ArticlePDF Available

Abstract and Figures

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.
Content may be subject to copyright.
RESEARCH ARTICLE
Insulin Resistance in PCOS Patients Enhances
Oxidative Stress and Leukocyte Adhesion:
Role of Myeloperoxidase
Victor M. Victor
1,2,3,4
*, Susana Rovira-Llopis
1,2
, Celia Bañuls
1,2
, Noelia Diaz-Morales
1
,
Arantxa Martinez de Marañon
1
, Cesar Rios-Navarro
3
, Angeles Alvarez
3,5
,
Marcelino Gomez
1
, Milagros Rocha
1,2,3
*, Antonio Hernández-Mijares
1,2,6
*
1Service of Endocrinology, University Hospital Doctor Peset, Foundation for the Promotion of Health and
Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain, 2Institute of Health Research
INCLIVA, University of Valencia, Valencia, Spain, 3CIBERehd - Department of Pharmacology and
Physiology, University of Valencia, Valencia, Spain, 4Department of Physiology, University of Valencia,
Valencia, Spain, 5General Foundation of the University of Valencia, Valencia, Spain, 6Department of
Medicine, University of Valencia, Valencia, Spain
*hernandez_antmij@gva.es (AH-M); milagros.rocha@uv.es (MR); Victor.Victor@uv.es (VMV)
Abstract
Cardiovascular diseases and oxidative stress are related to polycystic ovary syndrome
(PCOS) and insulin resistance (IR). We have evaluated the relationship between myeloper-
oxidase (MPO) and leukocyte activation in PCOS patients according to homeostatic model
assessment of IR (HOMA-IR), and have explored a possible correlation between these fac-
tors 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 sub-
jects. Increases in IL-6 and TNF-αand adhesion molecules (E-selectin, ICAM-1 and VCAM-
1) were also observed, particularly in the PCOS IR group, providing evidence that inflamma-
tion 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 par-
ticularly 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 condi-
tions are aggravated by the presence of IR.
PLOS ONE | DOI:10.1371/journal.pone.0151960 March 23, 2016 1/16
OPEN ACCESS
Citation: Victor VM, Rovira-Llopis S, Bañuls C, Diaz-
Morales N, Martinez de Marañon A, Rios-Navarro C,
et al. (2016) Insulin Resistance in PCOS Patients
Enhances Oxidative Stress and Leukocyte Adhesion:
Role of Myeloperoxidase. PLoS ONE 11(3):
e0151960. doi:10.1371/journal.pone.0151960
Editor: Wenjun Ding, University of Chinese Academy
of Sciences, CHINA
Received: September 3, 2015
Accepted: March 7, 2016
Published: March 23, 2016
Copyright: © 2016 Victor et al. This is an open
access article distributed under the terms of the
Creative Commons Attribution License, which permits
unrestricted use, distribution, and reproduction in any
medium, provided the original author and source are
credited.
Data Availability Statement: All relevant data are
within the paper and its Supporting Information file.
Funding: This study was financed by grants PI12/
1984, PI13/1025, PI13/0073, PI15/1424, CIBERehd
CB06/04/0071, PROMETEOII2014/035, SAF2010/
16030, UGP-14-93, UGP-14-95 and by the European
Regional Development Fund (ERDF A way to build
Europe). VMV and MR are recipients of contracts
from the Ministry of Health of the Valencian Regional
Government and Carlos III Health Institute (CES10/
030 and CP10/0360, respectively). SR-L is the
recipient of a predoctoral fellowship from Carlos III
Introduction
Polycystic ovary syndrome (PCOS) occurs in 620% of reproductive aged women [12]. Insu-
lin resistance (IR) is related to PCOS [3], and metabolic syndrome is reported in PCOS
patients, thus increasing the risk of major cardiovascular events, morbidity and diabetes and
affecting patient quality of life and overall health care costs [46].
Inflammation and oxidative stress have been related to the pathogenesis of PCOS [78],
including an increase in reactive oxygen species (ROS) production by peripheral blood leuko-
cytes [910], activation of leukocyte-endothelium interactions [11] and the proinflammatory
transcription factor nuclear κβ (NF-κβ), and a rise in the levels of proinflammatory cytokines
[12] and C-reactive proteins [13]. Oxidative stress has been implicated in the etiology of IR in
leukocytes from PCOS patients, and an increase in leukocytes has been highlighted as a putative
marker of low-grade chronic inflammation and early cardiovascular risk in these subjects [13].
In this sense, it has been suggested that some enzymes, such as myeloperoxidase (MPO), a
heme protein derived from leukocytes, play an important role in leukocyte-mediated endothe-
lium damage in inflammation and cardiovascular diseases [14]. MPO is released from activated
leukocytes at inflammatory sites, generating reactive oxygen species (ROS). However, the anti-
microbial activity of MPO can also produce oxidative damage in the endothelium and vessel
wall, thus promoting CVD and clinical complications [15]. In light of this, several studies have
explored the relationship between MPO and PCOS [1618].
Leukocytes can adhere to the endothelium and migrate to the bacterial focus, where the
pathogen is killed by ROS production and phagocytosis. Under some situations of IR, such as
type 2 diabetes, there is an increased recruitment of leukocytes [19] that is associated with
endothelial dysfunction. In this sense, arterial stiffness can be more pronounced in PCOS sub-
jects, independently of age, body mass index (BMI) and blood pressure [20]. PCOS is usually
linked to endothelial dysfunction due to the high levels of glucose present in patients, and this
process is normally associated with endothelial impairment and, in turn, leukocyte-endothe-
lium interactions.
The primary outcome of this study was to evaluate the relationship between MPO levels,
ROS production and leukocyte-endothelium interactions, adhesion molecules (E-Selectin,
ICAM-1 and VCAM-1) and proinflammatory cytokines (IL-6 and TNF-α) in PCOS according
to HOMA-IR. A secondary objective was to assess potential correlations between these factors
and endocrine and inflammatory parameters.
Materials and Methods
Subjects
The study was conducted in the Service of Endocrinology, University Hospital Dr. Peset,
Valencia, Spain. 101 women with PCOS (age 25.8 ± 5.3) and 105 controls (age 26.4 ± 5.6) were
selected according to age and BMI (Table 1). Controls were volunteers recruited from the Uni-
versity Hospital Dr Peset and the Faculty of Medicine (University of Valencia). PCOS subjects
were diagnosed using the Rotterdam criteria [21], which are as follows: oligoovulation (cycles
longer than 35 days or less than 26 days) [22]; elevated free testosterone levels (>0.5 ng/dl; the
cut-off level for free testosterone was the mean ± 2 SD according to normal levels in controls);
hirsutism (total Ferriman-Gallwey score>7) and polycystic ovaries, identified by transvaginal
ultrasonography and following the Rotterdam criteria; i.e. the presence of 12 or more small (2
to 9 mm) follicles in each ovary. Ultrasound scans were performed and scored independently
by one of two experienced and blinded reviewers. None of the subjects had any systemic or
endocrine disease or galactorrhea, or any condition which could have affected her reproductive
Myeloperoxidase, Insulin Resistance and PCOS
PLOS ONE | DOI:10.1371/journal.pone.0151960 March 23, 2016 2/16
Health Institute (FI11/00637). CB is the recipient of a
postdoctoral contract from Carlos III Health Institute
(CD14/00043). ND-M is the recipient of a predoctoral
contract from Carlos III Health Institute (FI14/00125).
CR-N was supported by Conselleria de Educación,
Cultura y Deporte (CPI-13-194). AA was supported
by Ministerio de Ciencia e Innovación (Ramón y Cajal
program RYC2005-002295 and I3 program).
Competing Interests: The authors have declared
that no competing interests exist.
Abbreviations: PCOS, polycystic ovary syndrome;
IR, insulin resistance; MPO, myeloperoxidase;
HOMA, homeostatic model assessment; ROS,
reactive oxygen species; ICAM-1, intercellular
adhesion molecule 1; VCAM-1, vascular cell
adhesion molecule 1; IL-6, interleukin 6; TNF-α,
tumor necrosis factor alpha; hsCRP, high sensitivity
c-reactive protein; CVD, cardiovascular disease; NF-
κβ, nuclear factor kappa B; BMI, body mass index;
LDLc, low density lipoproteins cholesterol; HDLc, high
density lipoproteins cholesterol; Apo, apolipoprotein;
LH, luteinizing hormone; FSH, follicle-stimulating
hormone; SHBG, sex hormone binding globulin;
DHEAS, dehydroepiandrosterone-sulfate; PMNs,
polymorphonuclear leukocytes; DCFH-DA,
dichlorodihydrofluorescein diacetate; HUVEC, human
umbilical vein endothelial cells; MetS, Metabolic
Syndrome.
physiology. Absence during the previous semester of any medication that might have affected
the hypothalamic-pituitary-gonadal axis was confirmed in all subjects.
Exclusion criteria were malignant neoplasia, anemia, active infectious diseases or thrombo-
embolism, stroke or history of ischaemic heart disease, diabetes mellitus and the taking of
lipid-lowering or antihypertensive drugs.
Biochemical determinations
An anthropometric and analytical evaluation was performed and weight (kg), height (m) and
waist (cm) measured in all subjects. Body mass index (BMI = weight (kg) / height (m)
2
) was then
calculated. Blood was collected from the antecubital vein at 810 a.m, after 12 hours of fasting,
during the follicular phase, on the second/third day of the menstrual cycle or after 3 months of
amenorrhea. In cases of very irregular cycles, blood was collected after progesterone deprivation.
Low density lipoproteins cholesterol (LDLc) concentration was calculated using the Friede-
wald method. Total cholesterol and triglycerides were measured by means of enzymatic assays,
and high density lipoproteins cholesterol (HDLc) concentrations were recorded with a Beck-
man LX-20 autoanalyser (Beckman Coulter, La Brea, CA, USA) using a direct method. The
intraserial variation coefficient was <3.5% for all determinations. HbA1c was determined with
Table 1. Anthropometric parameters, lipoprotein profile, hydrocarbonated metabolism parameters and circulating androgens of PCOS women
and control subjects.
Controls (n = 105) PCOS HOMA-IR<2.5 (n = 65) PCOS HOMA-IR2.5 (n = 36) P-value P-value BMI adjusted
Age (years) 26.4 ±5.6 25.3 ±5.3 26.6 ±5.9 0.671 0.249
Body weight (kg) 68.4 ±18.5 68.9 ±11.7 78.8 ±10.8 0.144 -- -
BMI (kg/m
2
)25.6 ±6.8 25.4 ±4.2 28.3 ±4.6 0.368 -- -
Waist (cm) 82.1 ±15.7 84.8 ±10.2 91.1 ±9.8 0.093 0.155
Systolic BP (mmHg) 110.3 ±14.9 111.6 ±13.7 111.2 ±11.5 0.859 0.993
Diastolic BP (mmHg) 67.6 ±12.0 71.0 ±9.3 75.9 ±11.6 0.016 0.038
Total cholesterol (mg/dl) 173.3 ±31.3 173.0 ±33.6 198.3 ±37.8*0.011 0.442
LDLc (mg/dl) 104.0 ±24.3 106.1 ±30.0 124.7 ±26.2*0.007 0.412
HDLc (mg/dl) 55.7 ±12.9 53.8 ±10.7 44.8 ±13.3*0.002 0.061
Triglycerides (mg/dl) 70.6 ±36.3 65.6 ±30.3 143.8 ±97.5
#
*<0.001 0.001
Apo AI (mg/dl) 152.9 ±37.1 148.1 ±22.6 135.3 ±27.9 0.108 0.316
Apo B (mg/dl) 77.0 ±19.3 76.3 ±23.9 96.4 ±28.1*0.004 0.090
hsCRP (mg/l) 2.07 ±2.46 3.29 ±3.42 3.72 ±3.19*0.018 0.137
Glucose (mg/dl) 83.1 ±10.5 81.5 ±8.9 85.6 ±7.8 0.280 0.483
Insulin (μIU/ml) 6.86 ±2.75 7.23 ±2.31 19.65 ±9.12
#
*<0.001 <0.001
HOMA-IR 1.38 ±0.56 1.75 ±0.34 4.17 ±2.04
#
*<0.001 <0.001
HbA1c (%) 5.16±0.27 5.19±0.21 5.26±0.28 0.444 0.873
FSH (mIU/ml) 4.31 ±2.36 4.50 ±1.36 4.59 ±1.37 0.871 0.503
LH (mIU/ml) 4.59 ±4.04 6.43 ±4.29*5.14 ±2.95 0.015 0.030
Testosterone (ng/ml) 0.45 ±0.21 0.77 ±0.44*0.68 ±0.46*<0.001 <0.001
DHEA-S (μg/dl) 244.7 ±101.8 331.9 ±161.1*273.4 ±145.3 0.005 0.011
Androstendione (ng/ml) 2.79 ±1.22 4.05 ±1.88*4.16 ±2.24*<0.001 <0.001
SHBG (nmol/l) 106.5 ±75.4 63.6 ±52.3*47.4 ±50.8*<0.001 <0.001
Data are expressed as mean ±SD. Statistical signicance (p<0.05) was considered when compared by an ANOVA followed by a post hoc test and using
BMI as covariate
(*p<0.05 when comparing PCOS vs Controls; # p<0.05 when comparing PCOS HOMA-IR2.5 vs HOMA-IR<2.5)
doi:10.1371/journal.pone.0151960.t001
Myeloperoxidase, Insulin Resistance and PCOS
PLOS ONE | DOI:10.1371/journal.pone.0151960 March 23, 2016 3/16
an Automatic Glycohemoglobin Analyzer (Arkray, Inc., 73 KYOTO, Japan). Apolipoprotein
(Apo) AI and B were determined by immunonephelometry (Dade Behring BNII, Marburg,
Germany) with an intra-assay variation coefficient of <5.5%. High sensitivity C-reactive pro-
tein (hsCRP) was evaluated by an immunonephelometric assay (Behring Nephelometer II,
Dade Behring, Inc., Newark, DE, USA) with an intra-assay coefficient of variation of 8.7% and
sensitivity of 0.01 mg/L. Glucose levels were measured using enzymatic techniques and a Dax-
72 autoanalyzer (Bayer Diagnostic, Tarrytown, New York, USA). The enzymatic luminescence
technique was employed to measure insulin. Samples were processed immediately in order to
avoid haemolysis and were frozen until analysis. IR was calculated by homeostasis model
assessment (HOMA) using baseline glucose and insulin: HOMA = (fasting insulin (μU/ml) ×
fasting glucose (mmol/L)/22.5. PCOS patients were classified as PCOS-IR when the HOMA
index was >than 2.5 and PCOS non-IR when the HOMA index was <2.5, as in a previous
study [23]. This cut-off point for IR has been established in our hospitals Endocrinology
Department based on the distribution of the HOMA index for women in our clinical setting
(University Hospital Dr. Peset, Valencia, Spain).
Serum luteinizing hormone (LH), follicle-stimulating hormone (FSH) were measured using
a 2-site monoclonal non-isotopic system (Architect, Abbott Laboratories, Abbott Park, IL). Sex
hormone binding globulin (SHBG), androstendione and testosterone were measured using spe-
cialised chemiluminiscence techniques in our hospitals Clinical Analysis Service. Dehydroepi-
androsterone-sulfate (DHEAS) was measured using a specific chemiluminescence technique.
Insulin resistance, medication and a documented history of vascular disease (ischemic car-
diopathy, peripheral arteriopathy, or cerebrovascular accident) or diabetes were ruled out in all
the volunteer control subjects. Umbilical cords were obtained from control women during nor-
mal delivery, and those damaged by hematoma or under pathological conditions were dis-
carded. Informed written consent was obtained from all subjects prior to participation. The
study was approved by the ethics committee of the University Hospital Dr. Peset and was per-
formed in accordance with the Helsinki declaration.
Cells
Human polymononuclear leukocytes (PMNs) were obtained from citrated blood samples and
incubated for 45 min with dextran (3%). The supernatant was centrifuged at 250g for 25 min
over Fycoll-Hypaque. Lysis buffer was added to the pellet and centrifuged at room temperature
(100g, 5 min). PMNs were evaluated in a Scepter device (Millipore, MA, USA), washed in
HBSS medium and stored in complete RPMI media.
Measurement of total and mitochondrial ROS production. Total ROS production was
evaluated in PMNs by two methods. Cells were incubated (30 min) with the fluorescent probe
(5 x 10
6
mol/L) 2,7-dichlorodihydrofluorescein diacetate (DCFH-DA) [24]. First, ROS pro-
duction was assessed by fluorimetry using a Synergy Mx plate reader (BioTek Instruments,
Winooski, VT). Second, it was assessed using a fluorescence microscope (IX81, Olympus,
Hamburg, Germany) coupled with the static cytometry software ScanRversion 2.03.2 (Olym-
pus). For static cytometry, PMNs from each subject were seeded in triplicate in 48-well plates
and 16 images per well were recorded and analyzed [24].
The fluorescent probe Mitosox Red (5μM) was employed to assess mitochondrial ROS pro-
duction. Leukocytes were seeded in 48-well plates and incubated for 30 min with the respective
fluorochrome and washed with HBSS. 16 images per well were recorded with an IX81 Olympus
fluorescence microscope (Olympus, Hamburg, Germany), and the static cytometry software
ScanRversion 2.03.2 (Olympus) was used to quantify fluorescence individually (per cell).
Fluorescent probes were purchased from Invitrogen (Life Technologies, Barcelona, Spain).
Myeloperoxidase, Insulin Resistance and PCOS
PLOS ONE | DOI:10.1371/journal.pone.0151960 March 23, 2016 4/16
MPO Assay
Plasma MPO concentrations were measured using an immunoassay based on a double-anti-
body sandwichtechnique according to the manufacturer's instructions (MPO EIA kit, Cay-
man Chemical) [14]. This is an immunometric assay in which a monoclonal antibody specific
for MPO is used to capture MPO in the plate wells and an HRP-labeled MPO monoclonal anti-
body is employed to detect the captured MPO. The concentration of the enzyme is determined
using the chromogenic substrate for HRP, 3,3,5,5-tetramethylbenzidine. The intra-assay coef-
ficient of variation (CV) of this immunoassay is 6.7% and the inter-assay CV is 8.3%.
Adhesion assay
The human umbilical vein endothelial cells (HUVEC) used in the adhesion studies were har-
vested from umbilical cords by treating them with collagenase. In short, umbilical cord veins
were rinsed of blood products with warm phosphate-buffered saline (PBS), after which the vein
was filled with collagenase (1 mg/mL) for 17 min at 37°C. The cords were then gently massaged
to ensure detachment of endothelial cells from the vessel wall. The digest was collected, centri-
fuged and pelleted. The pellet was resuspended in endothelial cell growth medium (EGM-2)
inside T25 culture flasks in which cells were cultured until confluence. After reaching conflu-
ence, primary cultures were detached with trypsin and transferred to 6-well plate culture dishes.
HUVEC were cultured on fibronectin (5 mg/mL)-coated 25-mm plastic coverslips until conflu-
ent (48 h). For the flow chamber in vitro model study, human leukocytes (1 x 10
6
cells/mL)
were resuspended in Dulbeccos PBS containing 20 x 10
3
mol/L HEPES and 0.1% human
serum albumin, and were drawn across the HUVEC monolayer under a flow rate of 0.36
mL/min (approximately shear stress of 0.7 dyne/cm
2
) under a microscope (Nikon Eclipse TE
2000-S; Amstleveen, The Netherlands) connected to a video camera (Sony Exware HAD; Koeln,
Germany) [25]. Images in a single field of view were recorded over a 5 min period and rolling
and adhesion parameters were evaluated. The rolling velocity in the field of focus was deter-
mined by measuring the time required by 20 consecutive leukocytes to cover a distance of
100 μm. Leukocyte rolling flux was estimated as the number of leukocytes rolling over 100 μm
2
of the endothelial monolayer during a 1 min period. Adhesion was evaluated by counting the
number of PMNs that maintained stable contact with HUVEC for 30 sec. Platelet-activating fac-
tor (1 μmol/L, 1 h) and TNF-α(10 ng/mL, 4 h) were used as positive controls for leukocytes and
HUVEC, respectively.
Adhesion molecules and levels TNF-αand IL-6 levels
A Luminex 200 flow analyser system was employed to analyse adhesion molecules (E-selectin,
ICAM-1 and VCAM-1) and proinflammatory cytokines (IL-6 and TNF-α) in serum from con-
trols and PCOS patients (Austin, TX, USA).
Drugs and solutions
Kits for glucose, insulin, total cholesterol, triglycerides, HDLc and LDLc were purchased from
Abbott Laboratories (Abbott Park, IL, USA). HsCRP kits were supplied by Beckman Corp
(Brea, A, USA). The HbA1c kit was purchased from Menarini Diagnostics (Florence, Italy).
The MPO kit was purchased from Cayman Chemical (Michigan, USA).
Glucose, trypan blue, arginine, glutathione reductase, H
2
O
2
, haemoglobin, RPMI1640 sup-
plemented with 20 mM HEPES, HBSS, TNF-α, human serum albumin (HSA, Albuminate
25%) and fibronectin were obtained from Sigma-Aldrich (Sigma Chem. Co., St. Louis, MO,
USA). Dextran was acquired from Fluka (St. Louis, MO, USA). HBSS was supplied by Cambrex
Myeloperoxidase, Insulin Resistance and PCOS
PLOS ONE | DOI:10.1371/journal.pone.0151960 March 23, 2016 5/16
(Verviers, Belgium). DCFH-DA and Mitosox tracker were provided by Calbiochem (San
Diego, CA, USA). Dulbeccos PBSwith (DPBS
+
) or without (DPBS
-
)Ca
2+
and Mg
2+
endothelial cell growth medium culture media and fetal bovine serum were obtained from
LONZA (Verviers, Belgium). Plastic coverslips (diameter of 25 mm) were purchased from
Nunc (Thermo Fisher Scientific). PBS, collagenase, and trypsin-EDTA were obtained from
Invitrogen (Eugene, OR, USA). Ficoll-Paque TM Plus was purchased from GE Healthcare (Lit-
tle Chalfont, Buckinghamshire,UK).
Statistical analysis
Data analysis was performed with SPSS 17.0. The values in Table 1 are mean ± SD. Bar graphs
show mean ± SEM. Data were compared with a one-way analysis of variance (ANOVA), fol-
lowed by a post hoc test. Analysis of covariance was employed to minimize the potential influ-
ence of BMI. Serum lipid and biochemical parameter changes were analyzed, using BMI as a
covariate. Correlations were calculated using Pearsons correlation coefficient. Significant dif-
ferences were considered when p<0.05.
Results
Metabolic and clinical characteristics
Baseline anthropometric characteristics of the PCOS and control groups were found to be simi-
lar, with no statistically significant differences regarding age, body weight, BMI, waist circum-
ference or systolic blood pressure, as shown in Table 1. However, as expected, there was a trend
towards higher BMI levels in PCOS women as their insulin resistance increased. There was a
significant increase in LH, testosterone, DHEA-S and androstendione levels and a decrease in
SHBG in patients with HOMA-IR<2.5 with respect to controls. Furthermore, patients with
HOMA-IR>2.5 exhibited a significant increase in diastolic BP, total cholesterol, LDLc, triglyc-
erides, hsCRP, insulin, testosterone and androstendione (p<0.05 for all) and a significant
reduction in HDLc and SHBG (p<0.05) with respect to the control group (Table 1). These dif-
ferences remained after adjustment for BMI, with the exception of diastolic BP, total choles-
terol, LDLc, HDLc, Apolipoprotein B and hsCRP. No changes were detected in the
biochemical parameters of any of the PCOS groups according to HOMA-IR, exception of tri-
glycerides and insulin (p<0.05).
The prevalence of MetS was 38.9%, 4.6% and 7.9% among PCOS IR, non-IR and controls,
respectively (p<0.001). When metabolic and oxidative stress parameters and MPO were analyzed
according to presence or absence of MetS in PCOS, we observed that patients with MetS had an
altered metabolic profile and higher levels of ROS production, hsCRP and ICAM-1 (S1 Table).
Total and mitochondrial ROS production
DCFH-DA fluorescence was significantly higher (p<0.001) in leukocytes from PCOS patients
(Fig 1A). When PCOS subjects were divided into insulin-resistant (PCOS IR group,
HOMA-IR>2.5) and non-insulin-resistant (PCOS non-IR group, HOMA-IR<2.5) groups, an
increase in DCFH fluorescence with respect to controls was observed in the PCOS non-IR
(p<0.05, Fig 1B and 1C) and PCOS IR (p<0.001, Fig 1B and 1C) groups, which was evidence
of enhanced ROS production and, consequently, oxidative stress. ROS production (p<0.001,
Fig 1B and 1C) was significantly higher in the PCOS IR group than in the PCOS non-IR group.
Representative images of fluorescence microscopy are shown in Fig 1D.
In addition, we evaluated levels of ROS depending on the levels of glucose and insulin in
each PCOS patient, finding a significant correlation between ROS production and HOMA
Myeloperoxidase, Insulin Resistance and PCOS
PLOS ONE | DOI:10.1371/journal.pone.0151960 March 23, 2016 6/16
(r = 0.621, p = 0.002), insulin (r = 0.581, p = 0.004) and glucose (r = 0.301, p = 0.163). More-
over, when we subdivided the populations of PCOS patients according to tertiles of insulin
(T1<7.6, T2: 7.615.6, T3 >15.6 μUI/ml), we observed that those with a higher tertile of insu-
lin presented higher levels of ROS production (T1 = 34.5±19.4, T2 = 36.4±22.5, T3 = 62.6
±12.9; p = 0.017), and when we subdivided them according to tertiles of glucose (T1<81.0, T2:
81.088.0, T3 >88.0 mg/dl), we observed that those with the higher tertile of glucose presented
an increase of ROS production (T1 = 45.9±23.8, T2 = 34.9±19.6, T3 = 61.2±15.2; p<0.05).
Mitosox Red fluorescence was significantly higher (p<0.05) in leukocytes from PCOS
patients (Fig 2A). When PCOS subjects were divided into insulin-resistant (PCOS IR group,
HOMA-IR>2.5) and non-insulin-resistant (PCOS non-IR group, HOMA-IR<2.5) groups, an
increase in Mitosox Red fluorescence with respect to controls was observed in the PCOS non-
IR (p<0.05, Fig 2B) and PCOS IR (p<0.05, Fig 2B) groups, which was evidence of enhanced
mitochondrial ROS production, thus indicating conditions of oxidative stress.
Myeloperoxidase levels
Therefore, we evaluated the levels of MPO in plasma from PCOS patients and controls and
found that the levels of MPO were higher among the former (p<0.01, Fig 3A). In addition, we
assessed MPO levels according to the presence or absence of IR and found them to be signifi-
cantly higher in the PCOS IR (p<0.01) and non-IR PCOS (p<0.05) groups than in controls
(Fig 3B). Levels of MPO were higher (p<0.05) in the PCOS IR group than in the PCOS non-IR
group.
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). *p<0.05 ***p<0.001 vs. Control;
c
p<0.001 between PCOS non-IR and PCOS IR subjects.
doi:10.1371/journal.pone.0151960.g001
Myeloperoxidase, Insulin Resistance and PCOS
PLOS ONE | DOI:10.1371/journal.pone.0151960 March 23, 2016 7/16
Leukocyte-endothelial interactions
We aimed to determine whether the IR control status of PCOS patients is related to leukocyte-
endothelial interactions. In PCOS patients, we found a decrease in PMN rolling velocity (Fig
4A,p<0.001) and an increase in PMN rolling flux (Fig 4C,p<0.001) and PMN adhesion (Fig
4E,p<0.001) with respect to controls. Furthermore, when these parameters were evaluated
according to the presence or absence of IR, leukocyte rolling velocity was found to be dimin-
ished in both PCOS groups dependently of IR control status (p<0.05 in the PCOS non-IR
group, p<0.001 in the PCOS IR group, Fig 4B). Leukocyte rolling flux (Fig 4D) was enhanced
in non-IR PCOS patients (p<0.05) and was significantly more pronounced in PCOS-IR
patients (p<0.001). Leukocyte adhesion to the endothelium was enhanced in both PCOS
groups, (p<0.05 in PCOS non-IR patients, and p<0.001 in PCOS-IR patients, Fig 4F) in com-
parison to controls. In addition, there was a significant decrease (p<0.01, Fig 4B) of PMN roll-
ing velocity and an increase of PMN rolling flux (p<0.05, Fig 4D) and PMN adhesion (p<0.01,
Fig 4F) in the PCOS IR group with respect to the PCOS non-IR group.
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<0.05 and **p<0.01 PCOS versus
control group.
doi:10.1371/journal.pone.0151960.g002
Fig 3. Plasma MPO concentrations in PCOS and control subjects (A) and in control, PCOS non-IR and PCOS IR subjects (B). *p<0.05 and **p<0.01
vs. Control;
a
p<0.05 between PCOS non-IR and PCOS IR subjects.
doi:10.1371/journal.pone.0151960.g003
Myeloperoxidase, Insulin Resistance and PCOS
PLOS ONE | DOI:10.1371/journal.pone.0151960 March 23, 2016 8/16
Levels of adhesion molecules and proinflammatory cytokines
PCOS patients exhibited increased levels of E-Selectin (Fig 5A,p<0.001), ICAM-1 (Fig 5C,
p<0.01) and VCAM-1 (Fig 5E,p<0.05). In addition, when these parameters were evaluated
according to the presence or absence of IR, an increase in the adhesion molecule ICAM-1 was
detected in PCOS non-IR patients (p<0.05, Fig 5D) and an increase in E-selectin (p<0.001, Fig
5B), ICAM-1 (p<0.01, Fig 5D) and VCAM-1 levels was observed in PCOS IR subjects (p<0.05,
Fig 5F) with respect to controls. In addition, there was a significant increase in E-selectin
(p<0.01) and VCAM-1 (p<0.05) in the PCOS IR group with respect to the non-IR group.
Finally, we found an increase in the proinflammatory cytokines IL-6 (Fig 6A,p<0.05) and
TNF-α(Fig 6C,p<0.01) with respect to controls. These effects were more evident among IR
patients (p<0.05 in PCOS non-IR for TNF-αand p<0.01 in IR subjects for IL-6 and TNF-α
Fig 6B and 6D). In addition, there was an increase of IL-6 (p<0.05) in the PCOS IR group with
respect to the PCOS non-IR group.
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.
a
p<0.05 and
b
p<0.01 between PCOS non-IR and PCOS IR
subjects.
doi:10.1371/journal.pone.0151960.g004
Myeloperoxidase, Insulin Resistance and PCOS
PLOS ONE | DOI:10.1371/journal.pone.0151960 March 23, 2016 9/16
Correlation studies
When we explored potential correlations among HOMA-IR and inflammation/adhesion
parameters we found that the former 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) (Fig 7).
Discussion
It has previously been described that PCOS subjects are in a proinflammatory state [26], and
that IR usually can coexist with PCOS, not only in obese patients, but also in lean subjects.
Although vascular dysfunction and endothelial/leukocyte interactions are key features of IR
[27] and are thought to be a major cause of IR-associated vascular complications, the underly-
ing molecular mechanisms are still not understood. In the present study, we have evaluated, as
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. *p<0.05, **p<0.01, ***p<0.001 with respect to the control group;
a
p<0.05 and
b
p<0.01
between PCOS non-IR and PCOS IR subjects.
doi:10.1371/journal.pone.0151960.g005
Myeloperoxidase, Insulin Resistance and PCOS
PLOS ONE | DOI:10.1371/journal.pone.0151960 March 23, 2016 10 / 16
primary outcome measures, the relationship between MPO, oxidative stress, endothelial/leuko-
cyte interactions and adhesion molecules in PCOS patients with different HOMA-IR indexes,
and have explored possible correlations between these factors and endocrine and inflammatory
parameters.
PCOS is a heterogeneous syndrome associated with a wide range of endocrine and meta-
bolic abnormalities, including hyperinsulinaemia, hyperglycaemia, dyslipidaemia and obesity,
which are regarded as hallmark components of MetS. In our population, the prevalence of
MetS in IR vs non-IR PCOS patients was significantly higher, which is consistent with previous
reports showing a higher incidence among approximately one-third of PCOS women, espe-
cially among those with the highest BMIs and insulin levels [28]. While obesity is regarded as
one of the putative factors leading to MetS, the link between PCOS and MetS would seem to be
attributable mainly to IR [29].
ROS can play an important role in hyperglycemia-mediated microvascular complications
and endothelial dysfunction in IR conditions [3031]. In fact, it has been demonstrated that
PCOS patients have an increased risk of developing metabolic syndrome, which may be related
to oxidative stress and cardiovascular events [6]. Our results confirm that PCOS is related to an
increase in ROS production in PMN, and show that this ROS production is related to the pres-
ence of IR. In the case of MPO, an important leukocyte-derived pro-oxidant enzyme, some
studies have focused specifically on its relationship with PCOS or conditions of IR. In the pres-
ent study, we have observed that levels of MPO are higher in the leukocytes of PCOS patients,
and that this increase is more pronounced in the presence of IR. Our results are in accordance
with those of Ribeiro et al., who reported increased levels of MPO in IR PCOS patients [16].
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.*p<0.05, **p<0.01 with respect to control group;
a
p<0.05 between PCOS non-IR and PCOS IR subjects.
doi:10.1371/journal.pone.0151960.g006
Myeloperoxidase, Insulin Resistance and PCOS
PLOS ONE | DOI:10.1371/journal.pone.0151960 March 23, 2016 11 / 16
MPO seems to play an important role in endothelial dysfunction [32], is released from gran-
ules of activated leukocytes, and can generate ROS as a system of defense against bacteria [33].
However, the antimicrobial activity of MPO can also lead to oxidative damage of the endothe-
lium and vessel wall. Activated leukocyte-released MPO binds to the vascular wall for a long
Fig 7. Correlation studies of HOMA-IR and inflammation and adhesion parameters in PCOS women.
doi:10.1371/journal.pone.0151960.g007
Myeloperoxidase, Insulin Resistance and PCOS
PLOS ONE | DOI:10.1371/journal.pone.0151960 March 23, 2016 12 / 16
time and can release ROS continuously, therefore increasing endothelial damage [15]. In this
context, our group has previously demonstrated an increase in MPO levels in diabetic patients
that correlated with the presence of nephropathy and leukocyte-endothelium interactions [14],
thus pointing to the importance of MPO in conditions of oxidative stress. However, other stud-
ies have shown that poor glycemic control in diabetic patients results in decreased histochemi-
cal MPO activity in neutrophils [34].
There are several mechanisms by which MPO can promote the atherosclerotic process: oxi-
dation of LDLc [35]; MPO-induced oxidation of HDLc [36]; and consumption and catabolism
of endothelial-derived nitric oxide, which can lead to endothelial dysfunction and plaque for-
mation [3739]. Furthermore, a correlation has been reported between plasma MPO concen-
tration and red blood cell rigidity index in type-2 diabetes patients with coronary heart disease
[40]. These findings suggest that MPO functions as a mediator of regulatory mechanism in
microcirculation.
Pathophysiological and inflammatory states such as hypertension and atherosclerosis are
characterized by leukocyte recruitment to the arterial wall [41]. To study this process, we have
used an in vitro model in which human leukocytes flow over a monolayer of human endothe-
lial cells with a shear stress similar to that observed in vivo [11]. This reproduces the process
that precedes inflammation in vivo (rolling and adhesion) and which is critical to homeostasis
and vascular cell integrity. If these interactions are exacerbated, the vascular dysfunction and
injury associated with many CVD can occur.
In the present study, we have demonstrated that the inflammatory state associated with
PCOS is exacerbated in PCOS IR patients, in which there is a decrease in PMN rolling velocity
and an increase in rolling flux and adhesion, thereby inducing leukocyte-endothelium interac-
tions. In relation to this, we have detected an increase in the adhesion molecule ICAM-1 in
PCOS non-IR subjects, and E-selectin, ICAM-1 and VCAM-1 levels in their PCOS IR counter-
parts. An increase of circulating ICAM-1 has previously been described in PCOS [42]. In addi-
tion, there was a significant increase in E-selectin and VCAM-1 (p<0.05) in the PCOS IR
group with respect to the non-IR group, which underlines the essential role of IR in increased
levels of adhesion molecules. In the case of proinflammatory cytokines, higher levels of these
markers were observed in the PCOS patients, particularly in those with IR, thus highlighting
the importance of IR in the development of inflammation in PCOS patients.
Finally, we explored potential correlations between HOMA and inflammation/adhesion
parameters and found that the former was positively correlated with hsCRP, ROS production,
leukocyte rolling flux, E-selectin and IL-6. These results suggest that IR is related to the inflam-
matory state that underlies PCOS and leads to an increase in MPO activity and ROS produc-
tion by leukocytes, which, in turn, enhances leukocyte-endothelium interactions and,
consequently, cardiovascular complications. In fact, the MPO molecule is thought to be capable
of attracting leukocytes to the vascular wall via its electrostatic characteristics [43], which
makes it a main player of inflammation [44].
We did not observe variations in HbA1c levels between the different groups of patients and
controls, but the evaluation of fasting glucose levels does not necessarily rule out abnormal glu-
cose tolerance in PCOS, which represents a potential limitation of this study.
Conclusions
In conclusion, we demonstrate an increase in the rate of ROS and in concentrations of MPO in
PCOS patients in general, and particularly in those with IR. Inflammation associated with
PCOS can induce leukocyte-endothelium interactions and a simultaneous increase in IL-6,
TNF-αand the adhesion molecules E-selectin, ICAM-1 and VCAM-1, conditions that are
Myeloperoxidase, Insulin Resistance and PCOS
PLOS ONE | DOI:10.1371/journal.pone.0151960 March 23, 2016 13 / 16
aggravated by the presence of IR. Enhanced levels of ROS and MPO in PCOS, especially in
PCOS-IR patients, may represent an important underlying factor in the clinical complications
seen in PCOS subjects.
Supporting Information
S1 Table. Metabolic and oxidative parameters in PCOS women according the presence of
metabolic syndrome. Data are expressed as mean ± SD. Statistical significance (p<0.05) was
considered when compared by an umpaired Students t-test.
(DOC)
Acknowledgments
We thank Isabel Soria for her work in the extraction of biological samples (University Hospital
Dr Peset), and B Normanly for his editorial assistance (University of Valencia).
Author Contributions
Conceived and designed the experiments: AH-M VMV. Performed the experiments: SR-L CB
ND-M AM CR-N AA. Analyzed the data: VMV AH-M SR-L CB MR. Contributed reagents/
materials/analysis tools: AA. Wrote the paper: VMV AH-M SR-L CB MR. Provided overall
supervision and patient follow-up: MG.
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: 40064011. PMID: 10566641
2. Dunaif A. Insulin resistance and the polycystic ovary syndrome: mechanism and implications for patho-
genesis. Endocr Rev. 1997; 18: 774800. PMID: 9408743
3. 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: 347363. doi: 10.1093/humupd/dmq001 PMID: 20159883
4. Ehrmann DA. Polycystic ovary syndrome. N Engl J Med. 2005; 352: 12231236. PMID: 15788499
5. Bajuk Studen K, Jensterle Sever M, Pfeifer M. Cardiovascular risk and subclinical cardiovascular dis-
ease in polycystic ovary syndrome. Front Horm Res. 2013; 40: 6482. doi: 10.1159/000341838 PMID:
24002406
6. Mahalingaiah S, Diamanti-Kandarakis E. Targets to treat metabolic syndrome in polycystic ovary syn-
drome. Expert Opin Ther Targets. 2015; 19: 15611574. doi: 10.1517/14728222.2015.1101067 PMID:
26488852
7. 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: 201211. doi: 10.1210/jc.2014-2419 PMID: 25303486
8. Escobar-Morreale HF, Luque-Ramírez M, González F. Circulating inflammatory markers in polycystic
ovary syndrome: a systematic review and metaanalysis. Fertil Steril. 2011; 95: 10481058. doi: 10.
1016/j.fertnstert.2010.11.036 PMID: 21168133
9. 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 Endocri-
nol Metab. 2006; 91: 336340. PMID: 16249279
10. 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 Endo-
crinol Metab. 2009; 94: 35053512. doi: 10.1210/jc.2009-0466 PMID: 19567514
11. 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: 31153122. doi: 10.1210/jc.2011-0651 PMID:
21778215
Myeloperoxidase, Insulin Resistance and PCOS
PLOS ONE | DOI:10.1371/journal.pone.0151960 March 23, 2016 14 / 16
12. 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: 521529. PMID: 16522732
13. Orio F Jr, Palomba S, Cascella T, Di Biase S, Manguso F, TauchmanovàL, et al. The increase of leuko-
cytes as a new putative marker of low-grade chronic inflammation and early cardiovascular risk in poly-
cystic ovary syndrome. J Clin Endocrinol Metab. 2005;90: 25.
14. 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: 14521458. doi: 10.1089/ars.2013.5307 PMID: 23521574
15. 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: 29502959. PMID: 15504976
16. Ribeiro AL, Scapinelli A, Tamanaha S, Oliveira RM, Kowastch I, Mathias W Jr, et al. Myeloperoxidases
and polycystic ovary syndrome. Gynecol Endocrinol. 2012; 28: 36. doi: 10.3109/09513590.2011.
579656 PMID: 21615238
17. 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: 107114. PMID:
23539878
18. Kurdoglu Z, Ozkol H, Kurdoglu M. Serum myeloperoxidase and adenosine deaminase activities in poly-
cystic ovary syndrome. Gynecol Endocrinol. 2012; 28:115118. doi: 10.3109/09513590.2011.583959
PMID: 21756068
19. 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: 455461. PMID: 11812755
20. 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 phe-
notype. J Hypertens. 2013; 31: 19982004. doi: 10.1097/HJH.0b013e3283630362 PMID: 24107731
21. The Rotterdam ESHRE/ASRM-Sponsored PCOS Consensus Workshop Group. Revised 2003 con-
sensus on diagnostic criteria and long-term health risks related to polycystic ovary syndrome. Fertil
Steril. 2004; 81: 1925.
22. 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 ClinEndocrinol Metab. 2004; 89: 453
462. PMID: 14764747
23. Hernández Mijares A, Riera Fortuny C, Solá Izquierdo E, Oliver Oliver MJ, Martínez Triguero ML, Moril-
las Ariño C, et al. Prevalence of metabolic syndrome in patients with coronary heart disease. Med Clin
(Barc) 2003; 121: 204208.
24. 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: 17591765. doi: 10.1089/ars.2014.6030 PMID: 25000244
25. Hernandez-Mijares A, Rocha M, Rovira-Llopis S, Bañuls C, Bellod L, de Pablo C, et al. Human leuko-
cyte/endothelial cell interactions and mitochondrial dysfunction in type 2 diabetic patients and their
association with silent myocardial ischemia. Diabetes Care 2013; 36: 16951702. doi: 10.2337/dc12-
1224 PMID: 23300290
26. 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 Endocri-
nol Metab. 2014; 99: 29422951. doi: 10.1210/jc.2014-1214 PMID: 24873996
27. 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-
βisoform in the endothelium. Circ Res. 2013; 113: 418427. doi: 10.1161/CIRCRESAHA.113.301074
PMID: 23759514
28. 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: 4853. PMID: 16249284
29. Grundy SM. Metabolic syndrome: a multiplex cardiovascular risk factor. J Clin Endocrinol Metab. 2007;
92: 399404. PMID: 17284640
30. Giacco F, Brownlee M. Oxidative stress and diabetic complications. Circ Res. 2010; 107: 10581070.
doi: 10.1161/CIRCRESAHA.110.223545 PMID: 21030723
31. Prieto D, Contreras C, Sánchez A. Endothelial dysfunction, obesity and insulin resistance. Curr Vasc
Pharmacol. 2014; 12:412426. PMID: 24846231
Myeloperoxidase, Insulin Resistance and PCOS
PLOS ONE | DOI:10.1371/journal.pone.0151960 March 23, 2016 15 / 16
32. Anatoliotakis N, Deftereos S, Bouras G, Giannopoulos G, Tsounis D, Angelidis C, etal. Myeloperoxi-
dase: expressing inflammation and oxidative stress in cardiovascular disease. Curr Top Med Chem.
2013; 13: 115138. PMID: 23470074
33. Klebanoff SJ. Myeloperoxidase: friend and foe. J Leukoc Biol. 2005; 77: 598625. PMID: 15689384
34. Unubol M, Yavasoglu I, Kacar F, Guney E, Omurlu IK, Ture M, et al. Relationship between glycemic
control and histochemicale M, et al.; 77: 598625.25. al. Myeloperoxidase: expressing inf diabetes.
Diabetol Metab Syndr. 2015; 7: 119.
35. Podrez EA, Schmitt D, Hoff HF, Hazen SL. Myeloperoxidase-generated reactive nitrogen species con-
vert LDL into an atherogenic form in vitro. J Clin Invest. 1999; 103: 15471560. PMID: 10359564
36. 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: 1303213037. PMID: 15326314
37. Eiserich JP, Baldus S, Brennan ML, Ma W, Zhang C, Tousson A, et al. Myeloperoxidase, a leukocyte-
derived vascular NO oxidase. Science 2002; 296: 23912394. PMID: 12089442
38. Vita JA, Brennan ML, Gokce N, Mann SA, Goormastic M, Shishehbor MH, et al. Serum myeloperoxi-
dase levels independently predict endothelial dysfunction in humans. Circulation 2004; 10: 11341139.
39. 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. PMID: 15304260
40. 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: 8797. doi: 10.1016/j.abb.2015.12.007
PMID: 26714302
41. Libby P, Ridker PM, Maseri A. Inflammation and atherosclerosis. Circulation 2002; 105: 11351143.
PMID: 11877368
42. 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 Endo-
crinol. 2004; 19:208215. PMID: 15724804
43. Klinke A, Nussbaum C, Kubala L, Friedrichs K, Rudolph TK, Rudolph V, et al. Myeloperoxidase attracts
neutrophils by physical forces. Blood 2011; 117: 13501358. doi: 10.1182/blood-2010-05-284513
PMID: 20980678
44. Nussbaum C, Klinke A, Adam M, Baldus S, Sperandio M. Myeloperoxidase: a leukocyte-derived pro-
tagonist of inflammation and cardiovascular disease. Antioxid Redox Signal. 2013; 18: 692713. doi:
10.1089/ars.2012.4783 PMID: 22823200
Myeloperoxidase, Insulin Resistance and PCOS
PLOS ONE | DOI:10.1371/journal.pone.0151960 March 23, 2016 16 / 16
... Oxidative stress refers to the imbalance of two opposite forces in the body, the production of ROS (reactive oxygen species) and antioxidants, in which the damaging effects of ROS are more powerful compared to the compensatory effect of antioxidants in the cells [12]. Two recent clinical trials noted that there is an increase in mitochondrial ROS production, as well as in the inflammatory enzyme myeloperoxidase (MPO), among females with PCOS, and the concentrations were even higher among those who presented with obesity and insulin resistance [13,14]. In more physiological implications, oxidative stress is thought to potentially lead to metabolic and hormonal imbalances, such conditions that are implicated in the development of PCOS [15]. ...
... It was observed that women with PCOS often present with higher serum concentration of tumor necrosis factor (TNF) and C-reactive protein (CRP), as well as increased monocyte and lymphocyte circulating levels, and inflammatory infiltration in ovarian tissue [16,17]. A recent study indicated that concentrations of tumor necrosis factor alpha (TNF-α) and interleukin 6 (IL-6) are higher in females with PCOS, especially among those cases that present with signs of insulin resistance [13]. Other inflammatory markers that have shown to be permanently elevated in PCOS include IL-8 and IL-18 [18]. ...
Article
Full-text available
Polycystic ovarian syndrome (PCOS) is an endocrine condition that impacts nutritional status, metabolic, and hormonal function in females of reproductive age. This condition is associated with increased androgen production (hyperandrogenism) and decreased insulin sensitivity, which often leads to insulin resistance and hyperinsulinemia. This increase in androgen production and insulin resistance is strongly associated with a high incidence of obesity, type-2 diabetes mellitus (T2DM), cardiovascular diseases (CVD), and certain types of gonad-related cancers among females who suffer from this condition. As research continues to grow, it has been demonstrated that PCOS is a complex condition, and some of its characteristics vary among the females that have this disorder. However, it has been suggested that oxidative stress and low-grade chronic inflammation could play an important role in the development of PCOS. Current evidence suggest that phytochemicals could potentially help with weight-loss by reducing oxidative stress and low-grade inflammation, as well as aid in metabolic and hormonal regulation due to their antioxidant properties. Some of the bioactive compounds found in plants that have shown positive effects in the attenuation of PCOS include flavonoids, polyphenols, phytoestrogen, and polyunsaturated fatty acids (PUFAs). Thus, a review of the current literature published on PCOS and phytochemicals was conducted in PubMed, Google Scholar, and the Academy of Nutrition and Dietetics databases for articles published between 2013 and 2023 with a study duration of 1 to 3 months and adequate sample sizes. The main purpose of this review of literature was to investigate the metabolic effects of phytochemical compounds and phytochemical-rich diets on females with PCOS by comparing the results of several randomized clinical trials.
... The present study are shawed that there were a significantly increase (P ˂ 0.05) in the serum of TNF-α in PCOS groups, this results agreed with [17], [16], [28], [6], [15], [30]. ...
... Furthermore, higher testosterone induces abdominal adiposity, insulin resistance, and secondary hyperinsulinemia (62), which in turn have been associated with sodium retention and increased blood pressure (95), impaired vascular function (96), and chronic low-grade inflammation (97). The inflammatory response of various cytokines such as high-sensitivity C-reactive protein, as well as adipokines including adiponectin (98-100), induce formation of reactive oxygen species as markers of oxidative stress and metabolic dysfunction (101,102). This immune response, strongly related to abdominal obesity, has been implicated in atherogenesis by causing endothelial inflammation and vascular dysfunction (99,100,102). ...
Article
Full-text available
Hyperandrogenism in women, such as polycystic ovary syndrome, ovarian hyperthecosis, congenital adrenal hyperplasia, and androgen-secreting tumors, are all associated with increased prevalence of cardiovascular risk factors that include type 2 diabetes, hypertension, dyslipidemia, and metabolic syndrome. However, it is not clear whether this also implies enhanced risk of cardiovascular disease and mortality. Furthermore, the involvement of obesity and menopausal status for cardiometabolic risk in these women has not been elucidated. Based on the most recent systematic reviews and meta-analyses, this review summarizes the latest scientific evidence. To conclude, hyperandrogenism in premenopausal women is associated with enhanced prevalence of cardiovascular risk factors, as well as increased risk of cardiovascular disease and mortality, independently of body mass index. In contrast, elevated cardiovascular risk factors and increased risk of myocardial infarction and stroke in hyperandrogenic postmenopausal women are dependent on obesity. Furthermore, the overall risk of cardiovascular disease and coronary artery disease in hyperandrogenic postmenopausal women is similar to controls. The reason for a reduced cardiometabolic risk after menopause in hyperandrogenic women compared to nonhyperandrogenic women is not clear. It can be speculated that the difference in endocrine balance and metabolic status between women with and without hyperandrogenism might decrease after menopause because hyperandrogenism usually improves with age, whereas menopausal transition itself is associated with androgen dominance and abdominal obesity. Although we have gained increased knowledge about cardiometabolic risks in women with hyperandrogenism, it must be acknowledged that the quality of data is overall low. More research is needed, especially longer and larger follow-up studies in women with hyperandrogenism of different etiologies and phenotypes.
... Also, oxidative imbalance plays a role in the pathogenesis of PCOS, e.g., the MPO G-463A variant is related to a higher risk of PCOS [88]. The co-presence of insulin resistance is associated with increased MPO activity and ROS production, potentiating leukocyte-endothelium interactions [89]. ...
Article
Full-text available
Salivary myeloperoxidase (MPO) is a key mediator of the oral immune system, acting as an enzyme that utilises H2O2 to generate molecules with high bactericidal activity. While MPO determination in plasma is quite common, the use of saliva is still rare. Our systematic review was designed to answer the question “Are salivary levels of myeloperoxidase altered in patients with systemic diseases?”. Following the inclusion and exclusion criteria, we included twenty-six studies. Altered MPO levels in saliva were most commonly found in patients with cardiovascular and gastrointestinal diseases. Most studies concerned unstimulated whole saliva, and only a few of them stimulated, mainly by chewing paraffin. Enzyme-linked immunosorbent assay (ELISA) was the most common method for determination of MPO concentrations in saliva. Increased salivary MPO levels were more often observed for inflammatory diseases, except patients with inflammatory bowel diseases who were eligible for biologic therapy. In conclusion, MPO could be altered in the saliva of patients with systematic diseases, especially cardiovascular or gastrointestinal diseases. However, further investigations are recommended to validate these outcomes.
... A large accumulation of oxygen free radicals causes lipid peroxidation damage in tissue cells, leads to insulin resistance, [29] and promotes the onset of PCOS. [30] Moreover, oxidative stress response is also able to damage follicular theca and granulosa cells, increasing the risk of anovulatory infertility in PCOS patients. [31] Currently, MDA, SOD and GSH-Px are the main indicators to evaluate the oxidative stress response of the body. ...
Article
Full-text available
Purpose: Clinical efficacy of Fuke Qianjin tablets combined with clomiphene citrate on infertility patients with polycystic ovary syndrome (PCOS) was expected to be retrospectively analyzed in this study. Methods: In this paper, 100 infertility patients with PCOS were selected and divided into the observation and control groups based on different medications. Firstly, clinical data of both groups of patients were acquired. Then, the uterine receptivity and ovarian status, the levels of sex hormones, inflammation and oxidative stress, and the pregnancy outcomes between the 2 groups were compared and analyzed before and after treatment. Results: After a variety of comparisons and analyses, Fuke Qianjin tablets combined with clomiphene citrate were confirmed to improve the uterine receptivity and ovarian status, levels of sex hormone, inflammation and oxidative stress, and pregnancy outcomes in infertility patients with PCOS. Conclusions: Overall, Fuke Qianjin tablets + clomiphene citrate treatment shows good clinical efficacy and is worth promotion in clinical practice.
Article
Introduction: Insulin resistance (IR) is confirmed as an important feature among polycystic ovary syndrome (PCOS) patients. Anti-Müllerian hormone (AMH), a vital marker of ovarian dysfunction, is proposed for inclusion in the diagnosis of PCOS in adolescents. We sought to investigate the relationship between the AMH level and IR in Chinese girls with PCOS. Material and methods: 92 girls with PCOS aged 14-18 years were enrolled and divided into 2 subgroups: PCOS with IR group (n = 25) and PCOS without IR group (n = 67). A homeostasis model assessment-insulin resistance (HOMA-IR) value ≥ 2.5 was defined as IR. Clinical data and biochemical indexes were compared between the 2 groups. Multivariate logistic regression analysis and multivariate linear regression analysis were performed to determine which clinical variables were independently associated with IR and AMH level, respectively. Results: PCOS girls with IR had higher levels of AMH than those of PCOS girls without IR (p < 0.01). Moreover, body mass index, triglyceride, and AMH values were shown to be independent risk factors for HOMA-IR after multivariate analysis. Meanwhile, age, insulin, and follicle-stimulating hormone levels were significantly related to AMH levels in those girls. Conclusions: Our findings show that AMH is an independent determinant of IR in PCOS adolescents, and the fasting insulin level is closely associated with the AMH level, which indicates that the AMH pathway might play a role in the development of IR in PCOS adolescents. The interaction between AMH and IR in PCOS girls warrants further large-scale evaluation.
Article
Significance: Metabolic syndrome (MetS) has become a major global public health problem and there is an urgent need to elucidate its pathogenesis and find more effective targets and modalities for intervention. Recent Advances: Oxidative stress and inflammation are two of the major causes of MetS-related symptoms such as insulin resistance and obesity. Nuclear factor erythroid 2 related factor 2 (Nrf2) is one of the important systems responding to oxidative stress and inflammation. As cells undergo stress, cysteines within Kelch-like ECH-associated protein 1 (Keap1) are oxidized or electrophilically modified, allowing Nrf2 to escape ubiquitination and be translocated from the cytoplasm to the nucleus, facilitating the initiation of the antioxidant transcriptional program. Meanwhile, a growing body of evidence points out a specific modulation of mitochondrial homeostasis by Nrf2. After nuclear translocation, Nrf2 activates downstream genes involved in various aspects of mitochondrial homeostasis, including mitochondrial biogenesis and dynamics, mitophagy, aerobic respiration, and energy metabolism. In turn, mitochondria reciprocally activate Nrf2 by releasing reactive oxygen species and regulating antioxidant enzymes. Critical Issues: In this review, we first summarize the interactions between Nrf2 and mitochondria in the modulation of oxidative stress and inflammation to ameliorate MetS, then propose that Nrf2 and mitochondria form a mutually regulating circuit critical to maintaining homeostasis during MetS. Future Directions: Targeting the Nrf2-mitochondrial circuit may be a promising strategy to ameliorate MetS, such as obesity, diabetes, and cardiovascular diseases.
Article
Background Vitamin E influences hormonal parameters in women, but the evidence concerning its effectiveness in polycystic ovarian syndrome (PCOS) are constrained. Objective The objective of this systematic review is to perceive the mechanism behind the hormonal effects of vitamin E in PCOS and explore the effectiveness of vitamin E by analyzing the clinical studies. Methods Electronic bibliographic databases, such as PubMed, Scopus, ProQuest, DOAJ, ScienceDirect, and Cochrane Library, were searched for clinical studies scrutinizing the effect of vitamin E supplementation on PCOS. The systematic review protocol is registered in PROSPERO (CRD42021272963) and was performed as per the PRISMA guidelines. Records analyzing the hormonal parameters were included. For individual study, the risk of bias was ascertained with NHLBI protocol for controlled randomized studies. Results There were 1438 records identified, of which seven records were retrieved which met the inclusion criteria. Bibliographic details, number of participants, the dose of vitamin E, duration of the treatment and trial, parameters analyzed, and outcome of each study were extracted comprehensively. In the review, the majority of the studies embraced high-quality studies, which included double-blinded controlled trials, open-label controlled studies, and cohort studies. Conclusion Vitamin E implies having a progesterone-like impression with estrogenic and androgenic properties. Furthermore, the ability of vitamin E as an antioxidant is also advantageous in PCOS women. However, there is a lack of empirical evidence on its mechanism, and the inadequacy of large-scale concerns about different age groups against PCOS is requisite.
Article
Full-text available
Polycystic ovary syndrome (PCOS) is the most common reproductive endocrine disorder affecting women, which can lead to infertility. Infertility, obesity, hirsutism, acne, and irregular menstruation are just a few of the issues that PCOS can be linked to. PCOS has a complicated pathophysiology and a range of clinical symptoms. Chronic low-grade inflammation is one of the features of PCOS. The inflammatory environment involves immune and metabolic disturbances. Numerous organ systems across the body, in addition to the female reproductive system, have been affected by the pathogenic role of immunological dysregulation in PCOS in recent years. Insulin resistance and hyperandrogenism are associated with immune cell dysfunction and cytokine imbalance. More importantly, obesity is also involved in immune dysfunction in PCOS, leading to an inflammatory environment in women with PCOS. Hormone, obesity, and metabolic interactions contribute to the pathogenesis of PCOS. Hormone imbalance may also contribute to the development of autoimmune diseases. The aim of this review is to summarize the pathophysiological role of immune dysregulation in various organ systems of PCOS patients and provide new ideas for systemic treatment of PCOS in the future.
Article
Full-text available
Background: Myeloperoxidase (MPO) is a lysosomal hemoprotein found in the azurophilic granules in neutrophils. Myeloperoxidase plays an important role in oxygen-dependent killing of bacteria, fungi, virus and malignant cells. Diabetes mellitus (DM) is listed among conditions that may lead to secondary MPO deficiency in neutrophils but inconsistent results concerning MPO activity in diabetic patients have been reported in the literature. In this study, we aimed to evaluate the relationship between glycemic control in patients with type 2 DM and MPO activity in neutrophils from a histochemical perspective. Methods: The study included 40 patients with type 2 DM with poor glycemic control, 30 patients with type 2 DM with good glycemic control and 31 healthy controls. Peripheral blood smears were analyzed for each patient included in the study. Myeloperoxidase dye was used for staining. Myeloperoxidase ratios in neutrophil were evaluated for proportions of staining with MPO in 100 neutrophils in each smear. SPSS 16.0 version was used for statistical analyses. Results: Myeloperoxidase ratios in neutrophils were 70 (58.5-80) in type 2 DM patients with poor glycemic control compared to 80 (73.75-90) in those with good glycemic control and 88 (78-92) in healthy controls. The DM group with poor glycemic control was statistically significantly different from the other groups (p < 0.001). Conclusions: Poor glycemic control in diabetic patients results in decreased MPO activity in neutrophils histochemically.
Article
Full-text available
Since the 1990 NIH‐sponsored conference on polycystic ovary syndrome (PCOS), it has become appreciated that the syndrome encompasses a broader spectrum of signs and symptoms of ovarian dysfunction than those defined by the original diagnostic criteria. The 2003 Rotterdam consensus workshop concluded that PCOS is a syndrome of ovarian dysfunction along with the cardinal features hyperandrogenism and polycystic ovary (PCO) morphology. PCOS remains a syndrome and, as such, no single diagnostic criterion (such as hyperandrogenism or PCO) is sufficient for clinical diagnosis. Its clinical manifestations may include: menstrual irregularities, signs of androgen excess, and obesity. Insulin resistance and elevated serum LH levels are also common features in PCOS. PCOS is associated with an increased risk of type 2 diabetes and cardiovascular events.
Article
Full-text available
Context: Polycystic ovary syndrome (PCOS) is a heterogeneous endocrine disorder accompanied by chronic low-grade inflammation, but the molecular mechanism remains unclear. Objective: We investigated the action of WNT5a in the development of chronic inflammation in PCOS and the related molecular signaling pathways. Design and setting: This was a prospective study conducted at the Division of Reproduction Center, Peking University Third Hospital. Patients: A total of 35 PCOS patients and 87 control women who reported to the clinic for the in vitro procedure and the cause of marital infertility was male azoospermia were included. Main outcome measures: Mural granulosa cells (GCs) of 35 PCOS patients and 37 controls were collected during oocyte retrieval and gene expression was analyzed. The human KGN cells and mural GCs from 50 control subjects (six to eight samples were pooled together for each experiment) were cultured in vitro. The regulation of inflammation and oxidative stress was confirmed by quantitative PCR, flow-cytometric assay, and dual-luciferase reporter assay after inflammatory stimuli or WNT5a overexpression. Relevant signaling pathways were identified using specific inhibitors. Results: Our data demonstrate significantly elevated WNT5a expression in the mural GCs of PCOS patients compared with the controls. Lipopolysaccharide stimulation increased WNT5a expression in KGN cells and mural GCs, and BAY-117082 and pyrrolidinedithiocarbamic acid [nuclear factor-κB (NF-κB) inhibitor] treatments suppressed WNT5a mRNA below the control level. WNT5a overexpression also enhanced the expression of inflammation-related genes and increased intracellular reactive oxygen species, whereas both BAY-117082 and LY-294002 (phosphatidylinositol 3-kinase inhibitor) significantly inhibited WNT5a-induced inflammation and oxidative stress. Conclusions: WNT5a acts as a proinflammatory factor in human ovarian GCs. The up-regulated expression of WNT5a in PCOS increases inflammation and oxidative stress predominantly via the phosphatidylinositol 3-kinase/AKT/NF-κB signaling pathway. The proinflammatory cytokines induced might further enhance WNT5a expression via NF-κB-dependent regulation, indicating a novel regulatory system for chronic inflammation in PCOS.
Article
Full-text available
Context: Polycystic ovary syndrome (PCOS) and pregnancy are conditions characterized by an increased low-grade chronic inflammation state. A higher incidence of pregnancy complications has been detected in pregnant PCOS women. Objective: The objective of the study was to test the hypothesis that the low-grade chronic inflammation state typical of PCOS patients persists during gestation and is exacerbated by pregnancy and contributes to the increased risk of obstetric/neonatal complications. Design: This was a prospective controlled clinical study. Setting: The study was conducted at the Academic Department of Obstetrics and Gynecology of the "Pugliese-Ciaccio" Hospital of Catanzaro (Catanzaro, Italy). Patients: One hundred fifty pregnant PCOS women and 150 age- and body mass index-matched healthy pregnant controls participated in the study. Interventions: INTERVENTIONS included serial clinical, biochemical, and ultrasonographic assessments before and throughout pregnancy. Main outcome measures: Serum levels of white blood cell count (WBC), C-reactive protein (CRP), and ferritin were measured. Results: Pregnant women with PCOS had higher WBC, CRP, and ferritin levels at study entry and at all gestational ages than controls. Changes in serum WBC and ferritin levels were significantly higher in PCOS than in controls starting from the 12th week of gestation whereas those in CRP from the 20th week of gestation. By multivariable Cox proportional hazard analysis, in the PCOS group, a significant association with the risk of adverse obstetric/neonatal outcomes was found for WBC [hazard ratio (HR) 1.52, 95% confidence interval (CI) 1.31-1.64; P = .010], CRP (HR 1.19, 95% CI 1.06-1.34; P = .019), and ferritin levels (HR 1.12, 95% CI 1.03-1.26; P = .011). Conclusions: In PCOS patients, the low-grade chronic inflammation persists during gestation and is exacerbated by pregnancy, and it is associated with adverse pregnancy outcomes.
Article
Myeloperoxidase (MPO) is an oxidant-producing enzyme that can also bind to cellular surface proteins. We found that band 3 protein and glycophorins A and B were the key MPO-binding targets of human red blood cells (RBCs). The interaction of MPO with RBC proteins was mostly electrostatic in nature because it was inhibited by desialation, exogenic sialic acid, high ionic strength, and extreme pH. In addition, MPO failed to interfere with the lectin-induced agglutination of RBCs, suggesting a minor role of glycan-recognizing mechanisms in MPO binding. Multiple biophysical properties of RBCs were altered in the presence of native (i.e., not hypochlorous acid-damaged) MPO. These changes included transmembrane potential, availability of intracellular Ca2+, and lipid organization in the plasma membrane. MPO-treated erythrocytes became larger in size, structurally more rigid, and hypersensitive to acidic and osmotic hemolysis. Furthermore, we found a significant correlation between the plasma MPO concentration and RBC rigidity index in type-2 diabetes patients with coronary heart disease. These findings suggest that MPO functions as a mediator of novel regulatory mechanism in microcirculation, indicating the influence of MPO-induced abnormalities on RBC deformability under pathological stress conditions.
Article
Introduction: Metabolic syndrome is comprised of a combination of the following states: increased insulin resistance, dyslipidemia, cardiovascular disease, and increased abdominal obesity. Women with polycystic ovary syndrome (PCOS) have an increased risk of developing metabolic syndrome over the course of their lives. Metabolic syndrome increases risk of major cardiovascular events, morbidity, quality of life, and overall health care costs. Though metabolic syndrome in women with PCOS is an area of great concern, there is no effective individual medical therapeutic to adequately treat this issue. Areas Covered: This article will review key aspects of metabolic syndrome in PCOS. We will discuss classic and novel therapeutics to address metabolic syndrome in women with PCOS. We will conclude with the importance of developing strategic interventions to increase the compliance to lifestyle and dietary modification, in addition to appreciation of the emerging pharmaceutical therapeutics available. Expert Opinion: Innovation in lifestyle modification, including diet, exercise, with and without dedicated stress reduction techniques is the future in treatment of metabolic syndrome in PCOS. Application of novel interventions, such as group medical care, may improve future adherence to lifestyle modification recommendations, in addition to or in combination with pharmaceutical therapeutics.
Article
Atherosclerosis, formerly considered a bland lipid storage disease, actually involves an ongoing inflammatory response. Recent advances in basic science have established a fundamental role for inflammation in mediating all stages of this disease from initiation through progression and, ultimately, the thrombotic complications of atherosclerosis. These new findings provide important links between risk factors and the mechanisms of atherogenesis. Clinical studies have shown that this emerging biology of inflammation in atherosclerosis applies directly to human patients. Elevation in markers of inflammation predicts outcomes of patients with acute coronary syndromes, independently of myocardial damage. In addition, low-grade chronic inflammation, as indicated by levels of the inflammatory marker C-reactive protein, prospectively defines risk of atherosclerotic complications, thus adding to prognostic information provided by traditional risk factors. Moreover, certain treatments that reduce coronary risk also limit inflammation. In the case of lipid lowering with statins, this anti-inflammatory effect does not appear to correlate with reduction in low-density lipoprotein levels. These new insights into inflammation in atherosclerosis not only increase our understanding of this disease, but also have practical clinical applications in risk stratification and targeting of therapy for this scourge of growing worldwide importance.
Article
Oxidative and endoplasmic reticulum (ER) stress are related to type 2 diabetes (T2D), but the influence of glycaemic control on these parameters and its relationship with leukocyte-endothelial interactions is not known. In our study population consisting of 164 diabetic patients, 102 with glycated haemoglobin (HbA1c) <7% and 62 with HbA1c >7%) and 84 non-diabetic controls, we have verified a common anthropometric and metabolic pattern of T2D with dyslipidemia. Inflammatory parameters (hsCRP and TNFα) and E-selectin levels were enhanced in the HbA1c >7% group with respect to controls. O2 consumption and mitochondrial membrane potential were lower in diabetic patients than in controls. ROS production was enhanced in diabetic patients than in controls and positively correlated with HbA1c levels. GRP78 levels were higher in both diabetic groups. However, whereas HbA1c <7% patients displayed higher levels of sXBP1, HbA1c >7% patients exhibited preferentially enhanced levels of CHOP and ATF6. Reduced leukocyte rolling velocity and increased rolling flux and adhesion were observed in diabetic patients. Our findings support the hypothesis of an association between poor glycaemic control in T2D and increased leukocyte ROS production and chronic ER stress that could finally promote leukocyte-endothelial interactions, which in turn poses a risk of vascular complications for these patients.