Genotoxic markers in patients with diabetes mellitus (Literature review)

Cover Page


Cite item

Full Text

Open Access Open Access
Restricted Access Access granted
Restricted Access Subscription or Fee Access

Abstract

This paper considers studies aimed at identifying markers of genotoxicity (chromosomal aberrations, micronuclei, and DNA damage assessed by the DNA comet assay) in patients with both gestational diabetes mellitus (GDM) and diabetes type 1 and 2 (T1DM and T2DM, respectively), as well as possible changes in the levels of these genotoxic markers under the influence of medicines and nutritions. Patients with T2DM are characterized by an increased level of genotoxicity markers. The results of genotoxicity markers in patients with T1DM and GDM studies are contradictory, however, they indicate the presence of an increased genotoxic load rather than its absence. The levels of genotoxic damage in diabetic patients may be reduced by physical exercises, diet, and/or hypoglycemic drugs. Metformin, Afobazole and Noopept are recommended for experimental and clinical studies as possible drug candidates that reduce the levels of genotoxic biomarkers in diabetic patients.

Full Text

INTRODUCTION

In 2017, there were 463 million people globally with diabetes mellitus (DM). According to forecasts the number of people with this pathology will increase steadily in the medium term and will reach 693 million by 2045 [1–3]. In the Russian Federation at the beginning of 2019, there were 4,584,575 patients with DM (3.12% of the population of the Russian Federation), including 256.2 thousand patients with type 1 diabetes (T1DM), 4.24 million people with type 2 diabetes (T2DM), (92.49% of the total number of DM patients), and 89.9 thousand patients with other types of DM. According to the statistics, since 2000, the number of DM patients in Russia has grown 2.2-fold [4].

General ideas about the pathogenesis of DM and its complications have been discussed repeatedly and in detail in the modern literature. They are summarized schematically in Figure 1 [5–9]. Oxidative stress caused by hyperglycemia and an integral part of the pathogenesis of DM, as well as the associated processes of lipid peroxidation, represents a source of reactive oxygen species and overoxidized lipids. Many studies represent free radical DNA damage as the main mechanism of endogenous mutagenesis [10].

 

Figure 1. Medical significance of oxidative damage to nucleic acids, lipids, and proteins in hyperglycemia ([9], significantly modified)

 

Against the background of existing ideas about the medical significance of genotoxic lesions, their role in the occurrence of neoplasms, hereditary diseases, and miscarriages [11], we can explain the interest in the study of genotoxicity markers in DM patients.

This review aimed to analyze the results of studies performed using the most common markers of genotoxicity in patients with T1DM, T2DM, and gestational diabetes and to discuss possible methods of reducing genotoxicity in such category of patients.

MATERIALS AND METHODS

Literature search was performed from January 1, 1990 to January 31, 2021 using the MedLine/PubMed scientific literature database (National Library of Medicine, National Institutes of Health, Bethesda, Maryland, USA, http://www.ncbi.nlm.nih.gov/PubMed) and the scientific electronic library of the Russian Science Citation Index (http://elibrary.ru). The search was performed using the keywords “diabetes”, “micronuclei”, “DNA comet”, “chromosomal aberrations”, and the corresponding Russian-language translations for Russian sources. When searching the PubMed database, we additionally used the “humans” filter. We considered studies published both in Russian and English languages for which full-text versions of articles were available, as well as those indexed by DOI for English sources.

Articles focused on identifying the most verified and widely used genotoxic markers in peripheral blood lymphocytes were recognized as acceptable for registration and analysis, subject to the following criteria:

1) the balance of the populations surveyed by gender and age in the group of patients and healthy volunteers, each of which exceeded 10 people;

2) the use of verified cytogenetic research methods [chromosomal aberrations (CA) test, the cytokinesis-block micronucleus (MN) cytome assay, registration of DNA damage by the DNA comet assay];

3) availability of acceptable statistical analysis, presentation of mean values for groups with standard errors or standard deviations;

4) compliance with ethical standards during the study, approval of the study protocol by the Ethics Committee.

From the full-text versions of the articles, information was selected about the research subjects (the number of subjects in each group, gender, age, duration of disease, concomitant diseases and medications, smoking status, alcohol consumption, and nutritional quality) and biomarkers studied, as well as the actual research results, such as “mean ± SD”. Only publications containing a detailed description of the study design and results were considered in detail.

During the discussion of the results, the data obtained in the analysis of genotoxicity markers in the cells of the buccal and lingual epithelium were also recorded, as well as other results of interest from the perspective of assessing the modification of genotoxicity in this category of patients.

RESULTS

An electronic search in the MedLine/PubMed database yielded a total of 598 publications, and among them, 50 original research articles were selected, on the basis of full-text reviews, that met the selected inclusion criteria. MN or CA in peripheral blood lymphocytes (PBL) were used as biomarkers of genotoxicity in 14 cases, and DNA damage assessed by the DNA comet assay in 36 cases. The information collected is summarized in Tables 1 and 2.

 

Table 1. Results of studies of cytogenetic markers of genotoxicity in lymphocytes of patients with diabetes mellitus

First author, country 1 and year of publication

Source

Group size

Average age, years

Disease duration, years

HbA1c, mg/dl

Characteristics of the studied cohort of patients (concomitant diseases, drugs taken), comments

“Number of MN per 1000 binuclear cells, ‰” or “Number of CA per cell” (M ± SD)

Multiplicity of excess

P (m/f)

C (m/f)

P

C

P

C

Patients with gestational diabetes mellitus

M. Toljic et al., Serbia, 2017

[14]

37 (0/37)

31 (0/31)

33 (24–40)

31 (20–44)

Not applicable

Pregnant women who had not previously been diagnosed with DM were included; blood was sampled in the period between weeks 24 and 28. A group of patients with mild gestational hyperglycemia was also included

12.76 ± 6.31**

6.32 ± 3.37

2.02

M. Witczak et al., Poland, 2017

[15]

35 (0/35)

30 (0/30)

32 ± 4

30 ± 7

Not applicable

5.2 ± 0.5

Pregnant women with GDM were included. The level of CA in the blood of their newborns was also assessed

0.0065 ± 0.0138 (CA)

0.0045 ± 0.009 (CA)

Patients with type 1 diabetes mellitus

O. Mihaljevic et al., Serbia, 2018

[21]

13 (6/7)

19 (9/10)

10.4 ± 3.9

11.9 ± 3.7

10.24 ± 2.03

7.77 ± 2.95**

2.10 ± 1.99

3.7

M. Witczak et al., Poland, 2014

[22]

17 (0/17)

40 (0/40)

30 ± 4

31 ± 6

16 ± 7.6

6.3 ± 1.0

The study included women with type 1 DM in trimester I of pregnancy, taking folic acid at a dose 400 µg. The umbilical cord blood lymphocytes from newborns were also sampled

2.35 ± 1.07** (1.42 ± 0.60* in newborns)

0.86 ± 0.90 (0.67 ± 0.79 in newborns)

2.73 (2.11 in newborns)

N. Cinkilic et al., Turkey, 2009

[23]

35 (20/15)

15 (9/6)

32 ± 10

39 ± 9

1–22

8.37 ± 1.36

The study included patients in whom type 1 DM was detected before the age of 30 years, insulin therapy for up to 1 year

0.009 ± 0.012 (CA),

1.83 ± 1.15 (MN)

0.007 ± 0.009 (CA)

1.53 ± 0.64 (MN)

Patients with type 2 diabetes mellitus

M. Salimi et al., Iran, 2016

[25]

50 (22/28)

50 (25/25)

58 ± 10

57 ± 10

10–20

9.22 ± 1.76 vs. 5.0 ± 0.5 (in the control group)

Information on the drugs taken was not provided. A group of patients with T2DM and nephropathy was included separately (50 patients, m/f ratio was 32/18; average age was 60 ± 11; the duration of the disease was 1–20 years; HbA1c was 7.5 ± 1.82) 2

4,4 ± 0,7** (in the group of patients with T2DM and nephropathy 7.54 ± 2.52)

2.5 ± 0.3

1.8

M. Prasad et al., India, 2015

[26]

22 (13/9)

42 (30/12)

60 ± 12

58 ± 11

0.08–20

Information on the drugs taken was not provided. The additional group included 20 patients with peripheral neuropathy, with a m/f ratio of 7/13; average age of 58 ± 11; and disease duration of 10–20 years

6 ± 7**

(in the group of patients with neuropathies 5 ± 0.5)

0.3 ± 0.5

20.0

M.K. Harishankar et al., India, 2015

[46]

147 (61/86)

54 (25/29)

52 ± 1

0.5–10

Metformin and/or glimepiride. Additionally, the MN level in the cells of the urothelial epithelium was assessed

16.1 ± 0.3*

2.5 ± 0.2

6.4

R. Saraswathy et al., India, 2014

[24]

50 (31/19)

50 (31/19)

52 ± 9

52 ± 9

9.02 ± 6.78

Information on the drugs taken was not provided. 50 patients with diabetic neuropathy were also included, with a m/f ratio of 31/19; average age of 58 ± 11; and disease duration of 0.5–30

0.03 ± 0.02* (CA)

(0.086 ± 0.04** in the group of patients with neuropathy)

0.014 ± 0.0001

2.1 (6.1)

S.C. Corbi et al., Brazil, 2014

[41]

30 (12/18)

30 (10/20)

30 (12/18)

48 ± 8

50 ± 7

39 ± 4

6.2 ± 4.2

5.2 ± 6.6

10.4 ± 1.9 (6.6 ± 0.9) vs. 5.4 ± 0.21 (in the control group)

Patients with dyslipidemia and periodontitis with high and low levels of glycated hemoglobin were included

5.42 ± 3.80** (8.17 ± 5.41)

1.57 ± 0.79

3.4

D.N. Binici et al., Turkey, 2013

[27]

50 (15/35)

30 (8/22)

58 ± 13

44 ± 14

5.4 ± 4.3

8.93 ± 2.56

Sulfonylurea and/or metformin

3.5 ± 1.0**

1.8 ± 0.7

1.9

S.K. Shettigar et al., India, 2012

[29]

25 (14/11)

24 (11/13)

58 ± 11 (m) / 51 ± 15 (f)

56 ± 10 (m) / 49 ± 7 (f)

Information was not provided

9.7 ± 1.3 vs. 7.9 ± 1 (in the control group)

Information was not provided

No differences were revealed

11.3 ± 3.5 (m) / 11.0 ± 3.9 (f)

9.3 ± 3.1 (m) / 11.8 ± 4.6 (f)

R.P. Palazzo et al., Brazil, 2012

[28]

22 (12/10)

22 (5/17)

63 ± 9

63 ± 8

1.5 ± 0.9

Patients receiving maintenance hemodialysis therapy were included; the intake of ACE inhibitors, hypoglycemic drugs, diuretics, in some cases, recombinant erythropoietin and iron hydroxide

5.5 ± 4.0*

3.5 ± 2.8

1.6

S.A. Supriya et al., India, 2011

[81]

46 (24/22)

25 (16/9)

55.2

50.5

Over 8 years

Patients with autonomic neuropathy (varying degrees of coronary artery disease) were included

15.2 ± 2.3*

10.6 ± 1.2

1.4

L.M. Martínez-Pérez et al. Mexico, 2007

[82]

15 (5/10)

10 (2/8)

49 ± 7

46 ± 4

Information was not provided

Oral hypoglycemic drugs

6.53 ± 2.03**

3.10 ± 1.79

2.1

* p < 0.05; ** p < 0.001. 1 State where patients were enrolled in the study. 2 Hereinafter, in brackets, the data are presented as the number of patients in the group (men/women); average age; diabetes duration; and the level of glycated hemoglobin, mg/dl.

Note. SD – standard deviation; P – patient group, C – control groups, PBL – peripheral blood lymphocytes, HbA1c – glycated hemoglobin, MN – micronucleus, CA – chromosomal aberrations, GDM – gestational diabetes mellitus. Studies that revealed no statistically significant difference between the groups of patients and healthy volunteers are highlighted in color.

 

Table 2. Results of studies of genotoxic markers in lymphocytes of patients with diabetes mellitus by the DNA comet assay (alkaline version, unless otherwise indicated)

First author and year of publication

Source

Group size

Age, years

Disease duration, years

HbA1c, mg/dl

Characteristics of the studied patient population (concomitant diseases, medications taken), comments

Multiplicity of excess of % DNA in the comet tail, significance of differences from the control group

patients (m/f)

control (m/f)

patients (m/f)

control (m/f)

Patients with gestational diabetes mellitus

R.B. Gelaleti et al., Brazil, 2015

[16, 83]

46 (0/46)

41 (0/41)

31 ± 5

30 ± 6

Not applicable

6.33 ± 0.90

Pregnant women who had not previously been diagnosed with DM were included; blood was sampled in the period between weeks 24 and 28. A group of female patients with mild gestational hyperglycemia was also included (24; 32 ± 4; 5.74 ± 0.67)

Only the presence of a difference was mentioned, without specifying the multiplicity of the excess *

J. Basu et al.,

India, 2018

[17]

114 (0/114)

114 (0/114)

26 ± 5

24 ± 4

Not applicable

6.00 ± 0.66

Pregnant women who had not previously been diagnosed with DM were included; blood was sampled in the period between weeks 24 and 28

The authors register an approximately 8–10-fold increase in the level of DNA damage, but the registration of the result differed from that recommended ** [63]

Patients with type 1 diabetes mellitus

O. Mihaljevic et al.,

Serbia, 2018

[21]

13

19 (9/10)

10.4 ± 3.9

11.9 ± 3.7

10.24 ± 2.03

The authors recorded an approximately 2–5-fold excess of the level of DNA damage; however, registration of the result differed from that recommended ** [63]

N. Wyatt et al.,

UK, 2006

[84]

11

24

18–77

18–77

23.7

Metformin (dosages not specified)

~1.5*

J. Varvarovská et al., Czech Republic, 2004

[85]

50 (29/21)

30

11.96 ± 4.69

4.25

9.5 ± 2.5

No differences were revealed

Y. Dinçer et al., Turkey, 2003

[39]

45 (21/24)

40 (17/23)

32 ± 8

35 ± 6

9 ± 6

7.04 ± 0.85

Among the complications, microangiopathies, retinopathies, and neuropathies were observed

The authors recorded an approximately 1.2–1.5-fold excess of the level of DNA damage; however, the registration of the result differed from that recommended ** [63]

S. Sardaş et al.,

Turkey, 2001

[51]

15

30

Information was not provided

Information was not provided

5.2 ± 8.1

The study included smokers (26 in the T2DM group, 11 in the control). No medication information was provided. Vitamin E supplementation reduced the level of DNA damage

The authors recorded an approximately 1.7-fold excess of the level of DNA damage; however, the registration of the result differed from that recommended * [63]

M.P. Hannon-Fletcher et al., UK, 2000

[43]

50 (30/20)

50 (28/22)

36 ± 2

38 ± 1

7.71 ± 0.03

Thirteen diabetic patients had at least one complication, namely retinopathy, nephropathy, neuropathy, and macrovascular diseases, eight patients were smokers. No significant differences were revealed in lymphocytes, monocytes, and whole blood; however, in the neutrophil fraction, the levels of DNA damage were significantly higher (p < 0.001) than those in the control group

No differences were revealed

S. Astley et al.,

UK, 1999

[42]

49 (31/18)

42 (20/22)

39 ± 7

40 ± 9

15.2 ± 5

9.0 ± 1.2

Thirteen patients had a history of retinopathy, and eight had patients had neuropathy. Vitamin E supplementation (400 IU/day) for 8 weeks did not have a significant effect on the level of DNA damage

No differences were revealed

A.R. Collins et al.,

Slovakia, 1998

[86]

10 (10/0)

10 (10/0)

48 ± 8

43 ± 6

15.6

11.0 ± 2.9

Eight patients had chronic complications (neuropathy and/or retinopathy); 2 patients had no diabetic complications

2.3 *

D. Anderson et al., UK, 1998

[61]

22 (11/11)

20 (9/11)

55 ± 7

50 ± 13

Over 5 years

7.61 ± 0.31

Information on the drugs taken was not provided

No differences were revealed

Patients with type 2 diabetes mellitus

A. Rao et al.,

India, 2020

[87]

50

50 (+50)

30–60

30–60

Information was not provided

<7

The experimental group included patients with T2DM and periodontitis without any concomitant systemic diseases. The control group consisted of both healthy volunteers and patients with periodontitis, but without T2DM or other systemic diseases

No differences were revealed (however, the mean values were higher in the T2DM group, p = 0.515)

A. Raghav et al.,

India, 2018

[88]

100

50

56 ± 10

57 ± 12

Information was not provided

Information was not provided. Half of the DM patients also had a history of chronic kidney disease

3.5–4.8 ** (depending on the absence or presence of kidney pathologies)

M. Pittaluga et al., Italy, 2015

[58]

12 (12/0)

12 (12/0)

62 ± 4

61 ± 4

Over 5 years

6.7 ± 0.2 vs. 5.5 ± 0.1 (in the control group)

Metformin as monotherapy or in combination with repaglinide or gliclazide. Significant (p < 0.05) decrease from baseline after 4 months of moderate training

No differences were revealed

S.A. Bukhari et al., Pakistan, 2015

[89]

80 (40/40)

80 (40/40)

Cohorts included young and elderly patients

Cohorts included young and elderly patients

Information was not provided

Over 7.5

Information on the drugs taken was not provided

The authors register an approximately 2–3-fold excess of the level of DNA damage; however, the registration of the result differed from that recommended ** [63]

A. Merecz et al., Poland, 2015

[90]

120 (67/53)

146 (77/69)

64 ± 12

65 ± 15

Information was not provided

Information on the drugs taken was not provided. A group with neuropathies was also included [89 (42/47); 66 ± 11], in which an increased level of DNA damage was revealed (p < 0.001)

1.2–2 ** (depending on the genotype)

D.J. Xavier et al., Brazil, 2015

[91]

27 (11/16)

16 (7/9)

53 ± 9

53 ± 7

10.1 ± 4.0

Metformin and/or insulin

The authors recorded an approximately 2.3-fold excess of the level of DNA damage; however, the registration of the result differed from that recommended * [63]

D.J. Xavier et al., Brazil, 2014

[56]

10 (3/7)

12 (7/9)

46 ± 12

53 ± 7

6.9

9.9 ± 2.0, after treatment 9.2 ± 2.1

Insulin and/or oral hypoglycemic drugs, as well as drugs for treatment of concomitant pathologies. For 7 days, the patients adhered to a diabetic diet, after which the level of DNA damage decreased significantly (p < 0.05)

The authors recorded an approximately 2.3-fold excess of the level of DNA damage, however, the registration of the result differed from that recommended * [63]

E. Müllner et al., Austria, 2013

[48]

76 (34/42)

21 (6/15)

65 ± 7

63 ± 6

Information was not provided

7.26 ± 1.08

Oral hypoglycemic drugs and/or insulin. After 4 weeks of increased consumption of vegetables and vegetable oil, a decrease in the level of DNA damage was observed

No differences were revealed

E. Tatsch et al., Brazil, 2012

[37]

32 (14/18)

30 (16/14)

57 ± 6

54 ± 7

10.7 ± 6

7.7 ± 1.7 vs. 5.2 ± 0.7 (in the control group)

in 9% of those treated with insulin alone, in 72% of those treated with oral hypoglycemic drugs, and in 19% of those treated with a combination thereof

The authors recorded an approximately 5–6-fold excess of the level of DNA damage, however, the registration of the result differed from that recommended * [63]

R.P. Palazzo et al., Italy, 2012

[28]

22 (12/10)

22 (5/17)

63 ± 9

63 ± 8

1.5 ± 0.9

Patients receiving maintenance hemodialysis therapy, ACE inhibitors, hypoglycemic drugs, diuretics, and in some cases, recombinant erythropoietin and iron hydroxide were included

2.2 * (the version of the DNA comet assay is not specified)

S.I. Salem et al., Egypt, 2012

[92]

28 (8/20)

25 (10/15)

53 ± 5

53 ± 7

9.7 ± 2.1

9.6 ± 2 vs. 3.4 ± 0.6 (in the control group)

Patients with a history of smoking, coronary heart disease, congestive heart failure, chronic liver diseases, diabetic nephropathy, rheumatic diseases, and cancer, as well as subjects who had recently undergone radiological procedures (1 month previously), were not included in the study. None of the patients took antioxidant supplements

12.2 **

J. Kasznicki et al., Poland, 2012

[93]

16 (6/10)

19 (10/9)

64 ± 12

65 ± 14

Information was not provided

9.44 ± 1.68 vs. 5.5 ± 0.3 (in the control group)

Information was not provided. An additional group included patients with T2DM and distal symmetric polyneuropathy, where DNA damage was significantly (p < 0.001) higher than in the group of patients with T2DM without neuropathy

3.1 *

M. Arif et al., Bangladesh, 2010

[38]

32 (18/14)

25 (15/10)

47 ± 5

49 ± 8

5.6 ± 1.8

8.1 ± 2.88 vs. 2.92 ± 0.64 (in the control group)

Information was not provided. Patients without systemic comorbidities and those not taking antioxidant drugs were included

1.8 ** (DNA comet assay is presented in a neutral environment)

V. Manfredini et al., Brazil, 2010

[94]

11

18

55 ± 7

49 ± 9

~3 years

7.8 ± 2

Metformin. Also included was a group of patients with T2DM with dyslipidemia who received simvastatin therapy (20 mg/day; n = 14) or did not receive it (n = 9); the level of DNA damage in group 1 was significantly lower than that in the group 2 and higher in both than in the control (p < 0.05)

4.0 *

E. Ibarra-Costilla et al., Mexico, 2010

[60]

71 (21/50)

14 (3/11)

40–70

40–50

Over 5 years

Over 8.88 ± 1.35

Information on the drugs taken was not provided

No differences were revealed

D.A. Vasiliev et al., Russia, 2008

[95]

17 (0/17)

14 (0/14)

57 ± 3

53 ± 1

Diagnosed for the first time

Information on the drugs taken was not provided

No differences were revealed

P.B. Bagatini et al., Brazil, 2008

[96]

25 (13/12)

20 (7/13)

63 ± 9

62 ± 9

2.13 ± 2.08

Patients who received maintenance hemodialysis therapy, ACE inhibitors, hypoglycemic drugs, diuretics, and, in some cases, recombinant erythropoietin and iron hydroxide were included

The authors recorded an approximately 1.5–2-fold excess of the level of DNA damage, however, the registration of the result differed from that recommended * [63]

M. Lodovici et al., Italy, 2008

[35]

39 (16/23)

18 (10/8)

41–79

35–70

Information was not provided

7.16 ± 0.19

Metformin as monotherapy or in combination with glibenclamide

2.1 *

A. Sliwinska et al., Poland, 2008

[97]

30 (15/15)

30 (12/18)

60 ± 10

64 ± 12

11.0 ± 7.6

8.1 ± 1.7

Metformin and/or insulin

3.4 **

F. Song et al., China, 2007

[98]

92 (48/44)

113 (64/51)

50 ± 10

52 ± 11

Diagnosed for the first time

7.73 ± 2.19 vs. 5.00 ± 0.46 (in the control group)

Information on the drugs taken was not provided

2.6 *

J. Blasiak et al.,

Poland, 2004

[99]

52 (41/11)

55 (43/12)

61 ± 5

59 ± 6

Information was not provided

8.5 ± 0.4

Oral hypoglycemic drugs (sulfonylurea and/or metformin) and/or insulin. Neither patients nor control groups took antioxidant supplements

1.6 *

V. Pitozzi et al., Italy, 2003

[100]

14 (9/5)

14 (7/7)

62 ± 5

61 ± 4

Information was not provided

7.0 ± 0.3

Metformin as monotherapy or in combination with other drugs

No differences were revealed

Y. Dinçer et al., Turkey, 2002

[36]

63 (33/30)

41 (21/20)

52 ± 6

50 ± 8

Over 10 years

8.4 ± 2.6 vs. 5.5 ± 0.3 (in the control group)

55/63 patients had a history of complications (retinopathy, neuropathy, nephropathy, angiopathy). No medication information was provided. The patients did not take antioxidant vitamins or supplements

The authors recorded an approximately 1.8-fold excess of the level of DNA damage; however, the registration of the result differed from that recommended ** [63]

S. Sardaş et al., Turkey, 2001

[51]

48

30

Information was not provided

Information was not provided

5.1 ± 5.2

The study included smokers (26 patients in the T2DM group, 11 patients in the control). No medication information was provided. Vitamin E supplementation reduced the level of DNA damage

The authors register an approximately 2.5-fold excess of the level of DNA damage; however, the registration of the result differed from that recommended * [63]

D. Anderson et al., UK, 1998

[61]

23 (15/8)

20 (9/11)

60.5 ± 5.4

50.1 ± 12.8

Over 5 years

7.1 ± 0.5 vs. 4.4 ± 0.1 (in the control group)

Information on the drugs taken was not provided

No differences were revealed

* p < 0.05; ** p < 0.001.

Note. HbA1c – glycated hemoglobin. Studies which revealed no statistically significant difference between the groups of patients and healthy volunteers are highlighted in color.

 

The subjects of the study were patients with type 1 or 2 diabetes or gestational diabetes. Unless otherwise indicated in the table, patients with bacterial infections, cancer, or hepatitis C or B and those who were HIV positive were excluded. Patients must not have taken immunosuppressants or antibiotics for at least 1–3 months before the study and must not have undergone X-ray examination or radiation therapy for more than 6 months before the study. Control groups consisted of healthy volunteers living in the same region, matched by age and gender to the patient groups, and the presence of gestational diabetes during pregnancy. Medical history and information about recent medical procedures and medications taken were collected from study participants. In some studies that used questionnaires, data were also collected on smoking status, alcohol consumption, nutritional quality, and lifestyle. Peripheral blood was collected for research by venipuncture. The protocols of all mentioned studies were approved by the Ethics Committees.

Patients with gestational diabetes mellitus (GDM)

In 2019, 16% of pregnancies (20 million births) ended in the birth of children with hyperglycemia, mainly due to gestational diabetes [12, 13]. The search identified only four studies of genotoxicity biomarkers in patients with gestational diabetes. Two of them were focused on the analysis of cytogenetic status [14, 15], and the assessment of DNA damage was discussed in two studies [16, 17].

The results of cytogenetic studies did not coincide; however, they shared a common tendency. Some researchers [14] revealed an increase in the levels of CA in patients compared with a comparable sample of healthy pregnant women. Other researchers [15] registered a moderate but statistically insignificant increase in the level of CA when compared between pregnant women with GDM and without DM and newborns in mothers with and without GDM. The authors explained the absence of the initially assumed cytogenetic effect by a relatively short period of exposure to an increased glucose level (the average gestational period for detecting GDM is 25.4 ± 5.6 weeks), as well as by proper control over the level of glycemia during pregnancy.

An increased level of DNA damage in PBL, defined as the average value of the percentage of DNA in the comet tail, was observed in two studies [16, 17], the total sample of which consisted of 160 patients and 155 healthy pregnant women. The discovered phenomenon was explained by the authors as a consequence of hyperglycemia, obesity, hypertension, and/or insulin resistance.

Lymphocytes from infants born from GDM mothers were also examined. As a result, a positive correlation was established between the mean level of glucose in the mother’s blood and an increased level of DNA damage in the children [18]. In addition, newborns of mothers with GDM had a higher level of DNA damage in umbilical cord blood cells compared with those of mothers with euglycemia [19].

Thus, at the present stage of research development, it can be concluded that the level of genotoxicity markers is increased both in pregnant women with GDM and their newborns. It is noteworthy that these data are aligned with the results of V.V. Zabrodina, who showed an increase in the levels of DNA damage in pregnant rats and their offspring in a model of streptozotocin-induced diabetes [20]. Nevertheless, it should be borne in mind that nowadays, data of a very limited range of clinical genotoxic studies are available, and a confident conclusion about the genotoxic profile of GDM patients can only be made on the basis of additional results of new independent studies.

Patients with type 1 diabetes mellitus

As a result of the search, 12 articles were selected, including 3 studies using cytogenetic parameters in the PBL of T1DM patients as a biomarker, and 9 studies using DNA damage detected using the DNA comet assay.

In 2 of 3 cytogenetic studies [21, 22], an increased value of MN levels in T1DM patients was revealed [65 patients (26 men, 39 women) were analyzed in total] compared with control groups (74 healthy volunteers, 18 men, 56 women). Study 3 presented only a tendency toward an increase in the CA level in T1DM patients, which was not supported by statistical significance [23].

It is noteworthy that in the group of pregnant T1DM patients, the average number of MN per 1000 cells was significantly higher (p < 0.001) than that in the control group of pregnant women. A similar effect was observed in the corresponding groups of newborns (p < 0.05). At the same time, there were no significant correlations between the incidence of MN in mothers with T1DM and that in their newborns, the duration of diabetes, or HbA1c levels [22]. This finding is clearly consistent with the data obtained in the study of patients with hyperglycemia due to GDM (see above).

Analysis of DNA damage in patients with T1DM by the DNA comet assay resulted in an ambiguous picture. In four out of nine studies, there were no significant differences between the groups of patients with T1DM (265 patients in total) and the control groups of healthy volunteers (265 subjects). High significance (p < 0.01) was revealed only in two out of five studies, indicating an increase in DNA damage in this category of patients. The authors associate the absence of an increased level of DNA damage with a high living standards and an appropriate drug therapy for patients. Thus, despite the formal predominance of studies indicating an increase in genotoxic markers in T1DM patients, the study of genotoxicity markers in this category of patients requires additional research.

Patients with type 2 diabetes mellitus

In comparison with other types of DM, T2DM is the most common disease. Most of the analyzed studies focused on the assessment of genotoxic markers in T2DM. MN were selected as a genotoxicity biomarker in 9 of 34 publications selected for the review, and DNA damage detected by the DNA comet assay was found in 25. A study that met all the inclusion criteria, in which the classical CA method was used, was presented by only one study [24].

In 10 studies focused on the analysis of MN or CA in PBL, 487 patients (219 men and 268 women) and 337 control subjects (165 men and 172 women) were examined. Summary data on age, duration of therapy, and the frequency of MN, as well as the multiplicity factor of the MN frequency in the group of patients compared with the control group are presented in Table 1. In 9 out of 10 studies focused on the analysis of MN or CA, the significance of the differences between groups of patients and the control group was shown, and the significance was high in 6 cases (p < 0.001). Two studies [25, 26] showed a correlation between the duration of disease and the incidence of MN, while two others [27, 28] did not justify this correlation. In one study [29], no differences were revealed; the authors explained this by the small size of the surveyed sample of patients, and a small difference between the groups in terms of the levels of glycated hemoglobin HbA1c (9.7 ± 1.3 vs. 7.9 ± 1 mg/dL).

These data are consistent with the results of studies, in which the authors chose the level of MN in the cells of the buccal and lingual epithelium as a biomarker as less invasive methods for obtaining a biomaterial. Thus, in a recent study [30], it was demonstrated that in T2DM patients, the MN level in the cells of the buccal (0.52 ± 0.27 vs. 0.07 ± 0.06‰; p < 0.001) and lingual (0.41 ± 0.21 vs. 0.06 ± 0.05‰; p < 0.001) epithelium was increased in comparison with that in a group of healthy volunteers of the same age. A similar result was obtained a little earlier by Russian scientists (0.52 ± 0.04 vs. 0.34 ± 0.05‰; p < 0.05) [31], as well as by other researchers [32] on a large group of female patients (n = 146), when the level of MN in the cells of the buccal epithelium was 1.85 ± 1.4 vs. 0.29 ± 0.4‰ in the control group.

In 25 studies focused on the analysis of the level of DNA damage, assessed by the DNA comet assay in PBL, 1,090 patients were examined (in several studies, the gender of the patients was not specified), and 945 were the control volunteers. A significant increase in the level of damage was revealed in 18 out of 25 publications focused on the study of DNA damage by the DNA comet assay in the PBL of T2DM patients, while in 8 cases the significance was high (p < 0.001). At the same time, in 7 out of 25 studies, such a result was not observed, which the authors explained by good control over glycemic status and/or low study power.

The first information on the possible induction of cytogenetic damage in T2DM patients appeared around half a century ago [33, 34]. Based on recent findings showing an increase in the levels of genotoxic markers in T2DM patients in most studies (27 out of 34), it is logical to infer an increased genotoxic load in these patients.

It is noteworthy that several of the cited authors set themselves the task of identifying correlations between the levels of glycated hemoglobin and genotoxicity biomarkers. Some of them indicated the presence of such a relationship with DNA damage [35–38] or MN [29, 32] in patients with T2DM or DNA damage in T1DM patients [39], while others did not reveal it in patients with T2DM [27, 40, 41] and T1DM [23, 42, 43]. Our own attempts to identify such correlations based on a generalized analysis were unsuccessful.

Thus, the question of the relationship between genotoxicity markers and levels of glycated hemoglobin remains open to date and can only be resolved with a significant expansion of the samples available for analysis.

GENOTOXIC BIOMARKERS IN NUTRITIOLOGICAL AND PHARMACOLOGICAL CORRECTION OF HYPERGLYCEMIA

An integral factor potentially capable of influencing the levels of genotoxic markers in DM patients is the use of drugs and diet to control patients’ glycemic status. Their effect can be expressed both as a direct genotoxic effect and as a comutagenic or antimutagenic modification of the action of endogenous genotoxicants, which are products of free radical reactions during oxidative stress, which inevitably accompanies hyperglycemia.

Currently, in published literature there are results of several studies focused on the targeted assessment of the cytogenetic status of patients undergoing therapy with hypoglycemic drugs and/or receiving the diet therapy.

The common design feature of most studies is the absence of a comparable group of T2DM patients not receiving therapy. This does not enable clear differentiation of the effects of endogenous genotoxicants, which are products of oxidative stress that are possible in T2DM, from the effects of exogenous medicinal or dietary factors. In all such cases, it is appropriate to suggest integrative genotoxic effect in the “therapeutic effect – disease” system, rather than the identification of a genotoxic or genotoxicant-modifying effect of a single drug and/or dietary factor.

An increase in the levels of genotoxic markers with drug therapy and/or diet therapy has been demonstrated in several studies. CA levels in PBL increased in T2DM patients receiving chlorpropamide therapy (daily doses were 100–400 mg) compared with a group of healthy, age- and gender-matched volunteers [44].

In T2DM patients who received daily therapy with a combination of pioglitazone (30 mg/day) and glimepiride (4 mg/day) for the long term (over 5 years), an increased frequency of micronuclei in buccal epithelium cells was observed compared with a group of healthy, age- and gender-matched volunteers (8.63 ± 2.23 vs. 2.93 ± 1.4‰; p < 0.001) [45].

A greater than 5-fold excess MN level was revealed in the cells of the urothelial epithelium in T2DM patients taking metformin and/or glibenclamide (doses not indicated) in comparison with a group of healthy volunteers (24.98 ± 2.87 vs. 5.02 ± 1.01‰; p < 0.001). At the same time, the analysis showed a significant increase in the number of MN in patients taking only metformin (23.02 ± 4.44) or a combination of metformin and glimepiride (24.98 ± 2.87‰) relative to that in subjects taking only glimepiride (17.52 ± 3.28) [46].

In T2DM patients treated with sitagliptin (100 mg/day), thiazolidinediones with pioglitazone (30 mg/day), or rosiglitazone (4 mg/day) or those receiving medical dietary therapy [50% of calories from carbohydrates, 30% from fat (6% saturated), and 20% from protein, with cholesterol not exceeding 300 mg/day, and fiber 35 g/day] for 6 months [47], the frequency of PBL with MN was significantly higher in the pharmacological treatment groups (1.5 ± 0.9, 2.6 ± 1.2 and 2.2 ± 1.1‰, respectively) in comparison with the nutritional control group (1.0 ± 0.6‰). At the same time, in a comparison of the drug treatment groups with each other, the frequency of MN was significantly lower (p < 0.01) in the sitagliptin therapy group, which may indicate a higher genotoxic potential of treatment with thiazolidinediones in comparison with the pyrazine inhibitor of dipeptidyl peptidase-4.

In this study, the number of cells with CA was also assessed. The above findings concerning a more intense cytogenetic status in patients receiving pharmacotherapy were also valid for the total number of CA (0.07% ± 0.02%, 0.14% ± 0.05%, 0.15% ± 0.05% vs. 0.04% ± 0.03%) [47]. It is fair to note that the last given indicators are not entirely clear, since they are an order of magnitude less than the values of the spontaneous level of chromosomal mutagenesis in humans, which is generally accepted to be estimated at 1%–3% of aberrant PBL.

Thus, evidence of an increase in genotoxic markers in patients under the influence of hypoglycemic drugs was presented in singular studies, which are unconfirmed and insufficiently convincing. The issue of the genotoxic effects of hypoglycemic drugs in treated patients requires more extensive research.

More convincing and numerous examples demonstrate a decrease in the levels of genotoxic markers under the influence of pharmacological and/or nutritional factors.

A decrease in DNA damage (p < 0.001) was revealed in T2DM patients after an 8-week period of additional daily consumption of 300 g of a mixture of vegetables, including various cruciferous species, as well as spinach, carrots, and legumes in combination with vegetable oil (25 ml per day) rich in polyunsaturated fatty acids. A decrease in DNA damage was registered as early as week 4 of the study and persisted for 8 weeks after diet cessation [48]. It is noteworthy that the same group of researchers did not reveal the effect of a similar diet on the MN levels in buccal epithelial cells in T2DM patients [49].

Regular consumption of green tea (twice daily, 150 ml 1% w/v) for 12 weeks led to a significant (p < 0.001) reduction in the level of DNA damage in T2DM patients in a placebo-controlled (water) study [50]. Vitamin E intake (900 mg/day) for 12 weeks resulted in a significant (p < 0.05) decrease in the level of DNA damage in PBL identified by the DNA comet assay in smokers and nonsmokers with T2DM [51]. A decrease in the frequency of MN in buccal epithelial cells was registered in two independent studies in T2DM patients with daily folic acid intake (5 mg orally three times a day) for 30 days [52–54]. No effect of regular consumption of low doses of the known antioxidant vitamin C on MN levels in peripheral blood cells of patients with T2DM or prediabetes was revealed [40]. However, this fact is not very informative, since the authors did not provide data on the initial provision of patients with this essential element of antioxidant protection in humans. Long term treatment with simvastatin (20 mg/day over 2 years) reduces oxidative damage to DNA in patients with dyslipidemic T2DM [55].

Information about the possibility of reducing the levels of genotoxic biomarkers was confirmed in studies where patients received a diet in combination with drugs. In particular, it was shown that, after a 7-day hospitalization during which T2DM patients adhered to a diabetic diet with a low sugar content under glycemic control (insulin, metformin), the level of DNA damage decreased significantly (p < 0.05), however, it did not reach the level of the control group of healthy volunteers (p < 0.05) [56]. Moreover, it was convincingly demonstrated that, with metformin monotherapy (on average 1.7 ± 0.9 g/day) for more than 5 months, the frequency of MN in the PBL of patients was inversely proportional to the concentration of metformin in the blood plasma of T2DM patients (p = 0.009) [57].

Thus, it can be stated that drugs used to control hyperglycemia or diet are not indifferent to the genotoxic status of DM patients. It is important to note that exercise and other environmental factors can also reduce the levels of genotoxic biomarkers. For example, 4 months of moderate physical training three times a week significantly reduces the levels of DNA damage (p < 0.05) [58], and bariatric surgery leads to a significant decrease in the level of MN in the peripheral lymphocytes of T2DM patients 1 year after surgery (p < 0.05) [59].

DISCUSSION

The results of the analysis of various genotoxic events in diabetes are heterogeneous, primarily in terms of DNA damage indices. Several authors [60, 61] suggest that this can be explained by the choice of cohorts of patients with different disease durations. Indeed, permanent oxidative stress can lead to activation of adaptive mechanisms on various levels, from the antioxidant defense system to DNA repair. Along with this, it should be mentioned that the methodology for registering DNA damage by the DNA comet assay was established relatively recently [62]. In this regard, the discrepancies revealed can be explained by the peculiarities of the instrumental part of the studies, which often cause interlaboratory discrepancies in the results of this test [63]. In addition, in the cited studies, various protocols for the obtaining and storage of the tested samples were used, which also has an impact on the estimated values [64].

Given the possibility of artifactual distortions in the results of the DNA comet assay and fewer studies denying an increase in DNA damage in hyperglycemia, as well as the obvious consistency among studies revealing a correlation of DNA damage in DM with cytogenetic biomarkers of CA and MN, we can reasonably infer an increase in DNA damage in diabetic patients.

The available pool of experimental data is obviously insufficient for a confident conclusion about an increase in genotoxicity markers in T1DM patients and, in part, GDM. The continuation of research in this field is all the more important because it is necessary to answer the question of whether antimutagenic protection and, consequently, efforts to develop it are needed for this category of patients.

The totality of the above information enables goals to be set for rationalizing the therapy of GDM, T2DM and, in the future, T1DM, considering the effect on genotoxicity markers. This is all the more important because DNA damage is a trigger for carcinogenesis and increases with the development of tumors. Diabetes, as shown for T2DM, is associated with an increased risk of developing cancer of the liver, pancreas, endometrium, colon and rectum, breast, and bladder. However, the link may be partly due to common risk factors for the two diseases, such as aging, obesity, diet, and physical inactivity [65, 66]. In addition to the direct or indirect effect of reactive oxygen species on DNA through lipid peroxidation and/or peroxynitrite, it should also be noted that, according to new data, the Akt/tuberin signaling pathway may be involved in the process of DNA damage [67, 68].

DNA damage initiates cell death and, therefore, can be considered as a factor in the development of cyto- and histopathogenesis in DM [69], which determines the need to develop methods of antimutagenic protection in DM not only in terms of prevention of oncogenesis, but also the prevention of degenerative processes.

From the point of view of rationalizing therapy, metformin attracts particular attention. First, this dimethylbiguanide derivative, an oral antiglycemic drug, is the most frequently prescribed first-line treatment for T2DM worldwide [70]. Secondly, there are experimental studies proving its antimutagenic properties. For example, oral single or daily 4- or 8-week administration of metformin at doses of 100, 500, and 2500 mg/kg significantly reduces the levels of bone marrow cells with CA and MN in a rat model of streptozotocin diabetes in a dose-dependent manner [71]. A similar effect was observed using the same experimental model of diabetes after daily 4-week oral administration of metformin at a dose of 50 mg/kg in combination with pioglitazone (1 mg/kg) when registering MN in bone marrow cells [72]. Metformin, at doses of 62.5, 125, and 250 mg/kg after 7-day daily administration, significantly dose-dependently reduced the frequency of polychromatophilic erythrocytes in the bone marrow of male Swiss albino mice 24, 48, or 72 hours after intraperitoneal administration of the cytostatic antitumor drug Adriamycin at a dose of 15 mg/kg [73]. Data from in vitro studies also indicate the protective effect of metformin against ionizing radiation and several chemical agents [74]. Third, the single but direct finding described above indicates its ability to reduce MN levels during monotherapy in T2DM patients [57]. It can also be contrasted with a single study [46], which showed the co-mutagenic effect of metformin. Certainly, inversion of the protective effect is not uncommon among antimutagens [75]. Nevertheless, this finding has not yet been confirmed either clinically or experimentally, while the evidence for the antimutagenic effects of metformin has been reproduced in the studies of various authors. Hence, there is an obvious recommendation for research aiming to extend the understanding of the antimutagenic/anticarcinogenic properties of metformin in experimental and clinical trials. As a result, hypoglycemic therapy may be enriched with new opportunities to prevent the genotoxic complications of diabetes.

It is possible that, for antigenotoxic prophylaxis in diabetic patients, it is advisable to use antimutagenic vitamin therapy, possibilities for which were discussed earlier [75, 76], as well as the well-known anxiolytic Afobazole®, which demonstrated experimentally antimutagenic properties [77] as well as antidiabetic and cytoprotective activity in a model of streptozotocin-induced diabetes [78] and a model of GDM in rats [79]. Noopept® is also an interesting drug, which has neuroprotective and nootropic properties and showed antidiabetic activity in a model of streptozotocin-induced diabetes in mice in combination with antigenotoxic activity in cells of the pancreas, liver, and kidneys [80].

CONCLUSION

Biomarkers of genotoxicity in diabetic patients are of significant interest to researchers. The summarized results are heterogeneous, and a more extensive study of biomarkers of genotoxicity in DM is required, with the use of modern approaches and generally accepted standardized protocols.

At the current stage, it is appropriate to state that T2DM patients are characterized by an increased level of genotoxicity markers, which indicates the risk of oncological diseases in them. The results of studies of genotoxicity markers in patients with T1DM and GDM are contradictory and represented by a small number of studies; however, they indicate an increased genotoxic load rather than its absence.

The levels of genotoxic damage, and therefore, the risk of carcinogenesis, can be reduced in diabetic patients under the influence of exercise, diet and/or hypoglycemic drugs. Metformin, Afobazol® and Noopept® may be recommended for experimental and clinical studies as possible drug candidates that reduce the levels of genotoxic biomarkers in diabetic patients.

×

About the authors

Natalya V. Eremina

Zakusov Research Institute of Pharmacology

Author for correspondence.
Email: neremina@panacelalabs.com
ORCID iD: 0000-0002-7226-5505
SPIN-code: 5224-1968

Cand. Sci. (Biol.), senior research associate

Russian Federation, 8 Baltiyskaya str., Moscow, 125315

Aliy K. Zhanataev

Zakusov Research Institute of Pharmacology

Email: zhanataev@academpharm.ru
ORCID iD: 0000-0002-7673-8672
SPIN-code: 7070-0510
Scopus Author ID: 6506103462

Cand. Sci. (Biol.), main researcher

Russian Federation, 8 Baltiyskaya str., Moscow, 125315

Artem A. Lisitsyn

Zakusov Research Institute of Pharmacology

Email: nordikal@yandex.ru
ORCID iD: 0000-0002-9597-6051
SPIN-code: 7857-1860

researcher

Russian Federation, 8 Baltiyskaya str., Moscow, 125315

Andrey D. Durnev

Zakusov Research Institute of Pharmacology

Email: addurnev@mail.ru
ORCID iD: 0000-0003-0218-8580
SPIN-code: 8426-0380
Scopus Author ID: 7006060753

Dr. Sci. (Med.), Professor, Corresponding Member of RAS

Russian Federation, 8 Baltiyskaya str., Moscow, 125315

References

  1. IDF Diabetes Atlas 9th edition. Available at: https://diabetesatlas.org/en/sections/worldwide-toll-of-diabetes.html. Accessed: 19.05.2021.
  2. van Dieren S, Beulens JW, van der Schouw YT, et al. The global burden of diabetes and its complications: an emerging pandemic. Eur J Cardiovasc Prev Rehabil. 2010;17 Suppl 1: S3–S8. doi: 10.1097/01.hjr.0000368191.86614.5a
  3. Dedov II, Shestakova MV, Maiorov A.Yu. Standards of specialized diabetes care. Edited by Dedov II, Shestakova MV, Mayorov AAYu. 9th edition. Diabet Mellitus. 2019;22(1S1):1–144 (In Russ.) doi: 10.14341/DM221S1
  4. Shestakova MV, Vikulova OK, Zheleznyakova AV, et al. Diabetes epidemiology in Russia: what has changed over the decade? Therapeutic Archive. 2019;91(10):4–13. (In Russ.) doi: 10.26442/00403660.2019.10.000364
  5. Demirbag R, Yilmaz R, Gur M, et al. DNA damage in metabolic syndrome and its association with antioxidative and oxidative measurements. Int J Clin Pract. 2006;60(10):1187–1193. doi: 10.1111/j.1742-1241.2006.01042.x
  6. Brownlee M. Biochemistry and molecular cell biology of diabetic complications. Nature. 2001;414(6865):813–820. doi: 10.1038/414813a
  7. Balabolkin MI. Rol’ glikirovaniya belkov, okislitel’nogo stressa v patogeneze sosudistykh oslozhnenii pri sakharnom diabete. Diabet Mellitus. 2002;4:8–16. (In Russ.)
  8. Darenskaya MA, Kolesnikova LI, Kolesnikov SI. Oxidative stress: pathogenetic role in diabetes mellitus and its complications development, therapeutic approaches to correction. BEBiM. 2021;171(2):136–149. (In Russ.) doi: 10.47056/0365-9615-2021-171-2-136-149
  9. Bigagli E, Lodovici M. Circulating Oxidative Stress Biomarkers in Clinical Studies on Type 2 Diabetes and Its Complications. Oxid Med Cell Longev. 2019;2019:5953685. doi: 10.1155/2019/5953685
  10. Durnev AD, Seredenin SB. Mutageny: skrining i farmakologicheskaya profilaktika vozdeistvii. Moscow: Meditsina; 1998, 328 p. (In Russ.)
  11. Durnev AD, Zhanataev AK, Shreder OV, Seredenina VS. Genotoxic events and diseases. Molekulyarnaya Meditsina. 2013;3:3–19 (In Russ.)
  12. Witczak M, Ferenc T, Gulczyńska E, et al Elevated frequencies of micronuclei in pregnant women with type 1 diabetes mellitus and in their newborns. Mutat Res Genet Toxicol Environ Mutagen. 2014;763:12–17. doi: 10.1016/j.mrgentox.2014.02.002
  13. International Diabetes Federation. IDF Diabetes Atlas, 9th edn. Brussels, Belgium: International Diabetes Federation, 2019. Available at: https://diabetesatlas.org/en/
  14. Toljic M, Egic A, Munjas J, et al. Increased oxidative stress and cytokinesis-block micronucleus cytome assay parameters in pregnant women with gestational diabetes mellitus and gestational arterial hypertension. Reprod Toxicol. 2017;71:55–62. doi: 10.1016/j.reprotox.2017.04.002
  15. Witczak M, Wilczyński J, Gulczyńska E, et al. What is the impact of gestational diabetes mellitus on frequency of structural chromosome aberrations in pregnant women and their offspring? Mutat Res. 2017;818:27–30. doi: 10.1016/j.mrgentox.2017.04.003
  16. Gelaleti RB, Damasceno DC, Lima PH, et al. Oxidative DNA damage in diabetic and mild gestational hyperglycemic pregnant women. Diabetol Metab Syndr. 2015;7:1. doi: 10.1186/1758-5996-7-1
  17. Basu J, Datta C, Chowdhury S, et al. Gestational Diabetes Mellitus in a Tertiary Care Hospital of Kolkata, India: Prevalence, Pathogenesis and Potential Disease Biomarkers. Exp Clin Endocrinol Diabetes. 2020;128(4):216–223. doi: 10.1055/a-0794-6057
  18. Gelaleti RB, Damasceno DC, Santos DP, et al. Increased DNA Damage is Related to Maternal Blood Glucose Levels in the Offspring of Women With Diabetes and Mild Gestational Hyperglycemia. Reprod Sci. 2016;23(3):318–323. doi: 10.1177/1933719115602766.
  19. Durga KD, Adhisivam B, Vidya G, et al. Oxidative stress and DNA damage in newborns born to mothers with hyperglycemia — a prospective cohort study. J Matern Fetal Neonatal Med. 2018;31(18):2396–2401. doi: 10.1080/14767058.2017.1344630
  20. Zabrodina VV, Shreder ED, Shreder OV, et al. Impaired prenatal development and glycemic status in the offspring of rats with experimental streptozotocin-induced diabetes and their correction with afobazole. BEBM. 2014;158(7):20–24. (In Russ.)
  21. Mihaljevic O, Zivancevic-Simonovic S, Milosevic-Djordjevic O, et al. Apoptosis and genome instability in children with autoimmune diseases. Mutagenesis. 2018;33(5–6):351–357. doi: 10.1093/mutage/gey037
  22. Witczak M, Ferenc T, Gulczyńska E, et al. Elevated frequencies of micronuclei in pregnant women with type 1 diabetes mellitus and in their newborns. Mutat Res Genet Toxicol Environ Mutagen. 2014;763:12–17. doi: 10.1016/j.mrgentox.2014.02.002
  23. Cinkilic N, Kiyici S, Celikler S, et al. Evaluation of chromosome aberrations, sister chromatid exchange and micronuclei in patients with type-1 diabetes mellitus. Mutat Res. 2009;676(1–2):1–4. doi: 10.1016/j.mrgentox.2009.02.014
  24. Saraswathy R, Anand S, Kunnumpurath SK, et al. Chromosomal Aberrations and Exon 1 Mutation in the AKR1B1 Gene in Patients with Diabetic Neuropathy. Ochsner J. 2014;14(3):339–342.
  25. Salimi M, Broumand B, Mozdarani H. Association of elevated frequency of micronuclei in peripheral blood lymphocytes of type 2 diabetes patients with nephropathy complications. Mutagenesis. 2016;31(6):627–633. doi: 10.1093/mutage/gew029
  26. Prasad M, Bronson SC, Warrier T, et al. Evaluation of DNA damage in Type 2 diabetes mellitus patients with and without peripheral neuropathy: A study in South Indian population. J Nat Sci Biol Med. 2015;6(1):80–84. doi: 10.4103/0976-9668.149096
  27. Binici DN, Karaman A, Coşkun M, et al. Genomic damage in patients with type-2 diabetes mellitus. Genet Couns. 2013;24(2):149–156.
  28. Palazzo RP, Bagatini PB, Schefer PB, et al. Genomic instability in patients with type 2 diabetes mellitus on hemodialysis. Rev Bras Hematol Hemoter. 2012;34(1):31–35. doi: 10.5581/1516-8484.20120011
  29. Shettigar SK, Shailaja C, Kulkarni RK. Elevated micronuclei frequency in type 2 diabetes with high glycosylated hemoglobin. Diabetes Res Clin Pract. 2012;95(2):246–250. doi: 10.1016/j.diabres.2011.10.025
  30. Quintero Ojeda JE, Aguilar-Medina M, Olimón-Andalón V, et al. Increased Micronuclei Frequency in Oral and Lingual Epithelium of Treated Diabetes Mellitus Patients. Biomed Res Int. 2018;2018:4898153. doi: 10.1155/2018/4898153
  31. Il’inskikh NN, Kostromeeva MS, Il’inskikh EN. Monitoring tsitogeneticheskikh posledstvii u bol’nykh kleshchevym entsefalitom, imeyushchikh sakharnyi diabet 2 tipa, v zavisimosti ot polimorfizma genov glutation-S-transferaz. Diabet Mellitus. 2019;22(3):225–232. (In Russ.) doi: 10.14341/DM9715
  32. Grindel A, Brath H, Nersesyan A, et al. Association of Genomic Instability with HbA1c levels and Medication in Diabetic Patients. Sci Rep. 2017;7:41985. doi: 10.1038/srep41985
  33. Vormittag W. Structural chromosomal aberration rates and sister-chromatid exchange frequencies in females with type 2 (non-insulin-dependent) diabetes. Mutat Res. 1985;143(3):117–119. doi: 10.1016/s0165-7992(85)80020-8
  34. Shaw JF, Kansal PC, Gatti RA. Letter: Diabetes mellitus, chromosomal aberration, and malignancy. Lancet. 1976;2(7980):315. doi: 10.1016/s0140-6736(76)90769-8
  35. Lodovici M, Giovannelli L, Pitozzi V, et al. Oxidative DNA damage and plasma antioxidant capacity in type 2 diabetic patients with good and poor glycaemic control. Mutat Res. 2008;638(1–2):98–102. doi: 10.1016/j.mrfmmm.2007.09.002
  36. Dinçer Y, Akçay T, Alademir Z, Ilkova H. Assessment of DNA base oxidation and glutathione level in patients with type 2 diabetes. Mutat Res. 2002;505(1–2):75–81. Corrected and republished from: Mutat Res. 2003;525(1–2):129–30. doi: 10.1016/s0027-5107(02)00143-4
  37. Tatsch E, Bochi GV, Piva SJ, et al. Association between DNA strand breakage and oxidative, inflammatory and endothelial biomarkers in type 2 diabetes. Mutat Res. 2012;732(1–2):16–20. doi: 10.1016/j.mrfmmm.2012.01.004
  38. Arif M, Islam MR, Waise TM, et al. DNA damage and plasma antioxidant indices in Bangladeshi type 2 diabetic patients. Diabetes Metab. 2010;36(1):51–57. doi: 10.1016/j.diabet.2009.05.007
  39. Dinçer Y, Akçay T, Ilkova H, et al. DNA damage and antioxidant defense in peripheral leukocytes of patients with Type I diabetes mellitus. Mutat Res. 2003;527(1–2):49–55. doi: 10.1016/s0027-5107(03)00073-3.
  40. Franke SI, Müller LL, Santos MC, et al. Vitamin C intake reduces the cytotoxicity associated with hyperglycemia in prediabetes and type 2 diabetes. Biomed Res Int. 2013;2013:896536. doi: 10.1155/2013/896536
  41. Corbi SC, Bastos AS, Orrico SR, et al. Elevated micronucleus frequency in patients with type 2 diabetes, dyslipidemia and periodontitis. Mutagenesis. 2014;29(6):433–439. doi: 10.1093/mutage/geu043
  42. Astley S, Langrish-Smith A, Southon S, Sampson M. Vitamin E supplementation and oxidative damage to DNA and plasma LDL in type 1 diabetes. Diabetes Care. 1999;22(10):1626–1631. doi: 10.2337/diacare.22.10.1626
  43. Hannon-Fletcher MP, O’Kane MJ, Moles KW, et al. Levels of peripheral blood cell DNA damage in insulin dependent diabetes mellitus human subjects. Mutat Res. 2000;460(1):53–60. doi: 10.1016/s0921-8777(00)00013-6
  44. Watson WA, Petrie JC, Galloway DB, et al. In vivo cytogenetic activity of sulphonylurea drugs in man. Mutat Res. 1976;38(1):71–80. doi: 10.1016/0165-1161(76)90080-7
  45. Shaik NA, Shaik JP, Ali S, et al. Increased frequency of micronuclei in diabetes mellitus patients using pioglitazone and glimepiride in combination. Food Chem Toxicol. 2010;48(12):3432–3435. doi: 10.1016/j.fct.2010.09.016
  46. Harishankar MK, Logeshwaran S, Sujeevan S, et al. Genotoxicity evaluation of metformin and glimepiride by micronucleus assay in exfoliated urothelial cells of type 2 diabetes mellitus patients. Food Chem Toxicol. 2015;83:146–150. doi: 10.1016/j.fct.2015.06.013
  47. Oz Gul O, Cinkilic N, Gul CB, et al. Comparative genotoxic and cytotoxic effects of the oral antidiabetic drugs sitagliptin, rosiglitazone, and pioglitazone in patients with type-2 diabetes: a cross-sectional, observational pilot study. Mutat Res. 2013;757(1):31–35. doi: 10.1016/j.mrgentox.2013.04.024
  48. Müllner E, Brath H, Pleifer S, et al. Vegetables and PUFA-rich plant oil reduce DNA strand breaks in individuals with type 2 diabetes. Mol Nutr Food Res. 2013;57(2):328–338. doi: 10.1002/mnfr.201200343
  49. Müllner E, Brath H, Nersesyan A, et al. Nuclear anomalies in exfoliated buccal cells in healthy and diabetic individuals and the impact of a dietary intervention. Mutagenesis. 2014;29(1):1–6. doi: 10.1093/mutage/get056
  50. Choi SW, Yeung VT, Collins AR, Benzie IF. Redox-linked effects of green tea on DNA damage and repair, and influence of microsatellite polymorphism in HMOX-1: results of a human intervention trial. Mutagenesis. 2015;30(1):129–137. doi: 10.1093/mutage/geu022
  51. Sardaş S, Yilmaz M, Oztok U, et al. Assessment of DNA strand breakage by comet assay in diabetic patients and the role of antioxidant supplementation. Mutat Res. 2001;490(2):123–129. doi: 10.1016/s1383-5718(00)00157-1
  52. Zúñiga-González GM, Batista-González CM, Gómez-Meda BC, et al. Micronuclei in diabetes: folate supplementation diminishes micronuclei in diabetic patients but not in an animal model. Mutat Res. 2007;634(1–2):126–134. doi: 10.1016/j.mrgentox.2007.06.006
  53. Lazalde-Ramos BP, Zamora-Perez AL, Sosa-Macías M, et al. DNA and oxidative damages decrease after ingestion of folic acid in patients with type 2 diabetes. Arch Med Res. 2012;43(6):476–481. doi: 10.1016/j.arcmed.2012.08.013
  54. Gómez-Meda BC, Zamora-Perez AL, Muñoz-Magallanes T, et al. Nuclear abnormalities in buccal mucosa cells of patients with type I and II diabetes treated with folic acid. Mutat Res Genet Toxicol Environ Mutagen. 2016;797:1–8. doi: 10.1016/j.mrgentox.2015.12.003
  55. Manfredini V, Biancini GB, Vanzin CS, et al. Simvastatin treatment prevents oxidative damage to DNA in whole blood leukocytes of dyslipidemic type 2 diabetic patients. Cell Biochem Funct. 2010;28(5):360–366. doi: 10.1002/cbf.1654
  56. Xavier DJ, Takahashi P, Manoel-Caetano FS, et al. One-week intervention period led to improvements in glycemic control and reduction in DNA damage levels in patients with type 2 diabetes mellitus. Diabetes Res Clin Pract. 2014;105(3):356–363. doi: 10.1016/j.diabres.2014.06.004
  57. da Silva BS, Rovaris DL, Bonotto RM, et al. The influence on DNA damage of glycaemic parameters, oral antidiabetic drugs and polymorphisms of genes involved in the DNA repair system. Mutagenesis. 2013;28(5):525–530. doi: 10.1093/mutage/get029
  58. Pittaluga M, Sgadari A, Dimauro I, et al. Physical exercise and redox balance in type 2 diabetics: effects of moderate training on biomarkers of oxidative stress and DNA damage evaluated through comet assay. Oxid Med Cell Longev. 2015;2015:981242. doi: 10.1155/2015/981242
  59. Bankoglu EE, Arnold C, Hering I, et al. Decreased Chromosomal Damage in Lymphocytes of Obese Patients After Bariatric Surgery. Sci Rep. 2018;8(1):11195. doi: 10.1038/s41598-018-29581-6
  60. Ibarra-Costilla E, Cerda-Flores RM, Dávila-Rodríguez MI, et al. DNA damage evaluated by comet assay in Mexican patients with type 2 diabetes mellitus. Acta Diabetol. 2010;47 Suppl 1:111–116. doi: 10.1007/s00592-009-0149-9
  61. Anderson D, Yu TW, Wright J, Ioannides C. An examination of DNA strand breakage in the comet assay and antioxidant capacity in diabetic patients. Mutation Research. 199;398(1–2):151–161. doi: 10.1016/s0027-5107(97)00271-6
  62. OECD Test No. 489: In Vivo Mammalian Alkaline Comet Assay. Available at: https://www.oecd.org/env/test-no-489-in-vivo-mammalian-alkaline-comet-assay-9789264264885-en.htm
  63. Sirota NP, Zhanataev AK, Kuznetsova EA, et al. Some causes of inter-labolatory variation in the results of comet assay. Mutation Research. 2014;770:16–22. doi: 10.1016/j.mrgentox.2014.05.003
  64. Gajski G, Gerić M, Živković Semren T, et al. Application of the comet assay for the evaluation of DNA damage from frozen human whole blood samples: Implications for human biomonitoring. Toxicol Lett. 2020;319:58–65. doi: 10.1016/j.toxlet.2019.11.010
  65. Habib SL, Rojna M. Diabetes and risk of cancer. ISRN Oncol. 2013;2013:583786. doi: 10.1155/2013/583786
  66. Giovannucci E, Harlan DM, Archer MC, et al. Diabetes and cancer: a consensus report. Diabetes Care. 2010;33(7):1674–1685. doi: 10.2337/dc10-0666
  67. Simone S, Gorin Y, Velagapudi C, et al. Mechanism of oxidative DNA damage in diabetes: tuberin inactivation and downregulation of DNA repair enzyme 8-oxo-7,8-dihydro-2’-deoxyguanosine-DNA glycosylase. Diabetes. 2008;57(10):2626–2636. doi: 10.2337/db07-1579
  68. Lee SC, Chan JC. Evidence for DNA damage as a biological link between diabetes and cancer. Chin Med J (Engl). 2015;128(11): 1543–1548. doi: 10.4103/0366-6999.157693
  69. Eremina NV, Zhanataev AK, Durnev AD. Indutsiruemaya kletochnaya gibel’ kak vozmozhnyi put’ antimutagennogo vozdeistviya. BEBM. 2021. T. 171. № 1. P. 4–22. (In Russ.) doi: 10.47056/0365-9615-2021-171-1-4-22
  70. van Staa TP, Patel D, Gallagher AM, de Bruin ML. Glucose-lowering agents and the patterns of risk for cancer: a study with the General Practice Research Database and secondary care data. Diabetologia. 2012;55(3):654–665. doi: 10.1007/s00125-011-2390-3
  71. Attia SM, Helal GK, Alhaider AA. Assessment of genomic instability in normal and diabetic rats treated with metformin. Chem Biol Interact. 2009;180(2):296–304. doi: 10.1016/j.cbi.2009.03.001
  72. Rabbani SI, Devi K, Khanam S. Role of Pioglitazone with Metformin or Glimepiride on Oxidative Stress-induced Nuclear Damage and Reproductive Toxicity in Diabetic Rats. Malays J Med Sci. 2010;17(1):3–11.
  73. Aleisa AM, Al-Rejaie SS, Bakheet SA, et al. Effect of metformin on clastogenic and biochemical changes induced by adriamycin in Swiss albino mice. Mutat Res. 2007;634(1–2):93–100. doi: 10.1016/j.mrgentox.2007.06.005
  74. Najafi M, Cheki M, Rezapoor S, et al. Metformin: Prevention of genomic instability and cancer: A review. Mutat Res Genet Toxicol Environ Mutagen. 2018;827:1–8. doi: 10.1016/j.mrgentox.2018.01.007
  75. Durnev AD. Antimutagenesis and antimutagens. Human Physiology. 2018;44(3):116–137. (In Russ.) doi: 10.7868/S013116461803013X
  76. Sidneva ES, Katosova LD, Platonova VI, et al. Otsenka spontannogo i khimicheski indutsirovannogo mutageneza v kletkakh cheloveka v zavisimosti ot vitaminnoi obespechennosti. Bulletin of Experimental Biology and Medicine. 2005;139(5):199–203. (In Russ.)
  77. Durnev AD, Zhanataev AK, Shreder OV, Seredenin SB. Antimutagenic and antiteratogenic properties of afobazole. Eksp Klin Farmakol. 2009;72(1):46–51. (In Russ.)
  78. Zabrodina VV, Shreder OV, Shreder ED, Durnev AD. Effect of Afobazole and Betaine on Cognitive Disorders in the Offspring of Rats with Streptozotocin-Induced Diabetes and Their Relationship with DNA Damage. Bull Exp Biol Med. 2016;161(3):359–366. doi: 10.1007/s10517-016-3414-2
  79. Zabrodina VV, Shreder ED, Shreder OV, et al. Effect of Afobazole and Betaine on DNA Damage in Placental and Embryonic Tissues of Rats with Experimental Streptozocin Diabetes. Bull Exp Biol Med. 2015;159(6):757–760. doi: 10.1007/s10517-015-3068-5
  80. Ostrovskaya RU, Yagubova SS, Zhanataev AK, et al. Neuroprotective Dipeptide Noopept Prevents DNA Damage in Mice with Modeled Prediabetes. Bull Exp Biol Med. 2019;168(2):233–237. doi: 10.1007/s10517-019-04681-z.
  81. Supriya Simon A, Dinesh Roy D, Jayapal V, Vijayakumar T. Somatic DNA damages in cardiovascular autonomic neuropathy. Indian J Clin Biochem. 2011;26(1):50–56. doi: 10.1007/s12291-010-0087-x
  82. Martínez-Pérez LM, Cerda-Flores RM, Gallegos-Cabriales EC, et al. Frequency of micronuclei in Mexicans with type 2 diabetes mellitus. Prague Med Rep. 2007;108(3):248–255
  83. Gelaleti RB, Damasceno DC, Salvadori DM, et al. IRS-1 gene polymorphism and DNA damage in pregnant women with diabetes or mild gestational hyperglycemia. Diabetol Metab Syndr. 2015;7:30. doi: 10.1186/s13098-015-0026-3
  84. Wyatt N, Kelly C, Fontana V, et al. The responses of lymphocytes from Asian and Caucasian diabetic patients and non-diabetics to hydrogen peroxide and sodium nitrite in the Comet assay. Mutat Res. 2006;609(2):154–164. doi: 10.1016/j.mrgentox.2006.06.029
  85. Varvarovská J, Racek J, Stetina R, et al. Aspects of oxidative stress in children with type 1 diabetes mellitus. Biomed Pharmacother. 2004;58(10):539–545. doi: 10.1016/j.biopha.2004.09.011
  86. Collins AR, Raslová K, Somorovská M, et al. DNA damage in diabetes: correlation with a clinical marker. Free Radic Biol Med. 1998;25(3):373–377. doi: 10.1016/s0891-5849(98)00053-7
  87. Rao A, Thomas B, Prasad RB, et al. A comparative evaluation of the DNA damage in the serum of chronic periodontitis patients with and without diabetes mellitus type II. Indian J Dent Res. 2020;31(2):169–174. doi: 10.4103/ijdr.IJDR_503_17
  88. Raghav A, Ahmad J, Alam K. Preferential recognition of advanced glycation end products by serum antibodies and low-grade systemic inflammation in diabetes mellitus and its complications. Int J Biol Macromol. 2018;118(Pt B):1884–1891. doi: 10.1016/j.ijbiomac.2018.07.033
  89. Bukhari SA, Javed S, Ali M, et al. Serum haematological and biochemical indices of oxidative stress and their relationship with DNA damage and homocysteine in Pakistani type II diabetic patients. Pak J Pharm Sci. 2015;28(3):881–889.
  90. Merecz A, Markiewicz L, Sliwinska A, et al. Analysis of oxidative DNA damage and its repair in Polish patients with diabetes mellitus type 2: Role in pathogenesis of diabetic neuropathy. Adv Med Sci. 2015;60(2):220–230. doi: 10.1016/j.advms.2015.04.001
  91. Xavier DJ, Takahashi P, Evangelista AF, et al. Assessment of DNA damage and mRNA/miRNA transcriptional expression profiles in hyperglycemic versus non-hyperglycemic patients with type 2 diabetes mellitus. Mutat Res. 2015;776:98–110. doi: 10.1016/j.mrfmmm.2015.01.016
  92. Salem SI, El-Toukhy SE, El-Saeed GSM, El-Wassef M. Correlation of DNA damage in type 2 diabetes to glycemic control. The Egyptian Journal of Hospital Medicine. 2012;48:472–482. doi: 10.21608/EJHM.2012.16249
  93. Kasznicki J, Kosmalski M, Sliwinska A, et al. Evaluation of oxidative stress markers in pathogenesis of diabetic neuropathy. Mol Biol Rep. 2012;39(9):8669–8678. doi: 10.1007/s11033-012-1722-9
  94. Manfredini V, Biancini GB, Vanzin CS, et al. Simvastatin treatment prevents oxidative damage to DNA in whole blood leukocytes of dyslipidemic type 2 diabetic patients. Cell Biochem Funct. 2010;28(5):360–366. doi: 10.1002/cbf.1654
  95. Vasil’ev DA, Poroshina TE, Kovalenko IG, et al. The dual (joker) function of glucose and its association with aging and glucose intoleranc. Advances in Gerontology. 2008;21(2):204–211 (In Russ.)
  96. Bagatini PB, Palazzo RP, Rodrigues MT, et al. Induction and removal of DNA damage in blood leukocytes of patients with type 2 diabetes mellitus undergoing hemodialysis. Mutat Res. 2008;657(2):111–115. doi: 10.1016/j.mrgentox.2008.08.004
  97. Sliwinska A, Blasiak J, Kasznicki J, Drzewoski J. In vitro effect of gliclazide on DNA damage and repair in patients with type 2 diabetes mellitus (T2DM). Chem Biol Interact. 2008;173(3):159–165. doi: 10.1016/j.cbi.2008.03.017
  98. Song F, Jia W, Yao Y, et al. Oxidative stress, antioxidant status and DNA damage in patients with impaired glucose regulation and newly diagnosed Type 2 diabetes. Clin Sci (Lond). 2007;112(12): 599–606. doi: 10.1042/CS20060323
  99. Blasiak J, Arabski M, Krupa R, et al. DNA damage and repair in type 2 diabetes mellitus. Mutat Res. 2004;554(1–2):297–304. doi: 10.1016/j.mrfmmm.2004.05.011
  100. Pitozzi V, Giovannelli L, Bardini G, et al. Oxidative DNA damage in peripheral blood cells in type 2 diabetes mellitus: higher vulnerability of polymorphonuclear leukocytes. Mutat Res. 2003;529(1–2): 129–133. doi: 10.1016/s0027-5107(03)00114-3

Supplementary files

Supplementary Files
Action
1. JATS XML
2. Figure 1. Medical significance of oxidative damage to nucleic acids, lipids, and proteins in hyperglycemia ([9], significantly modified)

Download (275KB)

Copyright (c) 2021 ООО "Эко-Вектор"



СМИ зарегистрировано Федеральной службой по надзору в сфере связи, информационных технологий и массовых коммуникаций (Роскомнадзор).
Регистрационный номер и дата принятия решения о регистрации СМИ: серия ПИ № ФС 77 - 65617 от 04.05.2016.


This website uses cookies

You consent to our cookies if you continue to use our website.

About Cookies