.2-fold) ↔ free fatty acid in OB vs. STD ↑ glucose in OB vs. C in OB vs. STD (1.0-fold) ↑ cholestrol in OB vs. C in OB vs. STD (1.6fold) ↑ triglyceride in OB vs. C in OB vs. STD (1.2fold) ↑ mitochondrial (ROS) in OB vs. STD (1.6fold) ↑ intracellular ROS in OB vs. STD (1.9-fold)

M

A

Animal/method/p arameter (indicator cells) Comet assay

C57BL/6J mice

PT

ED

comet assay, tail length, tail extent moment, olive tail moment and tail DNA (testes) Terminal deoxynucleotidyl transferase dUTP nick end labelling (TUNEL assay)

CC E

TUNEL assay, cell detection kit (testes)

A

C57BL/6J mice

TUNEL assay, cell detection kit (testes)

Table 12. Continued

87

[110]

I N U SC R

Abbreviations: AU, arbitrary unit; BMI, body mass index; DFI, DNA fragmentation index; N, number; n.s. , not specified; OB, obese; OW, over weight; SCSA, sperm chromatin structure assay; TUNEL, terminal deoxynucleotidyl transferase dUTP nick end labelling; UW, underweight; Y, year; high (H), medium (M) and low (L) refer to the classification

ED

SCSA, DNA fragmentation index (testes) C57BL/6J mice

♂, fed either STD (10% fat) or HFD (45 % fat) weight after 11 wks: HFD=33.6g, STD=29.4g n=9 to 14 per group

CC E

PT

comet assay (testes, blood and liver) SCSA, flow cytometry (sperm)

Results

QS

Remarks

Ref

↑ DFI in HFD vs. STD (1.1fold)

11 (M)

(+) correlation BMI and DFI (+) correlation between blood glucose and DFI

[112]

↑ DNA damage (single strand breaks and alkali-labile sites) in STD vs. HFD in liver ↔ DNA damage in testes and blood ↑ FPG in white blood cells and liver in STD vs. HFD (1.1-fold) ↔ DFI in HFD vs. STD

11 (M)

↑ haemoglobin glycidamide adduct levels in HFD vs. STD ↑ weight gain HFD vs. STD (1.5-fold)

[93]

M

C57BL/6J (wild type mice), C57BL/6J-ob/+ (leptin heterozygous mice) and C57BL/6J-ob/ob (Lepob; leptin- deficient mice)

A

Animal/method/parameter Experimental design (indicator cells) Sperm chromatin structure assay (SCSA)

n=6 per group ♂, 5-6 wks fed either STD (10% fat) or HFD (45 % fat) weight after 6 wks: HFD=31.7g, STD=28.1g

A

on the basis of quality scoring (QS) (For details see supplementary Table).

88