2,608
Views
98
CrossRef citations to date
0
Altmetric
Original Article

CXCL10/IP-10: A missing link between inflammation and anti-angiogenesis in preeclampsia?

, , MD, , , , , , , , , , , & show all
Pages 777-792 | Received 29 Jan 2007, Accepted 02 May 2007, Published online: 07 Jul 2009

Abstract

Objective. Interferon (IFN)-γ inducible protein, CXCL10/IP-10, is a member of the CXC chemokine family with pro-inflammatory and anti-angiogenic properties. This chemokine has been proposed to be a key link between inflammation and angiogenesis. The aim of this study was to determine whether preeclampsia and delivery of a small for gestational age (SGA) neonate are associated with changes in maternal serum concentration of CXCL10/IP-10.

Study design. This cross-sectional study included patients in the following groups: (1) non-pregnant women (N = 49); (2) women with normal pregnancies (N = 89); (3) patients with preeclampsia (N = 100); and (4) patients who delivered an SGA neonate (N = 78). SGA was defined as birth weight below the 10th percentile. Maternal serum concentrations of CXCL10/IP-10 were measured by sensitive immunoassay. Non-parametric statistics were used for analysis.

Results. (1) Patients with normal pregnancies had a significantly higher median serum concentration of CXCL10/IP-10 than non-pregnant women (median 116.1 pg/mL, range 40.7–1314.3 vs. median 90.3 pg/mL, range 49.2–214.7, respectively; p = 0.002); (2) no significant correlation was found between maternal serum concentration of CXCL10/IP-10 and gestational age (between 19 and 38 weeks); (3) there were no differences in median serum CXCL10/IP-10 concentrations between patients who delivered an SGA neonate and those with normal pregnancies (median 122.4 pg/mL, range 37.3–693.5 vs. median 116.1 pg/mL, range 40.7–1314.3, respectively; p > 0.05); (4) patients with preeclampsia had a higher median serum concentration of CXCL10/IP-10 than normal pregnant women (median 156.4 pg/mL, range 47.4–645.9 vs. median 116.1 pg/mL, range 40.7–1314.3, respectively; p < 0.05); (5) patients with preeclampsia had a higher median concentration of CXCL10/IP-10 than those who delivered an SGA neonate (median 156.4 pg/mL, range 47.4–645.9 vs. median 122.4 pg/mL, range 37.3–693.5, respectively; p < 0.05).

Conclusions. Patients with preeclampsia have significantly higher serum concentrations of CXCL10/IP-10 than both normal pregnant women and mothers who have SGA neonates. These results are likely to reflect an anti-angiogenic state as well as an enhanced systemic inflammatory response in patients with preeclampsia. Alternatively, since preeclampsia and SGA share several mechanisms of disease, it is possible that a higher concentration of this chemokine may contribute to the clinical presentation of preeclampsia in patients with a similar intrauterine insult.

Introduction

Patients with preeclampsia and those with a small for gestational age (SGA) neonate share a number of pathophysiological characteristics including: (1) abnormal physiologic transformation of the spiral arteries Citation[1-8]; (2) chronic uteroplacental ischemia Citation[9-16]; (3) endothelial cell dysfunction Citation[17-24]; (4) an anti-angiogenic state Citation[25-32]; and (5) intravascular inflammation Citation[18],Citation[33-37]. Furthermore, a biased Th1/Th2 (T-helper 1/T-helper 2) balance towards a Th1 response has been reported in preeclampsia Citation[38-46] and SGA Citation[47].

The human interferon-inducible protein 10 (IP-10 or CXCL10) is a chemokine of the CXC family Citation[48]. A unique feature of members of this chemokine family is that they have pro-inflammatory properties and act as modulators of angiogenesis in conditions such as wound healing, ischemia, and neoplasia. These dual properties are related to the shared expression of specific chemokine receptors by leukocytes and endothelial cells Citation[49-60].

IP-10 is inducible by pro-inflammatory stimuli such as interferon-γ (IFN-γ) Citation[61-72], tumor necrosis factor-α (TNF-α) Citation[70],Citation[73-79], viruses, and microbial products Citation[66],Citation[70],Citation[80-85], directly or through activation of nuclear factor-kappa B (NF-kB) Citation[81],Citation[82],Citation[86-89]. It has been proposed that this chemokine is also involved in recruitment and potentiation of Th1 responses as well as in the pathogenesis of allograft rejection Citation[90-103], multiple sclerosis Citation[104-108], diabetes mellitus type 1 Citation[109],Citation[110], Graves' disease Citation[111-114], autoimmune thyroiditis Citation[115],Citation[116], pulmonary fibrosis Citation[117-119], and cardiovascular diseases such as atherosclerosis Citation[120] and coronary syndromes Citation[121],Citation[122]. Importantly, IP-10 has potent anti-angiogenic activity, both in vitro and in vivoCitation[123-126].

The balance between angiogenic and anti-angiogenic (angiostatic) factors controlled by molecules involved in inflammatory processes may have an impact on the pathogenesis of many diseases Citation[54],Citation[58].

We propose that IP-10 is involved in the pathophysiology of preeclampsia because this condition is characterized by intravascular inflammation and an anti-angiogenic state. The objective of this study was to compare the maternal serum IP-10 concentrations in normal pregnancy, preeclampsia, and SGA.

Methods

Study design

This retrospective cross-sectional study included patients with preeclampsia (N = 100), women who delivered an SGA neonate (N = 78), normal pregnant women (N = 89), and non-pregnant women (N = 49). All patients were enrolled at Hutzel Hospital, Detroit, MI, USA. All women provided written informed consent for the collection of clinical data and biological materials under protocols approved by the Institutional Review Boards of both Wayne State University and the National Institute of Child Health and Human Development of the National Institute of Health (NIH/DHHS). Many of these samples have been previously employed to study the biology of inflammation, hemostasis, angiogenesis regulation, and growth factor concentrations in non-pregnant women, normal pregnant women, and those with pregnancy complications.

Preeclampsia was defined in the presence of hypertension (systolic blood pressure ≥140 mmHg or diastolic blood pressure ≥90 mmHg on at least two occasions, 4 hours to 1 week apart, after the 20th week of gestation) and proteinuria (≥300 mg in a 24-hour urine collection, or two random urine specimens obtained 4 hours to 1 week apart containing ≥1+ protein by dipstick Citation[127],Citation[128] or one dipstick measurement ≥2+) Citation[129]. Severe preeclampsia was diagnosed according to the criteria proposed by the American College of Obstetricians and Gynecologists (ACOG) committee Citation[128]. Patients with preeclampsia were sub-classified as either early-onset (<34 weeks) or late onset (≥34 weeks) disease according to the gestational age at which preeclampsia was diagnosed. A neonate was defined as SGA when the birth weight was below the 10th percentile for gestational age according to the reference range proposed by Alexander et al. Citation[130]. Patients were considered to have a normal pregnancy if they met the following criteria: (1) no medical, obstetrical, or surgical complications; (2) absence of labor at the time of venipuncture; and (3) delivery of a normal term (≥37 weeks) infant whose birth weight was between the 10th and 90th percentile for gestational age Citation[130]. The non-pregnant group consisted of healthy volunteers not taking oral contraceptives whose blood was drawn in the secretory phase of the menstrual cycle.

IP-10 (CXCL10) determinations

Specific and sensitive enzyme-linked immunoassays were used to determine concentrations of IP-10 in human maternal serum. Immunoassays for IP-10 were obtained from R&D Systems (Minneapolis, MN, USA). Briefly, maternal serum samples were incubated in duplicate wells of the microtiter plates, pre-coated with a monoclonal antibody specific for IP-10. During this incubation step, any IP-10 present in the standards or maternal serum is bound by the immobilized antibodies. After repeated washing and aspiration to remove all unbound substances, an enzyme-linked polyclonal antibody specific for IP-10 was added to the wells. Following a wash to remove excess and unbound materials, a substrate solution was added to the wells and color developed in proportion to the amount of IP-10 bound in the initial step. The color development was stopped with the addition of an acid solution and the intensity of color was read using a programmable spectrophotometer (SpectraMax M2, Molecular Devices, Sunnyvale, CA, USA). The concentrations of IP-10 in serum samples were determined by interpolation from individual standard curves composed of recombinant human IP-10. The calculated inter- and intra-assay coefficients of variation for IP-10 immunoassays in our laboratory were 7.99% and 4.12%, respectively. The lower limit of detection (sensitivity) was calculated to be 5.01 pg/mL.

Statistical analysis

The Kolmogorov–Smirnov test was used to determine whether the data were normally distributed. The Spearman's correlation test was used in assessing the relationship between maternal serum concentration of IP-10 and gestational age at blood draw in patients with normal pregnancies. Comparisons among groups were performed using Kruskal–Wallis tests with post-hoc analysis for continuous variables, and Chi-square or Fisher's exact test for categorical variables. A p value <0.05 was considered statistically significant. The statistical package used was SPSS v.12.0 (SPSS Inc., Chicago, IL, USA).

Results

Three hundred sixteen patients were included in this study. The demographic and clinical characteristics of the study groups are displayed in .

Table I.  Clinical and obstetrical characteristics of the study groups.

Among patients with preeclampsia, 63% (63/100) were classified as early-onset and 88% (88/100) as severe preeclampsia. In 75.6% (59/78) of patients who delivered an SGA neonate, the birth weight was below the 5th percentile. IP-10 was detectable in the serum of all subjects.

Serum concentration of IP-10 in pregnancy

Patients with normal pregnancies had a significantly higher median serum concentration of IP-10 than non-pregnant women (median 116.1 pg/mL, range 40.7–1314.3 vs. median 90.3 pg/mL, range 49.2–214.7, respectively; p = 0.002) (see ). There was no significant correlation between maternal serum IP-10 concentration and gestational age at blood draw (r = 0.039; p = 0.7).

Figure 1. Serum concentrations of CXCL10/IP-10 in non-pregnant women and in patients with normal pregnancies. Patients with normal pregnancies had a significantly higher median serum concentration of IP-10 than non-pregnant women (median 116.1 pg/mL, range 40.7–1314.3 vs. median 90.3 pg/mL, range 49.2–214.7, respectively; p = 0.002).

Figure 1. Serum concentrations of CXCL10/IP-10 in non-pregnant women and in patients with normal pregnancies. Patients with normal pregnancies had a significantly higher median serum concentration of IP-10 than non-pregnant women (median 116.1 pg/mL, range 40.7–1314.3 vs. median 90.3 pg/mL, range 49.2–214.7, respectively; p = 0.002).

IP-10 in preeclampsia

Patients with preeclampsia had a higher median serum concentration of IP-10 than normal pregnant women and those who delivered an SGA neonate (preeclampsia: median 156.4 pg/mL, range 47.4–645.9; normal pregnancy: median 116.1 pg/mL, range 40.7–1314.3; SGA: median 122.4 pg/mL, range 37.3–693.5; p < 0.05 for both comparisons). In contrast, there were no significant differences in the maternal serum median IP-10 concentrations between patients who delivered an SGA neonate and those with normal pregnancies (SGA: median 122.4 pg/mL, range 37.3–693.5 vs. normal pregnancy: median 116.1 pg/mL, range 40.7–1314.3; p > 0.05) (see ).

Figure 2. Maternal serum concentrations of CXCL10/IP-10 among the study groups. Patients with preeclampsia had a significantly higher median serum concentration of IP-10 than normal pregnant women (median 156.4 pg/mL, range 47.4–645.9 vs. median 116.1 pg/mL, range 40.7–1314.3, respectively; p < 0.05) and than patients who delivered a small for gestational age (SGA) neonate (median 156.4 pg/mL, range 47.4–645.9 vs. median 122.4 pg/mL, range 37.3–693.5, respectively; p < 0.05). No significant differences were found in maternal serum median IP-10 concentrations between patients who delivered an SGA neonate and those with normal pregnancies (median 122.4 pg/mL, range 37.3–693.5 vs. median 116.1 pg/mL, range 40.7–1314.3, respectively; p > 0.05).

Figure 2. Maternal serum concentrations of CXCL10/IP-10 among the study groups. Patients with preeclampsia had a significantly higher median serum concentration of IP-10 than normal pregnant women (median 156.4 pg/mL, range 47.4–645.9 vs. median 116.1 pg/mL, range 40.7–1314.3, respectively; p < 0.05) and than patients who delivered a small for gestational age (SGA) neonate (median 156.4 pg/mL, range 47.4–645.9 vs. median 122.4 pg/mL, range 37.3–693.5, respectively; p < 0.05). No significant differences were found in maternal serum median IP-10 concentrations between patients who delivered an SGA neonate and those with normal pregnancies (median 122.4 pg/mL, range 37.3–693.5 vs. median 116.1 pg/mL, range 40.7–1314.3, respectively; p > 0.05).

Among patients with preeclampsia, no significant differences in serum concentrations of IP-10 were observed between patients with early onset and late onset disease (early onset preeclampsia: median 155.6 pg/mL, range 47.5–645.9 vs. late onset preeclampsia: median 165.2 pg/mL, range 64.7–401.1; p = 0.4). Similarly, there were no significant differences between mild and severe preeclampsia (mild preeclampsia: median 177.7 pg/mL, range 79.4–401.1 vs. severe preeclampsia: median 155.5 pg/mL, range 47.5–645.9; p = 0.56).

Among patients who delivered an SGA neonate, there were no significant differences in serum concentrations of IP-10 between patients who delivered a neonate with a birth weight at <5th percentile and those who delivered a neonate with a birth weight between the 5th and 9th percentile (SGA <5th percentile: median 126.7 pg/mL, range 37.3–693.5 vs. SGA 5th–9th percentile: median 108.7 pg/mL, range 40.7–349.5; p = 0.2).

Using analysis of covariance (ANCOVA), only diagnostic groups but not storage time had a significant effect on the serum concentrations of CXCL10 (storage time: p = 0.5; diagnostic groups: p < 0.001). Storage time did not have a significant effect on the serum concentrations of CXCL10 even when gestational age at blood draw was included in the analysis (storage time: p = 0.72; diagnostic groups: p = 0.02; gestational age at blood draw: p = 0.5).

Discussion

Principal findings of this study

(1) The median serum concentration of IP-10 in normal pregnancy was higher than that of non-pregnant women; (2) there was no relationship between gestational age and the maternal serum concentration of IP-10; (3) preeclampsia, but not SGA, was associated with a higher median concentration of maternal serum IP-10. These results are novel and suggest that IP-10 may participate in the pathophysiology of preeclampsia.

What is CXCL10/IP-10?

IP-10 (CXCL10) is a chemokine of the CXC family Citation[48], which was first described as the product of a gene induced in response to recombinant IFN-γ in several cell populations, including U937 histiocytic lymphoma, human fibroblasts, mononuclear and endothelial cells Citation[61]. The principal biological activity of chemotactic cytokines, such as IP-10, is regulation and control of the basal homeostatic and inflammatory leukocyte movement Citation[57].

In addition, IP-10 has potent anti-angiogenic properties Citation[54],Citation[56],Citation[57],Citation[59], promotes adhesion, migration, and invasion of trophoblast cell Citation[131],Citation[132], has an inhibitory effect on early hematopoietic progenitors Citation[133], and regulates intestinal crypt cell renewal in both physiologic conditions and during mucosal regeneration following injury Citation[134].

The biological properties of IP-10 are mediated through the interaction with a transmembrane G protein-coupled receptor, CXCR3 Citation[135],Citation[136] shared by two other IFN-γ inducible CXC chemokines—CXCL9 (MIG) and CXCL11 (I-TAC)—whose distinct biological activities are related to different transduction pathways Citation[137-140].

Factors controlling the expression of IP-10 and cell sources

IP-10 gene and protein expression is modulated by pro-inflammatory stimuli. Indeed, IFN-γ is an inducer of the gene, and protein expression of this chemokine by mononuclear cells Citation[61], neutrophils Citation[69],Citation[70], eosinophils Citation[70],Citation[141], keratinocytes Citation[61],Citation[64],Citation[65],Citation[67], fibroblasts Citation[61], endothelial cells Citation[61-63], pancreatic β cells Citation[71],Citation[72], and animal astrocytes/microglia Citation[66],Citation[68], TNF-αCitation[70],Citation[73-79], interleukin (IL)-1βCitation[70],Citation[71], as well as viral Citation[66],Citation[81],Citation[82] and microbial products Citation[70],Citation[80],Citation[83-85], can also stimulate the production of IL-10.

Activation of the nuclear factor-kappa B (NF-kB) pathway Citation[81],Citation[82],Citation[86-89] through ligation of pattern recognition receptors (TLR4 Citation[83] and TLR3 Citation[89]) can also upregulate gene and protein expression of IP-10. Interestingly, IP-10 is considered an ‘NF-kB responsive gene’Citation[142].

IP-10 in inflammation

The pro-inflammatory activity of IP-10 includes: chemotaxis and endothelial adhesion of activated T cells Citation[143] as well as chemotaxis Citation[144] and enhancement of natural killer (NK) cell-mediated cytolysis Citation[144]. However, this chemokine is a poor neutrophil activator Citation[143],Citation[145] and there is controversy about its effects on both monocytes Citation[135],Citation[143] and B cells Citation[146],Citation[147].

CXCR3 receptor expression on T lymphocytes is selective for activated cells Citation[135],Citation[143],Citation[146],Citation[148-151]. Interestingly, analysis of polarized T lymphocytes using specific monoclonal antibodies has demonstrated high CXCR3 expression (mRNA and protein) on Th1 cells and low on Th2 cells Citation[149],Citation[152], and this receptor has, therefore, been proposed as a useful clinical marker of circulating Th1-type cells Citation[149],Citation[153]. Further evidence that IP-10 promotes a Th1-like dominance is the observation that, following stimulation with phorbol-12-myristate 13-acetate (PMA) and ionomycin, the production of IFN-γ (a Th1-type chemokine) resides exclusively in CXCR3-expressing memory CD4+ T cells, whereas the production of Th2-type cytokines is mainly observed in those CXCR3-negative Citation[153]. In addition, in vitro assays show that, in polyclonally stimulated T cells, recombinant IP-10 selectively enhances IFN-γ protein synthesis, while having no effect on IL-4 production Citation[154]. The capability of IP-10 to enhance its own inducer, IFN-γ supports the hypothesis that a positive amplification loop between IP-10 and IFN-γ also exists in vivoCitation[154].

IP-10 in pathologic states

IP-10 has been implicated in states characterized by a prominent T cell response Citation[155],Citation[156], particularly when a Th1/Th2 imbalance is involved, including: (1) multiple sclerosis, where the serum and cerebrospinal fluid concentrations of IP10 correlate with the disease activity and CXCR3 expression is detectable on the majority of CNS-infiltrating lymphocytes Citation[104-108]; (2) herpetic encephalitis in mice Citation[157],Citation[158]; (3) experimental autoimmune encephalomyelitis Citation[159-162]; (4) inflammatory bowel/colon disease Citation[134],Citation[163],Citation[164]; (5) chronic hepatitis C, in which IP-10 serum concentrations are related to the inflammatory activity and the response to therapy Citation[165]; (6) Sjogren's syndrome, where the expression of IP-10 mRNA is significantly up-regulated in salivary glands (p < 0.01) and IP-10 has a potential role in the accumulation of T cells infiltrates Citation[166]; (7) type 1 diabetes, whose immunopathogenesis is likely to be linked to the IP-10 Citation[109],Citation[110] property of enhancing the traffic of auto-aggressive cells to the pancreas Citation[167] and imprinting a pattern for the subsequent development of the autoimmune disease Citation[109]; (8) Graves' disease Citation[111-114] and autoimmune thyroiditis Citation[115],Citation[116] where high concentrations of IP-10 have been detected in the serum of affected individuals; and (9) systemic lupus erythematosus, where IP-10 plasma concentrations not only are higher than in non-diseased individuals but also correlate with the disease activity Citation[168].

IP-10 as an inhibitor of angiogenesis

IP-10 has potent anti-angiogenic properties Citation[54],Citation[56],Citation[57],Citation[59]. Several mechanisms have been proposed to mediate these activities: (1) interaction with the CXCR3 receptor Citation[169],Citation[170]; (2) binding to a cell surface heparan sulfate site shared with platelet factor 4 Citation[124]; (3) interference with the pro-angiogenic activity of basic fibroblast growth factor (bFGF) and IL-8 Citation[124]; and (4) through another high affinity receptor (different from CXCR3 and glycosaminoglycans) Citation[171].

The IP-10 receptor CXCR3 (mRNA and protein) has been localized on the endothelial cells of several human tissues (kidney, gut, liver, thyroid, and thymus) Citation[170],Citation[172]. Interestingly, the endothelial cell expression is cell cycle-dependent Citation[170]. Indeed, staining is remarkably more frequent in conditions of activation and in the presence of a high proliferative rate (such as in inflamed and neoplastic tissues rather than in normal tissues), particularly during the S/G2-M phase of the endothelial cell cycle Citation[170]. Romagnani et al. Citation[170] suggested that these results represent evidence that CXCR3 is mediating the angiostatic activity of IP-10, thus controlling endothelial cell proliferation. In addition, two distinct isoforms of this receptor, CXCR3-A and CXCR3-B (both products of alternative splicing) are likely to mediate opposite effects on cellular proliferation Citation[173],Citation[174].

A specific region of the IP-10 molecule has been linked to its anti-angiogenic effect. Indeed, an aminoterminal truncated form of IP-10, resulting from a post-translational processing, has impaired receptor signaling and lymphocyte chemotaxis, but retains anti-angiogenic properties Citation[175].

Evidence in support of the anti-angiogenic role of IP-10 includes: (1) IP-10, added to human umbilical cord vein endothelial cells (HUVECs), cultured on a matrigel substrate, inhibits their differentiation into tube-like structures in a dose-dependent fashion Citation[125] and reduces the extent of the neo-vascular network Citation[126]; (2) IP-10, both in vitroCitation[123],Citation[124] and in vivoCitation[123],Citation[125] inhibits in a dose-dependent manner the pro-angiogenic effects of IL-8 and bFGF, including endothelial cell chemotaxis and proliferation as well as neovascularization in animal models of angiogenesis (rat cornea Citation[123] or matrigel injected in the subcutaneous tissue of nude mice Citation[125]); (3) recombinant IP-10 can inhibit [3H]-thymidine incorporation into HUVECs cultured with bFGF Citation[124]. Further support of the anti-angiogenic properties of IP-10 is the role attributed to this chemokine in the pathogenesis of pulmonary fibrosis. Indeed, the lower production of IP-10 in lung tissue of patients with pulmonary fibrosis, as determined by ELISA, has been proposed to contribute to the greater angiogenic activity observed in these patients Citation[117],Citation[119]. Additionally, when IP-10 is administered systemically to bleomycin pre-treated mice, it inhibits fibroplasia and deposition of extracellular matrix through the regulation of the local angiogenesis, resulting in a significant attenuation of the severity of the pulmonary fibrosis induced by this chemical Citation[118]. This experimental evidence indicates that IP-10 has an important anti-angiogenic effect.

Evidence that the anti-angiogenic properties of IP-10 may have therapeutic value is the observation that it inhibits tumor growth. Observations in support of this include: (1) Burkitt's tumors implanted into athymic mice, once injected or transfected with IP-10, show histological evidence of tissue necrosis, capillary damage, intimal thickening, and vascular thrombosis Citation[176]; (2) high serum concentrations of IP-10 have been reported in patients with lymphoproliferative disorders Citation[177]; (3) tumors derived from IP-10 transduced melanoma cells have a reduced in vivo growth compared to those originating from parental or null-transduced cells (p = 0.0002) and the growth inhibition is associated with a marked reduction in microvessel density Citation[178]; (4) the angiostatic activity of IP-10 in human melanoma cell line appears to depend on binding to CXCR3 Citation[169]; (5) tumors originating from human fibrosarcoma cell lines, secreting IP-10 upon vector transduction, and implanted into mice, have histological evidence of a significantly lower number of microvessels than controls (p = 0.01) Citation[126]; (6) recombinant IP-10 injection into non-small cell lung cancers grown in SCID (severe combined immunodeficiency) mice reduces their angiogenesis, growth, and incidence of spontaneous metastasis, whilst IP-10 neutralization results in enhanced tumor-derived angiogenic activity. Furthermore, plasma or tumor-associated IP-10 concentrations are inversely correlated to tumor growth. Extracts from these tumors decrease the angiogenic activity in the corneal micropocket assay Citation[179]; and (7) IP-10 contributes to the anti-angiogenic and anti-tumor properties of IL-12 Citation[180-183].

In summary, this evidence indicates that IP-10 plays an important anti-angiogenic role.

IP-10 concentration in serum during normal pregnancy

The observation that normal pregnant women have a significantly higher median serum concentration of IP-10 than non-pregnant women is novel. This observation is consistent with the findings that normal pregnancy is associated with systemic intravascular inflammation Citation[35],Citation[184-187]. Additional evidence in support of this view is the observation that pregnancy is associated with higher (serum or plasma) concentrations of IL-12 Citation[186] and TNF-αCitation[188] than the non-pregnant state.

IP-10, inflammation, and preeclampsia

The observation that preeclampsia is associated with a higher median maternal serum concentration of IP-10 than SGA and normal pregnancy is novel, as well as being consistent with the view that preeclampsia is characterized by an exaggerated intravascular pro-inflammatory state Citation[18],Citation[33-37].

Furthermore, accumulating evidence indicates that preeclampsia is associated with a predominant Th1 immune response, including: (1) high maternal plasma or serum concentrations of IL-2 Citation[38], TNF-αCitation[40],Citation[41],Citation[189-191], IFN-γCitation[192], and IL-12 Citation[42],Citation[193]; (2) low maternal plasma or serum concentrations of IL-10 Citation[194] and of IL-4 Citation[192] (although this topic is subject to some controversy) Citation[195]; and (3) up-regulation of mRNA and protein expression of IL-1βCitation[196] and TNF-αCitation[196],Citation[197] in the placenta. Thus, the high maternal plasma concentration of this chemokine in patients with preeclampsia may represent yet another feature of a pro-inflammatory state or intravascular inflammation or even contribute to the generation of such state. Some evidence also indicates that SGA may be associated with intravascular inflammation Citation[47]. However, the results presented herein do not implicate IP-10 in such process.

IP-10, preeclampsia, and anti-angiogenesis

An imbalance between pro-angiogenic and anti-angiogenic factors is involved in the pathophysiology of preeclampsia Citation[25-27],Citation[198-218]. The group of Maynard and Karumanchi has recently made a major set of observations that favors this hypothesis Citation[219]. Evidence in support of this includes: (1) over-expression of sVEGFR-1 mRNA and protein in placenta of patients with preeclampsia Citation[27],Citation[219]; (2) higher median plasma/serum concentration of sVEGFR-1 in preeclampsia at the time of diagnosis than in patients with normal pregnancies Citation[27],Citation[219],Citation[220] and the plasma concentration correlates with the severity of the disease Citation[28]; (3) preeclampsia is associated with decreased plasma/serum concentrations of VEGF and PlGF Citation[25],Citation[211],Citation[219],Citation[221]; (4) serum of pregnant women with preeclampsia has anti-angiogenic effects in the endothelial cell tube formation bioassay and these effects can be restored by the addition of VEGF and PlGF Citation[219]; and (5) administration of sVEGFR-1 to pregnant animals can induce the clinical manifestation of preeclampsia, including hypertension and proteinuria Citation[219]. Moreover, these animals develop the pathologic finding of glomerular endotheliosis but not pathognomonic of preeclampsia Citation[222],Citation[223]; and (6) preeclampsia is associated with a higher maternal serum concentration of endoglin at the time of the diagnosis or before the recognition of the disease Citation[31],Citation[32].

Given the evidence that IP-10 has anti-angiogenic properties and that this is a feature of preeclampsia, we propose that elevated IP-10 maternal serum concentrations may contribute to generating an anti-angiogenic state along with sVEGFR-1 and endoglin.

IP-10, preeclampsia, and allograft rejection

Allograft rejection has also been proposed as a mechanism of disease in preeclampsia Citation[224]. Of note, there is growing evidence that IP-10 is also involved in the process of graft rejection, including: (1) CXCR3 deficient (−/−) mice are resistant to both acute and chronic allograft rejection Citation[91]; (2) grafts from IP-10 (−/−) donors are less likely to undergo graft injury Citation[90]; (3) treatment with anti-CXCR3 Citation[91] or anti-CXCL10 Citation[96] monoclonal antibodies results in prolongation of cardiac and small bowel allograft survival in mice; (4) clinical rejection of human renal Citation[92],Citation[100], lung Citation[93], cardiac Citation[94],Citation[95], small bowel Citation[99], and arteries Citation[102] transplants is associated with intra-graft over expression of IP-10 and/or with intra-graft recruitment of CXCR3 positive T cells; and (5) pre-transplant serum Citation[97],Citation[103] and urine Citation[98],Citation[101] CXCL10 cellular mRNA or protein concentrations identify patients at risk for the development of acute rejection and/or chronic allograft nephropathy. Collectively, this evidence indicates that IP-10 is involved in the pathogenesis of allograft rejection. Whether or not this mechanism of disease operates in preeclampsia remains an interesting concept. Our observation that IP-10 is elevated in preeclampsia provides a possible link between preeclampsia and this potential pathologic process.

IP-10, preeclampsia, and atherosclerosis

Striking parallels exist between preeclampsia and atherosclerosis (see bibliography for details). Patients who develop preterm preeclampsia are at increased risk of death from coronary artery disease later in life Citation[225]. Moreover, atherosis of the spiral arteries, a lesion observed in patients with preeclampsia, has striking similarities with that of coronary artery disease Citation[226-228].

The current view of atherosclerosis is that it is an inflammatory mediated process Citation[229-231]. Th1 biased responses are thought to be pro-atherogenic, while Th2 biased immunoresponses confer atheroprotection Citation[232]. Chemokine-dependent migration of cells through the inflamed endothelium into the arterial intima has been proposed to be critical in atherosclerosis Citation[233-237]. There is evidence of a specific involvement of IP-10 in the pathogenesis of atherosclerosis and cardiovascular diseases. Such evidence includes: (1) patients with stable angiographically confirmed coronary heart disease have a higher mean IP-10 serum concentration than control patients, and the magnitude of the elevation correlates with the concentration of several acute-phase proteins (C-reactive proteins) or cytokines known to be central in the pathogenesis of atherosclerosis Citation[122]; (2) serum IP-10 baseline concentrations are significantly higher in individuals who develop coronary heart disease (CHD) than in those who do not (follow-up of 11 years). After adjustment for cardiovascular and immunological risk factors, however, the observed relationship with IP-10 disappeared, suggesting that the association may be explained by other markers of inflammation and not be specific to IP-10. Nonetheless, this observation is important because it suggests that an elevation of IP-10 precedes the development of CHD, thus strengthening the case that inflammation has a causal role rather being a consequence of atherosclerosis Citation[121]; (3) CXCR3-bearing T cells and chemokines IP-10, I-TAC, and MIG (all three IFN-γ-induced) are present in atherosclerotic plaques Citation[120].

Conclusions

Preeclampsia is associated with higher maternal plasma concentrations of IP-10 than normal pregnancy and SGA. These results suggest that this chemokine may contribute to both the exaggerated systemic inflammation and the anti-angiogenic state that characterize preeclampsia.

Acknowledgement

This research was supported by the Intramural Research Program of the National Institute of Child Health and Human Development, NIH, DHHS.

References

  • 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
  • Brosens I, Dixon H G, Robertson W B. Fetal growth retardation and the arteries of the placental bed. Br J Obstet Gynaecol 1977; 84: 656–663
  • 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
  • Khong T Y, De Wolf F, Robertson W B, 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
  • 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
  • Meekins J W, Pijnenborg R, Hanssens M, McFadyen I R, van Assche A. A study of placental bed spiral arteries and trophoblast invasion in normal and severe pre-eclamptic pregnancies. Br J Obstet Gynaecol 1994; 101: 669–674
  • Brosens J J, Pijnenborg R, Brosens I A. The myometrial junctional zone spiral arteries in normal and abnormal pregnancies: A review of the literature. Am J Obstet Gynecol 2002; 187: 1416–1423
  • Espinoza J, Romero R, Kim Y M, 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
  • 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–S20
  • Bower S, Schuchter K, Campbell S. Doppler ultrasound screening as part of routine antenatal scanning: Prediction of pre-eclampsia and intrauterine growth retardation. Br J Obstet Gynaecol 1993; 100: 989–994
  • Harrington K, Cooper D, Lees C, Hecher K, Campbell S. Doppler ultrasound of the uterine arteries: The importance of bilateral notching in the prediction of pre-eclampsia, placental abruption or delivery of a small-for-gestational-age baby. Ultrasound Obstet Gynecol 1996; 7: 182–188
  • Dekker G A, Sibai B M. Etiology and pathogenesis of preeclampsia: Current concepts. Am J Obstet Gynecol 1998; 179: 1359–1375
  • Albaiges G, Missfelder-Lobos H, Lees C, Parra M, Nicolaides K H. One-stage screening for pregnancy complications by color Doppler assessment of the uterine arteries at 23 weeks' gestation. Obstet Gynecol 2000; 96: 559–564
  • 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
  • Papageorghiou A T, Yu C K, Nicolaides K H. The role of uterine artery Doppler in predicting adverse pregnancy outcome. Best Pract Res Clin Obstet Gynaecol 2004; 18: 383–396
  • 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
  • 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
  • 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
  • Poston L, Chappell L C. Is oxidative stress involved in the aetiology of pre-eclampsia?. Acta Paediatr Suppl 2001; 90: 3–5
  • 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
  • Roberts J M, Lain K Y. Recent insights into the pathogenesis of pre-eclampsia. Placenta 2002; 23: 359–372
  • Redman C W, Sargent I L. Latest advances in understanding preeclampsia. Science 2005; 308: 1592–1594
  • 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
  • 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
  • Taylor R N, Grimwood J, Taylor R S, McMaster M T, Fisher S J, North R A. Longitudinal serum concentrations of placental growth factor: Evidence for abnormal placental angiogenesis in pathologic pregnancies. Am J Obstet Gynecol 2003; 188: 177–182
  • Tsatsaris V, Goffin F, Munaut C, Brichant J F, Pignon M R, Noel A, Schaaps J P, Cabrol D, Frankenne F, Foidart J M. Overexpression of the soluble vascular endothelial growth factor receptor in preeclamptic patients: Pathophysiological consequences. J Clin Endocrinol Metab 2003; 88: 5555–5563
  • Chaiworapongsa T, Romero R, Espinoza J, Bujold E, Mee K Y, 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
  • Shibata E, Rajakumar A, Powers R W, Larkin R W, Gilmour C, Bodnar L M, Crombleholme W R, Ness R B, Roberts J M, Hubel C A. Soluble fms-like tyrosine kinase 1 is increased in preeclampsia but not in normotensive pregnancies with small-for-gestational-age neonates: Relationship to circulating placental growth factor. J Clin Endocrinol Metab 2005; 90: 4895–4903
  • 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
  • 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
  • Barden A, Graham D, Beilin L J, Ritchie J, Baker R, Walters B N, Michael C A. Neutrophil CD11B expression and neutrophil activation in pre-eclampsia. Clin Sci (Lond) 1997; 92: 37–44
  • Haller H, Ziegler E M, Homuth V, Drab M, Eichhorn J, Nagy Z, Busjahn A, Vetter K, Luft F C. Endothelial adhesion molecules and leukocyte integrins in preeclamptic patients. Hypertension 1997; 29: 291–296
  • 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
  • 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
  • Sunder-Plassmann G, Derfler K, Wagner L, Stockenhuber F, Endler M, Nowotny C, Balcke P. Increased serum activity of interleukin-2 in patients with pre-eclampsia. J Autoimmun 1989; 2: 203–205
  • Greer I A, Lyall F, Perera T, Boswell F, Macara L M. Increased concentrations of cytokines interleukin-6 and interleukin-1 receptor antagonist in plasma of women with preeclampsia: A mechanism for endothelial dysfunction?. Obstet Gynecol 1994; 84: 937–940
  • Vince G S, Starkey P M, Austgulen R, Kwiatkowski D, Redman C W. Interleukin-6, tumour necrosis factor and soluble tumour necrosis factor receptors in women with pre-eclampsia. Br J Obstet Gynaecol 1995; 102: 20–25
  • Conrad K P, Miles T M, Benyo D F. Circulating levels of immunoreactive cytokines in women with preeclampsia. Am J Reprod Immunol 1998; 40: 102–111
  • Daniel Y, Kupferminc M J, Baram A, Jaffa A J, Fait G, Wolman I, Lessing J B. Plasma interleukin-12 is elevated in patients with preeclampsia. Am J Reprod Immunol 1998; 39: 376–380
  • Saito S, Umekage H, Sakamoto Y, Sakai M, Tanebe K, Sasaki Y, Morikawa H. Increased T-helper-1-type immunity and decreased T-helper-2-type immunity in patients with preeclampsia. Am J Reprod Immunol 1999; 41: 297–306
  • Saito S, Sakai M, Sasaki Y, Tanebe K, Tsuda H, Michimata T. Quantitative analysis of peripheral blood Th0, Th1, Th2 and the Th1:Th2 cell ratio during normal human pregnancy and preeclampsia. Clin Exp Immunol 1999; 117: 550–555
  • Darmochwal-Kolarz D, Leszczynska-Gorzelak B, Rolinski J, Oleszczuk J. T helper 1- and T helper 2-type cytokine imbalance in pregnant women with pre-eclampsia. Eur J Obstet Gynecol Reprod Biol 1999; 86: 165–170
  • Sakai M, Ogawa K, Shiozaki A, Yoneda S, Sasaki Y, Nagata K, Saito S. Serum granulysin is a marker for Th1 type immunity in pre-eclampsia. Clin Exp Immunol 2004; 136: 114–119
  • Bartha J L, Romero-Carmona R, Comino-Delgado R. Inflammatory cytokines in intrauterine growth retardation. Acta Obstet Gynecol Scand 2003; 82: 1099–1102
  • Neville L F, Mathiak G, Bagasra O. The immunobiology of interferon-gamma inducible protein 10 kD (IP-10): A novel, pleiotropic member of the C-X-C chemokine superfamily. Cytokine Growth Factor Rev 1997; 8: 207–219
  • Strieter R M, Polverini P J, Kunkel S L, Arenberg D A, Burdick M D, Kasper J, Dzuiba J, Van Damme J, Walz A, Marriott D, et al. The functional role of the ELR motif in CXC chemokine-mediated angiogenesis. J Biol Chem 1995; 270: 27348–27357
  • Strieter R M, Polverini P J, Arenberg D A, Kunkel S L. The role of CXC chemokines as regulators of angiogenesis. Shock 1995; 4: 155–160
  • Strieter R M, Polverini P J, Arenberg D A, Walz A, Opdenakker G, Van Damme J, Kunkel S L. Role of C-X-C chemokines as regulators of angiogenesis in lung cancer. J Leukoc Biol 1995; 57: 752–762
  • Rollins B J. Chemokines. Blood 1997; 90: 909–928
  • Luster A D. Chemokines—chemotactic cytokines that mediate inflammation. N Engl J Med 1998; 338: 436–445
  • Belperio J A, Keane M P, Arenberg D A, Addison C L, Ehlert J E, Burdick M D, Strieter R M. CXC chemokines in angiogenesis. J Leukoc Biol 2000; 68: 1–8
  • Rossi D, Zlotnik A. The biology of chemokines and their receptors. Annu Rev Immunol 2000; 18: 217–242
  • Bernardini G, Ribatti D, Spinetti G, Morbidelli L, Ziche M, Santoni A, Capogrossi M C, Napolitano M. Analysis of the role of chemokines in angiogenesis. J Immunol Methods 2003; 273: 83–101
  • Rosenkilde M M, Schwartz T W. The chemokine system—a major regulator of angiogenesis in health and disease. APMIS 2004; 112: 481–495
  • Romagnani P, Lasagni L, Annunziato F, Serio M, Romagnani S. CXC chemokines: The regulatory link between inflammation and angiogenesis. Trends Immunol 2004; 25: 201–209
  • Strieter R M, Burdick M D, Gomperts B N, Belperio J A, Keane M P. CXC chemokines in angiogenesis. Cytokine Growth Factor Rev 2005; 16: 593–609
  • Charo I F, Ransohoff R M. The many roles of chemokines and chemokine receptors in inflammation. N Engl J Med 2006; 354: 610–621
  • Luster A D, Unkeless J C, Ravetch J V. Gamma-interferon transcriptionally regulates an early-response gene containing homology to platelet proteins. Nature 1985; 315: 672–676
  • Luster A D, Ravetch J V. Biochemical characterization of a gamma interferon-inducible cytokine (IP-10). J Exp Med 1987; 166: 1084–1097
  • Luster A D, Ravetch J V. Genomic characterization of a gamma-interferon-inducible gene (IP-10) and identification of an interferon-inducible hypersensitive site. Mol Cell Biol 1987; 7: 3723–3731
  • Kaplan G, Luster A D, Hancock G, Cohn Z A. The expression of a gamma interferon-induced protein (IP-10) in delayed immune responses in human skin. J Exp Med 1987; 166: 1098–1108
  • Gottlieb A B, Luster A D, Posnett D N, Carter D M. Detection of a gamma interferon-induced protein IP-10 in psoriatic plaques. J Exp Med 1988; 168: 941–948
  • Vanguri P, Farber J M. IFN and virus-inducible expression of an immediate early gene, crg-2/IP-10, and a delayed gene, I-A alpha in astrocytes and microglia. J Immunol 1994; 152: 1411–1418
  • Sarris A H, Esgleyes-Ribot T, Crow M, Broxmeyer H E, Karasavvas N, Pugh W, Grossman D, Deisseroth A, Duvic M. Cytokine loops involving interferon-gamma and IP-10, a cytokine chemotactic for CD4+ lymphocytes: An explanation for the epidermotropism of cutaneous T-cell lymphoma?. Blood 1995; 86: 651–658
  • Vanguri P. Interferon-gamma-inducible genes in primary glial cells of the central nervous system: Comparisons of astrocytes with microglia and Lewis with brown Norway rats. J Neuroimmunol 1995; 56: 35–43
  • Cassatella M A, Gasperini S, Calzetti F, Bertagnin A, Luster A D, McDonald P P. Regulated production of the interferon-gamma-inducible protein-10 (IP-10) chemokine by human neutrophils. Eur J Immunol 1997; 27: 111–115
  • Gasperini S, Marchi M, Calzetti F, Laudanna C, Vicentini L, Olsen H, Murphy M, Liao F, Farber J, Cassatella M A. Gene expression and production of the monokine induced by IFN-gamma (MIG), IFN-inducible T cell alpha chemoattractant (I-TAC), and IFN-gamma-inducible protein-10 (IP-10) chemokines by human neutrophils. J Immunol 1999; 162: 4928–4937
  • Frigerio S, Junt T, Lu B, Gerard C, Zumsteg U, Hollander G A, Piali L. Beta cells are responsible for CXCR3-mediated T-cell infiltration in insulitis. Nat Med 2002; 8: 1414–1420
  • Cardozo A K, Proost P, Gysemans C, Chen M C, Mathieu C, Eizirik D L. IL-1beta and IFN-gamma induce the expression of diverse chemokines and IL-15 in human and rat pancreatic islet cells, and in islets from pre-diabetic NOD mice. Diabetologia 2003; 46: 255–266
  • Narumi S, Yoneyama H, Inadera H, Nishioji K, Itoh Y, Okanoue T, Matsushima K. TNF-alpha is a potent inducer for IFN-inducible protein-10 in hepatocytes and unaffected by GM-CSF in vivo, in contrast to IL-1beta and IFN-gamma. Cytokine 2000; 12: 1007–1016
  • Kraft M, Riedel S, Maaser C, Kucharzik T, Steinbuechel A, Domschke W, Luegering N. IFN-gamma synergizes with TNF-alpha but not with viable H. pylori in up-regulating CXC chemokine secretion in gastric epithelial cells. Clin Exp Immunol 2001; 126: 474–481
  • Algood H M, Lin P L, Yankura D, Jones A, Chan J, Flynn J L. TNF influences chemokine expression of macrophages in vitro and that of CD11b+ cells in vivo during Mycobacterium tuberculosis infection. J Immunol 2004; 172: 6846–6857
  • Villagomez M T, Bae S J, Ogawa I, Takenaka M, Katayama I. Tumour necrosis factor-alpha but not interferon-gamma is the main inducer of inducible protein-10 in skin fibroblasts from patients with atopic dermatitis. Br J Dermatol 2004; 150: 910–916
  • Hardaker E L, Bacon A M, Carlson K, Roshak A K, Foley J J, Schmidt D B, Buckley P T, Comegys M, Panettieri R A, Jr, Sarau H M, et al. Regulation of TNF-alpha- and IFN-gamma-induced CXCL10 expression: Participation of the airway smooth muscle in the pulmonary inflammatory response in chronic obstructive pulmonary disease. FASEB J 2004; 18: 191–193
  • Sheng W S, Hu S, Ni H T, Rowen T N, Lokensgard J R, Peterson P K. TNF-alpha-induced chemokine production and apoptosis in human neural precursor cells. J Leukoc Biol 2005; 78: 1233–1241
  • Berthier-Vergnes O, Bermond F, Flacher V, Massacrier C, Schmitt D, Peguet-Navarro J. TNF-alpha enhances phenotypic and functional maturation of human epidermal Langerhans cells and induces IL-12 p40 and IP-10/CXCL-10 production. FEBS Lett 2005; 579: 3660–3668
  • Shin H S, Drysdale B E, Shin M L, Noble P W, Fisher S N, Paznekas W A. Definition of a lipopolysaccharide-responsive element in the 5′-flanking regions of MuRantes and crg-2. Mol Cell Biol 1994; 14: 2914–2925
  • Nazar A S, Cheng G, Shin H S, Brothers P N, Dhib-Jalbut S, Shin M L, Vanguri P. Induction of IP-10 chemokine promoter by measles virus: Comparison with interferon-gamma shows the use of the same response element but with differential DNA–protein binding profiles. J Neuroimmunol 1997; 77: 116–127
  • Cheng G, Nazar A S, Shin H S, Vanguri P, Shin M L. IP-10 gene transcription by virus in astrocytes requires cooperation of ISRE with adjacent kappaB site but not IRF-1 or viral transcription. J Interferon Cytokine Res 1998; 18: 987–997
  • Gasper N A, Petty C C, Schrum L W, Marriott I, Bost K L. Bacterium-induced CXCL10 secretion by osteoblasts can be mediated in part through toll-like receptor 4. Infect Immun 2002; 70: 4075–4082
  • Shen Q, Zhang R, Bhat N R. MAP kinase regulation of IP10/CXCL10 chemokine gene expression in microglial cells. Brain Res 2006; 1086: 9–16
  • Durand S H, Flacher V, Romeas A, Carrouel F, Colomb E, Vincent C, Magloire H, Couble M L, Bleicher F, Staquet M J, et al. Lipoteichoic acid increases TLR and functional chemokine expression while reducing dentin formation in in vitro differentiated human odontoblasts. J Immunol 2006; 176: 2880–2887
  • Ohmori Y, Hamilton T A. Cooperative interaction between interferon (IFN) stimulus response element and kappa B sequence motifs controls IFN gamma- and lipopolysaccharide-stimulated transcription from the murine IP-10 promoter. J Biol Chem 1993; 268: 6677–6688
  • Ohmori Y, Hamilton T A. The interferon-stimulated response element and a kappa B site mediate synergistic induction of murine IP-10 gene transcription by IFN-gamma and TNF-alpha. J Immunol 1995; 154: 5235–5244
  • Osawa Y, Iho S, Takauji R, Takatsuka H, Yamamoto S, Takahashi T, Horiguchi S, Urasaki Y, Matsuki T, Fujieda S. Collaborative action of NF-kappaB and p38 MAPK is involved in CpG DNA-induced IFN-alpha and chemokine production in human plasmacytoid dendritic cells. J Immunol 2006; 177: 4841–4852
  • Park C, Lee S, Cho I H, Lee H K, Kim D, Choi S Y, Oh S B, Park K, Kim J S, Lee S J. TLR3-mediated signal induces proinflammatory cytokine and chemokine gene expression in astrocytes: Differential signaling mechanisms of TLR3-induced IP-10 and IL-8 gene expression. Glia 2006; 53: 248–256
  • Hancock W W, Gao W, Csizmadia V, Faia K L, Shemmeri N, Luster A D. Donor-derived IP-10 initiates development of acute allograft rejection. J Exp Med 2001; 193: 975–980
  • Hancock W W, Lu B, Gao W, Csizmadia V, Faia K, King J A, Smiley S T, Ling M, Gerard N P, Gerard C. Requirement of the chemokine receptor CXCR3 for acute allograft rejection. J Exp Med 2000; 192: 1515–1520
  • Segerer S, Cui Y, Eitner F, Goodpaster T, Hudkins K L, Mack M, Cartron J P, Colin Y, Schlondorff D, Alpers C E. Expression of chemokines and chemokine receptors during human renal transplant rejection. Am J Kidney Dis 2001; 37: 518–531
  • Agostini C, Calabrese F, Rea F, Facco M, Tosoni A, Loy M, Binotto G, Valente M, Trentin L, Semenzato G. Cxcr3 and its ligand CXCL10 are expressed by inflammatory cells infiltrating lung allografts and mediate chemotaxis of T cells at sites of rejection. Am J Pathol 2001; 158: 1703–1711
  • Melter M, Exeni A, Reinders M E, Fang J C, McMahon G, Ganz P, Hancock W W, Briscoe D M. Expression of the chemokine receptor CXCR3 and its ligand IP-10 during human cardiac allograft rejection. Circulation 2001; 104: 2558–2564
  • Zhao D X, Hu Y, Miller G G, Luster A D, Mitchell R N, Libby P. Differential expression of the IFN-gamma-inducible CXCR3-binding chemokines, IFN-inducible protein 10, monokine induced by IFN, and IFN-inducible T cell alpha chemoattractant in human cardiac allografts: Association with cardiac allograft vasculopathy and acute rejection. J Immunol 2002; 169: 1556–1560
  • Zhang Z, Kaptanoglu L, Haddad W, Ivancic D, Alnadjim Z, Hurst S, Tishler D, Luster A D, Barrett T A, Fryer J. Donor T cell activation initiates small bowel allograft rejection through an IFN-gamma-inducible protein-10-dependent mechanism. J Immunol 2002; 168: 3205–3212
  • Rotondi M, Rosati A, Buonamano A, Lasagni L, Lazzeri E, Pradella F, Fossombroni V, Cirami C, Liotta F, La Villa G, et al. High pretransplant serum levels of CXCL10/IP-10 are related to increased risk of renal allograft failure. Am J Transplant 2004; 4: 1466–1474
  • Hu H, Aizenstein B D, Puchalski A, Burmania J A, Hamawy M M, Knechtle S J. Elevation of CXCR3-binding chemokines in urine indicates acute renal-allograft dysfunction. Am J Transplant 2004; 4: 432–437
  • Zhang Z, Kaptanoglu L, Tang Y, Ivancic D, Rao S M, Luster A, Barrett T A, Fryer J. IP-10-induced recruitment of CXCR3 host T cells is required for small bowel allograft rejection. Gastroenterology 2004; 126: 809–818
  • Panzer U, Reinking R R, Steinmetz O M, Zahner G, Sudbeck U, Fehr S, Pfalzer B, Schneider A, Thaiss F, Mack M, et al. CXCR3 and CCR5 positive T-cell recruitment in acute human renal allograft rejection. Transplantation 2004; 78: 1341–1350
  • Tatapudi R R, Muthukumar T, Dadhania D, Ding R, Li B, Sharma V K, Lozada-Pastorio E, Seetharamu N, Hartono C, Serur D, et al. Noninvasive detection of renal allograft inflammation by measurements of mRNA for IP-10 and CXCR3 in urine. Kidney Int 2004; 65: 2390–2397
  • Burns W R, Wang Y, Tang P C, Ranjbaran H, Iakimov A, Kim J, Cuffy M, Bai Y, Pober J S, Tellides G. Recruitment of CXCR3+ and CCR5+ T cells and production of interferon-gamma-inducible chemokines in rejecting human arteries. Am J Transplant 2005; 5: 1226–1236
  • Lazzeri E, Rotondi M, Mazzinghi B, Lasagni L, Buonamano A, Rosati A, Pradella F, Fossombroni V, La Villa G, Gacci M, et al. High CXCL10 expression in rejected kidneys and predictive role of pretransplant serum CXCL10 for acute rejection and chronic allograft nephropathy. Transplantation 2005; 79: 1215–1220
  • Sorensen T L, Tani M, Jensen J, Pierce V, Lucchinetti C, Folcik V A, Qin S, Rottman J, Sellebjerg F, Strieter R M, et al. Expression of specific chemokines and chemokine receptors in the central nervous system of multiple sclerosis patients. J Clin Invest 1999; 103: 807–815
  • Balashov K E, Rottman J B, Weiner H L, Hancock W W. CCR5(+) and CXCR3(+) T cells are increased in multiple sclerosis and their ligands MIP-1alpha and IP-10 are expressed in demyelinating brain lesions. Proc Natl Acad Sci U S A 1999; 96: 6873–6878
  • Simpson J E, Newcombe J, Cuzner M L, Woodroofe M N. Expression of the interferon-gamma-inducible chemokines IP-10 and Mig and their receptor, CXCR3, in multiple sclerosis lesions. Neuropathol Appl Neurobiol 2000; 26: 133–142
  • Franciotta D, Martino G, Zardini E, Furlan R, Bergamaschi R, Andreoni L, Cosi V. Serum and CSF levels of MCP-1 and IP-10 in multiple sclerosis patients with acute and stable disease and undergoing immunomodulatory therapies. J Neuroimmunol 2001; 115: 192–198
  • Trebst C, Ransohoff R M. Investigating chemokines and chemokine receptors in patients with multiple sclerosis: Opportunities and challenges. Arch Neurol 2001; 58: 1975–1980
  • Christen U, McGavern D B, Luster A D, Von Herrath M G, Oldstone M B. Among CXCR3 chemokines, IFN-gamma-inducible protein of 10 kDa (CXC chemokine ligand (CXCL) 10) but not monokine induced by IFN-gamma (CXCL9) imprints a pattern for the subsequent development of autoimmune disease. J Immunol 2003; 171: 6838–6845
  • Christen U, Von Herrath M G. IP-10 and type 1 diabetes: A question of time and location. Autoimmunity 2004; 37: 273–282
  • Romagnani P, Rotondi M, Lazzeri E, Lasagni L, Francalanci M, Buonamano A, Milani S, Vitti P, Chiovato L, Tonacchera M, et al. Expression of IP-10/CXCL10 and MIG/CXCL9 in the thyroid and increased levels of IP-10/CXCL10 in the serum of patients with recent-onset Graves' disease. Am J Pathol 2002; 161: 195–206
  • Antonelli A, Fallahi P, Rotondi M, Ferrari S M, Serio M, Miccoli P. Serum levels of the interferon-gamma-inducible alpha chemokine CXCL10 in patients with active Graves' disease, and modulation by methimazole therapy and thyroidectomy. Br J Surg 2006; 93: 1226–1231
  • Antonelli A, Fallahi P, Rotondi M, Ferrari S M, Romagnani P, Grosso M, Ferrannini E, Serio M. Increased serum CXCL10 in Graves' disease or autoimmune thyroiditis is not associated with hyper- or hypothyroidism per se, but is specifically sustained by the autoimmune, inflammatory process. Eur J Endocrinol 2006; 154: 651–658
  • Antonelli A, Rotondi M, Fallahi P, Romagnani P, Ferrari S M, Barani L, Ferrannini E, Serio M. Increase of interferon-gamma-inducible CXC chemokine CXCL10 serum levels in patients with active Graves' disease, and modulation by methimazole therapy. Clin Endocrinol (Oxf) 2006; 64: 189–195
  • Antonelli A, Rotondi M, Fallahi P, Romagnani P, Ferrari S M, Buonamano A, Ferrannini E, Serio M. High levels of circulating CXC chemokine ligand 10 are associated with chronic autoimmune thyroiditis and hypothyroidism. J Clin Endocrinol Metab 2004; 89: 5496–5499
  • Antonelli A, Rotondi M, Fallahi P, Romagnani P, Ferrari S M, Paolicchi A, Ferrannini E, Serio M. Increase of interferon-gamma inducible alpha chemokine CXCL10 but not beta chemokine CCL2 serum levels in chronic autoimmune thyroiditis. Eur J Endocrinol 2005; 152: 171–177
  • Keane M P, Arenberg D A, Lynch J P, III, Whyte R I, Iannettoni M D, Burdick M D, Wilke C A, Morris S B, Glass M C, DiGiovine B, et al. The CXC chemokines, IL-8 and IP-10, regulate angiogenic activity in idiopathic pulmonary fibrosis. J Immunol 1997; 159: 1437–1443
  • Keane M P, Belperio J A, Arenberg D A, Burdick M D, Xu Z J, Xue Y Y, Strieter R M. IFN-gamma-inducible protein-10 attenuates bleomycin-induced pulmonary fibrosis via inhibition of angiogenesis. J Immunol 1999; 163: 5686–5692
  • Strieter R M, Belperio J A, Keane M P. CXC chemokines in angiogenesis related to pulmonary fibrosis. Chest 2002; 122: 298S–301S
  • Mach F, Sauty A, Iarossi A S, Sukhova G K, Neote K, Libby P, Luster A D. Differential expression of three T lymphocyte-activating CXC chemokines by human atheroma-associated cells. J Clin Invest 1999; 104: 1041–1050
  • Herder C, Baumert J, Thorand B, Martin S, Lowel H, Kolb H, Koenig W. Chemokines and incident coronary heart disease: Results from the MONICA/KORA Augsburg case–cohort study, 1984–2002. Arterioscler Thromb Vasc Biol 2006; 26: 2147–2152
  • Rothenbacher D, Muller-Scholze S, Herder C, Koenig W, Kolb H. Differential expression of chemokines, risk of stable coronary heart disease, and correlation with established cardiovascular risk markers. Arterioscler Thromb Vasc Biol 2006; 26: 194–199
  • Strieter R M, Kunkel S L, Arenberg D A, Burdick M D, Polverini P J. Interferon gamma-inducible protein 10 (IP-10), a member of the C-X-C chemokine family, is an inhibitor of angiogenesis. Biochem Biophys Res Commun 1995; 210: 51–57
  • Luster A D, Greenberg S M, Leder P. The IP-10 chemokine binds to a specific cell surface heparan sulfate site shared with platelet factor 4 and inhibits endothelial cell proliferation. J Exp Med 1995; 182: 219–231
  • Angiolillo A L, Sgadari C, Taub D D, Liao F, Farber J M, Maheshwari S, Kleinman H K, Reaman G H, Tosato G. Human interferon-inducible protein 10 is a potent inhibitor of angiogenesis in vivo. J Exp Med 1995; 182: 155–162
  • Sun Y, Finger C, Alvarez-Vallina L, Cichutek K, Buchholz C J. Chronic gene delivery of interferon-inducible protein 10 through replication-competent retrovirus vectors suppresses tumor growth. Cancer Gene Ther 2005; 12: 900–912
  • Report of the National High Blood Pressure Education Program Working Group on High Blood Pressure in Pregnancy. Am J Obstet Gynecol 2000; 183: S1–S22
  • ACOG practice bulletin. Diagnosis and management of preeclampsia and eclampsia. Number 33, January 2002. Obstet Gynecol 2002; 99: 159–167
  • 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
  • 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
  • Nagaoka K, Nojima H, Watanabe F, Chang K T, Christenson R K, Sakai S, Imakawa K. Regulation of blastocyst migration, apposition, and initial adhesion by a chemokine, interferon gamma-inducible protein 10 kDa (IP-10), during early gestation. J Biol Chem 2003; 278: 29048–29056
  • Hanna J, Goldman-Wohl D, Hamani Y, Avraham I, Greenfield C, Natanson-Yaron S, Prus D, Cohen-Daniel L, Arnon T I, Manaster I, et al. Decidual NK cells regulate key developmental processes at the human fetal–maternal interface. Nat Med 2006; 12: 1065–1074
  • Sarris A H, Broxmeyer H E, Wirthmueller U, Karasavvas N, Cooper S, Lu L, Krueger J, Ravetch J V. Human interferon-inducible protein 10: Expression and purification of recombinant protein demonstrate inhibition of early human hematopoietic progenitors. J Exp Med 1993; 178: 1127–1132
  • Sasaki S, Yoneyama H, Suzuki K, Suriki H, Aiba T, Watanabe S, Kawauchi Y, Kawachi H, Shimizu F, Matsushima K, et al. Blockade of CXCL10 protects mice from acute colitis and enhances crypt cell survival. Eur J Immunol 2002; 32: 3197–3205
  • Loetscher M, Gerber B, Loetscher P, Jones S A, Piali L, Clark-Lewis I, Baggiolini M, Moser B. Chemokine receptor specific for IP10 and mig: Structure, function, and expression in activated T-lymphocytes. J Exp Med 1996; 184: 963–969
  • Liu L, Callahan M K, Huang D, Ransohoff R M. Chemokine receptor CXCR3: An unexpected enigma. Curr Top Dev Biol 2005; 68: 149–181
  • Cole K E, Strick C A, Paradis T J, Ogborne K T, Loetscher M, Gladue R P, Lin W, Boyd J G, Moser B, Wood D E, et al. Interferon-inducible T cell alpha chemoattractant (I-TAC): A novel non-ELR CXC chemokine with potent activity on activated T cells through selective high affinity binding to CXCR3. J Exp Med 1998; 187: 2009–2121
  • Cox M A, Jenh C H, Gonsiorek W, Fine J, Narula S K, Zavodny P J, Hipkin R W. Human interferon-inducible 10-kDa protein and human interferon-inducible T cell alpha chemoattractant are allotopic ligands for human CXCR3: Differential binding to receptor states. Mol Pharmacol 2001; 59: 707–715
  • Sauty A, Colvin R A, Wagner L, Rochat S, Spertini F, Luster A D. CXCR3 internalization following T cell–endothelial cell contact: Preferential role of IFN-inducible T cell alpha chemoattractant (CXCL11). J Immunol 2001; 167: 7084–7093
  • Colvin R A, Campanella G S, Sun J, Luster A D. Intracellular domains of CXCR3 that mediate CXCL9, CXCL10, and CXCL11 function. J Biol Chem 2004; 279: 30219–30227
  • Dajotoy T, Andersson P, Bjartell A, Lofdahl C G, Tapper H, Egesten A. Human eosinophils produce the T cell-attracting chemokines MIG and IP-10 upon stimulation with IFN-gamma. J Leukoc Biol 2004; 76: 685–691
  • Hoffmann A, Levchenko A, Scott M L, Baltimore D. The IkappaB-NF-kappaB signaling module: Temporal control and selective gene activation. Science 2002; 298: 1241–1245
  • Taub D D, Lloyd A R, Conlon K, Wang J M, Ortaldo J R, Harada A, Matsushima K, Kelvin D J, Oppenheim J J. Recombinant human interferon-inducible protein 10 is a chemoattractant for human monocytes and T lymphocytes and promotes T cell adhesion to endothelial cells. J Exp Med 1993; 177: 1809–1814
  • Taub D D, Sayers T J, Carter C R, Ortaldo J R. Alpha and beta chemokines induce NK cell migration and enhance NK-mediated cytolysis. J Immunol 1995; 155: 3877–3888
  • Dewald B, Moser B, Barella L, Schumacher C, Baggiolini M, Clark-Lewis I. IP-10, a gamma-interferon-inducible protein related to interleukin-8, lacks neutrophil activating properties. Immunol Lett 1992; 32: 81–84
  • Qin S, Rottman J B, Myers P, Kassam N, Weinblatt M, Loetscher M, Koch A E, Moser B, Mackay C R. The chemokine receptors CXCR3 and CCR5 mark subsets of T cells associated with certain inflammatory reactions. J Clin Invest 1998; 101: 746–754
  • Muehlinghaus G, Cigliano L, Huehn S, Peddinghaus A, Leyendeckers H, Hauser A E, Hiepe F, Radbruch A, Arce S, Manz R A. Regulation of CXCR3 and CXCR4 expression during terminal differentiation of memory B cells into plasma cells. Blood 2005; 105: 3965–3971
  • Farber J M. Mig and IP-10: CXC chemokines that target lymphocytes. J Leukoc Biol 1997; 61: 246–257
  • Sallusto F, Lenig D, Mackay C R, Lanzavecchia A. Flexible programs of chemokine receptor expression on human polarized T helper 1 and 2 lymphocytes. J Exp Med 1998; 187: 875–883
  • Piali L, Weber C, LaRosa G, Mackay C R, Springer T A, Clark-Lewis I, Moser B. The chemokine receptor CXCR3 mediates rapid and shear-resistant adhesion-induction of effector T lymphocytes by the chemokines IP10 and Mig. Eur J Immunol 1998; 28: 961–972
  • Loetscher M, Loetscher P, Brass N, Meese E, Moser B. Lymphocyte-specific chemokine receptor CXCR3: Regulation, chemokine binding and gene localization. Eur J Immunol 1998; 28: 3696–3705
  • Bonecchi R, Bianchi G, Bordignon P P, D'Ambrosio D, Lang R, Borsatti A, Sozzani S, Allavena P, Gray P A, Mantovani A, et al. Differential expression of chemokine receptors and chemotactic responsiveness of type 1 T helper cells (Th1s) and Th2s. J Exp Med 1998; 187: 129–134
  • Yamamoto J, Adachi Y, Onoue Y, Adachi Y S, Okabe Y, Itazawa T, Toyoda M, Seki T, Morohashi M, Matsushima K, et al. Differential expression of the chemokine receptors by the Th1- and Th2-type effector populations within circulating CD4+ T cells. J Leukoc Biol 2000; 68: 568–574
  • Gangur V, Simons F E, Hayglass K T. Human IP-10 selectively promotes dominance of polyclonally activated and environmental antigen-driven IFN-gamma over IL-4 responses. FASEB J 1998; 12: 705–713
  • Goldberg S H, van der Meer P, Hesselgesser J, Jaffer S, Kolson D L, Albright A V, Gonzalez-Scarano F, Lavi E. CXCR3 expression in human central nervous system diseases. Neuropathol Appl Neurobiol 2001; 27: 127–138
  • Christensen J E, Nansen A, Moos T, Lu B, Gerard C, Christensen J P, Thomsen A R. Efficient T-cell surveillance of the CNS requires expression of the CXC chemokine receptor 3. J Neurosci 2004; 24: 4849–4858
  • Wickham S, Lu B, Ash J, Carr D J. Chemokine receptor deficiency is associated with increased chemokine expression in the peripheral and central nervous systems and increased resistance to herpetic encephalitis. J Neuroimmunol 2005; 162: 51–59
  • Sellner J, Dvorak F, Zhou Y, Haas J, Kehm R, Wildemann B, Meyding-Lamade U. Acute and long-term alteration of chemokine mRNA expression after anti-viral and anti-inflammatory treatment in herpes simplex virus encephalitis. Neurosci Lett 2005; 374: 197–202
  • Narumi S, Kaburaki T, Yoneyama H, Iwamura H, Kobayashi Y, Matsushima K. Neutralization of IFN-inducible protein 10/CXCL10 exacerbates experimental autoimmune encephalomyelitis. Eur J Immunol 2002; 32: 1784–1791
  • Wildbaum G, Netzer N, Karin N. Plasmid DNA encoding IFN-gamma-inducible protein 10 redirects antigen-specific T cell polarization and suppresses experimental autoimmune encephalomyelitis. J Immunol 2002; 168: 5885–5892
  • Klein R S. Regulation of neuroinflammation: The role of CXCL10 in lymphocyte infiltration during autoimmune encephalomyelitis. J Cell Biochem 2004; 92: 213–222
  • Klein R S, Izikson L, Means T, Gibson H D, Lin E, Sobel R A, Weiner H L, Luster A D. IFN-inducible protein 10/CXC chemokine ligand 10-independent induction of experimental autoimmune encephalomyelitis. J Immunol 2004; 172: 550–559
  • Uguccioni M, Gionchetti P, Robbiani D F, Rizzello F, Peruzzo S, Campieri M, Baggiolini M. Increased expression of IP-10, IL-8, MCP-1, and MCP-3 in ulcerative colitis. Am J Pathol 1999; 155: 331–336
  • Singh U P, Singh S, Taub D D, Lillard J W, Jr. Inhibition of IFN-gamma-inducible protein-10 abrogates colitis in IL-10 −/− mice. J Immunol 2003; 171: 1401–1406
  • Narumi S, Tominaga Y, Tamaru M, Shimai S, Okumura H, Nishioji K, Itoh Y, Okanoue T. Expression of IFN-inducible protein-10 in chronic hepatitis. J Immunol 1997; 158: 5536–5544
  • Ogawa N, Ping L, Zhenjun L, Takada Y, Sugai S. Involvement of the interferon-gamma-induced T cell-attracting chemokines, interferon-gamma-inducible 10-kD protein (CXCL10) and monokine induced by interferon-gamma (CXCL9), in the salivary gland lesions of patients with Sjogren's syndrome. Arthritis Rheum 2002; 46: 2730–2741
  • Rhode A, Pauza M E, Barral A M, Rodrigo E, Oldstone M B, Von Herrath M G, Christen U. Islet-specific expression of CXCL10 causes spontaneous islet infiltration and accelerates diabetes development. J Immunol 2005; 175: 3516–3524
  • Lit L C, Wong C K, Tam L S, Li E K, Lam C W. Raised plasma concentration and ex vivo production of inflammatory chemokines in patients with systemic lupus erythematosus. Ann Rheum Dis 2006; 65: 209–215
  • Yang J, Richmond A. The angiostatic activity of interferon-inducible protein-10/CXCL10 in human melanoma depends on binding to CXCR3 but not to glycosaminoglycan. Mol Ther 2004; 9: 846–855
  • Romagnani P, Annunziato F, Lasagni L, Lazzeri E, Beltrame C, Francalanci M, Uguccioni M, Galli G, Cosmi L, Maurenzig L, et al. Cell cycle-dependent expression of CXC chemokine receptor 3 by endothelial cells mediates angiostatic activity. J Clin Invest 2001; 107: 53–63
  • Soejima K, Rollins B J. A functional IFN-gamma-inducible protein-10/CXCL10-specific receptor expressed by epithelial and endothelial cells that is neither CXCR3 nor glycosaminoglycan. J Immunol 2001; 167: 6576–6582
  • Salcedo R, Resau J H, Halverson D, Hudson E A, Dambach M, Powell D, Wasserman K, Oppenheim J J. Differential expression and responsiveness of chemokine receptors (CXCR1–3) by human microvascular endothelial cells and umbilical vein endothelial cells. FASEB J 2000; 14: 2055–2064
  • Datta D, Flaxenburg J A, Laxmanan S, Geehan C, Grimm M, Waaga-Gasser A M, Briscoe D M, Pal S. Ras-induced modulation of CXCL10 and its receptor splice variant CXCR3-B in MDA-MB-435 and MCF-7 cells: Relevance for the development of human breast cancer. Cancer Res 2006; 66: 9509–9518
  • Lasagni L, Francalanci M, Annunziato F, Lazzeri E, Giannini S, Cosmi L, Sagrinati C, Mazzinghi B, Orlando C, Maggi E, et al. An alternatively spliced variant of CXCR3 mediates the inhibition of endothelial cell growth induced by IP-10, Mig, and I-TAC, and acts as functional receptor for platelet factor 4. J Exp Med 2003; 197: 1537–1549
  • Proost P, Schutyser E, Menten P, Struyf S, Wuyts A, Opdenakker G, Detheux M, Parmentier M, Durinx C, Lambeir A M, et al. Amino-terminal truncation of CXCR3 agonists impairs receptor signaling and lymphocyte chemotaxis, while preserving antiangiogenic properties. Blood 2001; 98: 3554–3561
  • Sgadari C, Angiolillo A L, Cherney B W, Pike S E, Farber J M, Koniaris L G, Vanguri P, Burd P R, Sheikh N, Gupta G, et al. Interferon-inducible protein-10 identified as a mediator of tumor necrosis in vivo. Proc Natl Acad Sci U S A 1996; 93: 13791–13796
  • Teruya-Feldstein J, Jaffe E S, Burd P R, Kanegane H, Kingma D W, Wilson W H, Longo D L, Tosato G. The role of Mig, the monokine induced by interferon-gamma, and IP-10, the interferon-gamma-inducible protein-10, in tissue necrosis and vascular damage associated with Epstein–Barr virus-positive lymphoproliferative disease. Blood 1997; 90: 4099–4105
  • Feldman A L, Friedl J, Lans T E, Libutti S K, Lorang D, Miller M S, Turner E M, Hewitt S M, Alexander H R. Retroviral gene transfer of interferon-inducible protein 10 inhibits growth of human melanoma xenografts. Int J Cancer 2002; 99: 149–153
  • Arenberg D A, Kunkel S L, Polverini P J, Morris S B, Burdick M D, Glass M C, Taub D T, Iannettoni M D, Whyte R I, Strieter R M. Interferon-gamma-inducible protein 10 (IP-10) is an angiostatic factor that inhibits human non-small cell lung cancer (NSCLC) tumorigenesis and spontaneous metastases. J Exp Med 1996; 184: 981–992
  • Sgadari C, Angiolillo A L, Tosato G. Inhibition of angiogenesis by interleukin-12 is mediated by the interferon-inducible protein 10. Blood 1996; 87: 3877–3882
  • Kanegane C, Sgadari C, Kanegane H, Teruya-Feldstein J, Yao L, Gupta G, Farber J M, Liao F, Liu L, Tosato G. Contribution of the CXC chemokines IP-10 and Mig to the antitumor effects of IL-12. J Leukoc Biol 1998; 64: 384–392
  • Tannenbaum C S, Tubbs R, Armstrong D, Finke J H, Bukowski R M, Hamilton T A. The CXC chemokines IP-10 and Mig are necessary for IL-12-mediated regression of the mouse RENCA tumor. J Immunol 1998; 161: 927–932
  • Pertl U, Luster A D, Varki N M, Homann D, Gaedicke G, Reisfeld R A, Lode H N. IFN-gamma-inducible protein-10 is essential for the generation of a protective tumor-specific CD8 T cell response induced by single-chain IL-12 gene therapy. J Immunol 2001; 166: 6944–6951
  • Koumandakis E, Koumandaki I, Kaklamani E, Sparos L, Aravantinos D, Trichopoulos D. Enhanced phagocytosis of mononuclear phagocytes in pregnancy. Br J Obstet Gynaecol 1986; 93: 1150–1154
  • Naccasha N, Gervasi M T, Chaiworapongsa T, Berman S, Yoon B H, Maymon E, Romero R. Phenotypic and metabolic characteristics of monocytes and granulocytes in normal pregnancy and maternal infection. Am J Obstet Gynecol 2001; 185: 1118–1123
  • Sacks G P, Redman C W, Sargent I L. Monocytes are primed to produce the Th1 type cytokine IL-12 in normal human pregnancy: An intracellular flow cytometric analysis of peripheral blood mononuclear cells. Clin Exp Immunol 2003; 131: 490–497
  • Smarason A K, Gunnarsson A, Alfredsson J H, Valdimarsson H. Monocytosis and monocytic infiltration of decidua in early pregnancy. J Clin Lab Immunol 1986; 21: 1–5
  • Melczer Z, Banhidy F, Csomor S, Toth P, Kovacs M, Winkler G, Cseh K. Influence of leptin and the TNF system on insulin resistance in pregnancy and their effect on anthropometric parameters of newborns. Acta Obstet Gynecol Scand 2003; 82: 432–438
  • Kupferminc M J, Peaceman A M, Wigton T R, Rehnberg K A, Socol M L. Tumor necrosis factor-alpha is elevated in plasma and amniotic fluid of patients with severe preeclampsia. Am J Obstet Gynecol 1994; 170: 1752–1757
  • Meekins J W, McLaughlin P J, West D C, McFadyen I R, Johnson P M. Endothelial cell activation by tumour necrosis factor-alpha (TNF-alpha) and the development of pre-eclampsia. Clin Exp Immunol 1994; 98: 110–114
  • Teran E, Escudero C, Moya W, Flores M, Vallance P, Lopez-Jaramillo P. Elevated C-reactive protein and pro-inflammatory cytokines in Andean women with pre-eclampsia. Int J Gynaecol Obstet 2001; 75: 243–249
  • Arriaga-Pizano L, Jimenez-Zamudio L, Vadillo-Ortega F, Martinez-Flores A, Herrerias-Canedo T, Hernandez-Guerrero C. The predominant Th1 cytokine profile in maternal plasma of preeclamptic women is not reflected in the choriodecidual and fetal compartments. J Soc Gynecol Investig 2005; 12: 335–342
  • Dudley D J, Hunter C, Mitchell M D, Varner M W, Gately M. Elevations of serum interleukin-12 concentrations in women with severe pre-eclampsia and HELLP syndrome. J Reprod Immunol 1996; 31: 97–107
  • Hennessy A, Pilmore H L, Simmons L A, Painter D M. A deficiency of placental IL-10 in preeclampsia. J Immunol 1999; 163: 3491–3495
  • Omu A E, Al-Qattan F, Diejomaoh M E, Al-Yatama M. Differential levels of T helper cytokines in preeclampsia: Pregnancy, labor and puerperium. Acta Obstet Gynecol Scand 1999; 78: 675–680
  • Rinehart B K, Terrone D A, Lagoo-Deenadayalan S, Barber W H, Hale E A, Martin J N, Jr, Bennett W A. Expression of the placental cytokines tumor necrosis factor alpha, interleukin 1beta, and interleukin 10 is increased in preeclampsia. Am J Obstet Gynecol 1999; 181: 915–920
  • Wang Y, Walsh S W. TNF alpha concentrations and mRNA expression are increased in preeclamptic placentas. J Reprod Immunol 1996; 32: 157–169
  • Reynolds L P, Redmer D A. Utero-placental vascular development and placental function. J Anim Sci 1995; 73: 1839–1851
  • Kendall R L, Wang G, Thomas K A. Identification of a natural soluble form of the vascular endothelial growth factor receptor, FLT-1, and its heterodimerization with KDR. Biochem Biophys Res Commun 1996; 226: 324–328
  • Lyall F, Greer I A. The vascular endothelium in normal pregnancy and pre-eclampsia. Rev Reprod 1996; 1: 107–116
  • Cheung C Y. Vascular endothelial growth factor: Possible role in fetal development and placental function. J Soc Gynecol Investig 1997; 4: 169–177
  • 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
  • Lyall F, Greer I A, Boswell F, Fleming R. Suppression of serum vascular endothelial growth factor immunoreactivity in normal pregnancy and in pre-eclampsia. Br J Obstet Gynaecol 1997; 104: 223–228
  • Vuorela P, Hatva E, Lymboussaki A, Kaipainen A, Joukov V, Persico M G, Alitalo K, Halmesmaki E. Expression of vascular endothelial growth factor and placenta growth factor in human placenta. Biol Reprod 1997; 56: 489–494
  • Athanassiades A, Lala P K. Role of placenta growth factor (PIGF) in human extravillous trophoblast proliferation, migration and invasiveness. Placenta 1998; 19: 465–473
  • Clark D E, Smith S K, He Y, Day K A, Licence D R, Corps A N, Lammoglia R, Charnock-Jones D S. A vascular endothelial growth factor antagonist is produced by the human placenta and released into the maternal circulation. Biol Reprod 1998; 59: 1540–1548
  • Taylor R N, de Groot C J, Cho Y K, Lim K H. Circulating factors as markers and mediators of endothelial cell dysfunction in preeclampsia. Semin Reprod Endocrinol 1998; 16: 17–31
  • Desai J, Holt-Shore V, Torry R J, Caudle M R, Torry D S. Signal transduction and biological function of placenta growth factor in primary human trophoblast. Biol Reprod 1999; 60: 887–892
  • Hayman R, Brockelsby J, Kenny L, Baker P. Preeclampsia: The endothelium, circulating factor(s) and vascular endothelial growth factor. J Soc Gynecol Investig 1999; 6: 3–10
  • Lash G E, Cartwright J E, Whitley G S, Trew A J, Baker P N. The effects of angiogenic growth factors on extravillous trophoblast invasion and motility. Placenta 1999; 20: 661–667
  • Reuvekamp A, Velsing-Aarts F V, Poulina I E, Capello J J, Duits A J. Selective deficit of angiogenic growth factors characterises pregnancies complicated by pre-eclampsia. Br J Obstet Gynaecol 1999; 106: 1019–1022
  • Torry D S, Ahn H, Barnes E L, Torry R J. Placenta growth factor: Potential role in pregnancy. Am J Reprod Immunol 1999; 41: 79–85
  • Charnock-Jones D S, Burton G J. Placental vascular morphogenesis. Baillieres Best Pract Res Clin Obstet Gynaecol 2000; 14: 953–968
  • Hornig C, Barleon B, Ahmad S, Vuorela P, Ahmed A, Weich H A. Release and complex formation of soluble VEGFR-1 from endothelial cells and biological fluids. Lab Invest 2000; 80: 443–454
  • Ong S, Lash G, Baker P N. Angiogenesis and placental growth in normal and compromised pregnancies. Baillieres Best Pract Res Clin Obstet Gynaecol 2000; 14: 969–980
  • 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
  • Zhou Y, McMaster M, Woo K, Janatpour M, Perry J, Karpanen T, Alitalo K, Damsky C, Fisher S J. Vascular endothelial growth factor ligands and receptors that regulate human cytotrophoblast survival are dysregulated in severe preeclampsia and hemolysis, elevated liver enzymes, and low platelets syndrome. Am J Pathol 2002; 160: 1405–1423
  • 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, Epstein F H, Sukhatme V P, Karumanchi S A. 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
  • Koga K, Osuga Y, Yoshino O, Hirota Y, Ruimeng X, Hirata T, Takeda S, Yano T, Tsutsumi O, Taketani Y. Elevated serum soluble vascular endothelial growth factor receptor 1 (sVEGFR-1) levels in women with preeclampsia. J Clin Endocrinol Metab 2003; 88: 2348–2351
  • Livingston J C, Chin R, Haddad B, McKinney E T, Ahokas R, Sibai B M. Reductions of vascular endothelial growth factor and placental growth factor concentrations in severe preeclampsia. Am J Obstet Gynecol 2000; 183: 1554–1557
  • Gaber L W, Spargo B H, Lindheimer M D. Renal pathology in pre-eclampsia. Baillieres Clin Obstet Gynaecol 1987; 1: 971–995
  • Kincaid-Smith P. The renal lesion of preeclampsia revisited. Am J Kidney Dis 1991; 17: 144–148
  • Wilczynski J R. Immunological analogy between allograft rejection, recurrent abortion and pre-eclampsia—the same basic mechanism?. Hum Immunol 2006; 67: 492–511
  • Irgens H U, Reisaeter L, Irgens L M, Lie R T. Long term mortality of mothers and fathers after pre-eclampsia: Population based cohort study. BMJ 2001; 323: 1213–1217
  • Meekins J W, Pijnenborg R, Hanssens M, van Assche A, McFadyen I R. Immunohistochemical detection of lipoprotein(a) in the wall of placental bed spiral arteries in normal and severe preeclamptic pregnancies. Placenta 1994; 15: 511–524
  • Katabuchi H, Yih S, Ohba T, Matsui K, Takahashi K, Takeya M, Okamura H. Characterization of macrophages in the decidual atherotic spiral artery with special reference to the cytology of foam cells. Med Electron Microsc 2003; 36: 253–262
  • Harsem N K, Roald B, Braekke K, Staff A C. Acute atherosis in decidual tissue: Not associated with systemic oxidative stress in preeclampsia. Placenta 2007; 28(8–9)958–964
  • Plutzky J. Inflammatory pathways in atherosclerosis and acute coronary syndromes. Am J Cardiol 2001; 88: 10K–15K
  • Libby P. Inflammation in atherosclerosis. Nature 2002; 420: 868–874
  • Hansson G K. Inflammation, atherosclerosis, and coronary artery disease. N Engl J Med 2005; 352: 1685–1695
  • Binder C J, Chang M K, Shaw P X, Miller Y I, Hartvigsen K, Dewan A, Witztum J L. Innate and acquired immunity in atherogenesis. Nat Med 2002; 8: 1218–1226
  • Reape T J, Groot P H. Chemokines and atherosclerosis. Atherosclerosis 1999; 147: 213–225
  • Mach F. The role of chemokines in atherosclerosis. Curr Atheroscler Rep 2001; 3: 243–251
  • Burke-Gaffney A, Brooks A V, Bogle R G. Regulation of chemokine expression in atherosclerosis. Vascul Pharmacol 2002; 38: 283–292
  • Shin W S, Szuba A, Rockson S G. The role of chemokines in human cardiovascular pathology: Enhanced biological insights. Atherosclerosis 2002; 160: 91–102
  • Sheikine Y, Hansson G K. Chemokines and atherosclerosis. Ann Med 2004; 36: 98–118

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.