734
Views
27
CrossRef citations to date
0
Altmetric
Original Article

Maternal and neonatal circulating visfatin concentrations in patients with pre-eclampsia and a small-for-gestational age neonate

, , , , , , , , , , , , , , & show all
Pages 1119-1128 | Received 28 Nov 2009, Accepted 18 Dec 2009, Published online: 01 Feb 2010

Abstract

Objective. Maternal circulating visfatin concentrations are higher in patients with a small-for-gestational-age (SGA) neonate than in those who delivered an appropriate-for-gestational age (AGA) neonate or in those with pre-eclampsia. It has been proposed that enhanced transfer of visfatin from the foetal to maternal circulation may account for the high concentrations of maternal visfatin observed in patients with an SGA neonate. The aims of this study were: (1) to determine whether cord blood visfatin concentrations differ between normal neonates, SGA neonates and newborns of pre-eclamptic mothers; and (2) to assess the relationship between maternal and foetal circulating visfatin concentrations in patients with an SGA neonate and those with pre-eclampsia.

Study design. This cross-sectional study included 88 pregnant women and their neonates, as well as 22 preterm neonates in the following groups: (1) 44 normal pregnant women at term and their AGA neonates; (2) 22 normotensive pregnant women and their SGA neonates; (3) 22 women with pre-eclampsia and their neonates; and (4) 22 preterm neonates delivered following spontaneous preterm labour without funisitis or histologic chorioamnionitis, matched for gestational age with infants of pre-eclamptic mothers. Maternal plasma and cord blood visfatin concentrations were determined by ELISA. Non-parametric statistics were used for analyses.

Results. (1) The median visfatin concentration was lower in umbilical cord blood than in maternal circulation, in normal pregnancy, SGA and pre-eclampsia groups (p < 0.001 for all comparisons); (2) the median cord blood visfatin concentrations did not differ significantly between term AGA or SGA neonates, infants of mothers with pre-eclampsia and their gestational-age-matched preterm AGA neonates; (3) maternal and cord blood visfatin concentrations correlated only in the normal term group (r = 0.48, p = 0.04).

Conclusion. Circulating visfatin concentrations are lower in the foetal than in the maternal circulation and did not significantly differ between the study groups. Thus, it is unlikely that the foetal circulation is the source of the high maternal visfatin concentrations reported in patients with an SGA neonate.

Introduction

Pre-eclampsia and delivery of a small-for-gestational age (SGA) neonates, two of the ‘great obstetrical syndromes’ [Citation1], share several mechanisms of disease including failure of physiologic transformation of the spiral arteries [Citation2,Citation3], an anti-angiogenic state [Citation4–18], endothelial cell dysfunction [Citation19–21], and an increased maternal intravascular inflammatory response [Citation15,Citation22–32]. Despite these similarities, pre-eclampsia and pregnancies complicated by an SGA neonate have different clinical manifestations. While pre-eclampsia is characterised by hypertension, proteinuria and organ damage [Citation33,Citation34], SGA is usually defined as a birthweight below the 10th percentile for gestational age at birth according to the birthweight distribution of a particular population [Citation35]. Hypertension, proteinuria and organ damage are not clinical features of pregnancies with isolated SGA.

Several explanations have been proposed to reconcile this apparent disparity including exposure to infection during pregnancy [Citation36,Citation37], differences in the profile of angiogenic and anti-angiogenic response to intrauterine insults [Citation13,Citation17], altered activity of the coagulation system [Citation38], and changes in the concentrations of placental growth hormone [Citation39] and pro-inflammatory chemokines (CXCL10/IP-10) [Citation15]. Ness and Sibai [Citation19] have proposed that the presence of altered metabolic states (e.g. obesity, insulin resistance, dyslipidemia) predisposes pregnant women to develop pre-eclampsia, while the absence of these metabolic derangements will result in an SGA neonate.

Visfatin, a newly discovered 52 kDa adipokine, has been implicated in the regulation of glucose homeostasis [Citation40], Type-2 diabetes mellitus (Type-2 DM) [Citation41], gestational diabetes mellitus (GDM) [Citation42–46], as well as in foetal growth [Citation47]. Recently, we have reported that patients with an SGA neonate, but not those with pre-eclampsia, had a higher maternal plasma visfatin concentration than those with a normal pregnancy [Citation48] suggesting that perturbation of visfatin homeostasis may be implicated in the phenotypic distinction between pre-eclampsia and SGA. Nevertheless, the source of the higher maternal circulating visfatin concentrations in patients with an SGA neonate has not yet been determined. It has been hypothesised that enhanced transfer of visfatin from the foetal to the maternal circulation can account for the high concentrations of maternal visfatin reported in patients with an SGA neonate. Thus, the aims of this study were: (1) to determine whether cord blood visfatin concentration differ between normal neonates, SGA neonates and newborns of pre-eclamptic mothers; and (2) to assess the relationship between maternal and foetal circulating visfatin concentrations in patients with an SGA neonate and those with pre-eclampsia.

Materials and methods

Study population

A case–control study was conducted by searching our clinical database and bank of biological samples, and included 88 pregnant women and their neonates, as well as 22 preterm neonates, in the following groups: (1) 44 normal pregnant women at term and their appropriate-for-gestational age (AGA) neonates; (2) 22 normotensive pregnant women and their SGA neonates; (3) 22 women with pre-eclampsia and their neonates; and (4) 22 preterm AGA neonates delivered following spontaneous preterm labour without funisitis or histologic chorioamnionitis, matched for gestational age with neonates of the pre-eclampsia group.

Maternal plasma, umbilical cord blood and clinical and demographic data were retrieved from our bank of biological samples and clinical database. Many of these samples have previously been employed to study the biology of inflammation, homeostasis, angiogenesis regulation and adipokines concentrations in normal pregnant women and those with pregnancy complications. Maternal visfatin concentrations for the patients included in the present study have been previously reported [Citation48] and were included in this study in order to provide a complete picture to the reader.

All participating women provided written informed consent prior to enrolment and the collection of blood samples. The collection and use of blood for research purposes was approved by the Institutional Review Boards of the Sotero del Rio Hospital (Santiago, Chile) and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (NIH/DHHS, Bethesda, Maryland, USA).

Definitions

The inclusion criteria for normal pregnancy were: (1) no medical, obstetrical or surgical complications; (2) intact membranes; (3) delivery of a term neonate (>37 weeks) with a birthweight above the 10th percentile; [Citation49] and (4) a normal oral 75-g oral glucose tolerance test between 24 and 28 weeks of gestation based on the World Health Organization (WHO) criteria [Citation50].

Pre-eclampsia was defined as the presence of hypertension (systolic blood pressure ≥140 mmHg and/or diastolic blood pressure ≥90 mmHg on at least two occasions, 4 h to 1 week apart) first occurring after 20 weeks of gestation in a woman with previously normal blood pressure, and proteinuria (≥300 mg in a 24-h urine collection or at least one dipstick measurement ≥1+) [Citation51]. The diagnosis of SGA was based on ultrasonographic estimated foetal weight and confirmed by a birthweight below the 10th percentile for gestational age [Citation49]. The body mass index (BMI) was calculated according to the following formula: weight (kg)/height (m2). Normal weight women were defined as those with a BMI of 18.5–24.9 kg/m2 according to the definition of the WHO [Citation52]. Ponderal Index was calculated according to the following formula: weight (kg)/height (m3).

Sample collection and human visfatin C-terminal immunoassay

Maternal blood samples were collected during clinical visits and umbilical cord blood was obtained from the umbilical vein at the time of delivery. Blood was centrifuged at 1300g for 10 min at 4°C. The plasma obtained was stored at −80°C until analysis.

Comparison between maternal and foetal circulating visfatin concentrations, as well as correlation analysis was conducted only in cases in which the time interval between maternal blood sampling and delivery was <48 h. The 48-h interval was chosen to maintain a meaningful temporal relationship of visfatin concentrations between umbilical cord blood and maternal blood.

Concentrations of visfatin in maternal and foetal plasma were determined using specific and sensitive enzyme immunoassays (Phoenix Pharmaceuticals, Inc. Belmont, CA, USA). An initial assay validation was performed in our laboratory prior to the conduction of this study. A detailed description of the assay has been previously published [Citation46,Citation53–55]. The calculated inter- and intra-assay coefficients of variation for visfatin C-terminal immunoassays in our laboratory were 5.3% and 2.4%, respectively. The sensitivity was calculated to be 0.04 ng/ml.

Statistical analysis

Normality of the data was tested using the Kolmogorov–Smirnov and Shapiro–Wilk tests. Since maternal plasma and umbilical cord blood visfatin concentrations were not normally distributed, Kruskal–Wallis tests with post hoc analysis by Mann–Whitney U-tests were used for comparisons of continuous variables between the different groups. Comparison of proportions was performed by Fisher's test. Wilcoxon Signed ranks exact test was used to compare the visfatin concentrations between mother–neonate pairs. Spearman rank correlation was utilised to assess correlations between umbilical cord blood visfatin concentrations and birthweight, Ponderal Index, gestational age at delivery, maternal plasma visfatin concentration and maternal BMI. A p-value <0.05 was considered statistically significant. Analysis was performed with SPSS, version 14 (SPSS Inc., Chicago, IL, USA).

Results

Demographic and clinical characteristics of the study groups are presented in . The median gestational age at delivery (i.e. gestational age at cord blood sampling) was significantly lower in neonates of mothers with pre-eclampsia than in normal term AGA and SGA newborns. There was no significant difference in the median gestational age at delivery between neonate of mothers with pre-eclampsia and those with preterm labour ().

Table I.  Clinical and demographic characteristics of the study population.

Umbilical cord blood visfatin concentrations in pre-eclampsia, SGA and preterm labour

The median umbilical cord plasma concentrations did not differ significantly between AGA [7.2 ng/ml, interquartile range (IQR): 5.8–8.0], SGA newborns (7.1 ng/ml, IQR: 5.7–9.5) and infants of patients with pre-eclampsia (7.4 ng/ml, IQR: 5.6–8.2) (p = 0.8, Kruskal–Wallis; ).

Figure 1.  Comparison between umbilical cord visfatin concentration in normal neonates, SGA newborns, infants of patients with pre-eclampsia and preterm neonates. The median umbilical cord plasma concentrations of normal neonates (7.2 ng/ml, IQR 5.8–8.0), SGA newborns (7.1 ng/ml, IQR: 5.7–9.5) and infants of patients with pre-eclampsia (7.4 ng/ml, IQR: 5.6–8.2) did not differ significantly (p = 0.8, Kruskal–Wallis). Similarly, the median umbilical cord plasma visfatin concentrations did not differ significantly between neonates of patients with pre-eclampsia and those delivered preterm without pre-eclampsia matched for gestational age (7.4 ng/ml, IQR: 5.6–8.2 vs. 7.1 ng/ml, IQR: 6.4–7.7).

Figure 1.  Comparison between umbilical cord visfatin concentration in normal neonates, SGA newborns, infants of patients with pre-eclampsia and preterm neonates. The median umbilical cord plasma concentrations of normal neonates (7.2 ng/ml, IQR 5.8–8.0), SGA newborns (7.1 ng/ml, IQR: 5.7–9.5) and infants of patients with pre-eclampsia (7.4 ng/ml, IQR: 5.6–8.2) did not differ significantly (p = 0.8, Kruskal–Wallis). Similarly, the median umbilical cord plasma visfatin concentrations did not differ significantly between neonates of patients with pre-eclampsia and those delivered preterm without pre-eclampsia matched for gestational age (7.4 ng/ml, IQR: 5.6–8.2 vs. 7.1 ng/ml, IQR: 6.4–7.7).

Since gestational age at umbilical cord sampling was significantly lower in neonate of mothers with pre-eclampsia, circulating visfatin concentrations were determined in 22 preterm AGA neonates delivered following spontaneous preterm labour without funisitis or histologic chorioamnionitis, matched for gestational age with the infants of patients with pre-eclampsia. The median umbilical cord visfatin plasma concentrations did not differ significantly between neonates of patients with pre-eclampsia and those born preterm without pre-eclampsia (7.4 ng/ml, IQR: 5.6–8.2 vs. 7.1 ng/ml, IQR: 6.4–7.7, p = 0.9, ).

In contrast, the median maternal plasma visfatin concentrations differed significantly among groups (p = 0.04, Kruskal–Wallis). The median maternal plasma concentration of visfatin was significantly higher in patients with an SGA neonate than in those with either normal pregnancy (18 ng/ml, IQR: 16.2–23.3 vs. 16.1 ng/ml, IQR: 11.2–20.3; p = 0.02) or those with pre-eclampsia (15.9 ng/ml, IQR: 13–22; p = 0.04). The median maternal plasma visfatin concentration did not differ significantly between patients with pre-eclampsia and those with a normal pregnancy (p = 0.7).

Comparison between maternal plasma and umbilical cord blood visfatin concentrations

Comparison between maternal and foetal circulating visfatin concentrations was conducted only in cases in which the time interval between maternal and umbilical cord blood sampling was <48 h. Using this cut-off, maternal-neonatal paired samples were available for 18 patients in the normal pregnancy group, 20 in the SGA group and 19 in the pre-eclampsia group. The median plasma visfatin concentration was significantly higher in the maternal than in the umbilical blood in the normal pregnancy group (18.7 ng/ml, IQR: 12.7–21.3 vs. 7.3 ng/ml, IQR: 6.1–7.9, p < 0.001; ), SGA group (18 ng/ml, IQR: 16.4–23 vs. 6.7 ng/ml, IQR: 5.7–8.5, p < 0.001; ), and in the pre-eclampsia group (16.5 ng/ml, IQR: 13.1–22.6 vs. 7.6 ng/ml, IQR: 6.4–8.2, p < 0.001; ).

Figure 2.  Comparison between umbilical cord blood and maternal plasma visfatin concentrations in normal gestations (A), pregnancies complicated by an SGA neonate (B) or pre-eclampsia (C). The median maternal plasma visfatin concentration was higher than in the umbilical blood in the normal pregnancy group (18.7 ng/ml, IQR: 12.7–21.3 vs. 7.3 ng/ml, IQR: 6.1–7.9, p < 0.001), SGA group (18.0 ng/ml, IQR: 16.4–23.0 vs. 6.7 ng/ml, IQR: 5.7–8.5, p < 0.001), and in the pre-eclampsia group (16.5 ng/ml, IQR: 13.1–22.6 vs. 7.6 ng/ml, IQR: 6.4–8.2, p < 0.001).

Figure 2.  Comparison between umbilical cord blood and maternal plasma visfatin concentrations in normal gestations (A), pregnancies complicated by an SGA neonate (B) or pre-eclampsia (C). The median maternal plasma visfatin concentration was higher than in the umbilical blood in the normal pregnancy group (18.7 ng/ml, IQR: 12.7–21.3 vs. 7.3 ng/ml, IQR: 6.1–7.9, p < 0.001), SGA group (18.0 ng/ml, IQR: 16.4–23.0 vs. 6.7 ng/ml, IQR: 5.7–8.5, p < 0.001), and in the pre-eclampsia group (16.5 ng/ml, IQR: 13.1–22.6 vs. 7.6 ng/ml, IQR: 6.4–8.2, p < 0.001).

The median umbilical cord plasma visfatin concentration did not differ significantly between male and female neonates in a pooled analysis (p = 0.1) or within normal neonates (p = 0.5), SGA newborns (p = 0.8), neonates of mothers with pre-eclampsia (p = 0.1) or preterm neonates (p = 0.9, data not shown).

Maternal and cord blood plasma visfatin concentrations correlated only in the normal term group (r = 0.48, p = 0.04; ), but not in the SGA (r = 0.2, p = 0.3) or pre-eclampsia group (r = 0.5, p = 0.1). No significant correlation was found between cord blood visfatin concentrations and birthweight, Ponderal Index or gestational age at delivery.

Figure 3.  Correlation between umbilical cord blood and maternal plasma visfatin concentrations in the normal gestation. Maternal and cord blood plasma visfatin concentrations correlated only in the normal term group (r = 0.48, p = 0.04).

Figure 3.  Correlation between umbilical cord blood and maternal plasma visfatin concentrations in the normal gestation. Maternal and cord blood plasma visfatin concentrations correlated only in the normal term group (r = 0.48, p = 0.04).

Discussion

Principal findings of the study

(1) The median plasma visfatin concentration was significantly lower in cord blood than in the maternal circulation in normal pregnancy, SGA and pre-eclampsia; (2) the median cord blood visfatin concentrations did not differ significantly between term AGA, SGA and neonates of patients with pre-eclampsia; (3) maternal and cord blood visfatin concentrations correlated only in the normal term group.

The physiological role of visfatin

Visfatin, also known as Pre-B cell colony-enhancing factor (PBEF), was originally identified as a growth factor for early B cell [Citation56]. Subsequently, it was recognised as a novel adipokine which is preferentially produced by the visceral fat depot [Citation40]. Visfatin/PBEF has been implicated in the regulation of glucose homeostasis. Indeed, in vitro, adipocytes secrete visfatin in response to treatment with glucose [Citation57] and this protein can exert insulin-mimicking effects [Citation58]. In vivo, visfatin-deficient mice have impaired glucose tolerance [Citation59] and a polymorphism in the human visfatin gene promoter is associated with a susceptibility to type-2 DM [Citation60]. Moreover, high circulating concentrations of this adipokine characterise patients with insulin resistance [Citation44–46,Citation61,Citation62].

In addition to its metabolic effects, visfatin has pro-inflammatory properties. In vitro, visfatin synergises with interleukin (IL)- 7 and stem cell factors to promote the growth of B-cell precursors and treatment of human monocytes with visfatin results in an increased secretion of IL-6, tumour necrosis factor-α and IL-1β in a dose-dependent manner [Citation63]. In addition, patients with chronic inflammatory disorders such as inflammatory bowel disease [Citation63] and rheumatoid arthritis [Citation64] have a higher circulating visfatin concentration than normal subjects.

Visfatin in normal gestation and in complications of pregnancy

The rationale to study visfatin concentrations in human pregnancy rests on the association between alterations in adipokines concentrations and adaptations to gestation [Citation55,Citation65–73], as well as with complications of pregnancy such as pre-eclampsia [Citation48,Citation74–77], preterm labour [Citation53,Citation78], intra-amniotic infection/inflammation [Citation54,Citation79–81], delivery of an SGA neonate [Citation82,Citation83], macrosomia [Citation84,Citation85], GDM [Citation84] and pyelonephritis [Citation86]. Moreover, visfatin is expressed in the placenta, foetal membranes [Citation87–94] and the myometrium [Citation95]. Normal pregnancy is associated with high maternal circulating visfatin concentrations [Citation55,Citation96–100]. In addition, GDM is characterised by alterations in maternal concentrations of this adipokine [Citation42,Citation44–46,Citation101]. Recently, we have reported that intra-amniotic infection/inflammation is associated with higher amniotic fluid concentrations of visfatin than in the absence of infection [Citation54], and that preterm labour is characterised by high maternal circulating concentrations of this adipokine [Citation53].

Visfatin concentrations are lower in the foetal than in the maternal circulation

Only several studies have addressed umbilical cord blood visfatin concentrations [Citation102–106], and a comparison between maternal and neonatal concentrations of this adipokine was included in only two reports [Citation104,Citation105]. The results of the present study indicate that circulating visfatin concentrations are lower in the foetal than in the maternal circulation. This novel finding was demonstrated in all study groups, including normal neonates, SGA newborns and neonates of mothers with pre-eclampsia. The results reported herein are in contrast to the report by Malamitsi-Puchner et al. [Citation105] in which there were no significant differences between maternal and neonatal circulating visfatin concentrations. Ethnic origin, clinical definitions and gestational age at enrollment varied between the studies and may account for this discrepancy.

It is not clear why visfatin concentrations are lower in the foetal than in the maternal circulation. Increased production by the larger maternal fat depot compared to the newborn is a plausible explanation. In addition, visfatin, which is expressed in a considerable amount in the term human placenta [Citation97], may be preferentially released into the maternal systemic circulation. Disparity in placental secretion of adipokines into the maternal and foetal circulation has been demonstrated for other adipokines such as leptin [Citation107].

Our findings indicate that maternal and cord blood plasma visfatin concentrations were correlated only in the normal term group. This finding is in agreement with the reports by Ibánez et al. [Citation102] and Malamitsi-Puchner et al. [Citation104,Citation105]. Our findings are also in agreement with López-Bermejo et al. [Citation103] who found no association between umbilical cord blood visfatin concentrations and anthropometric indices of the newborn. The present study extends the aforementioned reports by demonstrating these finding not only to term or AGA neonates but also to those of mother with pre-eclampsia and premature neonates.

The foetal circulation is not the source for the elevated visfatin concentrations of mother with a small-for-gestational-age neonate

Two studies, conducted by Fasshauer et al. [Citation108] and Malamitsi-Puchner et al. [Citation104] have reported higher maternal circulating visfatin concentrations in patients with an SGA neonate than in those who delivered an AGA neonate. In accordance with these findings, we have recently reported that patients with an SGA neonate, but not those with pre-eclampsia, had a higher maternal plasma visfatin concentration than those with a normal pregnancy suggesting differential involvement of visfatin in SGA and pre-eclampsia [Citation48]. However, the mechanism by which an SGA neonate affects maternal visfatin circulation is not clear. It has been proposed that enhanced transfer of visfatin from the foetal to the maternal circulation can account for the high concentrations of maternal visfatin reported in patients with an SGA neonate [Citation48]. The results of the present study suggest that the foetal circulation is not the source for the increased maternal visfatin concentrations in patients with an SGA neonate as no significant differences were documented between the cord blood concentrations of the different study groups.

The finding reported herein concerning the lack of difference in umbilical cord blood visfatin concentrations between SGA neonates and those with a normal pregnancy is in contrast to the report by Malamitsi-Puchner et al. [Citation104] in which umbilical cord blood visfatin concentrations were higher in SGA than in AGA neonates matched for gestational age. Differences in the definition of an SGA neonate and ethnic origin may account for this discrepancy. Specifically, in the study by Malamitsi-Puchner et al. [Citation104] the SGA group included patients with pre-eclampsia, gestational hypertension, iron-deficiency anaemia, gestational diabetes, hypothyroidism, as well as cardiac arrhythmias, whereas in the present study mothers with these conditions were excluded from the SGA group.

In conclusion, circulating foetal visfatin concentrations are lower than maternal plasma concentrations. Despite the association between the presence of an SGA neonate and high maternal circulating visfatin concentrations, comparable visfatin concentrations were detected in the umbilical cord blood of normal neonates, SGA newborns, infants of patients with pre-eclampsia and preterm neonates. Collectively, these observations suggest that foetal–maternal transport of visfatin is not likely to account for the high maternal visfatin concentrations in patients with an SGA neonate.

Acknowledgement

This research was supported in part by the Perinatology Research Branch, Division of Intramural Research Program of the Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, DHHS.

Reference

  • Di Renzo GC. The great obstetrical syndromes. J Matern Fetal Neonatal Med 2009;22:633–635.
  • Gerretsen G, Huisjes HJ, Elema JD. Morphological changes of the spiral arteries in the placental bed in relation to pre-eclampsia and fetal growth retardation. Br J Obstet Gynaecol 1981;88:876–881.
  • Khong TY, De Wolf F, Robertson WB, Brosens I. Inadequate maternal vascular response to placentation in pregnancies complicated by pre-eclampsia and by small-for-gestational age infants. Br J Obstet Gynaecol 1986;93:1049–1059.
  • Chaiworapongsa T, Romero R, Espinoza J, Bujold E, Mee KY, Goncalves LF, Gomez R, Edwin S. Evidence supporting a role for blockade of the vascular endothelial growth factor system in the pathophysiology of preeclampsia. Young Investigator Award. Am J Obstet Gynecol 2004;190:1541–1547.
  • Chaiworapongsa T, Romero R, Kim YM, Kim GJ, Kim MR, Espinoza J, Bujold E, Goncalves L, Gomez R, Edwin S, et al Plasma soluble vascular endothelial growth factor receptor-1 concentration is elevated prior to the clinical diagnosis of pre-eclampsia. J Matern Fetal Neonatal Med 2005;17:3–18.
  • Espinoza J, Romero R, Nien JK, Kusanovic JP, Richani K, Gomez R, Kim CJ, Mittal P, Gotsh F, Erez O, et al A role of the anti-angiogenic factor sVEGFR-1 in the ‘mirror syndrome’ (Ballantyne's syndrome). J Matern Fetal Neonatal Med 2006;19:607–613.
  • Levine RJ, Maynard SE, Qian C, Lim KH, England LJ, Yu KF, Schisterman EF, Thadhani R, Sachs BP, Epstein FH, et al Circulating angiogenic factors and the risk of preeclampsia. N Engl J Med 2004;350:672–683.
  • Levine RJ, Thadhani R, Qian C, Lam C, Lim KH, Yu KF, Blink AL, Sachs BP, Epstein FH, Sibai BM, et al Urinary placental growth factor and risk of preeclampsia. JAMA 2005;293:77–85.
  • Levine RJ, Lam C, Qian C, Yu KF, Maynard SE, Sachs BP, Sibai BM, Epstein FH, Romero R, Thadhani R, et al Soluble endoglin and other circulating antiangiogenic factors in preeclampsia. N Engl J Med 2006;355:992–1005.
  • Maynard SE, Min JY, Merchan J, Lim KH, Li J, Mondal S, Libermann TA, Morgan JP, Sellke FW, Stillman IE, et al Excess placental soluble fms-like tyrosine kinase 1 (sFlt1) may contribute to endothelial dysfunction, hypertension, and proteinuria in preeclampsia. J Clin Invest 2003;111:649–658.
  • Bujold E, Romero R, Chaiworapongsa T, Kim YM, Kim GJ, Kim MR, Espinoza J, Goncalves LF, Edwin S, Mazor M. Evidence supporting that the excess of the sVEGFR-1 concentration in maternal plasma in preeclampsia has a uterine origin. J Matern Fetal Neonatal Med 2005;18:9–16.
  • Chaiworapongsa T, Romero R, Gotsch F, Espinoza J, Nien JK, Goncalves L, Edwin S, Kim YM, Erez O, Kusanovic JP, et al Low maternal concentrations of soluble vascular endothelial growth factor receptor-2 in preeclampsia and small for gestational age. J Matern Fetal Neonatal Med 2008;21:41–52.
  • Romero R, Nien JK, Espinoza J, Todem D, Fu W, Chung H, Kusanovic JP, Gotsch F, Erez O, Mazaki-Tovi S, et al A longitudinal study of angiogenic (placental growth factor) and anti-angiogenic (soluble endoglin and soluble vascular endothelial growth factor receptor-1) factors in normal pregnancy and patients destined to develop preeclampsia and deliver a small for gestational age neonate. J Matern Fetal Neonatal Med 2008;21:9–23.
  • Lindheimer MD, Romero R. Emerging roles of antiangiogenic and angiogenic proteins in pathogenesis and prediction of preeclampsia. Hypertension 2007;50:35–36.
  • Gotsch F, Romero R, Friel L, Kusanovic JP, Espinoza J, Erez O, Than NG, Mittal P, Edwin S, Yoon BH, et al CXCL10/IP-10: a missing link between inflammation and anti-angiogenesis in preeclampsia?J Matern Fetal Neonatal Med 2007;20:777–792.
  • Venkatesha S, Toporsian M, Lam C, Hanai J, Mammoto T, Kim YM, Bdolah Y, Lim KH, Yuan HT, Libermann TA, et al Soluble endoglin contributes to the pathogenesis of preeclampsia. Nat Med 2006;12:642–649.
  • Chaiworapongsa T, Espinoza J, Gotsch F, Kim YM, Kim GJ, Goncalves LF, Edwin S, Kusanovic JP, Erez O, Than NG, et al The maternal plasma soluble vascular endothelial growth factor receptor-1 concentration is elevated in SGA and the magnitude of the increase relates to Doppler abnormalities in the maternal and fetal circulation. J Matern Fetal Neonatal Med 2008;21:25–40.
  • Solomon CG, Seely EW. Preeclampsia – searching for the cause. N Engl J Med 2004;350:641–642.
  • Ness RB, Sibai BM. Shared and disparate components of the pathophysiologies of fetal growth restriction and preeclampsia. Am J Obstet Gynecol 2006;195:40–49.
  • Schiff E, Ben-Baruch G, Peleg E, Rosenthal T, Alcalay M, Devir M, Mashiach S. Immunoreactive circulating endothelin-1 in normal and hypertensive pregnancies. Am J Obstet Gynecol 1992;166:624–628.
  • Higgins JR, Papayianni A, Brady HR, Darling MR, Walshe JJ. Circulating vascular cell adhesion molecule-1 in pre-eclampsia, gestational hypertension, and normal pregnancy: evidence of selective dysregulation of vascular cell adhesion molecule-1 homeostasis in pre-eclampsia. Am J Obstet Gynecol 1998;179:464–469.
  • Girardi G, Yarilin D, Thurman JM, Holers VM, Salmon JE. Complement activation induces dysregulation of angiogenic factors and causes fetal rejection and growth restriction. J Exp Med 2006;203:2165–2175.
  • Schiff E, Friedman SA, Baumann P, Sibai BM, Romero R. Tumor necrosis factor-alpha in pregnancies associated with preeclampsia or small-for-gestational-age newborns. Am J Obstet Gynecol 1994;170:1224–1229.
  • Kusanovic JP, Romero R, Hassan SS, Gotsch F, Edwin S, Chaiworapongsa T, Erez O, Mittal P, Mazaki-Tovi S, Soto E, et al Maternal serum soluble CD30 is increased in normal pregnancy, but decreased in preeclampsia and small for gestational age pregnancies. J Matern Fetal Neonatal Med 2007;20:867–878.
  • Than NG, Erez O, Wildman DE, Tarca AL, Edwin SS, Abbas A, Hotra J, Kusanovic JP, Gotsch F, Hassan SS, et al Severe preeclampsia is characterized by increased placental expression of galectin-1. J Matern Fetal Neonatal Med 2008;21:429–442.
  • Gervasi MT, Chaiworapongsa T, Pacora P, Naccasha N, Yoon BH, Maymon E, Romero R. Phenotypic and metabolic characteristics of monocytes and granulocytes in preeclampsia. Am J Obstet Gynecol 2001;185:792–797.
  • Redman CW, Sacks GP, Sargent IL. Preeclampsia: an excessive maternal inflammatory response to pregnancy. Am J Obstet Gynecol 1999;180:499–506.
  • Sacks GP, Studena K, Sargent K, Redman CW. Normal pregnancy and preeclampsia both produce inflammatory changes in peripheral blood leukocytes akin to those of sepsis. Am J Obstet Gynecol 1998;179:80–86.
  • Chaiworapongsa T, Gervasi MT, Refuerzo J, Espinoza J, Yoshimatsu J, Berman S, Romero R. Maternal lymphocyte subpopulations (CD45RA+ and CD45RO+) in preeclampsia. Am J Obstet Gynecol 2002;187:889–893.
  • Than NG, Romero R, Erez O, Kusanovi JP, Tarca AL, Edwin SS, Kim JS, Hassan SS, Espinoza J, Mittal P, et al A role for mannose-binding lectin, a component of the innate immune system in pre-eclampsia. Am J Reprod Immunol 2008;60:333–345.
  • Bachmayer N, Sohlberg E, Sundstrom Y, Hamad RR, Berg L, Bremme K, Sverremark-Ekstrom E. Women with pre-eclampsia have an altered NKG2A and NKG2C receptor expression on peripheral blood natural killer cells. Am J Reprod Immunol 2009;62:147–157.
  • Lok CA, Jebbink J, Nieuwland R, Faas MM, Boer K, Sturk A, Van Der Post JA. Leukocyte activation and circulating leukocyte-derived microparticles in preeclampsia. Am J Reprod Immunol 2009;61:346–359.
  • Sibai B, Dekker G, Kupferminc M. Pre-eclampsia. Lancet 2005;365:785–799.
  • Solomon CG, Seely EW. Hypertension in pregnancy. Endocrinol Metab Clin North Am 2006;35:157–171, vii.
  • Seeds JW. Impaired fetal growth: definition and clinical diagnosis. Obstet Gynecol 1984;64:303–310.
  • Villar J, Carroli G, Wojdyla D, Abalos E, Giordano D, Ba'aqeel H, Farnot U, Bergsjo P, Bakketeig L, Lumbiganon P, et al Preeclampsia, gestational hypertension and intrauterine growth restriction, related or independent conditions?Am J Obstet Gynecol 2006;194:921–931.
  • von DP, Magee LA. Could an infectious trigger explain the differential maternal response to the shared placental pathology of preeclampsia and normotensive intrauterine growth restriction?Acta Obstet Gynecol Scand 2002;81:642–648.
  • Erez O, Romero R, Hoppensteadt D, Than NG, Fareed J, Mazaki-Tovi S, Espinoza J, Chaiworapongsa T, Kim SS, Yoon BH, et al Tissue factor and its natural inhibitor in pre-eclampsia and SGA. J Matern Fetal Neonatal Med 2008;21:855–869.
  • Mittal P, Espinoza J, Hassan S, Kusanovic JP, Edwin SS, Nien JK, Gotsch F, Than NG, Erez O, Mazaki-Tovi S, et al Placental growth hormone is increased in the maternal and fetal serum of patients with preeclampsia. J Matern Fetal Neonatal Med 2007;20:651–659.
  • Sethi JK, Vidal-Puig A. Visfatin: the missing link between intra-abdominal obesity and diabetes?Trends Mol Med 2005;11:344–347.
  • Chen MP, Chung FM, Chang DM, Tsai JC, Huang HF, Shin SJ, Lee YJ. Elevated plasma level of visfatin/pre-B cell colony-enhancing factor in patients with type 2 diabetes mellitus. J Clin Endocrinol Metab 2006;91:295–299.
  • Chan TF, Chen YL, Lee CH, Chou FH, Wu LC, Jong SB, Tsai EM. Decreased plasma visfatin concentrations in women with gestational diabetes mellitus. J Soc Gynecol Investig 2006;13:364–367.
  • Haider DG, Handisurya A, Storka A, Vojtassakova E, Luger A, Pacini G, Tura A, Wolzt M, Kautzky-Willer A. Visfatin response to glucose is reduced in women with gestational diabetes mellitus. Diabetes Care 2007;30:1889–1891.
  • Krzyzanowska K, Krugluger W, Mittermayer F, Rahman R, Haider D, Shnawa N, Schernthaner G. Increased visfatin concentrations in women with gestational diabetes mellitus. Clin Sci (Lond) 2006;110:605–609.
  • Lewandowski KC, Stojanovic N, Press M, Tuck SM, Szosland K, Bienkiewicz M, Vatish M, Lewinski A, Prelevic GM, Randeva HS. Elevated serum levels of visfatin in gestational diabetes: a comparative study across various degrees of glucose tolerance. Diabetologia 2007;50:1033–1037.
  • Mazaki-Tovi S, Romero R, Kusanovic JP, Vaisbuch E, Erez O, Than NG, Chaiworapongsa T, Nhan-Chang CL, Pacora P, Gotsch F, et al Visfatin in human pregnancy: maternal gestational diabetes vis-a-vis neonatal birthweight. J Perinat Med 2009;37:218–231.
  • Briana DD, Malamitsi-Puchner A. Intrauterine growth restriction and adult disease: the role of adipocytokines. Eur J Endocrinol 2009;160:337–347.
  • Mazaki-Tovi S, Romero R, Kim SK, Vaisbuch E, Kusanovi JP, Erez O, Chaiwaropongsa T, Gotsch F, Mittal P, Nhan-Chang C, et al Could alterations in maternal plasma visfatin concentration participate in the phenotype definition of preeclampsia and SGA? J Matern Fetal Neonatal Med 2009; PMID:19900033.
  • Gonzalez RP, Gomez RM, Castro RS, Nien JK, Merino PO, Etchegaray AB, Carstens MR, Medina LH, Viviani PG, Rojas IT. A national birth weight distribution curve according to gestational age in Chile from 1993 to 2000. Rev Med Chil 2004;132:1155–1165.
  • WHO Study Group. Prevention of diabetes mellitus. Report of a WHO Study Group. World Health Organ Tech Rep Ser 1994;844:1–100.
  • ACOG practice bulletin. Diagnosis and management of preeclampsia and eclampsia. Number 33, January 2002. Obstet Gynecol 2002;99:159–167.
  • WHO. Diet, nutrition and the prevention of chronic diseases. World Health Organ Tech Rep Ser 2003;916:i–149, backcover.
  • Mazaki-Tovi S, Romero R, Vaisbuch E, Erez O, Chaiwaropongsa T, Mittal P, Kim SK, Pacora P, Gotsch F, Dong Z, et al Maternal plasma visfatin in preterm labor. J Matern Fetal Neonatal Med 2009;22:693–704.
  • Mazaki-Tovi S, Romero R, Kusanovic JP, Erez O, Gotsch F, Mittal P, Than NG, Nhan-Chang CL, Hamill N, Vaisbuch E, et al Visfatin/pre-B cell colony-enhancing factor in amniotic fluid in normal pregnancy, spontaneous labor at term, preterm labor and prelabor rupture of membranes: an association with subclinical intrauterine infection in preterm parturition. J Perinat Med 2008;36:485–496.
  • Mazaki-Tovi S, Romero R, Kusanovic JP, Vaisbuch E, Erez O, Than NG, Chaiworapongsa T, Nhan-Chang CL, Pacora P, Gotsch F, et al Maternal visfatin concentration in normal pregnancy. J Perinat Med 2009;37:206–217.
  • Samal B, Sun Y, Stearns G, Xie C, Suggs S, McNiece I. Cloning and characterization of the cDNA encoding a novel human pre-B-cell colony-enhancing factor. Mol Cell Biol 1994;14:1431–1437.
  • Haider DG, Schaller G, Kapiotis S, Maier C, Luger A, Wolzt M. The release of the adipocytokine visfatin is regulated by glucose and insulin. Diabetologia 2006;49:1909–1914.
  • Xie H, Tang SY, Luo XH, Huang J, Cui RR, Yuan LQ, Zhou HD, Wu XP, Liao EY. Insulin-like effects of visfatin on human osteoblasts. Calcif Tissue Int 2007;80:201–210.
  • Revollo JR, Korner A, Mills KF, Satoh A, Wang T, Garten A, Dasgupta B, Sasaki Y, Wolberger C, Townsend RR, et al Nampt/PBEF/Visfatin regulates insulin secretion in beta cells as a systemic NAD biosynthetic enzyme. Cell Metab 2007;6:363–375.
  • Zhang YY, Gottardo L, Thompson R, Powers C, Nolan D, Duffy J, Marescotti MC, Avogaro A, Doria A. A visfatin promoter polymorphism is associated with low-grade inflammation and type 2 diabetes. Obesity 2006;14:2119–2126.
  • Lopez-Bermejo A, Chico-Julia B, Fernandez-Balsells M, Recasens M, Esteve E, Casamitjana R, Ricart W, Fernandez-Real JM. Serum visfatin increases with progressive beta-cell deterioration. Diabetes 2006;55:2871–2875.
  • Sandeep S, Velmurugan K, Deepa R, Mohan V. Serum visfatin in relation to visceral fat, obesity, and type 2 diabetes mellitus in Asian Indians. Metabolism 2007;56:565–570.
  • Moschen AR, Kaser A, Enrich B, Mosheimer B, Theurl M, Niederegger H, Tilg H. Visfatin, an adipocytokine with proinflammatory and immunomodulating properties. J Immunol 2007;178:1748–1758.
  • Otero M, Lago R, Gomez R, Lago F, Dieguez C, Gomez-Reino JJ, Gualillo O. Changes in plasma levels of fat-derived hormones adiponectin, leptin, resistin and visfatin in patients with rheumatoid arthritis. Ann Rheum Dis 2006;65:1198–1201.
  • Mazaki-Tovi S, Kanety H, Sivan E. Adiponectin and human pregnancy. Curr Diab Rep 2005;5:278–281.
  • Mazaki-Tovi S, Kanety H, Pariente C, Hemi R, Wiser A, Schiff E, Sivan E. Maternal serum adiponectin levels during human pregnancy. J Perinatol 2007;27:77–81.
  • Sivan E, Mazaki-Tovi S, Pariente C, Efraty Y, Schiff E, Hemi R, Kanety H. Adiponectin in human cord blood: relation to fetal birth weight and gender. J Clin Endocrinol Metab 2003;88:5656–5660.
  • Mazaki-Tovi S, Kanety H, Pariente C, Hemi R, Efraty Y, Schiff E, Shoham A, Sivan E. Determining the source of fetal adiponectin. J Reprod Med 2007;52:774–778.
  • Mazaki-Tovi S, Romero R, Kusanovic JP, Erez O, Vaisbuch E, Gotsch F, Mittal P, Than GN, Nhan-Chang C, Chaiworapongsa T, et al Adiponectin multimers in maternal plasma. J Matern Fetal Neonatal Med 2008;21:796–815.
  • Nien JK, Mazaki-Tovi S, Romero R, Erez O, Kusanovic JP, Gotsch F, Pineles BL, Gomez R, Edwin S, Mazor M, et al Plasma adiponectin concentrations in non-pregnant, normal and overweight pregnant women. J Perinat Med 2007;35:522–531.
  • Nien JK, Mazaki-Tovi S, Romero R, Kusanovic JP, Erez O, Gotsch F, Pineles BL, Friel LA, Espinoza J, Goncalves L, et al Resistin: a hormone which induces insulin resistance is increased in normal pregnancy. J Perinat Med 2007;35:513–521.
  • Briana DD, Malamitsi-Puchner A. Adipocytokines in normal and complicated pregnancies. Reprod Sci 2009;16:921–937.
  • Catalano PM, Hoegh M, Minium J, Huston-Presley L, Bernard S, Kalhan S, Hauguel-De MS. Adiponectin in human pregnancy: implications for regulation of glucose and lipid metabolism. Diabetologia 2006;49:1677–1685.
  • Mazaki-Tovi S, Romero R, Vaisbuch E, Kusanovic JP, Erez O, Gotsch F, Chaiworapongsa T, Than NG, Kim SK, Nhan-Chang CL, et al Maternal serum adiponectin multimers in preeclampsia. J Perinat Med 2009;37:349–363.
  • Nien JK, Mazaki-Tovi S, Romero R, Erez O, Kusanovic JP, Gotsch F, Pineles BL, Gomez R, Edwin S, Mazor M, et al Adiponectin in severe preeclampsia. J Perinat Med 2007;35:503–512.
  • Lewis DF, Canzoneri BJ, Wang Y. Maternal circulating TNF-alpha levels are highly correlated with IL-10 levels, but not IL-6 and IL-8 levels, in women with pre-eclampsia. Am J Reprod Immunol 2009;65:269–274.
  • Vaisbuch E, Romero R, Mazaki-Tovi S, Erez O, Kim SK, Chaiworapongsa T, Gotsch F, Than NG, Dong Z, Pacora P, et al Retinol binding protein 4 – a novel association with early-onset preeclampsia. J Perinat Med 2009; DOI: 10.1515/JPM.2009.140.
  • Mazaki-Tovi S, Romero R, Vaisbuch E, Erez O, Mittal P, Chaiwaropongsa T, Kim SK, Pacora P, Yeo L, Gotsch F, et al Dysregulation of maternal serum adiponectin in preterm labor. J Matern Fetal Neonatal Med 2009;22:887–904.
  • Kusanovic JP, Romero R, Mazaki-Tovi S, Chaiworapongsa T, Mittal P, Gotsch F, Erez O, Vaisbuch E, Edwin SS, Than NG, et al Resistin in amniotic fluid and its association with intra-amniotic infection and inflammation. J Matern Fetal Neonatal Med 2008;21:902–916.
  • Vaisbuch E, Mazaki-Tovi S, Kusanovic JP, Erez O, Than GN, Kim SK, Dong M, Gotsch F, Mittal P, Chaiworapongsa T, et al Retinol binding protein 4: an adipokine associated with intra-amniotic infection/inflammation. J Matern Fetal Neonatal Med 2009; PMID:19900011.
  • Mazaki-Tovi S, Romero R, Vaisbuch E, Kusanovic JP, Erez O, Mittal P, Gotsch F, Chaiworapongsa T, Than NG, Kim SK, et al Adiponectin in amniotic fluid in normal pregnancy, spontaneous labor at term, and preterm labor: a novel association with subclinical intrauterine infection/inflammation. J Matern Fetal Neonatal Med 2009; DOI:10. 1080/14767050903026481.
  • Mazaki-Tovi S, Romero R, Vaisbuch E, Erez O, Mittal P, Chaiwaropongsa T, Kim SK, Pacora P, Yeo L, Gotsch F, et al Maternal serum adiponectin multimers in patients with a small-for-gestational-age newborn. J Perinat Med 2009;37:623–635.
  • Mazaki-Tovi S, Kanety H, Pariente C, Hemi R, Yinon Y, Wiser A, Schiff E, Sivan E. Adiponectin and leptin concentrations in dichorionic twins with discordant and concordant growth. J Clin Endocrinol Metab 2009;94:892–898.
  • Mazaki-Tovi S, Romero R, Vaisbuch E, Erez O, Mittal P, Chaiwaropongsa T, Kim SK, Pacora P, Yeo L, Gotsch F, et al Maternal serum adiponectin multimers in gestational diabetes. J Perinat Med 2009;37:637–650.
  • Mazaki-Tovi S, Kanety H, Pariente C, Hemi R, Schiff E, Sivan E. Cord blood adiponectin in large-for-gestational age newborns. Am J Obstet Gynecol 2005;193:1238–1242.
  • Mazaki-Tovi S, Romero R, Vaisbuch E, Chaiworapongsa T, Erez O, Mittal P, Kim SK, Gotsch F, Lamont RF, Ogge G, et al Low circulating maternal adiponectin in patients with pyelonephritis: adiponectin at the crossroads of pregnancy and infection. J Perinat Med 2009; DOI:10.1515/JPM.2009. 134.
  • Marvin KW, Keelan JA, Eykholt RL, Sato TA, Mitchell MD. Use of cDNA arrays to generate differential expression profiles for inflammatory genes in human gestational membranes delivered at term and preterm. Mol Hum Reprod 2002;8:399–408.
  • Nemeth E, Tashima LS, Yu Z, Bryant-Greenwood GD. Fetal membrane distention: I. Differentially expressed genes regulated by acute distention in amniotic epithelial (WISH) cells. Am J Obstet Gynecol 2000;182:50–59.
  • Ognjanovic S, Bao S, Yamamoto SY, Garibay-Tupas J, Samal B, Bryant-Greenwood GD. Genomic organization of the gene coding for human pre-B-cell colony enhancing factor and expression in human fetal membranes. J Mol Endocrinol 2001;26:107–117.
  • Ognjanovic S, Bryant-Greenwood GD. Pre-B-cell colony-enhancing factor, a novel cytokine of human fetal membranes. Am J Obstet Gynecol 2002;187:1051–1058.
  • Ognjanovic S, Tashima LS, Bryant-Greenwood GD. The effects of pre-B-cell colony-enhancing factor on the human fetal membranes by microarray analysis. Am J Obstet Gynecol 2003;189:1187–1195.
  • Ognjanovic S, Ku TL, Bryant-Greenwood GD. Pre-B-cell colony-enhancing factor is a secreted cytokine-like protein from the human amniotic epithelium. Am J Obstet Gynecol 2005;193:273–282.
  • Nemeth E, Millar LK, Bryant-Greenwood G. Fetal membrane distention: II. Differentially expressed genes regulated by acute distention in vitro. Am J Obstet Gynecol 2000;182:60–67.
  • Kendal-Wright CE, Hubbard D, Bryant-Greenwood GD. Chronic stretching of amniotic epithelial cells increases pre-B cell colony-enhancing factor (PBEF/visfatin) expression and protects them from apoptosis. Placenta 2008;29:255–265.
  • Esplin MS, Fausett MB, Peltier MR, Hamblin S, Silver RM, Branch DW, Adashi EY, Whiting D. The use of cDNA microarray to identify differentially expressed labor-associated genes within the human myometrium during labor. Am J Obstet Gynecol 2005;193:404–413.
  • Mastorakos G, Valsamakis G, Papatheodorou DC, Barlas I, Margeli A, Boutsiadis A, Kouskouni E, Vitoratos N, Papadimitriou A, Papassotiriou I, et al The role of adipocytokines in insulin resistance in normal pregnancy: visfatin concentrations in early pregnancy predict insulin sensitivity. Clin Chem 2007;53:1477–1483.
  • Morgan SA, Bringolf JB, Seidel ER. Visfatin expression is elevated in normal human pregnancy. Peptides 2008;29:1382–1389.
  • Katwa LC, Seidel ER. Visfatin in pregnancy: proposed mechanism of peptide delivery. Amino Acids 2008;37:555–558.
  • Szamatowicz J, Kuzmicki M, Telejko B, Zonenberg A, Nikolajuk A, Kretowski A, Gorska M. Serum visfatin concentration is elevated in pregnant women irrespectively of the presence of gestational diabetes. Ginekol Pol 2009;80:14–18.
  • Hu W, Wang Z, Wang H, Huang H, Dong M. Serum visfatin levels in late pregnancy and pre-eclampsia. Acta Obstet Gynecol Scand 2008;87:413–418.
  • Akturk M, Altinova AE, Mert I, Buyukkagnici U, Sargin A, Arslan M, Danisman N. Visfatin concentration is decreased in women with gestational diabetes mellitus in the third trimester. J Endocrinol Invest 2008;31:610–613.
  • Ibanez L, Sebastiani G, Lopez-Bermejo A, Diaz M, Gomez-Roig MD, de ZF. Gender specificity of body adiposity and circulating adiponectin, visfatin, insulin, and insulin growth factor-I at term birth: relation to prenatal growth. J Clin Endocrinol Metab 2008;93:2774–2778.
  • Lopez-Bermejo A, de ZF, az-Silva M, Vicente MP, Valls C, Ibanez L. Cord serum visfatin at term birth: maternal smoking unmasks the relation to foetal growth. Clin Endocrinol (Oxf) 2008;68:77–81.
  • Malamitsi-Puchner A, Briana DD, Boutsikou M, Kouskouni E, Hassiakos D, Gourgiotis D. Perinatal circulating visfatin levels in intrauterine growth restriction. Pediatrics 2007;119:e1314–e1318.
  • Malamitsi-Puchner A, Briana DD, Gourgiotis D, Boutsikou M, Baka S, Hassiakos D. Blood visfatin concentrations in normal full-term pregnancies. Acta Paediatr 2007;96:526–529.
  • Briana DD, Boutsikou M, Gourgiotis D, Kontara L, Baka S, Iacovidou N, Hassiakos D, Malamitsi-Puchner A. Role of visfatin, insulin-like growth factor-I and insulin in fetal growth. J Perinat Med 2007;35:326–329.
  • Hauguel-De MS, Lepercq J, Catalano P. The known and unknown of leptin in pregnancy. Am J Obstet Gynecol 2006;194:1537–1545.
  • Fasshauer M, Bluher M, Stumvoll M, Tonessen P, Faber R, Stepan H. Differential regulation of visfatin and adiponectin in pregnancies with normal and abnormal placental function. Clin Endocrinol (Oxf) 2007;66:434–439.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.