1,292
Views
61
CrossRef citations to date
0
Altmetric
Original Article

Severe preeclampsia is characterized by increased placental expression of galectin-1

, MD, PhD, , , , , , , , , , , & show all
Pages 429-442 | Received 07 Sep 2007, Accepted 21 Feb 2008, Published online: 07 Jul 2009

Abstract

Objective. Galectin-1 is a major anti-inflammatory protein expressed by the placenta and immune cells that can bias the character of inflammatory responses toward the Th2 type. Galectin-1 is expressed in immune privileged sites, it can facilitate immune tolerance and tumor immune escape, and it has been successfully used for the suppression of experimental autoimmune diseases as well as graft-versus-host disease in murine models. We propose that an abnormal immune response in some pregnancy complications may be associated with changes in placental expression of galectin-1. To test this hypothesis, we studied placental galectin-1 mRNA and protein expression and localization in women with preeclampsia (PE) and in those who delivered a small-for-gestational age (SGA) neonate.

Study design. This cross-sectional study included pregnant women matched for gestational age at delivery in the following groups: (1) severe PE (n = 10), (2) severe PE complicated with SGA (n = 10), (3) SGA without PE (n = 10), and (4) controls (n = 10). Galectin-1 mRNA and protein were localized in placentas by in situ hybridization and immunofluorescence microscopy. Galectin-1 mRNA expression was determined by quantitative real-time reverse transcription-polymerase chain reaction (RT-PCR), and galectin-1 protein content by Western blot. Non-parametric statistics were used for analysis.

Results. (1) In normal term placentas, galectin-1 mRNA or immunofluorescence signals were detected in the trophoblasts, villous stromal cells, Hofbauer cells, endothelial cells of the villous blood vessels, and the villous stroma. (2) Placental galectin-1 mRNA expression was significantly higher in severe PE (with or without SGA) than in controls (1.47-fold, p = 0.004; 1.44-fold, p = 0.003, respectively) and in SGA (1.68-fold, p = 0.001; 1.64-fold, p = 0.001, respectively). (3) Trophoblasts in placentas of patients with severe PE had the most intense galectin-1 immunostaining.

Conclusions. (1) We report for the first time the placental expression and localization of galectin-1 mRNA and demonstrate that the protein is abundantly present in third trimester human placentas. (2) Placental galectin-1 expression is higher in severe PE than in normal pregnancy regardless of the presence of SGA. (3) However, it is not altered in SGA without PE. We propose that the increased placental expression of galectin-1 in patients with severe PE may represent a fetal response to an exaggerated systemic maternal inflammation; thus, galectin-1 may be implicated in maternal–fetal immune tolerance in humans.

Introduction

Preeclampsia (PE) is considered a maternal disease, which is associated with fetal growth restriction in 10–25% of the cases, while pregnancies complicated with growth-restricted (IUGR) fetuses or small-for-gestational age (SGA) neonates without PE usually have no appreciable clinical impact on the mother Citation[1],Citation[2]. These ‘great obstetrical syndromes’Citation[3] share similar pathophysiologic mechanisms Citation[2], such as generalized endothelial cell dysfunction Citation[4-8], abnormal placentation Citation[9-16], anti-angiogenic state Citation[17-35], chronic uteroplacental ischemia Citation[36-40], and an increased maternal systemic inflammatory response Citation[41-46]. The latter is highly exaggerated in PE and involves the activation of the innate immune system Citation[42],Citation[47-49].

Galectins are cytokine-like immunoregulatory proteins, members of an evolutionarily-conserved protein family that share similar structures, carbohydrate recognition domains (CRDs), and affinity for beta-galactosides present on cell surface glycoconjugates Citation[50-56]. Galectins exhibit preferential binding to a subset of ligands Citation[57-59] and are implicated in deciphering the high-density ‘glycocode’ stored in glycoproteins, proteoglycans, and glycolipids Citation[60-62]. Their versatile functions include the regulation of cell–cell/matrix interactions, cell cycle, apoptosis, cell migration, and the recognition of microbial glycosignatures Citation[50],Citation[53],Citation[55],Citation[56]. Galectins can affect both the innate and the adaptive arms of the immune system, inhibiting (e.g., galectin-1) Citation[56],Citation[63] or augmenting (e.g., galectin-3) Citation[56] the inflammatory response.

Galectin-1 was the first human galectin discovered, purified Citation[64] and cloned Citation[65],Citation[66] from the placenta Citation[63]. Subsequent studies presented the immunolocalization of galectin-1 in normal first and third trimester placentas; however, the described expression patterns lacked consistency Citation[67-71]. Galectin-1 is expressed in immune-privileged sites (e.g., testis and brain) Citation[72],Citation[73], and upregulated in tumors (e.g., melanoma), presumably to facilitate the escape of immunosurveillance Citation[63],Citation[74]. Galectin-1 has pleiotropic binding activity Citation[57],Citation[59],Citation[63] and mediates a wide variety of immune cell interactions () Citation[63], mainly promoting immune tolerance Citation[75] and down-regulating the innate and adaptive immune responses Citation[76]. Moreover, galectin-1 has potent anti-inflammatory effects including: (1) inhibition of acute inflammation Citation[76]; (2) suppression of T cell-mediated autoimmune diseases Citation[77-80]; (3) amelioration of graft-versus-host disease Citation[81]; and (4) biasing the character of the immune response to the Th2 type Citation[63].

Figure 1. Pleiotropic effects of galectin-1 on immune cells. Galectin-1 is up-regulated in inflammation and infection and contributes to the regulation of immune cells in physiological and pathological conditions. Galectin-1 is involved in both the adaptive and innate immune responses and predominantly exerts anti-inflammatory effects on different immune cell types. (X-ray crystallographic data of galectin-1 (1GZW) [56] was accessed at the MMDB Database (NCBI, NLM, NIH, Bethesda, MD, USA) and the ribbon diagram was generated with Cn3D and Adobe Photoshop 7.0. The jelly-roll structure of galectin-1 includes two antiparallel β-sheets (F1–F5 in yellow; S1–S6a/b in red); the carbohydrate-recognition domains encompass the S4–S6a/S6b sheets on the concave face of the subunits.)

Figure 1. Pleiotropic effects of galectin-1 on immune cells. Galectin-1 is up-regulated in inflammation and infection and contributes to the regulation of immune cells in physiological and pathological conditions. Galectin-1 is involved in both the adaptive and innate immune responses and predominantly exerts anti-inflammatory effects on different immune cell types. (X-ray crystallographic data of galectin-1 (1GZW) [56] was accessed at the MMDB Database (NCBI, NLM, NIH, Bethesda, MD, USA) and the ribbon diagram was generated with Cn3D and Adobe Photoshop 7.0. The jelly-roll structure of galectin-1 includes two antiparallel β-sheets (F1–F5 in yellow; S1–S6a/b in red); the carbohydrate-recognition domains encompass the S4–S6a/S6b sheets on the concave face of the subunits.)

The overexpression of galectin-1 has been observed in cultured human endothelial cells Citation[82], in the synovia of rheumatoid arthritis patients Citation[83], and in activated immune cells Citation[84-87]. Galectin-1 was proposed to regulate the extent of the immune response during inflammation Citation[88]; thus, it might also be involved in the complex inflammatory responses observed in pregnancy complications.

The aims of this study were to determine: (1) the cellular localization of galectin-1 mRNA and protein in normal third trimester placentas, and (2) the changes in placental galectin-1 expression in patients with severe PE with and without SGA, as well as in those patients without PE who delivered an SGA neonate.

Materials and methods

Study design and population

This cross-sectional study included pregnant women in the following groups: (1) severe PE (n = 10), (2) severe PE with SGA (n = 10), (3) SGA without PE (n = 10), and (4) pregnant women with preterm and term labor (control group, n = 10). Women with severe PE or SGA were matched for gestational age at delivery within two weeks of gestation to women in the control group. Patients with multiple pregnancies, preterm prelabor rupture of membranes, histologic chorioamnionitis, stillbirth, or fetal congenital or chromosomal abnormalities were excluded. Samples and data were retrieved from the bank of biological samples and clinical databases of the Perinatology Research Branch. All patients were enrolled at Hutzel Women's Hospital, Detroit, MI, USA, and provided written informed consent prior to the collection of samples. The utilization of samples for research purposes was approved by the institutional review boards of both Wayne State University and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD/NIH/DHHS). Many of these samples have been employed to study the biology of inflammation in normal pregnant women and those with pregnancy complications.

Definitions

PE was defined as hypertension (systolic blood pressure ≥140 mmHg or diastolic blood pressure ≥90 mmHg on at least two occasions, 4 hours to 1 week apart) associated with proteinuria (≥300 mg in a 24-hour urine collection, or two dipstick measurements of ≥1+Citation[89] or one dipstick measurement of ≥2+) Citation[90]. Severe PE was defined as systolic blood pressure ≥160 mmHg or diastolic blood pressure ≥110 mmHg and/or proteinuria greater than 5 g in a 24-hour collection or >3+ on dipstick Citation[1] and in the presence of multi-organ involvement Citation[1]. SGA was defined as neonatal birth weight below the 10th percentile for gestational age at birth according to the national birth weight distribution Citation[91]. Labor was defined by the presence of regular uterine contractions at a frequency of at least two contractions every 10 minutes with cervical changes resulting in delivery at <37 (preterm) or ≥37 (term) completed weeks of gestation. Control women with preterm or term labor delivered neonates with a birth weight appropriate-for-gestational age (≥10th and ≤90th percentile).

mRNA in situ hybridization

The 123-bp fragment of human galectin-1 cDNA generated by PCR (forward primer: CATCTCTCTCgggTggAgTC, reverse primer: gAAggCACTCTCCAggTTTg) was subcloned into pGEM-T Easy vector (Promega Corp., Madison, WI, USA) containing SP6 and T7 polymerase promoters. Digoxigenin-labeled anti-sense and sense riboprobes were generated with SP6 and T7 polymerases after linearization of the plasmid with Bam HI and Hind III, respectively. Five μm sections of paraffin-embedded villous tissues were deparaffinized, hydrated in xylene and graded ethanol, and then treated with proteinase K (15 μg/mL) in 0.1 M Tris buffer (pH 8.0) and 50 mM EDTA for 10 minutes at 37°C. Slides were fixed with 4% paraformaldehyde for 20 minutes and acetic anhydride for 10 minutes. Sections were incubated in a hybridization buffer containing digoxigenin-tagged galectin-1 riboprobe (2 μg/mL). Hybridization was carried out in a humidity chamber overnight at 55°C. After repeated post-hybridization washes, sections were incubated with alkaline phosphatase-conjugated anti-digoxigenin antibody (Roche Diagnostics, Indianapolis, IN, USA) for 1 hour at room temperature. Nitro-blue tetrazolium chloride and 5-bromo-4-chloro-3-indolyl phosphate p-toluidine salt were used for detection of the hybridization signal.

Total RNA extraction

Total RNA was isolated from snap-frozen placental villous tissues using TRIzol reagent (Invitrogen, Carlsbad, CA, USA) and then Qiagen RNeasy kit (Qiagen, Valencia, CA, USA) according to the manufacturers’ recommendations. The 28S/18S ratio and the RNA integrity number (RIN) were assessed using a Bioanalyzer 2100 (Agilent Technologies, Wilmington, DE, USA). An A260nm/A280nm ratio of 1.8, a 28S/18S ratio of 1.3, and a RIN of 6 were minimum requirements for inclusion in expression analysis.

Quantitative real-time reverse transcription-polymerase chain reaction (qRT-PCR)

Total RNA was reverse-transcribed with a TaqMan reverse transcription reagent kit using random hexamers (Applied Biosystems, Foster City, CA, USA). The standard curve was run with the LGALS1 TaqMan gene expression assay (Hs00169327_m1; Applied Biosystems, Foster City, CA, USA) to determine the quantity of cDNA needed for an approximate cycle threshold (Ct) of 25. The human RPLPO (large ribosomal protein) TaqMan endogenous control (part number: 4326314E) was used as the housekeeping gene for relative quantitation. The LGALS1 and RPLPO genes were then run in triplicate for each case to allow for the assessment of technical variability.

Immunofluorescence confocal microscopy

Five μm sections of snap-frozen villous tissues were fixed with 4% paraformaldehyde for 1 hour at room temperature and acetone for 1 minute at −20°C. Slides were preincubated with Image-it FX signal enhancer (Molecular Probes, Carlsbad, CA, USA) for 30 minutes and CAS blocking solution (Zymed, San Francisco, CA, USA) for 10 minutes. Sections were incubated with goat anti-human galectin-1 IgG (R&D Systems, Minneapolis, MN, USA) and goat isotype control primary IgG at 1:50 dilutions for 1 hour, and with Alexa Fluor 568 conjugated donkey anti-goat IgG (Invitrogen Co., Carlsbad, CA, USA) at 1:1000 dilution for 1 hour. Sytox Green nuclear counter stain (Cambrex, North Brunswick, NJ, USA) was applied at a 1:100,000 dilution for 3 minutes. Stainings were performed on an autostainer (Dako, Carpinteria, CA, USA). Fluorescent and differential interference contrast (DIC) images were captured with a Zeiss Axiovert 200 Ultra-View ERS rapid confocal imager equipped with an argon laser and a Zeiss Fluar 40×/1.3 oil objective. Images were evaluated by Perkin Elmer ImageSuite 3.0 version 14 (Perkin Elmer Inc., Waltham, MA, USA).

Western blot analysis

Placental villous tissues were lysed with RIPA buffer (Sigma, St Louis, MO, USA) containing protease inhibitor (Roche Diagnostics, Mannheim, Germany). Thirty μg of the protein lysates and 15 ng of recombinant human galectin-1 (R&D Systems, Minneapolis, MN, USA) were electrophoresed on 15% (w/v) SDS-polyacrylamide gels and electroblotted onto PVDF membranes (Bio-Rad, Hercules, CA, USA). The membranes were probed with a goat anti-human galectin-1 IgG (R&D Systems, Minneapolis, MN, USA) or with a murine monoclonal anti-β-actin antibody (Sigma, St Louis, MO, USA) at 1:2000 dilutions for 1 hour, then incubated with horseradish peroxidase conjugated donkey anti-goat IgG (R&D Systems, Minneapolis, MN, USA) or goat anti-mouse IgG (Jackson ImmunoResearch Laboratories Inc., West Grove, PA, USA) at 1:4000 dilutions for 1 hour. Protein bands were detected by ECL chemiluminescence (Amersham Biosciences, Piscataway, NJ, USA). The specificity of the anti-human galectin-1 IgG was validated by testing with human recombinant galectins-2, -3, -4, -7, and -8 in the same experimental conditions as galectin-1.

Statistical analysis

Comparisons among groups were performed using Fisher's exact test for proportions and one-way ANOVA test for normally distributed continuous variables, as well as Kruskal–Wallis test and Mann–Whitney U-test for non-normally distributed continuous variables. For the analysis of qRT-PCR data, pair-wise group comparisons were performed using ‘generalized estimating equations’Citation[92]. In parallel, the t-test was also applied by averaging over the three technical replicates (Ct values) of each subject. To determine the influence of the gestational age on LGALS1 gene expression within the groups, a linear model was fitted in which the gestational age was used as a predictor for the Ct values. An adjustment of p-values to account for the six different comparisons among the four groups was performed using the Bonferroni method Citation[93]. An adjusted p-value of <0.05 was considered to be statistically significant. The R statistical software (www.r-project.org) including required libraries and SPSS version 12.0 (SPSS Inc., Chicago, IL, USA) were used for the analyses.

Results

Demographic, clinical, and histopathologic data

displays the demographic and clinical characteristics of the study groups. A larger proportion of women with severe PE complicated by SGA had chronic hypertension than did patients in the other study groups. Placental histopathologic findings consistent with maternal underperfusion Citation[94],Citation[95] (e.g., increased syncytial knots, increased intervillous fibrin, distal villous hypoplasia) were more frequent in preterm than in term cases of SGA without PE and in severe PE with or without SGA.

Table I.  Demographic and clinical characteristics of the study population.

Localization of galectin-1 mRNA and protein in normal term placentas

Galectin-1 mRNA in situ hybridization signals were readily detectable in normal term villous placentas, and a similar pattern was observed with galectin-1 immunofluorescence. mRNA hybridization signals were distinct in the trophoblastic layers, especially in cytotrophoblasts, and were also detected in stromal cells (). Immunofluorescence microscopy demonstrated galectin-1 immunopositivity of trophoblasts, Hofbauer cells, stromal cells, and the endothelium of the villous blood vessels (capillaries, arterioles, and venules), as well as the syncytiotrophoblast apical membrane and the villous stroma ().

Figure 2. Localization of galectin-1 mRNA and protein in normal term placentas. (A) Galectin-1 mRNA hybridization signal was distinct in villous stromal cells and in the trophoblastic layers, especially in cytotrophoblasts. Inlet: there was no hybridization signal with antisense control (mRNA in situ hybridization, 20× magnification). (B) The syncytiotrophoblast apical membrane, villous capillary endothelial cells, and stromal cells were strongly immunopositive, while the villous stroma was weakly stained. Galectin-1 staining is shown in red, nuclei are in green. Inlet: there was only nuclear staining when applying isotype control primary antibody (immunofluorescence confocal microscopy, 40× magnification, fluorescence and differential interference contrast (DIC) combination images).

Figure 2. Localization of galectin-1 mRNA and protein in normal term placentas. (A) Galectin-1 mRNA hybridization signal was distinct in villous stromal cells and in the trophoblastic layers, especially in cytotrophoblasts. Inlet: there was no hybridization signal with antisense control (mRNA in situ hybridization, 20× magnification). (B) The syncytiotrophoblast apical membrane, villous capillary endothelial cells, and stromal cells were strongly immunopositive, while the villous stroma was weakly stained. Galectin-1 staining is shown in red, nuclei are in green. Inlet: there was only nuclear staining when applying isotype control primary antibody (immunofluorescence confocal microscopy, 40× magnification, fluorescence and differential interference contrast (DIC) combination images).

Placental galectin-1 mRNA expression is increased in severe PE

Placental galectin-1 mRNA expression was significantly higher in severe PE (1.44-fold, p = 0.003) and severe PE complicated by SGA (1.47-fold, p = 0.004) than in gestational age-matched controls. In addition, there was a significantly higher placental galectin-1 mRNA expression in severe PE with or without SGA than in SGA without PE (1.68-fold, p = 0.001; 1.64-fold, p = 0.001, respectively). However, there was no difference in galectin-1 mRNA expression between the SGA and control groups and between patients with severe PE and SGA and those with severe PE alone (). In these subsets of patients, gene expressions within the groups did not depend on gestational age.

Figure 3. Placental galectin-1 mRNA expression is increased in severe PE. Placental galectin-1 mRNA expression was significantly increased in severe PE (1.44-fold, p = 0.003) and in severe PE complicated by SGA (1.47-fold, p = 0.004) when compared to gestational age-matched controls. Galectin-1 expression was significantly higher in severe PE with or without SGA than in SGA without PE (1.68-fold, p = 0.001; 1.64-fold, p = 0.001; respectively). There was no difference in galectin-1 mRNA expression between SGA and controls (p = 1.00) and the two severe PE groups (p = 1.00).

Figure 3. Placental galectin-1 mRNA expression is increased in severe PE. Placental galectin-1 mRNA expression was significantly increased in severe PE (1.44-fold, p = 0.003) and in severe PE complicated by SGA (1.47-fold, p = 0.004) when compared to gestational age-matched controls. Galectin-1 expression was significantly higher in severe PE with or without SGA than in SGA without PE (1.68-fold, p = 0.001; 1.64-fold, p = 0.001; respectively). There was no difference in galectin-1 mRNA expression between SGA and controls (p = 1.00) and the two severe PE groups (p = 1.00).

Placental galectin-1 immunoreactivity is increased in severe PE

In control placentas, galectin-1 immunopositivity was detected in trophoblasts, stromal cells, Hofbauer cells, endothelial cells of the villous blood vessels, and the villous stroma (). In patients with SGA without PE, galectin-1 immunofluorescent staining was similar to that in controls (). Severe PE complicated with SGA was characterized by villous galectin-1 immunofluorescence signal present in all cell types, with the strongest signal in trophoblasts, stromal cells, and the stroma (). Similarly, in severe PE without SGA, trophoblasts, stromal cells, and stroma had the most intense immunofluorescent staining (). Immunoblots revealed a 15-kDa immunoreactive protein in all placentas, consistent with the size of galectin-1 monomer. Compared to gestational age-matched controls, the strongest signal was detected in the severe PE groups both preterm and term as represented in . When testing for cross-reactivity on Western blots, the primary antibody specifically recognized placental and recombinant galectin-1 but not recombinant galectins-2, -3, -4, -7, and -8.

Figure 4. Placental galectin-1 immunofluorescence signal is increased in severe PE. (A) In term controls (38 weeks), the syncytiotrophoblast apical membrane, capillary endothelial cells, and stromal cells had the strongest galectin-1 immunopositivity. (B) In term SGA (39 weeks), galectin-1 immunopositivity was similar in its extent and pattern to that seen in controls. (C) In severe PE complicated with SGA (39 weeks), galectin-1 immunofluorescence signal was intense, predominantly in the trophoblastic layer, stromal cells, and the villous stroma. (D) Similarly, in severe PE without SGA (37 weeks), trophoblastic, stromal cell and stromal galectin-1 immunofluorescence staining was the strongest. Galectin-1 staining is shown in red, nuclei are in green (immunofluorescence confocal microscopy, 40× magnifications, fluorescence and differential interference contrast (DIC) combination images).

Figure 4. Placental galectin-1 immunofluorescence signal is increased in severe PE. (A) In term controls (38 weeks), the syncytiotrophoblast apical membrane, capillary endothelial cells, and stromal cells had the strongest galectin-1 immunopositivity. (B) In term SGA (39 weeks), galectin-1 immunopositivity was similar in its extent and pattern to that seen in controls. (C) In severe PE complicated with SGA (39 weeks), galectin-1 immunofluorescence signal was intense, predominantly in the trophoblastic layer, stromal cells, and the villous stroma. (D) Similarly, in severe PE without SGA (37 weeks), trophoblastic, stromal cell and stromal galectin-1 immunofluorescence staining was the strongest. Galectin-1 staining is shown in red, nuclei are in green (immunofluorescence confocal microscopy, 40× magnifications, fluorescence and differential interference contrast (DIC) combination images).

Figure 5. Placental galectin-1 is increased in severe PE. Fifteen ng of recombinant human galectin-1 and 30 μg of villous tissue lysates were electrophoresed on 15% (w/v) SDS-polyacrylamide gel. Proteins were electroblotted and probed with anti-human galectin-1 IgG. All samples used for the representative image were taken from placentas delivered at 39 weeks of gestation. Lane 1: recombinant galectin-1; lane 2: control; lane 3: SGA; lane 4: severe PE; lane 5: severe PE complicated with SGA. Galectin-1 migrated as a single 15-kDa band in all lanes. Galectin-1 immunopositive signal was the strongest in severe PE complicated with SGA.

Figure 5. Placental galectin-1 is increased in severe PE. Fifteen ng of recombinant human galectin-1 and 30 μg of villous tissue lysates were electrophoresed on 15% (w/v) SDS-polyacrylamide gel. Proteins were electroblotted and probed with anti-human galectin-1 IgG. All samples used for the representative image were taken from placentas delivered at 39 weeks of gestation. Lane 1: recombinant galectin-1; lane 2: control; lane 3: SGA; lane 4: severe PE; lane 5: severe PE complicated with SGA. Galectin-1 migrated as a single 15-kDa band in all lanes. Galectin-1 immunopositive signal was the strongest in severe PE complicated with SGA.

Discussion

Principal findings of this study

(1) Galectin-1 mRNA is ubiquitously expressed in the villous placenta, and its expression pattern is similar to that of galectin-1 immunostaining. (2) Placental galectin-1 mRNA expression was significantly increased in severe PE with or without SGA compared to gestational age-matched controls and to SGA. (3) Increased placental galectin-1 immunofluorescence was detected in trophoblasts, stromal cells, and the villous stroma in cases of severe PE with or without SGA when compared to controls or to SGA.

Galectin-1 is abundantly expressed in third trimester villous placenta

This is the first study that localizes galectin-1 in the villous placenta with mRNA in situ hybridization along with sensitive immunofluorescence staining. The combination of these methods allowed us to identify galectin-1 in all cell types of the villi, as well as in the syncytiotrophoblast apical membrane at the maternal–fetal interface. Other studies localized galectin-1 with immunohistochemistry in first and third trimester normal human placentas Citation[67-71]; however, these reports gave less comprehensive and inconsistent results. Two studies demonstrated galectin-1 immunopositivity of mesenchymal cells in both trimesters Citation[67],Citation[69]. The villous stroma, where galectin-1 had been co-localized with fibronectin and laminin Citation[96], was also immunopositive in the first trimester Citation[67],Citation[70] and at term Citation[67]. Endothelial cells, vessel walls Citation[67],Citation[68], and the syncytiotrophoblast was shown to be galectin-1 positive in the first Citation[70] and third trimester Citation[68],Citation[71]; however, villous cytotrophoblast immunoreactivity was reported only at term Citation[68]. Overall, none of these reports were consistent with others in terms of the described expression patterns Citation[67-71]. This inconsistency might be the consequence of the differences in gestational age of the tissues, the applied methodologies, and the type of antibodies used. In our study, the specificity of the immunostaining was supported by the following: (1) There was no immunofluorescent signal (besides the nuclear counterstain) when the isotype control primary antibody was applied; (2) the antibody that recognized placental and recombinant galectin-1 did not cross-react with homologous galectins on Western blot analyses; and (3) in situ hybridization confirmed galectin-1 mRNA expression in immunopositive cells.

Galectin-1 is expressed in placentas of other mammals Citation[97],Citation[98]. Murine galectin-1, which has an 88% amino acid sequence identity to its human ortholog, was found to be ubiquitously expressed in the mouse placenta, including trophoblast cells in the labyrinth region and the spongy layer Citation[97]. Recent publications revealed that cytoplasmic galectin-1 can be translocated to the intra- Citation[63],Citation[99] and extracellular side of the cell membrane Citation[63],Citation[100], and that recombinant galectin-1 is capable of binding to human syncytiotrophoblast and extravillous trophoblast cell surface Citation[101]. Thus, the abundance and ubiquitous expression of galectin-1 by the villous tissues and its presence at the maternal–fetal interface suggest that this galectin might have several functions in the placenta.

What is the role of galectin-1 in the placenta?

The most important biological processes to which galectin-1 has been linked include connective tissue organization, tumor invasiveness and metastasis, regulation of cell proliferation and differentiation, and local immunomodulation Citation[63]. Currently, there are no functional data on the effect of galectin-1 on normal human placental cells. Based on its placental expression pattern and functional effects on other cell types, we propose that galectin-1 may participate in the following processes in the placenta.

Extracellular matrix (ECM) organization

The strong galectin-1 immunopositivity of villous stromal cells and the villous stroma found in this study is in accordance with the abundance of galectin-1 in cells of mesenchymal origin and their ECM, where it was proposed to have a pivotal role in the organization and presentation of connective tissue components and tissue development Citation[63],Citation[70]. Galectin-1 co-localizes, binds, and cross-links beta-integrins and poly-N-acetyl-lactosamine-rich components of the placental ECM (e.g., laminin and fibronectin) Citation[59],Citation[69],Citation[70],Citation[96],Citation[102], which are important in the control of cell attachment, migration, invasion, as well as in the assembly and remodeling of the ECM Citation[63]. Indeed, overexpression of galectin-1 decreases the incorporation of its ligands (vitronectin and chondroitin sulphate) into the ECM of smooth muscle cells Citation[103]. Based on these findings, we propose that the expression of galectin-1 may have importance in the cross-talk between trophoblasts, stromal cells, and the stroma during placentation and in the development and maintenance of villous tissues.

Immune regulation by villous endothelial cells

Our data demonstrated that galectin-1 is expressed by the endothelium of villous capillaries, arterioles, and veins in the villous placenta. This is consistent with previous studies demonstrating the expression of galectin-1 in endothelial cells of human umbilical vein and aorta, bovine aorta, and microvessels in rat lung and mouse lung and brain Citation[82],Citation[104-106]. Microvascular endothelial cells form specialized microcirculatory networks, which regulate coagulation, angiogenesis, and the distribution of activated immune cells, thus, innate and adaptive immune responses Citation[107]. Indeed, galectin-1 was demonstrated to inhibit polymorphonuclear cell chemotaxis and trans-endothelial migration in vitro and interleukin (IL)-1-induced polymorphonuclear cell recruitment into the mouse peritoneal cavity in vivoCitation[106]. Galectin-1 inhibited T cell migration across endothelial cells expressing increased amounts of the protein Citation[108], and induced apoptosis of susceptible T cells bound to cultured human aortic endothelial cells expressing high amounts of galectin-1 Citation[105]. These data suggest that galectin-1 expressed by the villous endothelium may also be part of an anti-inflammatory loop, regulating recruitment and transmigration of fetal immune cells in the villi.

Host–pathogen immune response

Hofbauer cells are also regarded as fetal macrophages capable of phagocytosis Citation[109-111] and production of cytokines (e.g., IL-1, IL-8, suppressor of cytokine signaling (SOCS) proteins) Citation[112-114], chemokines (e.g., macrophage inhibitory protein (MIP)-1-beta) Citation[115], and phagocytosis-related enzymes (acid phosphatase (ACP) and glucose-6-phosphate dehydrogenase (G6PD)) Citation[111]. Hofbauer cells exhibiting strong G6PD staining and ACP labeling in the phagosomes are phagocytic cells. The percentage of these activated macrophages is significantly higher in placentas of patients with infectious miscarriages than in gestational age-matched controls Citation[111]. Of interest, there is an up-regulation of galectin-1 expression in macrophages during their activation Citation[84],Citation[87] and in infection Citation[85]. In turn, galectin-1 inhibits macrophage microbicidal activity Citation[85], decreases iNOS expression and NO metabolism Citation[116], and regulates constitutive and inducible expression of high affinity FcγRI (CD64) and phagocytosis Citation[117]. Based on these data, galectin-1 may also have a role in the fetal response to pathogenic insults and in the regulation of the extent of the inflammatory response in the villi.

Maternal–fetal immune tolerance

This study confirmed the expression of galectin-1 in the syncytiotrophoblast Citation[68],Citation[70],Citation[71] and showed its sublocalization onto the apical membrane. The syncytiotrophoblast is a rich source of immunomodulatory molecules Citation[118-120], and those with immunosuppressive properties (e.g., human chorionic gonadotropin, human placental lactogen, indoleamine 2,3-dioxygenase, CD95L/Fas ligand, pregnancy-specific beta-1 glycoproteins, SOCS proteins, TNF-related apoptosis-inducing ligand (TRAIL)) Citation[114],Citation[121-126] are proposed to attenuate maternal immune responses and, thus, maintain tolerance to the fetus. The evidence supporting that placental galectin-1 may also have an important role in immune tolerance includes the following: (1) Galectin-1 reduces host alloreactivity and ameliorates graft-versus-host disease in mice Citation[81]; (2) it is capable of triggering the apoptosis of immature thymocytes Citation[105] and activated T cells Citation[75], biasing the immune responses to the Th2 type Citation[63],Citation[127],Citation[128], and suppressing experimental T cell-mediated autoimmune diseases (e.g., encephalomyelitis, retinal disease, arthritis, or hepatitis) in rats and mice Citation[77-80]; (3) galectin-1 plays a role in the immune escape of a wide variety of tumors (e.g., head and neck carcinoma, astrocytoma, glioma, melanoma) by possibly down-regulating tumor resident T cell survival and linking tumor hypoxia and immune privilege Citation[74],Citation[129-132]; (4) galectin-1 is up-regulated in mammary adenocarcinoma cells by transforming growth factor (TGF)-βCitation[133], an important molecule in tolerance Citation[125]; (5) it is highly expressed in uterine natural killer (NK) cells Citation[134] and activated CD4+CD25+ regulatory T cells Citation[86] that have been implicated in maternal–fetal tolerance Citation[125],Citation[135],Citation[136].

Moreover, a recent study reported that LGALS1-null mice show higher rates of fetal loss compared to wild-type mice in allogeneic matings, and treatment with recombinant galectin-1 prevents fetal loss and restores tolerance through various mechanisms, such as the induction of tolerogenic dendritic cells and the expansion of CD4+CD25+ IL-10 secreting regulatory T cells Citation[137]. Thus, galectin-1 expressed by uterine tissues (decidua, myometrium) has been suggested to have a pivotal role in conferring maternal–fetal tolerance Citation[137]. These results mainly relate the effects of galectin-1 to the maternal side of the fetal–maternal interface. However, based on the abundance of galectin-1 in the placenta, especially in the syncytiotrophoblast, our study suggests that galectin-1 may also be important in the fetal immune response.

Placental galectin-1 is up-regulated in severe PE but not in SGA

A novel finding described herein is that both galectin-1 mRNA expression and immunopositivity were significantly increased in the placentas of patients with severe PE. The up-regulation of galectin-1 in severe PE was independent of gestational age, placental histopathologic findings, and the presense or absence of SGA; thus, it is most likely to be associated with the exaggerated maternal systemic inflammatory response Citation[41-46],Citation[138], which is generally found in PE but may be less developed in pregnancies complicated by SGA or IUGR Citation[2]. The semi-quantitative results of our immunostainings are in accord with those described by a recent study Citation[71]; however, we also quantitatively verified these results by measuring galectin-1 mRNA expression in the villi.

An analogy between preeclampsia and allograft rejection has recently been proposed Citation[139]. It was reported that in rejected human kidney allograft, galectin-1 was up-regulated in endothelial cells of peritubular capillaries and large vessels in inflammatory regions, at the sites of direct contact between host immune cells and the rejected graft Citation[140]. In the light of this finding, it is not surprising that we found up-regulation of galectin-1 expression in the syncytiotrophoblast, which is also in direct contact with activated maternal leukocytes Citation[42]. Moreover, galectin-1 is a pattern-recognition molecule, which operates as a ‘cell stress sensor’ under physiological and pathological conditions Citation[141]. It is up-regulated during cell activation and at the time and loci of acute and chronic inflammation and infection Citation[82-87], where it is proposed to regulate the extent of the immune responses Citation[88]. ‘Danger signals’ and pattern-recognition receptors at the maternal–fetal interface have recently been proposed to create an abnormal placental cytokine milieu Citation[142] and link the activation of the innate immune system and PE Citation[48],Citation[49]. Hence, the increased expression of galectin-1 in trophoblasts of patients with severe PE may reflect an enlarged ‘cellular stress’ response.

Recently, an analogy between immune responses to infections and transplants, which involves terminating mechanisms that may help to avoid damage to either normal or infected tissues, has been proposed Citation[143]. The newly evolved ‘danger model’ of immunity suggests that tissues may have control over immune cells and restrict the class and extent of immune reactions Citation[144,145]. The current approach in the immunology of pregnancy that has challenged the traditional transplantation paradigm also focuses on the unique uterine immune response to the placenta and on the local interactions between placental and maternal immune cells Citation[146]. The increased expression of galectin-1 in the placenta of patients with severe PE is in agreement with these concepts, and may represent a mechanism by which the placenta may control exaggerated immune responses, which include both the fine-tuning of host–pathogen interactions and the maintenance of maternal–fetal immune tolerance.

Conclusions

We report for the first time the placental expression and localization of galectin-1 mRNA, and demonstrate that the protein is abundantly present in third trimester placentas. Placental galectin-1 expression is higher in severe PE than in normal pregnancy, regardless of the presence of SGA. This finding was not observed in patients with SGA alone. We propose that the increased placental expression of galectin-1 in patients with severe PE may represent a local response to systemic maternal inflammation, suggesting that galectin-1 may be implicated in maternal–fetal tolerance.

Acknowledgements

The authors thank Dr Susan Land, Daniel Lott, and Ms Sarah McNorton at the Applied Genomics Technology Center of Wayne State University for performing the qRT-PCR reactions. We wish to acknowledge the invaluable contributions of Dr Yu Mi Han, Dr Sung-Su Kim, Ms Lorri McLuckie, Ms Rona Wang, Adam Pitt, Ms Sandy Field, Ms Nancy Hauff, Gerardo Rodriguez, and the nursing staff of the Perinatology Research Branch and the Detroit Medical Center to this manuscript.

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

Declaration of interest: The authors report no conflicts of interest. The authors alone are responsible for the content and writing of the paper.

Notes

This paper was presented in part as a poster (abstract No. 442) at the 27th Annual Meeting of the Society for Maternal–Fetal Medicine in San Francisco, CA, USA, February 9, 2007.

References

  • Sibai B, Dekker G, Kupferminc M. Pre-eclampsia. Lancet 2005; 365: 785–799
  • Ness R B, Sibai B M. Shared and disparate components of the pathophysiologies of fetal growth restriction and preeclampsia. Am J Obstet Gynecol 2006; 195: 40–49
  • Romero R. Prenatal medicine: The child is the father of the man. Prenat Neonatal Med 1996; 1: 8–11
  • Roberts J M, Taylor R N, Musci T J, Rodgers G M, Hubel C A, McLaughlin M K. Preeclampsia: An endothelial cell disorder. Am J Obstet Gynecol 1989; 161: 1200–1204
  • Bretelle F, Sabatier F, Blann A, D'Ercole C, Boutiere B, Mutin M, Boubli L, Sampol J, Dignat-George F. Maternal endothelial soluble cell adhesion molecules with isolated small for gestational age fetuses: Comparison with pre-eclampsia. BJOG 2001; 108: 1277–1282
  • Johnson M R, Anim-Nyame N, Johnson P, Sooranna S R, Steer P J. Does endothelial cell activation occur with intrauterine growth restriction. BJOG 2002; 109: 836–839
  • Redman C W, Sargent I L. Latest advances in understanding preeclampsia. Science 2005; 308: 1592–1594
  • Poston L. Endothelial dysfunction in pre-eclampsia. Pharmacol Rep 2006; 58(Suppl)69–74
  • Brosens I A, Robertson W B, Dixon H G. The role of the spiral arteries in the pathogenesis of pre-eclampsia. J Pathol 1970; 101: vi
  • De Wolf F, Brosens I, Renaer M. Fetal growth retardation and the maternal arterial supply of the human placenta in the absence of sustained hypertension. Br J Obstet Gynaecol 1980; 87: 678–685
  • Pijnenborg R, Anthony J, Davey D A, Rees A, Tiltman A, Vercruysse L, van Assche A. Placental bed spiral arteries in the hypertensive disorders of pregnancy. Br J Obstet Gynaecol 1991; 98: 648–655
  • Myatt L. Role of placenta in preeclampsia. Endocrine 2002; 19: 103–111
  • Burton G J, Jauniaux E. Placental oxidative stress: From miscarriage to preeclampsia. J Soc Gynecol Investig 2004; 11: 342–352
  • Fisher S J. The placental problem: Linking abnormal cytotrophoblast differentiation to the maternal symptoms of preeclampsia. Reprod Biol Endocrinol 2004; 2: 53
  • Lyall F. Priming and remodelling of human placental bed spiral arteries during pregnancy – a review. Placenta 2005; 26(Suppl A)S31–36
  • Espinoza J, Romero R, Kim YM, Kusanovic J P, Hassan S, Erez O, Gotsch F, Than N G, Papp Z, Kim C J. Normal and abnormal transformation of the spiral arteries during pregnancy. J Perinat Med 2006; 34: 447–458
  • Kupferminc M J, Daniel Y, Englender T, Baram A, Many A, Jaffa A J, Gull I, Lessing J B. Vascular endothelial growth factor is increased in patients with preeclampsia. Am J Reprod Immunol 1997; 38: 302–306
  • Torry D S, Wang H S, Wang T H, Caudle M R, Torry R J. Preeclampsia is associated with reduced serum levels of placenta growth factor. Am J Obstet Gynecol 1998; 179: 1539–1544
  • Tidwell S C, Ho H N, Chiu W H, Torry R J, Torry D S. Low maternal serum levels of placenta growth factor as an antecedent of clinical preeclampsia. Am J Obstet Gynecol 2001; 184: 1267–1272
  • Torry D S, Mukherjea D, Arroyo J, Torry R J. Expression and function of placenta growth factor: Implications for abnormal placentation. J Soc Gynecol Investig 2003; 10: 178–188
  • Maynard S E, Min J Y, Merchan J, Lim K H, Li J, Mondal S, Libermann T A, Morgan J P, Sellke F W, Stillman I E, 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
  • Chaiworapongsa T, Romero R, Espinoza J, Bujold E, Kim Y M, Goncalves L F, 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
  • Levine R J, Maynard S E, Qian C, Lim K H, England L J, Yu K F, Schisterman E F, Thadhani R, Sachs B P, Epstein F H, et al. Circulating angiogenic factors and the risk of preeclampsia. N Engl J Med 2004; 350: 672–683
  • Malamitsi-Puchner A, Boutsikou T, Economou E, Sarandakou A, Makrakis E, Hassiakos D, Creatsas G. Vascular endothelial growth factor and placenta growth factor in intrauterine growth-restricted fetuses and neonates. Mediators Inflamm 2005; 2005: 293–297
  • Bujold E, Romero R, Chaiworapongsa T, Kim Y M, Kim G J, Kim M R, Espinoza J, Goncalves L F, 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, Kim Y M, Kim G J, Kim M R, 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
  • Padavala S, Pope N, Baker P, Crocker I. An imbalance between vascular endothelial growth factor and its soluble receptor in placental villous explants of intrauterine growth-restricted pregnancies. J Soc Gynecol Investig 2006; 13: 40–47
  • Levine R J, Lam C, Qian C, Yu K F, Maynard S E, Sachs B P, Sibai B M, Epstein F H, Romero R, Thadhani R, et al. Soluble endoglin and other circulating antiangiogenic factors in preeclampsia. N Engl J Med 2006; 355: 992–1005
  • Crispi F, Dominguez C, Llurba E, Martin-Gallan P, Cabero L, Gratacos E. Placental angiogenic growth factors and uterine artery Doppler findings for characterization of different subsets in preeclampsia and in isolated intrauterine growth restriction. Am J Obstet Gynecol 2006; 195: 201–207
  • Venkatesha S, Toporsian M, Lam C, Hanai J, Mammoto T, Kim Y M, Bdolah Y, Lim K H, Yuan H T, Libermann T A, et al. Soluble endoglin contributes to the pathogenesis of preeclampsia. Nat Med 2006; 12: 642–649
  • Wallner W, Sengenberger R, Strick R, Strissel P L, Meurer B, Beckmann M W, Schlembach D. Angiogenic growth factors in maternal and fetal serum in pregnancies complicated by intrauterine growth restriction. Clin Sci (Lond) 2007; 112: 51–57
  • Espinoza J, Romero R, Nien J K, Gomez R, Kusanovic J P, Goncalves L F, Medina L, Edwin S, Hassan S, Carstens M, et al. Identification of patients at risk for early onset and/or severe preeclampsia with the use of uterine artery Doppler velocimetry and placental growth factor. Am J Obstet Gynecol 2007; 196: 326.e1–326.e13
  • Chaiworapongsa T, Espinoza J, Gotsch F, Kim Y M, Kim G J, Goncalves L F, Edwin S, Kusanovic J P, Erez O, Than N G, 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
  • Erez O, Romero R, Espinoza J, Fu W, Todem D, Kusanovic J P, Gotsch F, Edwin S, Nien J K, Chaiworapongsa T, et al. The change in concentrations of angiogenic and anti-angiogenic factors in maternal plasma between the first and second trimesters in risk assessment for the subsequent development of preeclampsia and small-for-gestational age. J Matern Fetal Neonatal Med 2008; 21: 279–287
  • Romero R, Nien J K, Espinoza J, Todem D, Fu W, Chung H, Kusanovic J P, Gotsch F, Erez O, Mazaki-Tovi S, et al. A longitudinal study of angiogenic (placental growth factor) and anti-angiogenic (soluble endoglin and soluble VEGF 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
  • Robertson W B, Brosens I, Dixon G. Maternal uterine vascular lesions in the hypertensive complications of pregnancy. Perspect Nephrol Hypertens 1976; 5: 115–127
  • Campbell S, Diaz-Recasens J, Griffin D R, Cohen-Overbeek T E, Pearce J M, Willson K, Teague M J. New Doppler technique for assessing uteroplacental blood flow. Lancet 1983; 1: 675–677
  • Harrington K F, Campbell S, Bewley S, Bower S. Doppler velocimetry studies of the uterine artery in the early prediction of pre-eclampsia and intra-uterine growth retardation. Eur J Obstet Gynecol Reprod Biol 1991; 42(Suppl)S14–20
  • Papageorghiou A T, Yu C K, Bindra R, Pandis G, Nicolaides K H. Multicenter screening for pre-eclampsia and fetal growth restriction by transvaginal uterine artery Doppler at 23 weeks of gestation. Ultrasound Obstet Gynecol 2001; 18: 441–449
  • Granger J P, Alexander B T, Llinas M T, Bennett W A, Khalil R A. Pathophysiology of preeclampsia: Linking placental ischemia/hypoxia with microvascular dysfunction. Microcirculation 2002; 9: 147–160
  • Sacks G P, Studena K, Sargent K, Redman C W. 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
  • Gervasi M T, Chaiworapongsa T, Pacora P, Naccasha N, Yoon B H, Maymon E, Romero R. Phenotypic and metabolic characteristics of monocytes and granulocytes in preeclampsia. Am J Obstet Gynecol 2001; 185: 792–797
  • Redman C W, Sargent I L. The pathogenesis of pre-eclampsia. Gynecol Obstet Fertil 2001; 29: 518–522
  • Chaiworapongsa T, Yoshimatsu J, Espinoza J, Kim Y M, Berman S, Edwin S, Yoon B H, Romero R. Evidence of in vivo generation of thrombin in patients with small-for-gestational-age fetuses and pre-eclampsia. J Matern Fetal Neonatal Med 2002; 11: 362–367
  • Chaiworapongsa T, Gervasi M T, 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
  • Bartha J L, Romero-Carmona R, Comino-Delgado R. Inflammatory cytokines in intrauterine growth retardation. Acta Obstet Gynecol Scand 2003; 82: 1099–1102
  • Redman C W, Sacks G P, Sargent I L. Preeclampsia: An excessive maternal inflammatory response to pregnancy. Am J Obstet Gynecol 1999; 180: 499–506
  • Kim Y M, Romero R, Oh S Y, Kim C J, Kilburn B A, Armant D R, Nien J K, Gomez R, Mazor M, Saito S, et al. Toll-like receptor 4: A potential link between “danger signals”, the innate immune system, and preeclampsia. Am J Obstet Gynecol 2005; 193: 921–927
  • Bonney E A. Preeclampsia: A view through the danger model. J Reprod Immunol 2007; 76: 68–74
  • Barondes S H, Castronovo V, Cooper D N, Cummings R D, Drickamer K, Feizi T, Gitt M A, Hirabayashi J, Hughes C, Kasai K, et al. Galectins: A family of animal beta-galactoside-binding lectins. Cell 1994; 76: 597–598
  • Leonidas D D, Elbert B L, Zhou Z, Leffler H, Ackerman S J, Acharya K R. Crystal structure of human Charcot–Leyden crystal protein, an eosinophil lysophospholipase, identifies it as a new member of the carbohydrate-binding family of galectins. Structure 1995; 3: 1379–1393
  • Visegrady B, Than N G, Kilar F, Sumegi B, Than G N, Bohn H. Homology modelling and molecular dynamics studies of human placental tissue protein 13 (galectin-13). Protein Eng 2001; 14: 875–880
  • Leffler H, Carlsson S, Hedlund M, Qian Y, Poirier F. Introduction to galectins. Glycoconj J 2004; 19: 433–440
  • Houzelstein D, Goncalves I R, Fadden A J, Sidhu S S, Cooper D N, Drickamer K, Leffler H, Poirier F. Phylogenetic analysis of the vertebrate galectin family. Mol Biol Evol 2004; 21: 1177–1187
  • Buzas E I, Gyorgy B, Pasztoi M, Jelinek I, Falus A, Gabius H J. Carbohydrate recognition systems in autoimmunity. Autoimmunity 2006; 39: 691–704
  • Lopez-Lucendo M F, Solis D, Andre S, Hirabayashi J, Kasai K, Kaltner H, Gabius H J, Romero A. Growth-regulatory human galectin-1: Crystallographic characterisation of the structural changes induced by single-site mutations and their impact on the thermodynamics of ligand binding. J Mol Biol 2004; 343: 957–970
  • Brewer F C. Binding and cross-linking properties of galectins. Biochim Biophys Acta 2002; 1572: 255–262
  • Rabinovich G A, Baum L G, Tinari N, Paganelli R, Natoli C, Liu F T, Iacobelli S. Galectins and their ligands: Amplifiers, silencers or tuners of the inflammatory response. Trends Immunol 2002; 23: 313–320
  • Elola M T, Chiesa M E, Alberti A F, Mordoh J, Fink N E. Galectin-1 receptors in different cell types. J Biomed Sci 2005; 12: 13–29
  • Kasai K, Hirabayashi J. Galectins: A family of animal lectins that decipher glycocodes. J Biochem (Tokyo) 1996; 119: 1–8
  • Villalobo A, Gabius H. Signaling pathways for transduction of the initial message of the glycocode into cellular responses. Acta Anat (Basel) 1998; 161: 110–129
  • Gabius H J, Andre S, Kaltner H, Siebert H C. The sugar code: Functional lectinomics. Biochim Biophys Acta 2002; 1572: 165–177
  • Camby I, Le Mercier M, Lefranc F, Kiss R. Galectin-1: A small protein with major functions. Glycobiology 2006; 16: 137–157
  • Hirabayashi J, Kasai K. Human placenta beta-galactoside-binding lectin. Purification and some properties. Biochem Biophys Res Commun 1984; 122: 938–944
  • Hirabayashi J, Kasai K. Complete amino acid sequence of a beta-galactoside-binding lectin from human placenta. J Biochem (Tokyo) 1988; 104: 1–4
  • Couraud P O, Casentini-Borocz D, Bringman T S, Griffith J, McGrogan M, Nedwin G E. Molecular cloning, characterization, and expression of a human 14-kDa lectin. J Biol Chem 1989; 264: 1310–1316
  • Bevan B H, Kilpatrick D C, Liston W A, Hirabayashi J, Kasai K. Immunohistochemical localization of a beta-D-galactoside-binding lectin at the human maternofetal interface. Histochem J 1994; 26: 582–586
  • Walzel H, Neels P, Bremer H, Kohler H, Raab N, Barten M, Brock J. Immunohistochemical and glycohistochemical localization of the beta-galactoside-binding S-type lectin in human placenta. Acta Histochem 1995; 97: 33–42
  • Maquoi E, van den Brule F A, Castronovo V, Foidart J M. Changes in the distribution pattern of galectin-1 and galectin-3 in human placenta correlates with the differentiation pathways of trophoblasts. Placenta 1997; 18: 433–439
  • Vicovac L, Jankovic M, Cuperlovic M. Galectin-1 and -3 in cells of the first trimester placental bed. Hum Reprod 1998; 13: 730–735
  • Jeschke U, Mayr D, Schiessl B, Mylonas I, Schulze S, Kuhn C, Friese K, Walzel H. Expression of galectin-1, -3 (gal-1, gal-3) and the Thomsen–Friedenreich (TF) antigen in normal, IUGR, preeclamptic and HELLP placentas. Placenta 2007; 28: 1165–1173
  • Dettin L, Rubinstein N, Aoki A, Rabinovich G A, Maldonado C A. Regulated expression and ultrastructural localization of galectin-1, a proapoptotic beta-galactoside-binding lectin, during spermatogenesis in rat testis. Biol Reprod 2003; 68: 51–59
  • Sakaguchi M, Shingo T, Shimazaki T, Okano H J, Shiwa M, Ishibashi S, Oguro H, Ninomiya M, Kadoya T, Horie H, et al. A carbohydrate-binding protein, galectin-1, promotes proliferation of adult neural stem cells. Proc Natl Acad Sci U S A 2006; 103: 7112–7117
  • Rubinstein N, Alvarez M, Zwirner N W, Toscano M A, Ilarregui J M, Bravo A, Mordoh J, Fainboim L, Podhajcer O L, Rabinovich G A. Targeted inhibition of galectin-1 gene expression in tumor cells results in heightened T cell-mediated rejection: A potential mechanism of tumor-immune privilege. Cancer Cell 2004; 5: 241–251
  • He J, Baum L G. Presentation of galectin-1 by extracellular matrix triggers T cell death. J Biol Chem 2004; 279: 4705–4712
  • Rabinovich G A, Sotomayor C E, Riera C M, Bianco I, Correa S G. Evidence of a role for galectin-1 in acute inflammation. Eur J Immunol 2000; 30: 1331–1339
  • Offner H, Celnik B, Bringman T S, Casentini-Borocz D, Nedwin G E, Vandenbark A A. Recombinant human beta-galactoside binding lectin suppresses clinical and histological signs of experimental autoimmune encephalomyelitis. J Neuroimmunol 1990; 28: 177–184
  • Rabinovich G A, Daly G, Dreja H, Tailor H, Riera C M, Hirabayashi J, Chernajovsky Y. Recombinant galectin-1 and its genetic delivery suppress collagen-induced arthritis via T cell apoptosis. J Exp Med 1999; 190: 385–398
  • Santucci L, Fiorucci S, Cammilleri F, Servillo G, Federici B, Morelli A. Galectin-1 exerts immunomodulatory and protective effects on concanavalin A-induced hepatitis in mice. Hepatology 2000; 31: 399–406
  • Toscano M A, Commodaro A G, Ilarregui J M, Bianco G A, Liberman A, Serra H M, Hirabayashi J, Rizzo L V, Rabinovich G A. Galectin-1 suppresses autoimmune retinal disease by promoting concomitant Th2- and T regulatory-mediated anti-inflammatory responses. J Immunol 2006; 176: 6323–6332
  • Baum L G, Blackall D P, Arias-Magallano S, Nanigian D, Uh S Y, Browne J M, Hoffmann D, Emmanouilides C E, Territo M C, Baldwin G C. Amelioration of graft versus host disease by galectin-1. Clin Immunol 2003; 109: 295–307
  • Baum L G, Seilhamer J J, Pang M, Levine W B, Beynon D, Berliner J A. Synthesis of an endogenous lectin, galectin-1, by human endothelial cells is up-regulated by endothelial cell activation. Glycoconj J 1995; 12: 63–68
  • Kim C W, Cho E H, Lee Y J, Kim Y H, Hah Y S, Kim D R. Disease-specific proteins from rheumatoid arthritis patients. J Korean Med Sci 2006; 21: 478–484
  • Rabinovich G A, Iglesias M M, Modesti N M, Castagna L F, Wolfenstein-Todel C, Riera C M, Sotomayor C E. Activated rat macrophages produce a galectin-1-like protein that induces apoptosis of T cells: Biochemical and functional characterization. J Immunol 1998; 160: 4831–4840
  • Zuniga E, Gruppi A, Hirabayashi J, Kasai K I, Rabinovich G A. Regulated expression and effect of galectin-1 on Trypanosoma cruzi-infected macrophages: Modulation of microbicidal activity and survival. Infect Immun 2001; 69: 6804–6812
  • Garin M I, Chu C C, Golshayan D, Cernuda-Morollon E, Wait R, Lechler R I. Galectin-1: A key effector of regulation mediated by CD4+CD25+ T cells. Blood 2007; 109: 2058–2065
  • Gil C D, Cooper D, Rosignoli G, Perretti M, Oliani S M. Inflammation-induced modulation of cellular galectin-1 and -3 expression in a model of rat peritonitis. Inflamm Res 2006; 55: 99–107
  • Rabinovich G A, Gruppi A. Galectins as immunoregulators during infectious processes: From microbial invasion to the resolution of the disease. Parasite Immunol 2005; 27: 103–114
  • Sibai B M, Ewell M, Levine R J, Klebanoff M A, Esterlitz J, Catalano P M, Goldenberg R L, Joffe G. Risk factors associated with preeclampsia in healthy nulliparous women. The Calcium for Preeclampsia Prevention (CPEP) Study Group. Am J Obstet Gynecol 1997; 177: 1003–1010
  • Report of the National High Blood Pressure Education Program Working Group on High Blood Pressure in Pregnancy. Am J Obstet Gynecol 2000; 183: S1–22
  • Alexander G R, Himes J H, Kaufman R B, Mor J, Kogan M. A United States national reference for fetal growth. Obstet Gynecol 1996; 87: 163–168
  • Fu W J, Hu J, Spencer T, Carroll R, Wu G. Statistical models in assessing fold change of gene expression in real-time RT-PCR experiments. Comput Biol Chem 2006; 30: 21–26
  • Bonferroni C E. Il calcolo delle assicurazioni su gruppi di teste. Studi in Onore del Professore Salvatore Orto Carboni. Rome 1935; 13–60
  • Redline R W, Boyd T, Campbell V, Hyde S, Kaplan C, Khong T Y, Prashner H R, Waters B L. Maternal vascular underperfusion: Nosology and reproducibility of placental reaction patterns. Pediatr Dev Pathol 2004; 7: 237–249
  • Redline R W, Heller D, Keating S, Kingdom J. Placental diagnostic criteria and clinical correlation—a workshop report. Placenta 2005; 26(Suppl A)S114–117
  • Ozeki Y, Matsui T, Yamamoto Y, Funahashi M, Hamako J, Titani K. Tissue fibronectin is an endogenous ligand for galectin-1. Glycobiology 1995; 5: 255–261
  • Phillips B, Knisley K, Weitlauf K D, Dorsett J, Lee V, Weitlauf H. Differential expression of two beta-galactoside-binding lectins in the reproductive tracts of pregnant mice. Biol Reprod 1996; 55: 548–558
  • Iglesias M M, Rabinovich G A, Ivanovic V, Sotomayor C, Wolfenstein-Todel C. Galectin-1 from ovine placenta—amino-acid sequence, physicochemical properties and implications in T-cell death. Eur J Biochem 1998; 252: 400–407
  • Prior I A, Muncke C, Parton R G, Hancock J F. Direct visualization of Ras proteins in spatially distinct cell surface microdomains. J Cell Biol 2003; 160: 165–170
  • Nickel W. Unconventional secretory routes: Direct protein export across the plasma membrane of mammalian cells. Traffic 2005; 6: 607–614
  • Jeschke U, Karsten U, Wiest I, Schulze S, Kuhn C, Friese K, Walzel H. Binding of galectin-1 (gal-1) to the Thomsen–Friedenreich (TF) antigen on trophoblast cells and inhibition of proliferation of trophoblast tumor cells in vitro by gal-1 or an anti-TF antibody. Histochem Cell Biol 2006; 126: 437–444
  • Moiseeva E P, Williams B, Goodall A H, Samani N J. Galectin-1 interacts with beta-1 subunit of integrin. Biochem Biophys Res Commun 2003; 310: 1010–1016
  • Peters D G, Kassam A B, Feingold E, Heidrich-O’Hare E, Yonas H, Ferrell R E, Brufsky A. Molecular anatomy of an intracranial aneurysm: Coordinated expression of genes involved in wound healing and tissue remodeling. Stroke 2001; 32: 1036–1042
  • Lotan R, Belloni P N, Tressler R J, Lotan D, Xu X C, Nicolson G L. Expression of galectins on microvessel endothelial cells and their involvement in tumour cell adhesion. Glycoconj J 1994; 11: 462–468
  • Perillo N L, Pace K E, Seilhamer J J, Baum L G. Apoptosis of T cells mediated by galectin-1. Nature 1995; 378: 736–739
  • La M, Cao T V, Cerchiaro G, Chilton K, Hirabayashi J, Kasai K, Oliani S M, Chernajovsky Y, Perretti M. A novel biological activity for galectin-1: Inhibition of leukocyte–endothelial cell interactions in experimental inflammation. Am J Pathol 2003; 163: 1505–1515
  • Danese S, Dejana E, Fiocchi C. Immune regulation by microvascular endothelial cells: Directing innate and adaptive immunity, coagulation, and inflammation. J Immunol 2007; 178: 6017–6022
  • He J, Baum L G. Endothelial cell expression of galectin-1 induced by prostate cancer cells inhibits T-cell transendothelial migration. Lab Invest 2006; 86: 578–590
  • Frauli M, Ludwig H. Demonstration of the ability of Hofbauer cells to phagocytose exogenous antibodies. Eur J Obstet Gynecol Reprod Biol 1987; 26: 135–144
  • Zaccheo D, Pistoia V, Castellucci M, Martinoli C. Isolation and characterization of Hofbauer cells from human placental villi. Arch Gynecol Obstet 1989; 246: 189–200
  • Matsubara S, Takayama T, Yamada T, Usui R, Izumi A, Watanabe T, Ohkuchi A, Shibahara H, Sato I, Suzuki M. Hofbauer cell activation and its increased glucose-6-phosphate dehydrogenase activity in second trimester-spontaneous abortion: An ultrastructural dual staining enzyme-cytochemical study. Am J Reprod Immunol 2003; 49: 202–209
  • Taniguchi T, Matsuzaki N, Kameda T, Shimoya K, Jo T, Saji F, Tanizawa O. The enhanced production of placental interleukin-1 during labor and intrauterine infection. Am J Obstet Gynecol 1991; 165: 131–137
  • Saito S, Kasahara T, Sakakura S, Umekage H, Harada N, Ichijo M. Detection and localization of interleukin-8 mRNA and protein in human placenta and decidual tissues. J Reprod Immunol 1994; 27: 161–172
  • Blumenstein M, Keelan J A, Bowen-Shauver J M, Mitchell M D. Suppressors of cytokine signaling proteins in human preterm placental tissues. J Mol Endocrinol 2005; 35: 165–175
  • Moussa M, Mognetti B, Dubanchet S, Menu E, Roques P, Dormont D, Barre-Sinoussi F, Chaouat G. Expression of beta chemokines in explants and trophoblasts from early and term human placentae. Am J Reprod Immunol 2001; 46: 309–317
  • Correa S G, Sotomayor C E, Aoki M P, Maldonado C A, Rabinovich G A. Opposite effects of galectin-1 on alternative metabolic pathways of L-arginine in resident, inflammatory, and activated macrophages. Glycobiology 2003; 13: 119–128
  • Barrionuevo P, Beigier-Bompadre M, Ilarregui J M, Toscano M A, Bianco G A, Isturiz M A, Rabinovich G A. A novel function for galectin-1 at the crossroad of innate and adaptive immunity: Galectin-1 regulates monocyte/macrophage physiology through a nonapoptotic ERK-dependent pathway. J Immunol 2007; 178: 436–445
  • Than G N, Bohn H, Szabo D G. Advances in pregnancy-related protein research. CRC Press, Boca Raton, FL 1993; 1–333
  • Chaouat G, Ledee-Bataille N, Zourbas S, Ostojic S, Dubanchet S, Martal J, Frydman R. Cytokines, implantation and early abortion: Re-examining the Th1/Th2 paradigm leads to question the single pathway, single therapy concept. Am J Reprod Immunol 2003; 50: 177–186
  • Keelan J A, Mitchell M D. Placental cytokines and preeclampsia. Front Biosci 2007; 12: 2706–2727
  • Hunt J S, Vassmer D, Ferguson T A, Miller L. Fas ligand is positioned in mouse uterus and placenta to prevent trafficking of activated leukocytes between the mother and the conceptus. J Immunol 1997; 158: 4122–4128
  • Munn D H, Zhou M, Attwood J T, Bondarev I, Conway S J, Marshall B, Brown C, Mellor A L. Prevention of allogeneic fetal rejection by tryptophan catabolism. Science 1998; 281: 1191–1193
  • Phillips T A, Ni J, Pan G, Ruben S M, Wei Y F, Pace J L, Hunt J S. TRAIL (Apo-2L) and TRAIL receptors in human placentas: Implications for immune privilege. J Immunol 1999; 162: 6053–6059
  • Kayisli U A, Selam B, Guzeloglu-Kayisli O, Demir R, Arici A. Human chorionic gonadotropin contributes to maternal immunotolerance and endometrial apoptosis by regulating Fas–Fas ligand system. J Immunol 2003; 171: 2305–2313
  • Niederkorn J Y. See no evil, hear no evil, do no evil: The lessons of immune privilege. Nat Immunol 2006; 7: 354–359
  • Lei Z M, Yang M, Li X, Takikawa O, Rao C V. Upregulation of placental indoleamine 2,3-dioxygenase by human chorionic gonadotropin. Biol Reprod 2007; 76: 639–644
  • Rabinovich G A, Ariel A, Hershkovitz R, Hirabayashi J, Kasai K I, Lider O. Specific inhibition of T-cell adhesion to extracellular matrix and proinflammatory cytokine secretion by human recombinant galectin-1. Immunology 1999; 97: 100–106
  • van der Leij J, van den Berg A, Harms G, Eschbach H, Vos H, Zwiers P, van Weeghel R, Groen H, Poppema S, Visser L. Strongly enhanced IL-10 production using stable galectin-1 homodimers. Mol Immunol 2007; 44: 506–513
  • Rorive S, Belot N, Decaestecker C, Lefranc F, Gordower L, Micik S, Maurage C A, Kaltner H, Ruchoux M M, Danguy A, et al. Galectin-1 is highly expressed in human gliomas with relevance for modulation of invasion of tumor astrocytes into the brain parenchyma. Glia 2001; 33: 241–255
  • Camby I, Belot N, Rorive S, Lefranc F, Maurage C A, Lahm H, Kaltner H, Hadari Y, Ruchoux M M, Brotchi J, et al. Galectins are differentially expressed in supratentorial pilocytic astrocytomas, astrocytomas, anaplastic astrocytomas and glioblastomas, and significantly modulate tumor astrocyte migration. Brain Pathol 2001; 11: 12–26
  • Le Q T, Shi G, Cao H, Nelson D W, Wang Y, Chen E Y, Zhao S, Kong C, Richardson D, O'Byrne K J, et al. Galectin-1: A link between tumor hypoxia and tumor immune privilege. J Clin Oncol 2005; 23: 8932–8941
  • Saussez S, Lorfevre F, Lequeux T, Laurent G, Chantrain G, Vertongen F, Toubeau G, Decaestecker C, Kiss R. The determination of the levels of circulating galectin-1 and -3 in HNSCC patients could be used to monitor tumor progression and/or responses to therapy. Oral Oncol 2008; 44: 86–93
  • Daroqui C M, Ilarregui J M, Rubinstein N, Salatino M, Toscano M A, Vazquez P, Bakin A, Puricelli L, Bal de Kier J E, Rabinovich G A. Regulation of galectin-1 expression by transforming growth factor beta1 in metastatic mammary adenocarcinoma cells: Implications for tumor-immune escape. Cancer Immunol Immunother 2007; 56: 491–499
  • Koopman L A, Kopcow H D, Rybalov B, Boyson J E, Orange J S, Schatz F, Masch R, Lockwood C J, Schachter A D, Park P J, et al. Human decidual natural killer cells are a unique NK cell subset with immunomodulatory potential. J Exp Med 2003; 198: 1201–1212
  • Aluvihare V R, Kallikourdis M, Betz A G. Regulatory T cells mediate maternal tolerance to the fetus. Nat Immunol 2004; 5: 266–271
  • Trowsdale J, Betz A G. Mother's little helpers: Mechanisms of maternal–fetal tolerance. Nat Immunol 2006; 7: 241–246
  • Blois S M, Ilarregui J M, Tometten M, Garcia M, Orsal A S, Cordo-Russo R, Toscano M A, Bianco G A, Kobelt P, Handjiski B, et al. A pivotal role for galectin-1 in fetomaternal tolerance. Nat Med 2007; 13: 1450–1457
  • Than N G, Wildman D E, Erez O, Edwin S S, Espinoza J, Kim C J, Han Y M, Mazaki-Tovi S, Kusanovic J P, Hassan S, et al. Trophoblast, galectin-1 and pre-eclampsia. Am J Obstet Gynecol 2006; 195: S138
  • Wilczynski J R. Immunological analogy between allograft rejection, recurrent abortion and pre-eclampsia—the same basic mechanism. Hum Immunol 2006; 67: 492–511
  • van der Leij J, van den Berg A, Blokzijl T, Harms G, van Goor H, Zwiers P, van Weeghel R, Poppema S, Visser L. Dimeric galectin-1 induces IL-10 production in T-lymphocytes: An important tool in the regulation of the immune response. J Pathol 2004; 204: 511–518
  • Fuertes M B, Molinero L L, Toscano M A, Ilarregui J M, Rubinstein N, Fainboim L, Zwirner N W, Rabinovich G A. Regulated expression of galectin-1 during T-cell activation involves Lck and Fyn kinases and signaling through MEK1/ERK, p38 MAP kinase and p70S6 kinase. Mol Cell Biochem 2004; 267: 177–185
  • Keelan J A, Marvin K W, Sato T A, Coleman M, McCowan L M, Mitchell M D. Cytokine abundance in placental tissues: Evidence of inflammatory activation in gestational membranes with term and preterm parturition. Am J Obstet Gynecol 1999; 181: 1530–1536
  • Starzl T E, Zinkernagel R M. Antigen localization and migration in immunity and tolerance. N Engl J Med 1998; 339: 1905–1913
  • Matzinger P. The danger model: A renewed sense of self. Science 2002; 296: 301–305
  • Sato S. Galectins as molecules of danger signal, which could evoke an immune response to infection. Trends Glycosci Glyc 2002; 14: 285–301
  • Moffett A, Loke C. Immunology of placentation in eutherian mammals. Nat Rev Immunol 2006; 6: 584–594

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.