718
Views
19
CrossRef citations to date
0
Altmetric
Original Article

Maternal serum concentrations of the chemokine CXCL10/IP-10 are elevated in acute pyelonephritis during pregnancy

, , MD, , , , , , , , & show all
Pages 735-744 | Received 23 Feb 2007, Accepted 30 May 2007, Published online: 07 Jul 2009

Abstract

Objective. Acute pyelonephritis is one of the most frequent medical complications of pregnancy, as well as a common cause of antepartum hospitalization. Interferon (IFN)-γ inducible protein, CXCL10/IP-10, is a member of the CXC chemokine family with pro-inflammatory and anti-angiogenic properties. The purpose of this study was to determine whether maternal serum concentrations of CXCL10/IP-10 change in patients with acute pyelonephritis during pregnancy.

Study design. This cross-sectional study was conducted to determine the difference in maternal serum concentrations of CXCL10/IP-10 in pregnant women with acute pyelonephritis (N = 41) and normal pregnant women (N = 89). Pyelonephritis was defined in the presence of a positive urine culture, fever, and maternal clinical signs; blood cultures were performed in 36 cases. Maternal serum concentrations of CXCL10/IP-10 were measured by a sensitive immunoassay. Non-parametric statistics were used for analysis.

Results. (1) The median serum concentration of CXCL10/IP-10 in pregnant patients with pyelonephritis was significantly higher than in normal pregnant women (median 318.5 pg/mL, range 78.8–2459.2 vs. median 116.1 pg/mL, range 40.7–1314.3, respectively; p < 0.001); (2) maternal median serum concentrations of CXCL10/IP-10 did not differ significantly among patients with acute pyelonephritis with and without bacteremia (positive blood cultures: median 362.6 pg/mL, range 100.2–2459.2 vs. negative blood cultures: median 298.9 pg/mL, range 108.5–1148.7, respectively; p = 0.3).

Conclusions. Pyelonephritis in pregnant women is associated with an increased maternal serum concentration of the chemokine CXCL10/IP-10.

Introduction

Acute pyelonephritis is one of the most frequent medical complications in pregnancy Citation[1], and a common cause of antepartum hospitalization Citation[2], with most of the cases occurring during the second trimester Citation[3]. Complications following an event of acute pyelonephritis during pregnancy include anemia Citation[3-5], transient renal dysfunction Citation[3], preterm labor Citation[1],Citation[6], sepsis Citation[3],Citation[7], and septic shock Citation[7-11]. Moreover, pregnant women with pyelonephritis are at increased risk of developing acute respiratory distress syndrome (ARDS) Citation[12-14].

The human interferon-inducible protein 10 (IP-10 or CXCL10) is a chemokine of the CXC family Citation[15]. A unique feature of some members of this type of chemokine 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[16-27].

IP-10 is inducible by pro-inflammatory stimuli such as interferon-γ (IFN-γ) Citation[28-39], tumor necrosis factor-α (TNF-α) Citation[37],Citation[40-46], viruses, and microbial products Citation[33],Citation[37],Citation[47-52], either directly or through nuclear factor-κB (NF-κB) Citation[48],Citation[49],Citation[53-56]. It has been proposed that this chemokine is also involved in the recruitment and potentiation of T helper 1 (Th1) responses Citation[57-59].

The objective of this study was to determine whether the maternal serum concentrations of IP-10 are different in normal pregnancies and pregnant women with acute pyelonephritis.

Methods

Study design

This cross-sectional study included normal pregnant women (N = 89) and pregnant patients with acute pyelonephritis (N = 41). 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[60]. Acute pyelonephritis was diagnosed in the presence of fever (temperature ≥38°C), clinical signs or symptoms of upper urinary infection (e.g., flank pain, costovertebral angle tenderness), pyuria, and a positive urine culture. Blood cultures were performed in 36 patients with pyelonephritis.

Patients were enrolled at the Detroit Medical Center/Hutzel Women's Hospital, MI, USA. All 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 employed to study the biology of inflammation (i.e., complement, flow cytometry), hemostasis, angiogenesis regulation, and growth factor concentrations in normal pregnant women and those with complications. Samples from these patients have been used for other studies of the biology of pyelonephritis and inflammation.

IP-10 (CXCL10) determinations

A specific and sensitive enzyme-linked immunoassay was used to determine concentrations of CXCL10/IP-10 in human maternal serum. Immunoassays for CXCL10/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 monoclonal antibodies specific for CXCL10/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 CXCL10/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 CXCL10/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 CXCL10/IP-10. The calculated inter- and intra-assay coefficients of variation (CVs) for CXCL10/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 employed to determine whether the data were normally distributed. Comparisons among groups were performed using the Mann–Whitney test. A p-value of <0.05 was considered statistically significant. The statistical package used was SPSS v.14.0 (SPSS Inc., Chicago, IL, USA).

Results

Clinical and obstetrical characteristics of women in each group are displayed in . The median serum concentration of CXCL10/IP-10 was significantly higher in pregnant patients with acute pyelonephritis than in normal pregnant women (acute pyelonephritis: median 318.5 pg/mL, range 78.8–2459.2 vs. normal pregnancy: median 116.1 pg/mL, range 40.7–1314.3; p < 0.001) ().

Figure 1. Maternal serum concentrations of CXCL10/IP-10 in patients with normal pregnancies and those with pyelonephritis. The median serum concentration of CXCL10/IP-10 in pregnant patients with pyelonephritis was significantly higher than in normal pregnant women (median 318.5 pg/mL, range 78.8–2459.2 vs. median 116.1 pg/mL, range 40.7–1314.3, respectively; p < 0.001).

Figure 1. Maternal serum concentrations of CXCL10/IP-10 in patients with normal pregnancies and those with pyelonephritis. The median serum concentration of CXCL10/IP-10 in pregnant patients with pyelonephritis was significantly higher than in normal pregnant women (median 318.5 pg/mL, range 78.8–2459.2 vs. median 116.1 pg/mL, range 40.7–1314.3, respectively; p < 0.001).

Table I.  Clinical and obstetrical characteristics of the study groups

The most common microorganism isolated from urine cultures of patients with pyelonephritis was Escherichia coli (30 cases). Other microorganisms less frequently isolated included mixed flora (2), Klebsiella pneumoniae (2), E. coli and Streptococcus viridans (1), Enterobacter aerogenes (1), Pseudomonas aeruginosa (1), Gram-negative bacilli (1), Proteus mirabilis (1), Streptococcus agalactiae (1), and Citrobacter koseri (1).

Among the 36 women with acute pyelonephritis in which blood cultures were performed, the results were positive in 16 cases (44.4%). The following microorganisms were isolated: E. coli (11), coagulase negative Staphylococcus (2), Gram-positive cocci (1), Klebsiella pneumoniae (1), and Enterobacter aerogenes (1).

Among patients with acute pyelonephritis, no significant difference in the median serum concentration of CXCL10/IP-10 was observed between those who had positive blood cultures and those who did not (positive blood cultures: median 362.6 pg/mL, range 100.2–2459.2 vs. negative blood cultures: median 298.9 pg/mL, range 108.5–1148.7, respectively; p = 0.3) ().

Figure 2. Maternal serum concentrations of CXCL10/IP-10 in patients with pyelonephritis. No significant differences in maternal median serum concentrations of CXCL10/IP-10 were observed between patients with positive and negative blood cultures (median 362.6 pg/mL, range 100.2–2459.2 vs. median 298.9 pg/mL, range 108.5–1148.7, respectively; p = 0.3).

Figure 2. Maternal serum concentrations of CXCL10/IP-10 in patients with pyelonephritis. No significant differences in maternal median serum concentrations of CXCL10/IP-10 were observed between patients with positive and negative blood cultures (median 362.6 pg/mL, range 100.2–2459.2 vs. median 298.9 pg/mL, range 108.5–1148.7, respectively; p = 0.3).

Patients with acute pyelonephritis delivered neonates with a lower birth weight than those in the control group (pyelonephritis: median 3140 g, range 1080–4090 g vs. normal pregnancy: median 3342 g, range 2550–4050 g; p = 0.02). However, the mean gestational age at delivery was not different between the two groups.

Discussion

Principal findings of this study

(1) The median serum concentration of CXCL10/IP-10 in pregnant women with acute pyelonephritis was significantly higher than that of normal pregnant women; (2) among patients with pyelonephritis, no significant difference in the median serum concentration of CXCL10/IP-10 was observed between those who had positive blood cultures and those who did not. This is the first report documenting an increase in the maternal serum concentration of CXCL10/IP-10 in women with an acute infection during pregnancy.

The chemokine family

The superfamily of chemokines includes secreted proteins with a molecular weight of 8–10 kD that selectively target and activate specific cell populations and attract them to inflamed tissues in which microbial invasion has occurred Citation[19],Citation[20],Citation[22],Citation[61],Citation[62].

Until recently, there was much discussion about the specificity of subsets of chemokines for a particular leukocyte population (e.g., neutrophils, lymphocytes, etc.), and whether or not a particular set of chemokines was involved in the disease processes with a characteristic type of white blood cell infiltration. Recent findings indicate that chemokines and their receptors are expressed by a wide variety of cells of non-hematopoietic origin, and that the functions of chemokines extend far beyond white blood cell attraction. A fundamental observation, which has added complexity to the field, is the ‘promiscuity’ of this family of molecules and their receptors. Indeed, chemokines can bind several different receptors, while chemokine receptors can bind more than one chemokine Citation[19].

Traditionally, two main subfamilies of chemokines have been described on the basis of the number and arrangement of the conserved cystine residues, the CC and the CXC chemokines Citation[19],Citation[20],Citation[22],Citation[61],Citation[62]. The CC chemokines preferentially attract monocytes, basophils, eosinophils, and lymphocytes, but have no effect on neutrophils Citation[63]. The CXC chemokines are subdivided according to the presence or absence of the glutamic acid–leucine–arginine (ELR) sequence. ELR-containing CXC chemokines (ELR+) are chemotactic for neutrophils and promoters of angiogenesis. In contrast, non-ELR containing CXC chemokines (ELR−) are chemotactic mainly for lymphocytes, and characterized by anti-angiogenic activity Citation[21],Citation[23-26],Citation[64],Citation[65].

The chemokine CXCL10/IP-10

IP-10 (CXCL10) is an ELR− CXC chemokine Citation[15] first described as the product of a gene induced in response to recombinant IFN-γ in several cell populations Citation[28].

The biological properties of IP-10 are mediated through the interaction with a trans-membrane G protein-coupled receptor, CXCR3 Citation[66],Citation[67], shared by two other IFN-γ-inducible CXC chemokines, CXCL9 (MIG) and CXCX11 (ITAC), whose distinct biological activities are related to different signal transduction pathways Citation[68-71].

CXCL10/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[28], neutrophils Citation[36],Citation[37], eosinophils Citation[37],Citation[72], keratinocytes Citation[28],Citation[31],Citation[32],Citation[34], fibroblasts Citation[28], endothelial cells Citation[28-30], pancreatic β cells Citation[38],Citation[39], and animal astrocytes/microglia Citation[33],Citation[35]. TNF-αCitation[37],Citation[40-46], interleukin (IL)-1βCitation[37],Citation[38], as well as viral Citation[33],Citation[48],Citation[49] and microbial products Citation[37],Citation[47],Citation[50-52], can also stimulate the production of IP-10. Interestingly, in vitro infection of epithelium of the urinary tract with E. coli results in an increase (as measured in the conditioned media with ELISA) in both CXCL10/IP-10 secretion and epithelial cell CXCL10/IP-10 mRNA expression Citation[73]. Activation of the NF-κB pathway Citation[48],Citation[49],Citation[53-56] after ligation of pattern recognition receptors (TLR4 Citation[50] and TLR3 Citation[56]) can also upregulate gene and protein expression of IP-10. Thus, IP-10 is considered an ‘NF-κB responsive gene’Citation[74].

IP-10 in inflammation

The pro-inflammatory activities of CXCL10/IP-10 include chemotaxis and endothelial adhesion of activated T cells Citation[75], as well as chemotaxis Citation[76] and enhancement of natural killer (NK) cell-mediated cytolysis Citation[76]. However, this chemokine is a poor neutrophil activator Citation[75],Citation[77], and its effects on monocytes Citation[66],Citation[75] and B cells Citation[78],Citation[79] are a matter of controversy.

Serum IP-10 in the presence of infections

Infection is associated with an increase in serum and/or plasma concentrations of CXCL10/IP-10. Evidence in support of this statement includes: (1) median serum concentrations of CXCL10/IP-10, determined on the first postnatal day, are significantly higher in neonates with perinatal infections than in controls (p < 0.001) Citation[80]; (2) median serum concentrations of CXCL10/IP-10 are significantly higher in neonates with nosocomial infections as compared to controls Citation[80]; (3) neonatal plasma IP-10 ≥1250 pg/mL has been reported to identify all preterm infants with sepsis and necrotizing enterocolitis Citation[81]; (4) it has been proposed that the severity of neonatal infection is associated with the magnitude of upregulation of circulating CXCL10/IP-10 (indeed, CXCL10/IP-10 plasma concentrations in infants who died of sepsis were all markedly elevated (range 23 817–34 415 pg/mL)) Citation[81]; and (5) in adults, plasma CXCL10/IP-10 concentration has been used to monitor the disease activity and the response to therapy of Mycobacterium tuberculosis infection Citation[82].

Interestingly, serum concentrations of CXCL10/IP-10 have been reported to be significantly higher in those with severe ARDS than in control patients (patients with bacterial pneumonia). Chemokine and cytokine dysregulation has been implicated in the pathogenesis of ARDS Citation[81],Citation[83],Citation[84], and CXCL10/IP-10 has been proposed as a useful marker of lung injury in patients with severe acute respiratory syndrome (SARS) Citation[85].

IP-10 in urinary tract infections

Previous studies have reported higher serum Citation[86] and urine Citation[86],Citation[87] CXCL10/IP-10 concentrations (determined by ELISA) in the presence of both culture-proven Gram-negative urosepsis Citation[86] and febrile urinary tract infections than in control patients Citation[73],Citation[87]. Furthermore, serum CXCL10/IP-10 concentrations did not change significantly during the follow-up (72 h after beginning of treatment). However, a significant decrease in the urinary concentrations was observed Citation[86]. In serum and in urine, CXCL10/IP-10 concentrations were not different in patients with positive or negative blood cultures Citation[86],Citation[87]. Of interest is that in experimental endotoxemia in humans, CXCL10/IP-10 concentrations were elevated in both serum and urine Citation[86]. The finding of increased concentrations in urine raises questions about how the changes in the peripheral circulation are reflected in urine. It is possible that this reflects renal excretion of increased available CXCL10/IP-10 in the peripheral circulation, local production, or both.

IP-10: Maternal serum concentrations increase in pregnancies complicated with pyelonephritis

This is the first report on the topic of serum CXCL10/IP-10 concentrations in pregnant women with pyelonephritis. Our finding that pyelonephritis in pregnancy is associated with a significant increase in the serum concentration of this chemokine is consistent with what has been previously reported in non-pregnant patients Citation[86]. The mechanisms responsible for the increase in CXCL10/IP-10 during normal pregnancy Citation[88] and in acute infection during gestation are unknown. In addition, the physiologic function of CXCL10/IP-10 during pregnancy as well as the role of CXCL10/IP-10 in acute infection during gestation remain to be determined. Recently, it has been demonstrated that CXCL10/IP-10 has antimicrobial properties Citation[89]. Thus, it is tempting to postulate that this chemokine participates in host defense not only by orchestrating leukocyte chemotaxis, but also by inducing other biological activities that may have a protective effect during infection. In this study, no significant differences were observed in the median maternal serum concentrations of CXCL10/IP-10 between patients with and without positive blood cultures. Otto et al. Citation[87] reported similar results when quantifying urinary CXCL10 in response to febrile urinary tract infections. Indeed, only some chemokines such as CXCL1, CXCL3, CXCL5, CXCL8, and CCL2, but not CXCL10/IP-10, were significantly higher in the presence of bacteremia. One interpretation is that the elevation in the serum concentration of CXCL10/IP-10 in the context of pyelonephritis results from systemic inflammation and it is not affected by the presence of microorganisms in the circulation. However, one possible explanation for this negative result is a limited statistical power.

IP-10: A local first line defense mechanism in the urinary tract mucosae

Migration of immunocompetent cells to sites of mucosal infection requires expression of molecules by the epithelial cells of the affected tissues Citation[90]. Chemokines, particularly those belonging to the CXC family, have been shown to play an important role in this process Citation[20],Citation[90-94]. Indeed, infections of the mucosae are associated with a local increase in CXCL10/IP-10. Evidence in support of this statement includes: (1) exposure of uroepithelial cells to E. coli is followed by a significant increase in CXCL10/IP-10 mRNA expression (quantification with an RNA protection assay) and protein secretion (ELISA) Citation[73]; (2) intravesical instillation of Mycobacterium bovis bacillus Calmette–Guerin (BCG) is followed by detection of urinary CXCL10/IP-10 in patients undergoing immunotherapy for bladder cancer (by ELISA) Citation[95],Citation[96]; (3) conditioned media collected from culture of human uroepithelial cell lines exposed to BCG or BCG-derived cytokines such as IFN-γ, IL-1β, and TNF-α contains a significantly higher amount of CXCL10/IP-10 (ELISA) than control media Citation[95]; and (4) intrapulmonary administration of K. pneumoniae in mice is followed by upregulation of CXCL10/IP-10 mRNA expression in the lung. Moreover, neutralization of CXCL10/IP-10 activity with an antibody results in reduced bacterial clearance and decreased survival, while in contrast, over-expression of CXCL10/IP-10 (via adenovirus) results in improved bacterial clearance Citation[97].

Antimicrobial activity of IP-10

Recent studies have reported structural similarities between chemokines and defensins (a family of antimicrobial peptides serving as the first line of defense in epithelia before the activation of adaptive immunity) Citation[98-105]. The following evidence suggests an overlap between the biological functions of defensins and CXCL10/IP-10: (1) chemokines share many properties with defensins such as structure, size, charge, disulfide bonding, and IFN-inducibility Citation[89]; (2) the CXC motif, present in CXCL10/IP-10, is contained and conserved in α-defensins-1 and -2; these defensins, as well as a structurally-related β-defensin, have been shown to be chemotactic for leukocytes at subnanomolar concentrations Citation[106-109]; (3) CXCL10/IP-10 has antimicrobial properties similar to those of defensins: in a radial diffusion assay, CXCL10/IP-10 has been demonstrated to have antimicrobial activity against E. coli and Listeria monocytogenesCitation[89]. Moreover, this antimicrobial activity, similarly to the activity of defensins and many other antimicrobial peptides Citation[110], is salt-dependent (inhibited by NaCl) Citation[89]; and (4) calculations of the amount of CXCL10/IP-10 produced by infiltrating inflammatory cells indicate that the tissue concentrations of this chemokine may reach the threshold necessary for microbicidal activity locally, and directly inactivate microbes before attracting other host defense cells to the area Citation[89].

IP-10, pyelonephritis, and lung injury

Compared to non-pregnant subjects with pyelonephritis, mothers with pyelonephritis are at an increased risk for ARDS Citation[7],Citation[12-14],Citation[111-115]. This has been attributed to an enhanced baseline activation of the innate immunoresponse during pregnancy Citation[116],Citation[117], although other explanations may be possible Citation[118]. Indeed, an excessive inflammatory response (in particular neutrophil activation) has been implicated in the pathogenesis of ARDS Citation[119]. Nevertheless, the precise mechanism for the association between pyelonephritis and ARDS remains to be determined. Neumann et al. Citation[119] caused lung injury in female mice by inducing continuous leakage of bacteria into the peritoneal cavity. The investigators demonstrated that this procedure causes, in as early as 3 hours, massive recruitment of granulocytes to the lung and a significant increase in lung CXCL10/IP-10 mRNA. The authors concluded that CXCL10/IP-10 may contribute to the accumulation of mononuclear phagocytes in the lung Citation[119]. Thus, CXCL10/IP-10 may be involved in the pathogenesis of ARDS in pregnant women with sepsis and/or pyelonephritis. This hypothesis is strengthened by the observation that in patients with SARS the serum concentrations of CXCL10/IP-10 increase before the development of chest involvement, reaching a peak concentration before abnormalities are detected in the chest X-ray Citation[85].

Conclusion

Maternal serum concentrations of CXCL10/IP-10 are higher in the context of acute bacterial infection (pyelonephritis) than in normal pregnancy. The role of this chemokine in host defense during infection requires further study.

Acknowledgements

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

References

  • Pitukkijronnakorn S, Chittacharoen A, Herabutya Y. Maternal and perinatal outcomes in pregnancy with acute pyelonephritis. Int J Gynaecol Obstet 2005; 89: 286–287
  • Wing D A. Pyelonephritis in pregnancy: Treatment options for optimal outcomes. Drugs 2001; 61: 2087–2096
  • Hill J B, Sheffield J S, McIntire D D, Wendel G D, Jr. Acute pyelonephritis in pregnancy. Obstet Gynecol 2005; 105: 18–23
  • Cox S M, Shelburne P, Mason R, Guss S, Cunningham F G. Mechanisms of hemolysis and anemia associated with acute antepartum pyelonephritis. Am J Obstet Gynecol 1991; 164: 587–590
  • Cavenee M R, Cox S M, Mason R, Cunningham F G. Erythropoietin in pregnancies complicated by pyelonephritis. Obstet Gynecol 1994; 84: 252–254
  • Gilstrap L C, Leveno K J, Cunningham F G, Whalley P J, Roark M L. Renal infection and pregnancy outcome. Am J Obstet Gynecol 1981; 141: 709–716
  • Cunningham F G, Lucas M J. Urinary tract infections complicating pregnancy. Baillieres Clin Obstet Gynaecol 1994; 8: 353–373
  • Cavanagh D, Rao P S. Septic shock (endotoxic shock). Clin Obstet Gynecol 1973; 16: 25–39
  • Dilworth E E, Ward J V. Bacteremic shock in pyelonephritis and criminal abortion. Obstet Gynecol 1961; 17: 160–167
  • Cavanagh D, Knuppel R A, Shepherd J H, Anderson R, Rao P S. Septic shock and the obstetrician/gynecologist. South Med J 1982; 75: 809–813
  • Mabie W C, Barton J R, Sibai B. Septic shock in pregnancy. Obstet Gynecol 1997; 90: 553–561
  • Cunningham F G, Leveno K J, Hankins G D, Whalley P J. Respiratory insufficiency associated with pyelonephritis during pregnancy. Obstet Gynecol 1984; 63: 121–125
  • Catanzarite V A, Willms D. Adult respiratory distress syndrome in pregnancy: Report of three cases and review of the literature. Obstet Gynecol Surv 1997; 52: 381–392
  • Cunningham F G, Lucas M J, Hankins G D. Pulmonary injury complicating antepartum pyelonephritis. Am J Obstet Gynecol 1987; 156: 797–807
  • 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: 27 348–27 357
  • 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 D, 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; 11: 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
  • 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
  • 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
  • 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
  • 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
  • Baggiolini M, Dewald B, Moser B. Human chemokines: An update. Annu Rev Immunol 1997; 15: 675–705
  • Sallusto F, Mackay C R, Lanzavecchia A. The role of chemokine receptors in primary, effector, and memory immune responses. Annu Rev Immunol 2000; 18: 593–620
  • Baggiolini M. Chemokines in pathology and medicine. J Intern Med 2001; 250: 91–104
  • 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
  • Strieter R M, Belperio J A, Keane M P. CXC chemokines in angiogenesis related to pulmonary fibrosis. Chest 2002; 122: 298S–301S
  • 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: 2009–2021
  • 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: 30 219–30 227
  • 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
  • Godaly G, Otto G, Burdick M D, Strieter R M, Svanborg C. Fimbrial lectins influence the chemokine repertoire in the urinary tract mucosa. Kidney Int 2007; 71: 778–786
  • 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
  • Fotopoulos S, Mouchtouri A, Xanthou G, Lipsou N, Petrakou E, Xanthou M. Inflammatory chemokine expression in the peripheral blood of neonates with perinatal asphyxia and perinatal or nosocomial infections. Acta Paediatr 2005; 94: 800–806
  • Ng P C, Li K, Chui K M, Leung T F, Wong R P, Chu W C, Wong E, Fok T F. IP-10 is an early diagnostic marker for identification of late-onset bacterial infection in preterm infants. Pediatr Res 2007; 61: 93–98
  • Azzurri A, Sow O Y, Amedei A, Bah B, Diallo S, Peri G, Benagiano M, D'Elios M M, Mantovani A, Del Prete, G. IFN-gamma-inducible protein 10 and pentraxin 3 plasma levels are tools for monitoring inflammation and disease activity in Mycobacterium tuberculosis infection. Microbes Infect 2005; 7: 1–8
  • Puneet P, Moochhala S, Bhatia M. Chemokines in acute respiratory distress syndrome. Am J Physiol Lung Cell Mol Physiol 2005; 288: L3–15
  • Strieter R M, Kunkel S L. Acute lung injury: The role of cytokines in the elicitation of neutrophils. J Investig Med 1994; 42: 640–651
  • Chien J Y, Hsueh P R, Cheng W C, Yu C J, Yang P C. Temporal changes in cytokine/chemokine profiles and pulmonary involvement in severe acute respiratory syndrome. Respirology 2006; 11: 715–722
  • Olszyna D P, Prins J M, Dekkers P E, De Jonge, E, Speelman P, Van Deventer S J, Van Der Poll, T. Sequential measurements of chemokines in urosepsis and experimental endotoxemia. J Clin Immunol 1999; 19: 399–405
  • Otto G, Burdick M, Strieter R, Godaly G. Chemokine response to febrile urinary tract infection. Kidney Int 2005; 68: 62–70
  • Gotsch F, Romero R, Friel L, Kusanovic J P, Espinoza J, Erez O, Than N G, Mittal P, Edwin S, Yoon B H. Cxcl10/Ip-10: A missing link between inflammation and Anti-Angiogenesis in preeclampsia?. Matern Fetal Med 2007, 167
  • Cole A M, Ganz T, Liese A M, Burdick M D, Liu L, Strieter R M. Cutting edge: IFN-inducible ELR-CXC chemokines display defensin-like antimicrobial activity. J Immunol 2001; 167: 623–627
  • Godaly G, Bergsten G, Hang L, Fischer H, Frendeus B, Lundstedt A C, Samuelsson M, Samuelsson P, Svanborg C. Neutrophil recruitment, chemokine receptors, and resistance to mucosal infection. J Leukoc Biol 2001; 69: 899–906
  • Hang L, Haraoka M, Agace W W, Leffler H, Burdick M, Strieter R, Svanborg C. Macrophage inflammatory protein-2 is required for neutrophil passage across the epithelial barrier of the infected urinary tract. J Immunol 1999; 162: 3037–3044
  • Olszyna D P, Florquin S, Sewnath M, Branger J, Speelman P, Van Deventer S J, Strieter R M, Van Der Poll T. CXC chemokine receptor 2 contributes to host defense in murine urinary tract infection. J Infect Dis 2001; 184: 301–307
  • Frendeus B, Godaly G, Hang L, Karpman D, Lundstedt A C, Svanborg C. Interleukin 8 receptor deficiency confers susceptibility to acute experimental pyelonephritis and may have a human counterpart. J Exp Med 2000; 192: 881–890
  • Godaly G, Hang L, Frendeus B, Svanborg C. Transepithelial neutrophil migration is CXCR1 dependent in vitro and is defective in IL-8 receptor knockout mice. J Immunol 2000; 165: 5287–5294
  • Luo Y, Chen X, O'donnell M A. Mycobacterium bovis bacillus Calmette–Guerin (BCG) induces human CC- and CXC-chemokines in vitro and in vivo. Clin Exp Immunol 2007; 147: 370–378
  • Poppas D P, Pavlovich C P, Folkman J, Voest E E, Chen X, Luster A D, O'donnell M A. Intravesical bacille Calmette–Guerin induces the antiangiogenic chemokine interferon-inducible protein 10. Urology 1998; 52: 268–275
  • Zeng X, Moore T A, Newstead M W, Deng J C, Kunkel S L, Luster A D, Standiford T J. Interferon-inducible protein 10, but not monokine induced by gamma interferon, promotes protective type 1 immunity in murine Klebsiella pneumoniae pneumonia. Infect Immun 2005; 73: 8226–8236
  • Yoshio H, Lagercrantz H, Gudmundsson G H, Agerberth B. First line of defense in early human life. Semin Perinatol 2004; 28: 304–311
  • Espinoza J, Chaiworapongsa T, Romero R, Edwin S, Rathnasabapathy C, Gomez R, Bujold E, Camacho N, Kim Y M, Hassan S, et al. Antimicrobial peptides in amniotic fluid: Defensins, calprotectin and bacterial/permeability-increasing protein in patients with microbial invasion of the amniotic cavity, intra-amniotic inflammation, preterm labor and premature rupture of membranes. J Matern Fetal Neonatal Med 2003; 13: 2–21
  • Svinarich D M, Gomez R, Romero R. Detection of human defensins in the placenta. Am J Reprod Immunol 1997; 38: 252–255
  • Svinarich D M, Wolf N A, Gomez R, Gonik B, Romero R. Detection of human defensin 5 in reproductive tissues. Am J Obstet Gynecol 1997; 176: 470–475
  • Cole A M, Ganz T. Human antimicrobial peptides: Analysis and application. Biotechniques 2000; 29: 822–826, 828,830–831
  • Papagianni M. Ribosomally synthesized peptides with antimicrobial properties: Biosynthesis, structure, function, and applications. Biotechnol Adv 2003; 21: 465–499
  • Gallo R L, Murakami M, Ohtake T, Zaiou M. Biology and clinical relevance of naturally occurring antimicrobial peptides. J Allergy Clin Immunol 2002; 110: 823–831
  • Chen H, Xu Z, Peng L, Fang X, Yin X, Xu N, Cen P. Recent advances in the research and development of human defensins. Peptides 2006; 27: 931–940
  • Territo M C, Ganz T, Selsted M E, Lehrer R. Monocyte-chemotactic activity of defensins from human neutrophils. J Clin Invest 1989; 84: 2017–2020
  • Yang D, Chen Q, Chertov O, Oppenheim J J. Human neutrophil defensins selectively chemoattract naive T and immature dendritic cells. J Leukoc Biol 2000; 68: 9–14
  • Chertov O, Michiel D F, Xu L, Wang J M, Tani K, Murphy W J, Longo D L, Taub D D, Oppenheim J J. Identification of defensin-1, defensin-2, and CAP37/azurocidin as T-cell chemoattractant proteins released from interleukin-8-stimulated neutrophils. J Biol Chem 1996; 271: 2935–2940
  • Yang D, Chertov O, Bykovskaia S N, Chen Q, Buffo M J, Shogan J, Anderson M, Schroder J M, Wang J M, Howard O M, et al. Beta-defensins: Linking innate and adaptive immunity through dendritic and T cell CCR6. Science 1999; 286: 525–528
  • Ganz T. Defensins and host defense. Science 1999; 286: 420–421
  • Elkington K W, Greb L C. Adult respiratory distress syndrome as a complication of acute pyelonephritis during pregnancy: Case report and discussion. Obstet Gynecol 1986; 67: 18S–20S
  • Soisson A P, Eldridge E, Kopelman J N, Duff P. Acute pyelonephritis complicated by respiratory insufficiency. A case report. J Reprod Med 1986; 31: 525–527
  • Towers C V, Kaminskas C M, Garite T J, Nageotte M P, Dorchester W. Pulmonary injury associated with antepartum pyelonephritis: Can patients at risk be identified?. Am J Obstet Gynecol 1991; 164: 974–978
  • Yazigi R, Lerner S, Tejani N. Association of acute pyelonephritis with pulmonary complications in pregnancy. A report of two cases. J Reprod Med 1990; 38: 562–564
  • Pruett K, Faro S. Pyelonephritis associated with respiratory distress. Obstet Gynecol 1987; 69: 444–446
  • Soto E, Richani K, Romero R, Espinoza J, Chaiworapongsa T, Nien J K, Edwin S, Kim Y M, Hong J S, Goncalves L, et al. Increased concentration of the complement split product C5a in acute pyelonephritis during pregnancy. J Matern Fetal Neonatal Med 2005; 17: 247–252
  • 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
  • Windsor A C, Mullen P G, Fowler A A, Sugerman H J. Role of the neutrophil in adult respiratory distress syndrome. Br J Surg 1993; 80: 10–17
  • Neumann B, Zantl N, Veihelmann A, Emmanuilidis K, Pfeffer K, Heidecke C D, Holzmann B. Mechanisms of acute inflammatory lung injury induced by abdominal sepsis. Int Immunol 1999; 11: 217–227

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.