Insulin Resistance in PCOS Patients Enhances Oxidative Stress and Leukocyte Adhesion: Role of Myeloperoxidase

Victor M Victor, Susana Rovira-Llopis, Celia Bañuls, Noelia Diaz-Morales, Arantxa Martinez de Marañon, Cesar Rios-Navarro, Angeles Alvarez, Marcelino Gomez, Milagros Rocha, Antonio Hernández-Mijares, Victor M Victor, Susana Rovira-Llopis, Celia Bañuls, Noelia Diaz-Morales, Arantxa Martinez de Marañon, Cesar Rios-Navarro, Angeles Alvarez, Marcelino Gomez, Milagros Rocha, Antonio Hernández-Mijares

Abstract

Cardiovascular diseases and oxidative stress are related to polycystic ovary syndrome (PCOS) and insulin resistance (IR). We have evaluated the relationship between myeloperoxidase (MPO) and leukocyte activation in PCOS patients according to homeostatic model assessment of IR (HOMA-IR), and have explored a possible correlation between these factors and endocrine and inflammatory parameters. This was a prospective controlled study conducted in an academic medical center. The study population consisted of 101 PCOS subjects and 105 control subjects. We divided PCOS subjects into PCOS non-IR (HOMA-IR<2.5) and PCOS IR (HOMA-IR>2.5). Metabolic and anthropometric parameters, total and mitochondrial reactive oxygen species (ROS) production, MPO levels, interactions between human umbilical vein endothelial cells and leukocytes, adhesion molecules (E-selectin, ICAM-1 and VCAM-1) and proinflammatory cytokines (IL-6 and TNF-α) were evaluated. Oxidative stress was observed in PCOS patients, in whom there was an increase in total and mitochondrial ROS production and MPO levels. Enhanced rolling flux and adhesion, and a decrease in polymorphonuclear cell rolling velocity were also detected in PCOS subjects. Increases in IL-6 and TNF-α and adhesion molecules (E-selectin, ICAM-1 and VCAM-1) were also observed, particularly in the PCOS IR group, providing evidence that inflammation and oxidative stress are related in PCOS patients. HOMA-IR was positively correlated with hsCRP (p<0.001, r = 0.304), ROS production (p<0.01, r = 0.593), leukocyte rolling flux (p<0.05, r = 0.446), E-selectin (p<0.01, r = 0.436) and IL-6 (p<0.001, r = 0.443). The results show an increase in the rate of ROS and MPO levels in PCOS patients in general, and particularly in those with IR. Inflammation in PCOS induces leukocyte-endothelium interactions and a simultaneous increase in IL-6, TNF-α, E-selectin, ICAM-1 and VCAM-1. These conditions are aggravated by the presence of IR.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Fig 1. Effects of PCOS on levels…
Fig 1. Effects of PCOS on levels of ROS in PMN.
(A) Levels of DCFH-DA fluorescence measured by fluorimetry in controls and PCOS subjects; (B) Levels of DCFH fluorescence in controls and in PCOS non-IR and PCOS IR subjects; (C) Mean DCFH fluorescence assessed by static cytometry; % vs control. (D) Representative images of DCFH-DA fluorescence in PMNs assessed by fluorescence microscopy; nuclei: Hoechst 33342 signal (blue); ROS: DCFH-DA signal (green). *pcp<0.001 between PCOS non-IR and PCOS IR subjects.
Fig 2. Leukocyte mitochondrial ROS production in…
Fig 2. Leukocyte mitochondrial ROS production in PCOS non-IR and IR patients and control women.
A) Mitochondrial ROS (Mitosox Red Fluorescence) in PCOS versus control subjects. B) Mitochondrial ROS (Mitosox Red Fluorescence) in PCOS non IR and IR patients versus control subjects. Values in the bar graphs were obtained by calculating the percentage of fluorescence intensity relative to the control. *p

Fig 3. Plasma MPO concentrations in PCOS…

Fig 3. Plasma MPO concentrations in PCOS and control subjects (A) and in control, PCOS…

Fig 3. Plasma MPO concentrations in PCOS and control subjects (A) and in control, PCOS non-IR and PCOS IR subjects (B).
*pap<0.05 between PCOS non-IR and PCOS IR subjects.

Fig 4. Leukocyte/endothelium interactions in PCOS and…

Fig 4. Leukocyte/endothelium interactions in PCOS and control subjects.

PMN rolling velocity (μsecond -1 )…

Fig 4. Leukocyte/endothelium interactions in PCOS and control subjects.
PMN rolling velocity (μsecond-1) (A), rolling flux (PMN per minute) (C), and PMN adhesion (PMN per square millimetre) (E); and PCOS non-IR and PCOS IR subjects: PMN rolling velocity (μsecond-1) (B), rolling flux (PMN per minute) (D), and PMN adhesion (PMN per square millimetre) (F). *p<0.05 and ***p<0.001 vs. Control. ap<0.05 and bp<0.01 between PCOS non-IR and PCOS IR subjects.

Fig 5. Adhesion molecules in the serum…

Fig 5. Adhesion molecules in the serum of PCOS and control subjects.

(A) E-selectin, (C)…

Fig 5. Adhesion molecules in the serum of PCOS and control subjects.
(A) E-selectin, (C) ICAM-1 and (E) VCAM-1; and PCOS non-IR, PCOS IR and control subjects. (B) E-selectin, (D) ICAM-1 and (F) VCAM-1. *pap<0.05 and bp<0.01 between PCOS non-IR and PCOS IR subjects.

Fig 6. Proinflammatory cytokines in the serum…

Fig 6. Proinflammatory cytokines in the serum of PCOS and control subjects.

(A) IL-6, and…

Fig 6. Proinflammatory cytokines in the serum of PCOS and control subjects.
(A) IL-6, and (C) TNF-α; and PCOS non-IR, PCOS IR and control subjects. (B) IL-6, and (D) TNF-α. *pap<0.05 between PCOS non-IR and PCOS IR subjects.

Fig 7. Correlation studies of HOMA-IR and…

Fig 7. Correlation studies of HOMA-IR and inflammation and adhesion parameters in PCOS women.

Fig 7. Correlation studies of HOMA-IR and inflammation and adhesion parameters in PCOS women.
All figures (7)
Fig 3. Plasma MPO concentrations in PCOS…
Fig 3. Plasma MPO concentrations in PCOS and control subjects (A) and in control, PCOS non-IR and PCOS IR subjects (B).
*pap<0.05 between PCOS non-IR and PCOS IR subjects.
Fig 4. Leukocyte/endothelium interactions in PCOS and…
Fig 4. Leukocyte/endothelium interactions in PCOS and control subjects.
PMN rolling velocity (μsecond-1) (A), rolling flux (PMN per minute) (C), and PMN adhesion (PMN per square millimetre) (E); and PCOS non-IR and PCOS IR subjects: PMN rolling velocity (μsecond-1) (B), rolling flux (PMN per minute) (D), and PMN adhesion (PMN per square millimetre) (F). *p<0.05 and ***p<0.001 vs. Control. ap<0.05 and bp<0.01 between PCOS non-IR and PCOS IR subjects.
Fig 5. Adhesion molecules in the serum…
Fig 5. Adhesion molecules in the serum of PCOS and control subjects.
(A) E-selectin, (C) ICAM-1 and (E) VCAM-1; and PCOS non-IR, PCOS IR and control subjects. (B) E-selectin, (D) ICAM-1 and (F) VCAM-1. *pap<0.05 and bp<0.01 between PCOS non-IR and PCOS IR subjects.
Fig 6. Proinflammatory cytokines in the serum…
Fig 6. Proinflammatory cytokines in the serum of PCOS and control subjects.
(A) IL-6, and (C) TNF-α; and PCOS non-IR, PCOS IR and control subjects. (B) IL-6, and (D) TNF-α. *pap<0.05 between PCOS non-IR and PCOS IR subjects.
Fig 7. Correlation studies of HOMA-IR and…
Fig 7. Correlation studies of HOMA-IR and inflammation and adhesion parameters in PCOS women.

References

    1. Diamanti-Kandarakis E, Kouli CR, Bergiele AT, Filandra FA, Tsianateli TC, Spina GG, et al. A survey of the polycystic ovary syndrome in the Greek island of Lesbos: hormonal and metabolic profile. J Clin Endocrinol Metab. 1999;84: 4006–4011.
    1. Dunaif A. Insulin resistance and the polycystic ovary syndrome: mechanism and implications for pathogenesis. Endocr Rev. 1997;18: 774–800.
    1. Moran LJ, Misso ML, Wild RA, Norman RJ. Impaired glucose tolerance, type 2 diabetes and metabolic syndrome in polycystic ovary syndrome: a systematic review and meta-analysis. Hum Reprod Update 2010;16: 347–363. 10.1093/humupd/dmq001
    1. Ehrmann DA. Polycystic ovary syndrome. N Engl J Med. 2005;352: 1223–1236.
    1. Bajuk Studen K, Jensterle Sever M, Pfeifer M. Cardiovascular risk and subclinical cardiovascular disease in polycystic ovary syndrome. Front Horm Res. 2013;40: 64–82. 10.1159/000341838
    1. Mahalingaiah S, Diamanti-Kandarakis E. Targets to treat metabolic syndrome in polycystic ovary syndrome. Expert Opin Ther Targets. 2015;19: 1561–1574. 10.1517/14728222.2015.1101067
    1. Zhao Y, Zhang C, Huang Y, Yu Y, Li R, Li M, et al. Up-Regulated Expression of WNT5a Increases Inflammation and Oxidative Stressvia PI3K/AKT/NF-κB Signaling in the Granulosa Cells of PCOS Patients. J Clin Endocrinol Metab. 2015;100: 201–211. 10.1210/jc.2014-2419
    1. Escobar-Morreale HF, Luque-Ramírez M, González F. Circulating inflammatory markers in polycystic ovary syndrome: a systematic review and metaanalysis. Fertil Steril. 2011;95: 1048–1058. 10.1016/j.fertnstert.2010.11.036
    1. Gonzalez F, Rote NS, Minium J, Kirwan JP. Reactive oxygen species-induced oxidative stress in the development of insulin resistance and hyperandrogenism in polycystic ovary syndrome. J Clin Endocrinol Metab. 2006;91: 336–340.
    1. Victor VM, Rocha M, Bañuls C, Sanchez-Serrano M, Sola E, Gomez M, et al. Mitochondrial complex I impairment in leukocytes from polycystic ovary syndrome patients with insulin resistance. J Clin Endocrinol Metab. 2009;94: 3505–3512. 10.1210/jc.2009-0466
    1. Victor VM, Rocha M, Bañuls C, Alvarez A, de Pablo C, Sanchez-Serrano M, et al. Induction of oxidative stress and human leukocyte/endothelial cell interactions in polycystic ovary syndrome patients with insulin resistance. J Clin Endocrinol Metab. 2011;96: 3115–3122. 10.1210/jc.2011-0651
    1. Gonzalez F, Rote NS, Minium J, Kirwan JP. In vitro evidence that hyperglycemia stimulates tumor necrosis factor alpha release in obese women with polycystic ovary syndrome. J Endocrinol. 2006;188: 521–529.
    1. Orio F Jr, Palomba S, Cascella T, Di Biase S, Manguso F, Tauchmanovà L, et al. The increase of leukocytes as a new putative marker of low-grade chronic inflammation and early cardiovascular risk in polycystic ovary syndrome. J Clin Endocrinol Metab. 2005;90: 2–5.
    1. Rovira-Llopis S, Rocha M, Falcon R, de Pablo C, Alvarez A, Jover A, et al. Is myeloperoxidase a key component in the ROS-induced vascular damage related to nephropathy in type 2 diabetes? Antioxid Redox Signal. 2013;19: 1452–1458. 10.1089/ars.2013.5307
    1. Zhang C, Yang J, Jennings LK. Leukocyte-derived myeloperoxidase amplifies high-glucose-induced endothelial dysfunction through interaction with high-glucose-stimulated, vascular non-leukocyte-derived reactive oxygen species. Diabetes 2004;53: 2950–2959.
    1. Ribeiro AL, Scapinelli A, Tamanaha S, Oliveira RM, Kowastch I, Mathias W Jr, et al. Myeloperoxidases and polycystic ovary syndrome. Gynecol Endocrinol. 2012;28: 3–6. 10.3109/09513590.2011.579656
    1. Blair SA, Kyaw-Tun T, Young IS, Phelan NA, Gibney J, McEneny J. Oxidative stress and inflammation in lean and obese subjects with polycystic ovary syndrome. J Reprod Med. 2013;58: 107–114.
    1. Kurdoglu Z, Ozkol H, Kurdoglu M. Serum myeloperoxidase and adenosine deaminase activities in polycystic ovary syndrome. Gynecol Endocrinol. 2012;28:115–118. 10.3109/09513590.2011.583959
    1. Vozarova B, Weyer C, Lindsay RS, Pratley RE, Bogardus C, Tataranni PA. High white blood cell count is associated with a worsening of insulin sensitivity and predicts the development of type 2 diabetes. Diabetes 2002;51: 455–461.
    1. Armeni E, Stamatelopoulos K, Rizos D, Georgiopoulos G, Kazani M, Kazani A, et al. Arterial stiffness is increased in asymptomatic nondiabetic postmenopausal women with a polycystic ovary syndrome phenotype. J Hypertens. 2013;31: 1998–2004. 10.1097/HJH.0b013e3283630362
    1. The Rotterdam ESHRE/ASRM-Sponsored PCOS Consensus Workshop Group. Revised 2003 consensus on diagnostic criteria and long-term health risks related to polycystic ovary syndrome. Fertil Steril. 2004;81: 19–25.
    1. Azziz R, Sanchez LA, Knochenhauer ES, Moran C, Lazenby J, Stephens KC, et al. Androgen excess in women: experience with over 1000 consecutive patients. J Clin Endocrinol Metab. 2004;89: 453–462.
    1. Hernández Mijares A, Riera Fortuny C, Solá Izquierdo E, Oliver Oliver MJ, Martínez Triguero ML, Morillas Ariño C, et al. Prevalence of metabolic syndrome in patients with coronary heart disease. Med Clin (Barc) 2003;121: 204–208.
    1. Rovira-Llopis S, Bañuls C, Apostolova N, Morillas C, Hernandez-Mijares A, Rocha M, et al. Is glycemic control modulating endoplasmic reticulum stress in leukocytes of type 2 diabetic patients? Antioxid Redox Signal. 2014;21: 1759–1765. 10.1089/ars.2014.6030
    1. Hernandez-Mijares A, Rocha M, Rovira-Llopis S, Bañuls C, Bellod L, de Pablo C, et al. Human leukocyte/endothelial cell interactions and mitochondrial dysfunction in type 2 diabetic patients and their association with silent myocardial ischemia. Diabetes Care 2013;36: 1695–1702. 10.2337/dc12-1224
    1. Palomba S, Falbo A, Chiossi G, Orio F, Tolino A, Colao A, et al. Low-grade chronic inflammation in pregnant women with polycystic ovary syndrome: a prospective controlled clinical study. J Clin Endocrinol Metab. 2014;99: 2942–2951. 10.1210/jc.2014-1214
    1. Li Q, Park K, Li C, Rask-Madsen C, Mima A, Qi W, et al. Induction of vascular insulin resistance and endothelin-1 expression and acceleration of atherosclerosis by the overexpression of protein kinase C-β isoform in the endothelium. Circ Res. 2013;113: 418–427. 10.1161/CIRCRESAHA.113.301074
    1. Ehrmann DA, Liljenquist DR, Kasza K, Azziz R, Legro RS, Ghazzi MN, et al. Prevalence and predictors of the metabolic syndrome in women with polycystic ovary syndrome. J Clin Endocrinol Metab. 2006;91: 48–53.
    1. Grundy SM. Metabolic syndrome: a multiplex cardiovascular risk factor. J Clin Endocrinol Metab. 2007;92: 399–404.
    1. Giacco F, Brownlee M. Oxidative stress and diabetic complications. Circ Res. 2010;107: 1058–1070. 10.1161/CIRCRESAHA.110.223545
    1. Prieto D, Contreras C, Sánchez A. Endothelial dysfunction, obesity and insulin resistance. Curr Vasc Pharmacol. 2014;12:412–426.
    1. Anatoliotakis N, Deftereos S, Bouras G, Giannopoulos G, Tsounis D, Angelidis C, et al. Myeloperoxidase: expressing inflammation and oxidative stress in cardiovascular disease. Curr Top Med Chem. 2013;13: 115–138.
    1. Klebanoff SJ. Myeloperoxidase: friend and foe. J Leukoc Biol. 2005;77: 598–625.
    1. Unubol M, Yavasoglu I, Kacar F, Guney E, Omurlu IK, Ture M, et al. Relationship between glycemic control and histochemicale M, et al.; 77: 598–625.25. al. Myeloperoxidase: expressing inf diabetes. Diabetol Metab Syndr. 2015;7: 119.
    1. Podrez EA, Schmitt D, Hoff HF, Hazen SL. Myeloperoxidase-generated reactive nitrogen species convert LDL into an atherogenic form in vitro. J Clin Invest. 1999;103: 1547–1560.
    1. Bergt C, Pennathur S, Fu X, Byun J, O'Brien K, McDonald TO, et al. The myeloperoxidase product hypochlorous acid oxidizes HDL in the human artery wall and impairs ABCA1-dependent cholesterol transport. Proc Natl Acad Sci USA 2004;101: 13032–13037.
    1. Eiserich JP, Baldus S, Brennan ML, Ma W, Zhang C, Tousson A, et al. Myeloperoxidase, a leukocyte-derived vascular NO oxidase. Science 2002;296: 2391–2394.
    1. Vita JA, Brennan ML, Gokce N, Mann SA, Goormastic M, Shishehbor MH, et al. Serum myeloperoxidase levels independently predict endothelial dysfunction in humans. Circulation 2004;10: 1134–1139.
    1. Baldus S, Heitzer T, Eiserich JP, Lau D, Mollnau H, Ortak M, et al. Myeloperoxidase enhances nitric oxide catabolism during myocardial ischemia and reperfusion. Free Radic Biol Med. 2004;37: 902–911.
    1. Gorudko IV, Sokolov AV, Shamova EV, Grigorieva DV, Mironova EV, Kudryavtsev IV, et al. Binding of human myeloperoxidase to red blood cells: Molecular targets and biophysical consequences at the plasma membrane level. Arch Biochem Biophys. 2015;591: 87–97. 10.1016/j.abb.2015.12.007
    1. Libby P, Ridker PM, Maseri A. Inflammation and atherosclerosis. Circulation 2002;105: 1135–1143.
    1. Nasiek M, Kos-Kudla B, Ostrowska Z, Marek B, Kajdaniuk D, Sieminska L, et al. Plasma concentration of soluble intercellular adhesion molecule-1 in women with polycystic ovary syndrome. Gynecol Endocrinol. 2004;19:208–215.
    1. Klinke A, Nussbaum C, Kubala L, Friedrichs K, Rudolph TK, Rudolph V, et al. Myeloperoxidase attracts neutrophils by physical forces. Blood 2011;117: 1350–1358. 10.1182/blood-2010-05-284513
    1. Nussbaum C, Klinke A, Adam M, Baldus S, Sperandio M. Myeloperoxidase: a leukocyte-derived protagonist of inflammation and cardiovascular disease. Antioxid Redox Signal. 2013;18: 692–713. 10.1089/ars.2012.4783

Source: PubMed

3
Abonner