Skip to main content

Main menu

  • Home
  • Content
    • Published Ahead of Print
    • Current Issue
    • JASN Podcasts
    • Article Collections
    • Archives
    • ASN Meeting Abstracts
    • Saved Searches
  • Authors
    • Submit a Manuscript
    • Author Resources
  • Editorial Team
  • Editorial Fellowship
    • Editorial Fellowship Team
    • Editorial Fellowship Application Process
  • More
    • About JASN
    • Advertising
    • Alerts
    • Feedback
    • Impact Factor
    • Reprints
    • Subscriptions
  • ASN Kidney News
  • Other
    • CJASN
    • Kidney360
    • Kidney News Online
    • American Society of Nephrology

User menu

  • Subscribe
  • My alerts
  • Log in
  • My Cart

Search

  • Advanced search
American Society of Nephrology
  • Other
    • CJASN
    • Kidney360
    • Kidney News Online
    • American Society of Nephrology
  • Subscribe
  • My alerts
  • Log in
  • My Cart
Advertisement
American Society of Nephrology

Advanced Search

  • Home
  • Content
    • Published Ahead of Print
    • Current Issue
    • JASN Podcasts
    • Article Collections
    • Archives
    • ASN Meeting Abstracts
    • Saved Searches
  • Authors
    • Submit a Manuscript
    • Author Resources
  • Editorial Team
  • Editorial Fellowship
    • Editorial Fellowship Team
    • Editorial Fellowship Application Process
  • More
    • About JASN
    • Advertising
    • Alerts
    • Feedback
    • Impact Factor
    • Reprints
    • Subscriptions
  • ASN Kidney News
  • Follow JASN on Twitter
  • Visit ASN on Facebook
  • Follow JASN on RSS
  • Community Forum
REVIEW
You have accessRestricted Access

Chemokines, Chemokine Receptors, and Renal Disease: From Basic Science To Pathophysiologic and Therapeutic Studies

STEPHAN SEGERER, PETER J. NELSON and DETLEF SCHLÖNDORFF
JASN January 2000, 11 (1) 152-176;
STEPHAN SEGERER
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
PETER J. NELSON
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
DETLEF SCHLÖNDORFF
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data Supps
  • Info & Metrics
  • View PDF
Loading

Abstract

Abstract. Leukocyte trafficking from peripheral blood into affected tissues is an essential component of the inflammatory reaction to virtually all forms of injury and is an important factor in the development of many kidney diseases. Advances in the past few years have highlighted the central role of a family of chemotactic cytokines called chemokines in this process. Chemokines help to control the selective migration and activation of inflammatory cells into injured renal tissue. Chemokines and their receptors are expressed by intrinsic renal cells as well as by infiltrating cells during renal inflammation. This study summarizes the in vitro and in vivo data on chemokines and chemokine receptors in renal diseases with a special focus on potential therapeutic effects on inflammatory processes.

“In the injured glomerulus increased capillary permeability is associated, as in other examples of inflammation, with a so-called increased stickiness of the endothelial cells. Circulating polymorphonuclear neutrophils adhere to these sticky walls and thus accumulate in the glomerulus” (1). This observation by Jones in the early 1950s describes a fundamental premise of this review. Namely, what makes the endothelium “sticky” at a specific site of the vasculature to a specific subset of inflammatory cells and then controls their subsequent transmigration and entry into renal tissue? Developments over the past 10 years have highlighted the roles of specific adhesion molecules and chemokines in this process (2,3,4,5,6).

Chemokines are a family of chemotactic cytokines that were first identified on the basis of their ability to induce the migration of different cell types, particularly those of lymphoid origin (7,8,9). A wealth of data has demonstrated that chemokines working in concert with selectins and integrins act as directional signals to sort and direct effector leukocyte migration (4,10,11). In addition, chemokines have also been shown to activate leukocytes, influence hematopoiesis, and modulate angiogenesis (12,13,14).

The receptors for chemokines are expressed in a cell type-specific manner and are restricted primarily to subsets of leukocytes (15). The discovery that some chemokine receptors act as coreceptors for HIV entry into target cells has accelerated research in this field (16,17,18,19,20,21,22). Advances over the past few years have included the discovery of new chemokines, receptors, and antagonists, and a greater appreciation for the diverse biologic functions displayed by this cytokine family (15,23,24,25). Several recent reviews have dealt with the basic biology and the roles of chemokines in various disease processes (2,3,4,9,15,26,27,28). This review will focus on the biology of the chemokine and chemokine receptor families in the context of renal diseases.

Chemokines: Classification and Mechanisms of Action

The Chemokine Superfamily Is Comprised of Four Members

More than 40 chemokines and 17 chemokine receptors have been described to date, with additional candidates currently under investigation (Tables 1 and 2) (3,4,15,27,29). Chemokines are characterized by a series of shared structural determinants including conserved cysteine residues that form disulfide bonds in the chemokine tertiary structure (29,30). The chemokines were initially subdivided into two branches based on the position of these cysteine residues (9,15,27). In the CXC chemokine family, the first two cysteine residues in the primary amino acid sequence are separated by a single amino acid (X represents any intervening amino acid residue). In the CC subfamily, the cysteine residues lie next to each other. Although most of the chemokines belong to one of these two classes, two additional branches of the chemokine superfamily each containing a single member have been described in recent years (31,32). The C chemokine lymphotactin lacks both the first and third cysteine in the “4 cysteine motif,” but shares homology at its carboxyl end with the CC chemokines (32). Fractalkine has three intervening amino acids between the first two cysteine residues (31). This CX3C chemokine is tethered directly to the cell membrane via a long mucin stalk and has recently been shown to combine the function of a chemokine and an adhesion molecule (33).

View this table:
  • View inline
  • View popup
Table 1.

CC chemokine receptors, their tissue distribution, and ligandsa

View this table:
  • View inline
  • View popup
Table 2.

DARC, CXC, CX3C, and C chemokine receptors, their tissue distribution, and ligandsa

Some CXC chemokines display an additional structural designation, namely, the amino acid motif E-L-R-CXC (glutamic acid-leucine-arginine-cysteine-X-cysteine) just proximal to their first two cysteine residues. The E-L-R-CXC chemokines act primarily as neutrophil chemoattractants (30). In contrast, the CXC chemokines that lack the E-L-R motif bind different CXC receptors and are active on lymphocytes (9,30).

Chemokines are involved in more than the control of cell migration. Melanoma growth stimulating activity/growth-related oncogene-α (GRO-α) was originally identified as an autocrine growth factor for malignant melanoma cells (30,34). Interferon-inducible protein-10 (IP-10) has been shown to induce the proliferation of mesangial cells (35). Interleukin-8 (IL-8) can cause release of granules and respiratory burst in neutrophils (12). Regulated upon activation, normal T cell expressed and secreted (RANTES) can induce eosinophil and basophil degranulation, respiratory burst in eosinophils (36), and has been shown to augment T cell proliferation (37). Platelet factor 4 (PF4) inhibits megakaryopoiesis (38) and can be bactericidal (39). In addition, some chemokines are involved in hematopoiesis (13,40,41,42). A novel role for some classes of chemokines and their receptors as anti-inflammatory mediators has recently been suggested (43). The CXC chemokines IL-8, epithelial cell-derived neutrophil attractant 78 (ENA-78), and GRO-α,β,γ, which contain the E-L-R-CXC motif, have been shown to act as angiogenic agents (44), while PF4, IP-10, monokine induced by interferon-γ (MIG), and stromal cell-derived factor-1 (SDF-1), which lack this motif, can act as angiostatic factors (14). During embryogenesis, wound healing, chronic inflammation, and tumor growth, the expression of chemokines may help to determine the micro-vascularization within the tissue.

Regulation of Chemokine Expression

Chemokines are regulated at transcriptional, posttranscriptional, translational, and posttranslational levels (2). Many, but not all, of the proinflammatory chemokines are induced by IL-1β or tumor necrosis factor-α (TNF-α). Some CXC chemokines (MIG, IP-10) were initially identified on the basis of their specific upregulation by interferon-γ (IFN-γ) (45). Other chemokines, especially those that play a role in normal leukocyte trafficking, appear to be constitutively expressed (SDF-1, B cell attracting chemokine-1, thymus-expressed chemokine, secondary lymphoid tissue chemokine, EBI1 ligand chemokine, hemofiltrate CC chemokine-1, liver and activation-regulated chemokine) (28).

Some of the best-studied proinflammatory chemokines (e.g., IL-8, RANTES, monocyte chemoattractant protein-1 [MCP-1]) are controlled at the transcriptional level by the transcription factors nuclear factor-κB (NF-κB), CAAT enhancer binding protein, and activator protein-1 (21,46,47). Their activation requires a complex cascade of steps including phosphorylation by multiple kinases and phosphatases, degradation of transcriptional inhibitors, translocation of transcription factors from cytoplasm to nucleus, etc. (4). These signaling pathways can be different for each stimulus and transcription factor and are further complicated by “cross-talk” between the various pathways (48). Although at first glance this may appear to be an unnecessarily complicated system, it allows the fine-tuning and integration of the multiple signals required for a complex signal- and tissue-specific biologic response. These biochemical events are potential targets for therapeutic intervention. For example, glucocorticoids interfere with NF-κB activation (49,50). Similarly, agents that influence cAMP levels often modulate the stimulatory signals for chemokine expression (51,52,53). The inhibition of chemokine expression seen after treatment with free radical scavengers may be due to interference with NF-κB activation, but may also involve other redox-related steps (54).

Selective Expression of Chemokine Receptors Contributes to Cell Specificity of Chemokine Action

The biologic actions of chemokines (Table 3) are mediated through a family of G protein-coupled receptors with seven transmembrane domains (4,25,55,56). The nomenclature used to describe these receptors is based on the class of chemokine ligands that interact with the receptor (i.e., C, CC, CXC, and CX3C receptors) (27). Many chemokines bind to several different receptors (Tables 1 and 2) (57). The differential expression of the receptors by distinct leukocyte subsets is an important component of the specificity of chemokine action (27,58). The complexity and apparent redundancy of the system is thought to provide a high degree of effectiveness and flexibility in vivo (57).

View this table:
  • View inline
  • View popup
Table 3.

Principles of chemokine action

Binding of a chemokine to its receptor activates a signal transduction cascade leading to activation of phospholipase Cα1 and α2, and the production of inositol (1,4,5)-trisphosphate and diacylglycerol (2,4). In addition, a rapid and transient increase in intracellular calcium, and the activation of protein kinase C are observed (59,60,61). A variety of kinases may be involved in signal transduction, including serine/threonine kinases (e.g., members of the mitogen-activated protein kinase cascade) as well as tyrosine protein kinases (59). The various stimulatory and inhibitory pathways involved in these activation cascades are providing insight into the design of potential pharmacologic agents.

The Duffy Antigen Receptor for Chemokines

The Duffy antigen receptor for chemokines (DARC) is a “promiscuous” receptor-like structure that binds the CXC family proteins IL-8 (62,63), GRO (63), PF4 (64), neutrophil activating peptide-2, as well as the CC chemokines MCP-1 (62,63) and RANTES (63). DARC, first identified as the Duffy blood group antigen, is expressed on erythrocytes and endothelial cells of postcapillary venules of the kidney (64,65,66). DARC mediates Plasmodium vivax entry into red blood cells, and DARC-“negative” individuals (generally of central African lineage) are resistant to this form of malaria but still express DARC on their endothelial cells (67). At present, a function for the erythroid-expressed form of DARC is not known. Chemokine-induced signal transduction through DARC has not been demonstrated, and DARC is not coupled to G proteins. One hypothesis is that DARC may act as a scavenger that helps to clear chemokines from the circulation (62). DARC protein expressed on postcapillary venules may act as a presentation-like structure for chemokines on these surfaces, and hypothetically could contribute to induced leukocyte adhesion and transmigration at these sites.

Chemokines, Chemokine Receptors, and Th1, Th2 Immune Responses

The immune system mounts distinct and selective immune responses to different types of infection or antigenic challenge. These responses have been termed Th1-like and Th2-like after the two classes of T helper cells (Th) involved (68,69,70). Th1 and Th2 responses appear to represent the extremes of a spectrum of immune responses. Th1 responses are stimulated by pathogens that invade and inhabit cells and result in activation of cytotoxic T lymphocytes and delayed-type hypersensitivity. The Th1 subtype produces cytokines that stimulate strong cellular immune responses (IFN-γ, IL-2, leukotriene A, granulocyte macrophage colony-stimulating factor). The Th2 subtypes produce cytokines that evoke strong antibody responses (IL-3, -4, -5, -6, -10, and -13). In addition, Th2 cytokines can inhibit the inflmmatory reactions induced by Th1 cytokines. A third subtype called Th0 secretes cytokines of both types and is believed to give rise to the “polarized” Th1 and Th2 lineages. The recently characterized Th3/T-regulatory-1 T cell subset is thought to downregulate antigen-presenting cells, possibly via transforming growth factor-β (TGF-β) (71).

Growing evidence suggests that differential chemokine receptor expression may be crucial for the generation of a Th1- or Th2-type immune response, because specific chemokine receptor expression has been shown to characterize these T helper cell subtypes (72). Th1 cells appear to preferentially express the chemokine receptors CXCR3 and CCR5, while Th2 cells display CCR4, CCR8, and some CCR3 (28,72,73). The chemokine receptors expressed by different populations of T cells may thus dictate to a significant degree the tissue infiltration of Th1 and Th2 cells and the direction of the eventual immune response.

This is an important issue in the interpretation of results obtained from animal models. For example, different inbred mouse strains often demonstrate a more pronounced Th1- or Th2-like response. After an immunologic insult, C57BL/6 mice show a more Th1-like bias compared with Balb/C mice, which show a more Th2-like response (74,75). This can influence the composition of the cellular infiltrate and subsequently the course of the renal disease model (75).

Chemokines Control Important Aspects of Acute and Chronic Inflammation

Inflammation is a process involving changes in hemodynamics, vascular reaction of endothelial cells, leukocyte adhesion, activation, and migration (76). The nature of the inflammatory response is dictated by the pathogenic insult. The process of leukocyte trafficking from the peripheral circulation into tissue spaces involves a series of interactions between soluble mediators and surface molecules expressed by the endothelium and leukocyte, as well as subsequent interactions with the extra-cellular matrix (77). Chemokines mediate events at multiple stages in this process (78,79). The early events in this process entail the rolling of leukocytes along the microvascular surface through transient interactions between vascular addressins and selectins (80). The addressin/selectin-dependent cell-cell interaction is essential in leukocyte homing and promotes transient surface contact between the rolling leukocyte and the endothelial surface. The endothelium provides a selective interface between the peripheral circulation and extravascular space and ultimately acts as a discriminator of leukocyte infiltration.

During inflammation, the endothelium upregulates chemokine presentation structures (such as specific glucosaminoglycans), selectin ligands (addressins), selectins, and the Ig partners of leukocyte-expressed integrins (11,81). Thus, inflamed microvascular endothelium increases its capacity to bind chemokines and upregulates the expression of molecules required for the efficient rolling and firm adhesion of leukocytes.

Early in inflammation, as the microvascular endothelium becomes activated, chemokines generated by endothelial cells, subendothelial tissue, or released after platelet activation bind to the “activated” endothelial surface (78,82). Thus situated chemokines act as directional signals for leukocytes as they roll across the endothelium. A notable exception appears to be fractalkine, which is a membrane-bound chemokine that also enables direct leukocyte adhesion via its receptor CX3CR1 (33). Chemokines can trigger activation of leukocyte-expressed integrins, resulting in the arrest and firm adhesion of leukocytes to the activated endothelial surface. Important adhesion molecule pairs in this process include the selectins (E, L, and P), the Ig-like molecules intercellular adhesion molecule-1 and vascular cell adhesion molecule-1, and the β2 and β1 integrins (e.g., leukocyte function-associated antigen-1 and very late antigen-4) (80). (Note: The nomenclature of these factors is often confusing as it reflects the manner in which the proteins were first identified rather than their structural characteristics.)

After firm adhesion, leukocytes undergo spreading, diapedesis, extravasation, and migration into interstitial spaces. Although some chemokines are important for the control of leukocyte arrest, other chemokines appear to influence the subsequent events associated with leukocyte emigration. Leukocyte emigration in vivo occurs in a complex background of chemotactic signals where several receptors may be activated simultaneously or successively. Thus, the migrating leukocyte must distinguish among the various chemotactic signals within the three-dimensional environment of the tissue to move both up and down gradients to eventually reach the site of inflammation (21,83)

Chemokines May Play a Role in the Resolution or Progression of Inflammatory Processes

As leukocytes reach the site of inflammation and undergo activation, they can produce additional proinflammatory factors resulting in propagation and amplification of the inflammatory response. The accumulation of specific leukocytes at the site of injury normally results in removal of the initiating insult (e.g., phagocytosis of bacteria, immune complexes, apoptotic cells, cell debris, etc.) and tissue repair. Inactivation and removal of inflammatory effector cells once their goal has been accomplished are important to prevent chronic inflammation and progressive tissue destruction. Factors that govern the termination of the inflammatory response are thought to involve the downregulation of chemokine synthesis by local factors (e.g., prostaglandins, TGF-β) or a specific combination of inflammatory mediators leading to local apoptosis of the leukocytes involved (84).

During renal diseases, the infiltration of monocytes/macrophages and T cells into kidneys is thought to play a central role in progressive interstitial fibrosis and the progression of chronic renal failure (85). During this process, a cross-talk between cytokines, vasoactive substances, chemokines, and their respective target cells takes place. This interaction contributes to the outcome, i.e., healing or progression of the renal disorder. Chemokines appear to be integral players in this complex and dynamic process.

Transgenic and Knockout Mice: Models of Chemokine and Chemokine Receptor Action

The targeted disruption of chemokine and chemokine receptor genes as well as the expression of chemokines as transgenes has identified important roles for these molecules in specific aspects of the inflammatory response.

The ability of chemokines to direct leukocyte infiltration depends on a low regional expression level. In studies with MCP-1 transgenic mice, the systemic overexpression of a mouse mammary tumor virus long terminal repeat-MCP-1 transgene led to a general paralysis of the response to this chemokine (86). Only the local production as seen in insulin promoter-MCP-1 transgenic mice was shown to be effective in selectively recruiting monocytes (86). Besides persistent MCP-1 expression and insulitis, the macrophage infiltrate did not result in progressive tissue destruction. Therefore, additional costimulatory signals are required for the activation of macrophages.

Knockout mice with a targeted disruption for either MCP-1 or the MCP-1 receptor CCR2 gene show a reduced capacity to recruit monocytes and have suggested a fundamental role for these molecules in the generation of atherosclerotic lesions (87,88,89). In addition, CCR2(-/-) mice show significant defects in delayed-type hypersensitivity responses and in the production of Th1-type cytokines (89). Mice deficient for CCR5 show an increased humoral response to T cell-dependent antigenic challenge, suggesting a novel role for CCR5 in downmodulating T cell-dependent immune responses (90). Targeted disruption of the eotaxin gene demonstrated that eotaxin enhances the magnitude of early but not late eosinophil recruitment after antigen challenge (91). CXCR4 is broadly expressed by cells of the immune and central nervous system and mediates the migration of resting leukocytes and hematopoietic progenitors in response to its ligand SDF-1 (92). Essentially identical lethal defects were seen in mice deficient for either CXCR4 or SDF-1 (93,94,95). These include severely reduced B lymphopoiesis, reduced myelopoiesis in bone marrow, defective formation of the large vessels supplying the gastrointestinal tract, as well as cardiac defects and abnormal cerebellar development (93,94,95). In normal mice, CXCR5 is expressed on mature B cells and a subpopulation of T helper cells. CXCR5 knockout mice lack inguinal lymph nodes and possessed little or no normal Peyer's patches. Lymphocyte migration into splenic follicles of these mice were significantly impaired (96). Information obtained with chemokine and chemokine receptor knockout in models of renal diseases is discussed in later sections.

Chemokines, Chemokine Receptors, and Viral Biology

Given the important roles of chemokines in diverse immune processes, it is not surprising that viruses have exploited chemokine biology through the course of evolution. This phenomenon is emphasized by the large number of chemokine receptors, chemokines, and general inhibitors or modulators of chemokine action that are encoded by different viral genomes (21,60,97,98,99,100,101).

One of the most dramatic developments in the viral/chemokine connection is the observation that select chemokine receptors act as coreceptors for HIV entry into target cells (16,17,18,19,20). The chemokine ligands for these receptors can suppress infection by some strains of HIV-1 and may act as important moderators of HIV replication in vivo (21). CCR5 represents the major coreceptor for macrophage-tropic strains of HIV-1, whereas CXCR4 acts as a coreceptor for T celltropic strains. CCR5 appears to play a unique role in viral pathogenesis, as individuals homozygous for a nonfunctional allele of CCR5 (CCR5▵32) are highly protected from HIV-1 infection (21). These results have opened new avenues of research into the pathogenesis of HIV and have led to the development of new strategies to block viral fusion.

Chemokines, Chemokine Receptors, and the Kidney

All Types of Renal Cells Can Express Chemokines upon Stimulation

Nearly 10 years ago, we and others described the induced expression of chemokines by mesangial cells (Tables 4 and 5). An expanding body of data now strongly suggests that chemokines contribute to inflammatory glomerular as well as tubulointerstitial diseases (2,53,102,103,104,105,106,107,108,109,110). All types of renal cells (i.e., endothelial, mesangial, tubular epithelial, interstitial cells, and podocytes) are able to produce chemokines in a cell- and stimulus-specific manner. Details of the in vitro regulation of various chemokines can be found in Tables 4 and 5 and in several recent reviews (2,3). In general, proinflammatory stimuli such as TNF-α, IL-1β, IFN-γ, and lipopolysaccharide (LPS), especially when used in combination, rapidly (within a few hours) induce MCP-1, IL-8, and IP-10 (53, 104, 111,112,113). The induction of RANTES occurs in a more delayed manner (12 to 48 h). IgG and IgA immune complexes can also induce upregulation of MCP-1, RANTES, IL-8, and IP-10 expression by mesangial cells (Tables 4 and 5). Reactive oxygen species are able to upregulate chemokine expression and may represent a common mechanism of injury-induced chemokine generation (106).

View this table:
  • View inline
  • View popup
Table 4.

CXC chemokines produced by renal cellsa

View this table:
  • View inline
  • View popup
Table 5.

CC chemokines produced by renal cellsa

Growth factors such as platelet-derived growth factor and basic fibroblast growth factor can induce MCP-1 and RANTES expression by mesangial cells (114,115). This expression may be related to the macrophage influx seen during proliferative responses related to tissue repair, regeneration, and remodeling (114). MCP-1 expression by proximal tubular cells is seen after exposure to hyaluronan, a glucosaminoglycan degradation product of extracellular matrix that accumulates in the interstitium during kidney diseases (116). The interaction of CD40 with its ligand (CD154), together with IL-4 and IL-13, results in MCP-1, RANTES, and IL-8 generation by cultured proximal tubular cells (117), observations that may be relevant for tubulointerstitial inflammatory cell infiltrates in transplant rejection and other forms of interstitial diseases.

The generation of chemokines by proximal tubular cells can be induced by albumin, which is thought to mimic the effects of proteinuria and may be related to the tubulointerstitial damage seen in glomerular disease (118,119,120). Wang et al. described an increase of MCP-1 mRNA expression in proximal tubular cells in response to delipidated bovine serum albumin (BSA), holotransferrin, and apotransferrin, which was mediated by NF-κB (118,120). Lysine, an inhibitor of protein uptake, reduced the MCP-1 expression (118). Zoja et al. found a dose-dependent increase of RANTES expression by proximal tubular epithelial cells exposed to BSA (119). However, this effect required very high albumin concentrations (i.e., 10 to 30 mg/ml), probably not achieved in proximal tubular fluids. Although the significance of these in vitro effects of albumin on tubular epithelial cell chemokine production for the nephrotic syndrome remains hypothetical, it does suggest that the prolonged proteinuria and proximal tubular epithelial overload of proteins (“nephrosis”) may lead to tubular cell activation and chemokine induction. Furthermore, the role of lipids in the nephrotic syndrome and of lipid droplets in proximal tubules deserves attention in this context, as lipid oxidation may occur and oxidized lipids can stimulate chemokine production (121).

Vasoactive Agents Can Stimulate Chemokine Expression

Vasoactive agents and chemokines may directly interact. Wolf et al. found that angiotensin II via the AT2 receptor could stimulate RANTES production by glomerular endothelial cells (122). Moreover, the infusion of angiotensin II into rats led to an influx of macrophages into the glomerulus, which was attenuated by the administration of an AT2 receptor antagonist (122). The differential regulation of RANTES and MCP-1 could explain the apparent divergent reports from other groups implicating the AT1 rather than the AT2 receptor in “chemokine” stimulation (122,123,124,125).

The Production of Proinflammatory Chemokines Is Transitory

Some chemokines appear to be constitutively expressed (i.e., those that control normal leukocyte trafficking). The proinflammatory chemokines appear to be kept under tight regulatory control and are expressed only in response to specific stimuli (Tables 4 and 5). The basal expression of chemokines often seen in cultured renal cells may represent a tissue-culture artifact, as serum and endotoxin (a common contaminant of growth media) are potent stimuli for chemokine induction (112).

It is thought that the expression of proinflammatory chemokines normally follows a self-limited course. Interestingly, different time frames for the expression of various chemokines in response to the same stimulus have been reported (112,126). In general, chemokines that are rapidly induced (e.g., MCP-1) return to “baseline” within a day. Stimulation with a combination of cytokines (e.g., TNF-α, IL-1β, and IFN-γ) often results in a more prolonged expression (114,115). RANTES expression can be slower with an increase seen after 12 to 24 h, but the levels may remain elevated for days (114). The different time courses suggest different signal transduction events used for the individual chemokines.

Inhibition of Chemokine Expression

The expression of inflammatory chemokine genes can be inhibited by glucocorticoids (127,128), cytokines such as TGF-β (129), and prostaglandins (e.g., PGE2) (53). The inhibitory effect of prostaglandin appears to involve modulation of the second messenger cAMP (51,52,53,130). Because TGF-β and prostaglandins are generated at sites of tissue injury, the net effect on local chemokine generation may depend on the balance between proinflammatory and anti-inflammatory agents. The dual character of TGF-β is again illustrated by the report that this cytokine can inhibit MCP-1 expression by proximal tubular cells and at the same time enhanced synthesis of the neutrophil-specific chemokine IL-8 (129).

Renal Cells May Also be Targets for Chemokines

Renal cells also express chemokine receptors. Thymus-derived chemotactic agent-4 (TCA4), a mouse chemokine, can bind to mouse mesangial cells and mediate chemotaxis and proliferation (131). The TCA4-specific receptor expressed by mesangial cells has not yet been identified (131,132). In a human mesangial cell line, stimulation with IFN-γ led to induction of functional CCR1 expression (112). The activated mesangial cells were found to migrate in response to RANTES (112). In these experiments, no expression of CCR3, CCR4, CCR5, and CCR8 could be detected (112).

Romagnani et al. demonstrated the expression of CXCR3 both in cultured human mesangial cells and renal biopsies (35). The mesangial cells were shown to flux Ca2+, migrate in response to IP-10, and proliferate in response to MIG (35). The overall function of chemokine receptor expression by intrinsic renal cells remains to be defined, but could play a role in mesangial cell migration-proliferation during mesangial repair and could even function as local immune modulator. It is hoped that these questions can be addressed with receptor knockout models.

Expression of Chemokines and Chemokine Receptors in Experimental Models of Renal Diseases

Animal models have been helpful in identifying potential roles for chemokines in the initiation and propagation of renal disease. The results of these experiments are sometimes difficult to interpret due to differences in the genetic backgrounds of the animals used. The genetic background can profoundly influence the susceptibility, type of inflammatory infiltrate, and the eventual course of renal disease (133). The importance of adequate backcrossing cannot be overstated because a significant part of experimental findings assigned to the knockout phenotype are often explainable by the different genetic backgrounds used (134,135). The genetic background may also be important because of polymorphisms in the chemokine gene that may influence the expression of the chemokine (136,137).

Nephrotoxic Serum Nephritis

Many groups have used the classic model of nephrotoxic serum nephritis (NTS-GN) induced by the injection of antibodies against glomerular basement membranes for the study of chemokine expression and action (138,139,140,141). Mice with accelerated NTS-GN show increased expression of RANTES (138,139,141,142), MCP-1 (138,139,140,141,142), IP-10 (141,142), and MCP-3 (140) mRNA, whereas no change was seen in macrophage inflammatory protein-1α (MIP-1α) expression (139).

The biologic action of different chemokines has been studied through the use of neutralizing antibodies and specific chemokine antagonists. Lloyd et al. blocked RANTES-associated events in NTS-GN in CD1 mice (an outbred mouse strain) by the use of the antagonist Met-RANTES. Animals treated with Met-RANTES showed reduced proteinuria, reduced T cell infiltration, and mononuclear cell infiltration. However, glomerular crescent formation was not significantly reduced. Treatment with a neutralizing MCP-1 antibody led to a reduction in proteinuria, infiltrating macrophages, and a striking decrease in crescent formation and collagen production, suggesting that MCP-1, and not RANTES, is involved in crescent formation and early interstitial fibrosis (139). This issue was also examined in MCP-1 knockout mice. Tesch et al. induced NTS-GN in F1 generation littermates of MCP-1 knockout 129SV/J and wild-type C57BL/6 (i.e., a mixed genetic background). The animals developed a glomerular injury consisting of hyalinosis, capillary dilation and thickening, focal segmental sclerosis, and crescents (140). Glomerular hypertrophy or hypercellularity was not present (140). The lack of glomerular chemokine expression in this model is consistent with the lack of glomerular leukocyte infiltration, while the predominant tubular MCP-1 expression was associated with tubulointerstitial cell infiltrate (140). In the MCP-1-deficient F1 generation (129SV/JXC57BL/6), a reduced macrophage infiltrate adjacent to tubules was seen. The authors suggest that tubular epithelial cells represent the primary source of MCP-1 protein in this model. MCP-1 so positioned could act to recruit peritubular macrophages that could then promote tubular interstitial injury (140). Hisada et al. used the NTS-GN model in mice to evaluate the role of angiotensin II type 1a receptor (AT1a) by use of AT1a receptor-deficient mice (AT1a(-/-)) (125). AT1a-deficient mice were backcrossed with C57BL/6 for five generations (representing adequate backcrossing) (125). In these animals, a peak of MCP-1 mRNA expression was seen after 6 h. An additional increase in MCP-1 after 14 d, which was stronger in the AT1a(+/+) than in the AT1a(-/-) mice, was also seen (125). The higher expression of MCP-1 in wild-type mice was associated with a more severe disease course (125). The authors propose that angiotensin II via the AT1a receptor mediates the late MCP-1 expression and may play a role in progression of immune-mediated renal injury.

NTS-GN results in induction of CCR1, CCR2, and CCR5 that is associated with the expression of corresponding chemokine mRNA for MCP-1, RANTES in CD1 mice (142). Another recent abstract indicates that mice deficient in CCR1 show more severe renal impairment and proteinuria than the wildtype mice, suggesting a novel immune modulatory role for CCR1 (141).

In Wistar-Kyoto rats, the injection of NTS-GN antibodies leads to crescentic glomerulonephritis in which CD8+ T cells are thought to play a major role (143). In this model, an upregulation of MCP-1 (143,144,145,146,147,148,149,150,151,152), MIP-1α (143,148,149,151), MIP-1β (143,148,151), RANTES (145,150,151), MCP-3 (151), CINC (148,149), MIP-2 (148,149,153), PF4 (149), TCA3 (151), fractalkine (154), lymphotactin (155), and IP-10 mRNA (149) was detected (Table 6).

View this table:
  • View inline
  • View popup
Table 6.

Expression of chemokines in animal models of kidney diseases

A correlation between the expression of neutrophil-attracting chemokines and an early neutrophil influx and the later expression of macrophage-attracting chemokines followed by a macrophage influx was described (148,149). The MCP-1 mRNA was localized to intrinsic glomerular cells after 3 h. After 24 h, the predominant source of MCP-1 was from infiltrating monocytes/macrophages (156). MCP-1 protein (156) was demonstrated in glomeruli (144,150,152,157), vascular endothelial cells (157), and tubular epithelial cells (157). Expression of RANTES (150), MIP-1α (148), MIP-1β (150), and MIP-2 (153) protein was also demonstrated. Recently, the upregulation of fractalkine mRNA, as well as an induction of the fractalkine receptor (CX3CR1) mRNA, was described in NTS-GN. Fractalkine protein was localized to glomerular endothelium (154). Antibodies against CX3CR1 markedly attenuated the crescentic nephritis (154). A neutralizing antibody against MCP-1 decreased macrophage infiltration of the glomeruli and reduced proteinuria (146,156,157). This beneficial effect was shown to persist to day 56 after induction of the disease, with lower proteinuria and blood urea nitrogen seen compared with untreated rats (157).

Neutralizing antibodies against other chemokines have also shown partial “positive” effects. Blocking MIP-1α and MIP-1β resulted in a 60% reduction in proteinuria after 24 h, but the animals did not show a corresponding reduction in leukocyte influx (148). Blocking CINC (the rat homologue of GRO) also resulted in decreased proteinuria and reduced early glomerular cell infiltration by polymorphonuclear neutrophils (148). The combination of anti-CINC and anti-MIP treatment did not show an additive beneficial effect (148). A single injection of an anti-MIP-2 (CXC chemokine) antibody resulted in a reduced neutrophil influx and a significant decrease in proteinuria (153).

A natural and broadly acting chemokine receptor antagonist of viral origin, vMIP-II, can block both CC and CXC receptors (99,150). Treatment with vMIP-II decreased infiltration with CD8+ T cells, macrophages and reduced proteinuria to onethird of the control group (150).

Agents that modulate expression of chemokines have beneficial effects on the outcome of the disease. Treatment with dexamethasone can limit glomerular MCP-1 expression, as well as polymorphonuclear neutrophil and monocyte/macrophage infiltration (149). MCP-1 mRNA expression is controlled to a significant extent by the transcription factor NF-κB (147). Treatment of rats with the NF-κB inhibitor pyrrolidine dithiocarbamate led to a decrease in MCP-1 expression, reduced proteinuria, and preserved renal function (147). Treatment of animals with a nonselective cyclo-oxygenase (COX) inhibitor (indomethacin) or selective COX-2 inhibitors (meloxicam, SC 58125) resulted in an upregulation of MCP-1 and RANTES (145). The nonselective COX inhibitor led to a stronger induction of chemokines compared with the group treated with the COX-2 inhibitor. COX products (i.e., prostaglandins) may serve as endogenous chemokine repressors (Table 6) (53,145).

In summary, the animal models of nephrotoxic serum glomerulonephritis suggest a role for chemokines in the initiation and propagation of this disease. The blockade of select chemokines, or their receptors, can lead to a partial improvement of the disease. Surprisingly, recent abstracts indicate that interference with lymphotactin (43), or the elimination of CCR1 (141), may worsen the disease, suggesting “anti-inflammatory” functions for some chemokines.

Anti-Thy-1 Nephritis

In rats the injection of anti-thymocyte antiserum can lead to complement-dependent mesangiolysis followed by monocyte influx and a mesangial proliferative glomerulonephritis (107,158). These lesions heal spontaneously over several weeks. The type of renal disease is dependent on the rat strain used. Wistar and Lewis rats develop a transient influx of macrophages (159). By contrast, F344 rats do not show an influx of macrophages, but have a pronounced proliferative glomerulopathy (159).

MCP-1 (107,123,145,160,161,162) and RANTES (145,162) are upregulated in this model, and MCP-1 protein localizes to glomeruli (107,159). An important role for MCP-1 in this disease process was suggested in blocking experiments using MCP-1 neutralizing antibodies, which led to a decreased infiltration of monocytes/macrophages after 24 h (160). Treatment with AOP-RANTES (a partially “blocking” RANTES derivative) as described in a recent abstract showed a 60% decrease of glomerular macrophage infiltration and ameliorated collagen type IV deposition (Table 6) (162).

Prostaglandin E (PGE) infusion (161), depletion of complement (107), and AT1 antagonists have been shown to significantly decrease MCP-1 expression and glomerular macrophage infiltration (123). As seen in the NTS-GN model, COX inhibitors enhance production of MCP-1 and RANTES (Table 6) (145).

Models of Systemic Lupus Erythematosus

New Zealand black mice crossed with New Zealand white (NZB/W) mice serve as a model for lupus nephritis. The F1 hybrid develops circulating antibodies to nucleic acid, renal immune deposits, and proteinuria (163). MCP-1 mRNA expression is upregulated during the initial 6 mo of the disease (163). In situ hybridization localized MCP-1 mRNA to intrinsic glomerular cells, infiltrating mononuclear cells, and tubular epithelium (163). At later stages of the disease, MCP-1 expression in tubuli corresponded to an “adjacent” leukocyte infiltration (163). Mice treated with cyclophosphamide showed better survival, lower proteinuria, as well as less severe glomerular and interstitial changes, and reduced MCP-1 expression (163). Bindarit, a novel immunosuppressive drug, was recently tested in this model at different time points and in combination with low-dose steroids (164). Starting at 2 mo of age, bindarit administration significantly reduced expression of MCP-1, proteinuria, renal impairment, and prolonged animal survival (Table 6) (164).

The MRL-Faslpr mouse serves as an additional model of systemic lupus. These animals show an upregulation of RANTES and develop a nephritis with glomerular, perivascular, and interstitial inflammation (165). Tubular epithelial cells, genetically manipulated to express RANTES protein, were injected into kidneys of MRL-Faslpr mice. This led to an interstitial infiltrate composed mainly of CD4+ T cells and to a lesser extent CD8+ T cells and macrophages. By contrast, when colony-stimulating factor-1 (CSF-1) was overexpressed in kidneys using the same approach, an equal number of CD4+ and CD8+ T infiltrating cells were seen. The authors postulated complementary roles for RANTES and CSF-1 during autoimmune nephritis in MRL-Faslpr mice.

Immune Complex Glomerulonephritis

A model of immune complex glomerulonephritis in the rabbit is induced by the administration of BSA in combination with LPS (166). An influx of neutrophils into glomeruli, fusion of foot processes, and proteinuria by day 8 characterize this model (166). IL-8 expression by affected glomeruli was associated with an increased urinary IL-8 excretion (166). Injection of an anti-IL-8 antibody reduced glomerular neutrophil infiltration, prevented fusion of foot processes, and normalized proteinuria (166).

In Wistar rats, an immune complex nephritis was induced by the injection of ovalbumin over a period of several weeks. This resulted in an increase of MCP-1 mRNA expression in the renal cortex (167). Immunohistochemistry showed intense MCP-1 staining in glomerular capillaries, mesangial areas, proximal tubular epithelium, and interstitial mononuclear infiltrates (167). Treatment with an angiotensin-converting enzyme inhibitor reduced MCP-1 expression (167). These data suggest a possible link between renal immune injury, the angiotensin system, and specific chemokines (Table 6) (122,123,125,167).

A recent abstract described increased MCP-1, RANTES, CCR1, CCR2, and CCR5 mRNA during the early phase of apoferritin-induced immune complex nephritis in Balb/c mice (168). This upregulation of chemokines preceded the glomerular infiltration of leukocytes. Chemokine expression and the leukocyte infiltration disappeared after discontinuation of antigen exposure and the initiation of healing (168).

Puromycin Aminonucleoside Nephrosis

In rats the injection of puromycin aminonucleoside leads to proteinuria. A marked mononuclear interstitial infiltrate (predominantly macrophages) occurs together with an increase of IP-10, MCP-1, MCP-3, and TCA3 mRNA expression (169,170,171). Expression of CINC, MIP-2, and MIP-1α was not detected, and RANTES expression was low and did not change (171). A neutralizing MCP-1 antibody reduced interstitial infiltration by macrophages (169).

The hepatic hydroxymethylglutaryl CoA reductase inhibitor lovastatin suppressed MCP-1 expression by mesangial cells (172) and significantly reduced the interstitial influx of macrophages in the puromycin model (170). These findings are of particular interest, because interstitial infiltrates are a prognostic factor for the progression of human disease (85).

Tubulointerstitial Nephritis

Immunization of Brown Norway rats with bovine tubular basement membrane induces a severe tubular interstitial nephritis (173). In this model, MCP-1 mRNA expression precedes an influx of mononuclear cells on day 8 but is no longer detectable by day 10 (173). The early increase in CINC and MIP-2 mRNA expression was associated with neutrophil infiltration. A later expression of MCP-1, MIP-1α, and MIP-1β mRNA correlated with a monocyte influx (174). Treatment with a neutralizing MCP-1 antibody or an MCP-1 receptor antagonist reduced macrophage influx (20 and 60%, respectively) (Table 6) (174).

Other Renal Disease Models

Interstitial infiltrate and fibrosis in rats can be induced by daily injections of BSA. In a study by Eddy and Giachelli, Lewis rats received intraperitoneal injections of BSA after nephrectomy of the left kidney (175). MCP-1 mRNA expression was significantly elevated after 2 and 3 wk in whole kidney preparations (175). MCP-1 protein was found in glomeruli and occasionally in tubular cells (175). Because a significant macrophage infiltrate is seen in this model before MCP-1 expression, the infiltrate appears to be due to additional chemoattractants (175).

Injection of diethylaminoethyl dextran leads to proteinuria in Lewis rats but without significant cellular infiltration and without immune complex deposition (176). In this model, strong glomerular localization of RANTES was seen, but apparently was insufficient to cause a cell infiltrate (176).

A model of endotoxemia by LPS injection in Wistar rats leads to glomerular RANTES expression and an influx of macrophages (177). Supplementation with L-arginine, a precursor for nitric oxide (NO) synthesis, reduced the RANTES expression. A nonspecific NO synthase inhibitor resulted in increased RANTES expression and a glomerular infiltrate (177). These studies suggest that the NO pathway may be a counter regulator for LPS-induced RANTES expression and macrophage infiltration.

Chemokine Expression in Unilateral Ureteral Obstruction

Interstitial macrophage infiltration is a prominent feature of unilateral ureteral obstruction (178). MCP-1 mRNA is induced by tubular cells in the obstructed kidney (178,179). Angiotensin-converting enzyme inhibition and an AT1 antagonist decreased MCP-1 expression and reduced the macrophage infiltrate and fibrosis (178).

Chemokine Expression in Renal Ischemia

As early as 1991, Safirstein et al. described the expression of mRNA of the “early response genes” JE (MCP-1) and KC (GRO-α) during renal ischemia (180). The induction of MCP-1/JE can be prevented by treatment with methylprednisolone (128). Furthermore, renal ischemia causes an upregulation of IL-8 that can be inhibited by treatment with α-melanocyte-stimulating hormone, raising the possibility that α-melanocyte-stimulating hormone may act as a counter-regulatory hormone for chemokine induction (181).

Temporary renal pedicle occlusion with simultaneous right nephrectomy resulted in expression of RANTES and MCP-1 after 2 d (182,183). Treatment with bioflavonoids, agents with potential immunosuppressive and renoprotective properties, preserved histologic integrity and renal function, and prevented MCP-1 and RANTES upregulation (183). Blocking the T cell costimulatory molecule B7 by a soluble CTLA4Ig protein resulted in nearly complete suppression of RANTES and MCP-1 (182). Thus, non-immune-mediated injuries can be potent inductors of chemokines, which appear to play an integral role in tissue injury and repair and the associated leukocyte influx, regardless of the initiating insult.

Renal Transplantation

Rat models of transplantation have allowed a functional analysis of the role of chemokines in acute and chronic renal rejection (81,184,185). In acute renal transplant rejection in the rat, Nagano et al. found that RANTES mRNA was upregulated by 6 h during the initial transplant rejection phase and again after 3 to 6 d (185). The authors speculated that expression of E-selectin and RANTES within the first few hours after engraftment may occur secondary to ischemic injury and could help trigger subsequent immunologic events (185). In addition, they concluded that macrophages and their products may play a larger role in the process than previously appreciated (185).

The functional role of RANTES and its receptors was recently evaluated in rat models of acute renal allograft rejection using the RANTES receptor antagonist Met-RANTES (81). Treatment with Met-RANTES significantly reduced the vascular and tubular damage associated with acute rejection and suppressed mononuclear infiltration (81). Treatment with Met-RANTES and low-dose cyclosporin A markedly attenuated the damage to vessels, tubules, and the interstitial rejection (81). The potential clinical advantage of chemokine antagonists such as Met-RANTES may lie in their early effects on the suppression of cellular infiltration and subsequent graft damage, which may help lower the inclination toward the development of chronic transplant dysfunction.

Chemokines in Human Kidney Diseases

Much has been learned about the relationships between chemokine expression and the progression of human renal disorders. Obviously, biopsy studies can provide only a snap-shot of a given stage in the development of a disease. Recent developments suggest that monitoring chemokines in the urine may provide a more dynamic picture of the inflammatory state of the kidney (186,187,188,189,190,191,192).

Chemokines and Chemokine Receptors in Glomerular Diseases

The expression of IL-8 and MCP-1 (108,186,188,193) has been reported In IgA nephropathy. IL-8 mRNA was localized to glomeruli (188), and MCP-1 protein was detected in vascular endothelial cells (188), tubular epithelial cells (108,186,188,193), infiltrating mononuclear cells (186,188), and parietal cells of Bowman's capsule (186). MCP-1 was not seen in glomeruli that did not show proliferative nephropathy (144,194). In mild disease courses, MCP-1 was rarely expressed, whereas severe cases with interstitial infiltrates showed strong expression of MCP-1, which correlated well with monocyte infiltration and interstitial damage (186). Renal function was found to be significantly worse in patients with detectable MCP-1 expression (194). In IgA nephropathy, CCR1, CCR2a, and CCR2b were detected in biopsy tissue by reverse transcription-PCR (194). A strong glomerular expression of CXCR3, primarily by mesangial cells, was found by immunohistochemistry (35). CXCR3 was also found on infiltrating leukocytes, and endothelial and vascular smooth muscle cells (35). On the other hand, CCR5-positive cells are not present in glomeruli, but were a prominent part of the interstitial infiltrate (195). In summary, in IgA nephropathy MCP-1 is expressed by tubulointerstitial cells, and the chemokine receptors CCR2 and CCR5 are found on inflammatory cells. CXCR3 is expressed on intrinsic glomerular cells and may act as a mechanism for glomerular mesangial proliferation (35).

MCP-1 expression has not been found in glomeruli from nonproliferating diseases, such as membranous nephropathy, minimal change disease, thin basement membrane disease, and diabetic nephropathy (144). In contrast to the absence of glomerular chemokine expression in membranous nephropathy, MCP-1 and RANTES were expressed by tubular epithelial cells during proteinuria, and their expression was associated with interstitial cell infiltration and fibrosis (193,196). No CCR5-positive cells were detected within glomeruli during membranous nephropathy, but CCR5-positive mononuclear cells—predominantly CD3-positive T cells—were seen in the interstitium (195). This CCR5-positive T cell infiltrate may be related to the tubulointerstitial damage noted in these biopsies (195).

Chemokines and Chemokine Receptors in Proliferative and Crescentic Glomerulonephritis

Rovin et al. described the distribution of MCP-1 by immunohistochemistry in patients with idiopathic crescentic glomerulonephritis, Wegener's granulomatosis, and lupus nephritis (144). A focal, granular staining for MCP-1 with a mesangial distribution (and in some crescents) was detected (144). Cock-well et al. studied 20 patients with glomerulonephritis due to different forms of vasculitis by in situ hybridization (197). MCP-1, MIP-1α, and MIP-1β were expressed in some glomeruli at similar levels and in crescents (197). RANTES expression was described in 11% of the glomeruli (197). MCP-1 was not detected in the glomeruli of patients with lupus nephritis (189). By in situ hybridization, patients with lupus nephritis showed MCP-1 expression on endothelial cells, cortical tubules, and infiltrating mononuclear cells in the interstitium (189,197). In patients with cryoglobulinemic glomerulonephritis, a significant upregulation of MCP-1 expression was seen in glomeruli and in areas of tubulointerstitial damage (191). The MCP-1 was localized to tubular cells, parietal cells, and cells of the glomerular tuft, and correlated with glomerular and interstitial macrophage infiltration (191). The expression of MIP-1β and MIP-1α fits with the preliminary observation that crescents contain CCR5-positive cells (198). In crescentic glomerulonephritis, the expression of MIP-1α and MCP-1 was found in tubules, peritubular capillaries, and infiltrating leukocytes. MIP-1α was also present in crescents (199). Cockwell et al. described the distribution of IL-8 mRNA and protein in patients with antineutrophil cytoplasmic antibody-associated glomerulonephritis (200). About 30% of the glomeruli showed IL-8 expression at sites of inflammation. Outside the glomeruli, IL-8 mRNA was detected in interstitial infiltrates and proximal tubular epithelial cells. IL-8 protein was present in 50% of the glomeruli and was prominent in crescents and parietal cells. Intraglomerular leukocyte accumulation correlated with the IL-8 expression (200).

Chemokines and Chemokine Receptors in Tubulointerstitial Diseases

Biopsy samples from patients with acute interstitial nephritis were studied by In situ hybridization and immunohistochemistry for MCP-1 expression (186). MCP-1 was expressed by tubular as well as infiltrating cells and was detected in parietal cells of Bowman's capsule but not within the glomerular tuft. Eitner et al. described expression of CCR5 mRNA in interstitial infiltrates (201) that was confirmed by immunohistochemistry, where CCR5-positive cells correlated with the presence of an interstitial T cell infiltrate (195).

Chemokines and Chemokine Receptors during Renal Transplant Rejection

Acute allograft rejection is characterized by a mononuclear cell infiltrate consisting primarily of T cells, macrophages, and occasional eosinophils (202,203). The chemokines IL-8, ENA-78, MCP-1, MIP-1α, MIP-1β, and RANTES have all been implicated in the pathogenesis of acute rejection (204,205,206,207,208,209). One of the most studied chemokines in this regard is the CC chemokine RANTES (205,210). During cell-mediated rejection, in situ hybridization localized RANTES mRNA to infiltrating cells and tubular epithelium (210). RANTES protein was found on mononuclear cells, tubular epithelium, and the vascular endothelium (205). This expression mirrors the distribution of CCR5-positive leukocytes found in areas of endothelialitis, tubulitis, and interstitial infiltrates during cellular rejection (195). Strehlau and coworkers used reverse transcription-PCR to evaluate gene expression in human renal allograft biopsies and concluded that RANTES and IL-8 expression are sensitive but not specific markers of allograft rejection (211). Grandaliano et al. found an increased amount of MCP-1 in uring during rejection corresponding to elevated renal MCP-1 expression (204). An increased amount of urinary IL-8 was described during acute rejection, which returned to normal levels after successful treatment (206).

DARC Expression in Kidney Disease

A study of children with HIV-associated nephropathy, HIV-associated hemolytic uremic syndrome (HUS), and classic HUS (212) reported DARC mRNA and protein localized to endothelial cells of postcapillary renal venules in normal kidney. The biopsies of the patients showed increased DARC expression on glomerular capillaries, collecting duct epithelial cells, and interstitial inflammatory cells (212). DARC upregulation on peritubular endothelium was described during transplant rejection especially at sites of inflammation in a recent abstract (213). The role of this upregulation of DARC during renal diseases is unknown.

Quantification of Urinary Chemokines as a Measure of Renal Production

Elevated urinary MCP-1 levels occur in proliferative lupus nephritis, IgA nephropathy, proliferative glomerulonephritis associated with endocarditis, membranoproliferative glomerulonephritis, crescentic glomerulonephritis, and in transplant rejection (187,188,189,190,191,199). Only moderately increased amounts of MCP-1 have been measured in membranous nephropathy, focal segmental glomerulosclerosis, and diabetic nephropathy, consistent with moderate glomerular cell infiltration in these diseases (190). Urinary MCP-1 levels correlated with urinary protein excretion and macrophage infiltration of the glomeruli (190). No correlation between serum and urine MCP-1 was seen (187).

High urinary MCP-1 excretion was found in patients with active lupus nephritis compared with healthy control subjects (189,190,192). Urinary MCP-1 levels were reduced by highdose glucocorticoid therapy and remained low during remission (187,189,192). Elevated urinary excretion of MCP-1 and MIP-1α was also reported in patients with crescentic glomerulonephritis, and treatment reduced excretion of both chemokines (199). Similar results were reported for IL-8, with high levels seen during severe lupus nephritis, and a significant reduction in IL-8 levels was seen after glucocorticoid therapy (187,189).

A correlation between urinary MCP-1 levels, mesangial proliferation, and interstitial infiltrate has been described in IgA nephropathy (186,188). Patients with severe IgA nephropathy showed higher MCP-1 levels compared with patients with mild disease (186). IL-8 levels were only elevated during the acute phase and correlated with glomerular endocapillary proliferation and the degree of hematuria (188).

Other renal diseases that present elevated levels of IL-8 include acute postinfectious glomerulonephritis, membranoproliferative glomerulonephritis, and cryoglobulinemia (187). The urinary IL-8 levels were higher in patients with glomerular leukocyte infiltration than in those without infiltration (187). Children with HUS showed a significant increase in both IL-8 and MCP-1 excretion (214). Thus, urinary excretion of chemokines may reflect the intrarenal inflammatory cell infiltrate and may be of prognostic value as a measure of continued intrarenal inflammation.

A Model for Chemokines in Renal Diseases

The emerging picture of chemokine action allows us to construct models of potential roles for chemokines in various pathophysiologic processes thought to contribute to renal disorders. In this model (Figure 1A), a pathologic insult to the glomerular or tubulointerstitial compartment activates intrinsic renal cells and leads to the local generation of proinflammatory mediators (initiation phase) (Figure 1B). This insult may be immunologic, toxic, ischemic, or mechanical, and may target a specific renal cell type (e.g., endothelial, mesangial, and epithelial or interstitial cell). The expression of select chemokines and chemokine receptors, together with the local release of inflammatory mediators (such as IL-1, TNF-α, IFN-γ, plateletactivating factor, reactive oxygen species, etc.), promotes the upregulation and activation of selectins and integrins on leukocytes and endothelial cells leading to adhesion, transendothelial migration, and infiltration of specific subsets of leukocytes. T cells, monocytes, and polymorphonuclear leukocytes appear to preferentially adhere to different microvascular compartments in the kidney. For example, T cells are rare in glomeruli but are common in interstitial infiltrates. The specialized endothelia found in the kidney, e.g., glomerular, peritubular, have different characteristics, but at present information on the surface expression of selectin ligands is lacking (215). The role of this endothelial heterogeneity in the kidney for the leukocyte adhesion and infiltration process remains to be elucidated.

Figure 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1.

Hypothetical model of the role of chemokines and chemokine receptors during progressive renal diseases. (A) Normal renal tissue. (B) Initiation phase. (C) Amplification phase. (D) Progression phase.

An additional consequence of local cell activation may include the induced expression of chemokine receptors such as CCR1 and CXCR3 by mesangial cells, and other renal cell types. The expression of CXCR3 by mesangial cells in concert with local production of the chemokine IP-10 could lead to mesangial cell proliferation (35).

During the “amplification phase,” spillover of proinflammatory factors from affected glomeruli could reach the peritubular capillary circulation, as well as the tubular lumen via ultrafiltration, especially in the context of proteinuria with loss of the ultrafiltration barrier. In addition, protein and lipids escaping through damaged glomeruli could “stress” proximal tubular cells. In concert with inflammatory mediators, this could lead to an activation of tubular and interstitial cells and the production of additional chemokines, resulting in interstitial mononuclear cell infiltration. Similarly, the parietal cells of Bowman's capsule apparent bystander cells in this process could become activated and thus release chemokines into the surrounding interstitium. This sort of event could help explain the accentuated periglomerular infiltrate seen in some renal diseases. The periglomerular infiltrate may play a role in the eventual rupture of Bowman's capsule, which would open the door for T cells, macrophages, and fibroblasts to invade the urinary space and damage the parietal and visceral epithelial cells. This could be an example of the “point of no return,” in which amplification of a glomerular injury results in irreversible sclerosis. Thus, a cycle initiated through glomerular damage could, in the absence of sufficient counter-regulatory signals, result in downstream tubular-interstitial injury, interstitial inflammation, and fibrosis (progression phase) (Figure 1D).

Of course, renal inflammation does not always end with chronic damage. The events initiated during inflammation normally end in resolution of the disease process. Recent results suggest that some chemokines and their receptors may also assist in downmodulating the inflammatory response, a hypothesis that deserves further investigation.

Conclusions, Future Directions, and Therapeutic Outlook

Current therapies used for the treatment of renal diseases can influence chemokine expression. The production of chemokines by various renal and infiltrating cells can be suppressed by glucocorticoids in vitro (126,216). Glucocorticoids function in part via an inhibition of transcription factor NF-κB (217), and blockade of NF-κB function leads to suppression of chemokine release by renal cells (51,218). Elevated urinary chemokines in human lupus nephritis are decreased by glucocorticoid therapy (187,189,190,192). Glucocorticoid suppression of chemokine expression may be one component of the general beneficial effects of glucocorticoid therapy seen in lupus nephritis.

In animal models, a connection between angiotensin II, chemokine production by renal cells (122), and beneficial effects of angiotensin blockade were described (122,125,178). Thus, a reduction in chemokine production may be one beneficial effect of angiotensin-blocking drugs in the treatment of renal disease.

The side effects of nonsteroidal anti-inflammatory drugs (NSAID) are well established and are mostly related to alterations in renal hemodynamics and transport (219). The rare NSAID-related nephrotic syndrome and interstitial nephritis are thought to result from immunologic effects of these drugs (219). In this context, it is of interest that PGE suppresses chemokine production in mesangial cells and chemokine receptor expression on monocytes-macrophages (53,220). Therefore, PGE and possibly PGI2 may also inhibit chemokine production. This is supported by studies in the Thy 1.1 model, in which PGE administration decreases MCP-1 generation and cyclo-oxygenase inhibitors enhance MCP-1 and RANTES expression (145,161). In addition, PGE has been shown to decrease monocyte-macrophage infiltration (161), whereas NSAID can enhance it (145). Thus, NSAID may enhance renal chemokine production and hence renal inflammation, and additional reason for avoiding their use in patients with renal disease.

Taken together, these studies shed light on therapeutic protocols that may influence the regulation of chemokine/chemokine receptor expression and suggest efficacy for the development of agents that selectively target chemokine biology. In general, receptor-blocking drugs have proven to be very successful therapeutically. The development of chemokine receptor antagonists for the treatment of inflammatory disorders and HIV is a major focus of the pharmaceutical industry. The redundancy of the chemokines, which creates a robust system, could be a drawback for therapeutic interventions (57). Therefore, blockade of multiple chemokine receptors simultaneously might be necessary. This strategy has already been successfully exploited by nature, as viruses can produce broad-spectrum chemokine antagonists, e.g., vMIP-II (150). On the other hand, the redundancy might be of potential benefit for the treatment with antagonists, as this might lead to a reduction of side effects.

Given the large number of chemokines and chemokine receptors, the intricate control of their expression, and their diverse biologic actions, the emerging story reveals a biologic system that is extraordinarily complex. The therapeutic intervention into the system might depend on the underlying disease, the time point, and comedication. Based on experimental models and the immunohistologic findings in human biopsies, blockade of CCR5-positive infiltrating cells might prove to be a promising therapeutic strategy for inflammatory renal diseases including transplant rejection. Clearly, antagonists for chemokines and their receptors have the potential to become additional therapeutic tools in inflammatory disorders, including kidney diseases.

Acknowledgments

We thank Holger Maier for help with graphics. Our special thanks go to Bruno Luckow, Bernhard Banas, Matthias Mack, and Joachim Anders for carefully reading the manuscript.

Footnotes

  • This review is dedicated to our collaborators in Japan, the United States, and Europe.

  • © 2000 American Society of Nephrology

References

  1. ↵
    Jones DB: Glomerulonephritis. Am J Pathol29 : 33-43,1953
  2. ↵
    Schlondorff D, Nelson PJ, Luckow B, Banas B: Chemokines and renal disease. Kidney Int 51:610 -621, 1997
    OpenUrlCrossRefPubMed
  3. ↵
    Rovin BH, Phan LT: Chemotactic factors and renal inflammation. Am J Kidney Dis 31:1065 -1084, 1998
    OpenUrlCrossRefPubMed
  4. ↵
    Baggiolini M: Chemokines and leukocyte traffic. Nature 392:565 -568, 1998
    OpenUrlCrossRefPubMed
  5. ↵
    del Pozo MA, Sanchez Mateos P, Nieto M, Sanchez Madrid F: Chemokines regulate cellular polarization and adhesion receptor redistribution during lymphocyte interaction with endothelium and extracellular matrix: Involvement of cAMP signaling pathway. J Cell Biol131 : 495-508,1995
    OpenUrlAbstract/FREE Full Text

References

  1. 6.↵
    Muller GA, Muller CA, Markovic Lipkovski J: Adhesion molecules in renal diseases. Renal Failure18 : 711-724,1996
    OpenUrlPubMed
  2. 7.↵
    Yoshimura T, Matsushima K, Tanaka S, Robinson EA, Appella E, Oppenheim JJ, Leonard EJ: Purification of a human monocyte derived neutrophil chemotactic factor that has peptide sequence similarity to other host defense cytokines. Proc Natl Acad Sci USA84 : 9233-9237,1987
    OpenUrlAbstract/FREE Full Text
  3. 8.↵
    Baggiolini M, Clark Lewis I: Interleukin-8, a chemotactic and inflammatory cytokine. FEBS Lett307 : 97-101,1992
    OpenUrlCrossRefPubMed
  4. 9.↵
    Zlotnik A, Morales J, Hedrick JA: Recent advances in chemokines and chemokine receptors. Crit Rev Immunol19 : 1-47,1999
    OpenUrlPubMed
  5. 10.↵
    Wiedermann CJ, Kowald E, Reinisch N, Kaehler CM, von Luettichau I, Pattison JM, Huie P, Sibley RK, Nelson PJ, Krensky AM: Monocyte haptotaxis induced by the RANTES chemokine. Curr Biol3 : 735-739,1993
    OpenUrlCrossRefPubMed
  6. 11.↵
    Weber KS, von Hundelshausen P, Clark Lewis I, Weber PC, Weber C: Differential immobilization and hierarchical involvement of chemokines in monocyte arrest and transmigration on inflamed endothelium in shear flow. Eur J Immunol 29:700 -712, 1999
    OpenUrlCrossRefPubMed
  7. 12.↵
    Peveri P, Walz A, Dewald B, Baggiolini M: A novel neutrophil-activating factor produced by human mononuclear phagocytes. J Exp Med 167:1547 -1559, 1988
    OpenUrlAbstract/FREE Full Text
  8. 13.↵
    Quackenbush EJ, Aguirre V, Wershil BK, Gutierrez Ramos JC: Eotaxin influences the development of embryonic hematopoietic progenitors in the mouse. J Leukocyte Biol 62:661 -666, 1997
    OpenUrlPubMed
  9. 14.↵
    Moore BB, Keane MP, Addison CL, Arenberg DA, Strieter RM: CXC chemokine modulation of angiogenesis: The importance of balance between angiogenic and angiostatic members of the family. J Invest Med 46: 113-120,1998
    OpenUrlPubMed
  10. 15.↵
    Baggiolini M, Dewald B, Moser B: Human chemokines: An update. Annu Rev Immunol 15:675 -705, 1997
    OpenUrlCrossRefPubMed
  11. 16.↵
    Dragic T, Litwin V, Allaway GP, Martin SR, Huang Y, Nagashima KA, Cayanan C, Maddon PJ, Koup RA, Moore JP, Paxton WA: HIV-1 entry into CD4+ cells is mediated by the chemokine receptor CC-CKR-5. Nature 381:667 -673, 1996
    OpenUrlCrossRefPubMed
  12. 17.↵
    Deng H, Liu R, Ellmeier W, Choe S, Unutmaz D, Burkhart M, Di Marzio P, Marmon S, Sutton RE, Hill CM, Davis CB, Peiper SC, Schall TJ, Littman DR, Landau NR: Identification of a major co-receptor for primary isolates of HIV-1. Nature 381:661 -666, 1996
    OpenUrlCrossRefPubMed
  13. 18.↵
    Choe H, Farzan M, Sun Y, Sullivan N, Rollins B, Ponath PD, Wu L, Mackay CR, LaRosa G, Newman W, Gerard N, Gerard C, Sodroski J: The beta-chemokine receptors CCR3 and CCR5 facilitate infection by primary HIV-1 isolates. Cell 85:1135 -1148, 1996
    OpenUrlCrossRefPubMed
  14. 19.↵
    Doranz BJ, Rucker J, Yi Y, Smyth RJ, Samson M, Peiper SC, Parmentier M, Collman RG, Doms RW: A dual-tropic primary HIV-1 isolate that uses fusin and the beta-chemokine receptors CKR-5, CKR-3, and CKR-2b as fusion cofactors. Cell 85:1149 -1158, 1996
    OpenUrlCrossRefPubMed
  15. 20.↵
    Alkhatib G, Combadiere C, Broder CC, Feng Y, Kennedy PE, Murphy PM, Berger EA: CC CKR5: A RANTES, MIP-1alpha, MIP-1beta receptor as a fusion cofactor for macrophage-tropic HIV-1. Science272 : 1955-1958,1996
    OpenUrlAbstract/FREE Full Text
  16. 21.↵
    Nelson PJ, Krensky AM: Chemokines, lymphocytes and viruses: What goes around, comes around. Curr Opin Immunol10 : 265-270,1998
    OpenUrlCrossRefPubMed
  17. 22.↵
    Moore JP, Trkola A, Dragic T: Co-receptors for HIV-1 entry. Curr Opin Immunol 9:551 -562, 1997
    OpenUrlCrossRefPubMed
  18. 23.↵
    Baggiolini M, Moser B: Blocking chemokine receptors. J Exp Med 186:1189 -1191, 1997
    OpenUrlFREE Full Text
  19. 24.↵
    Horuk R, Peiper SC: Chemokines: Molecular double agents. Curr Biol 6:1581 -1582, 1996
    OpenUrlCrossRefPubMed
  20. 25.↵
    Horuk R: Molecular properties of the chemokine receptor family. Trends Pharmacol Sci 15:159 -165, 1994
    OpenUrlCrossRefPubMed
  21. 26.↵
    Baggiolini M, Loetscher P, Moser B: Interleukin-8 and the chemokine family. Int J Immunopharmacol17 : 103-108,1995
    OpenUrlCrossRefPubMed
  22. 27.↵
    Luster AD: Chemokines: Chemotactic cytokines that mediate inflammation. N Engl J Med 338:436 -445, 1998
    OpenUrlCrossRefPubMed
  23. 28.↵
    Sallusto F, Lanzavecchia A, Mackay CR: Chemokines and chemokine receptors in T-cell priming and Th1/Th2-mediated responses. Immunol Today 19:568 -574, 1998
    OpenUrlCrossRefPubMed
  24. 29.↵
    Mackay CR: Chemokines: What chemokine is that? Curr Biol 7:R384 -R386, 1997
    OpenUrlCrossRefPubMed
  25. 30.↵
    Baggiolini M, Dewald B, Moser B: Interleukin-8 and related chemotactic cytokines: CXC and CC chemokines. Adv Immunol 55:97 -179, 1994
    OpenUrlCrossRefPubMed
  26. 31.↵
    Bazan JF, Bacon KB, Hardiman G, Wang W, Soo K, Rossi D, Greaves DR, Zlotnik A, Schall TJ: A new class of membrane-bound chemokine with a CX3C motif. Nature 385:640 -644, 1997
    OpenUrlCrossRefPubMed
  27. 32.↵
    Kelner GS, Kennedy J, Bacon KB, Kleyensteuber S, Largaespada DA, Jenkins NA, Copeland NG, Bazan JF, Moore KW, Schall TJ: Lymphotactin: A cytokine that represents a new class of chemokine. Science 266:1395 -1399, 1994
    OpenUrlAbstract/FREE Full Text
  28. 33.↵
    Haskell CA, Cleary MD, Charo IF: Molecular uncoupling of fractalkine-mediated cell adhesion and signal transduction: Rapid flow arrest of CX3CR1-expressing cells is independent of G-protein activation. J Biol Chem 274:10053 -10058, 1999
    OpenUrlAbstract/FREE Full Text
  29. 34.↵
    Richmond A, Balentien E, Thomas HG, Flaggs G, Barton DE, Spiess J, Bordoni R, Francke U, Derynck R: Molecular characterization and chromosomal mapping of melanoma growth stimulatory activity, a growth factor structurally related to beta-thromboglobulin. EMBO J7 : 2025-2033,1988
    OpenUrlPubMed
  30. 35.↵
    Romagnani P, Beltrame C, Annunziato F, Lasagni L, Luconi M, Galli G, Cosmi L, Maggi E, Salvadori M, Pupilli C, Serio M: Role for interactions between IP-10/MIG and CXCR3 in proliferative glomerulonephritis. J Am Soc Nephrol 10:2518 -2526, 1999
    OpenUrlAbstract/FREE Full Text
  31. 36.↵
    Kuna P, Reddigari SR, Schall TJ, Rucinski D, Viksman MY, Kaplan AP: RANTES, a monocyte and T lymphocyte chemotactic cytokine releases histamine from human basophils. J Immunol149 : 636-642,1992
    OpenUrlAbstract/FREE Full Text
  32. 37.↵
    Taub DD, Turcovski Corrales SM, Key ML, Longo DL, Murphy WJ: Chemokines and T lymphocyte activation. I. Beta chemokines costimulate human T lymphocyte activation in vitro. J Immunol156 : 2095-2103,1996
    OpenUrlAbstract
  33. 38.↵
    Gewirtz AM, Zhang J, Ratajczak J, Ratajczak M, Park KS, Li C, Yan Z, Poncz M: Chemokine regulation of human megakaryocytopoiesis. Blood 86:2559 -2567, 1995
    OpenUrlAbstract/FREE Full Text
  34. 39.↵
    Darveau RP, Blake J, Seachord CL, Cosand WL, Cunningham MD, Cassiano Clough L, Maloney G: Peptides related to the carboxyl terminus of human platelet factor IV with antibacterial activity. J Clin Invest 90:447 -455, 1992
  35. 40.↵
    Broxmeyer HE, Cooper S, Hague N, Benninger L, Sarris A, Cornetta K, Vadhan Raj S, Hendrie P, Mantel C: Human chemokines: Enhancement of specific activity and effects in vitro on normal and leukemic progenitors and a factor-dependent cell line and in vivo in mice. Ann Hematol 71:235 -246, 1995
    OpenUrlCrossRefPubMed
  36. 41.↵
    Broxmeyer HE, Sherry B, Cooper S, Lu L, Maze R, Beckmann MP, Cerami A, Ralph P: Comparative analysis of the human macrophage inflammatory protein family of cytokines (chemokines) on proliferation of human myeloid progenitor cells: Interacting effects involving suppression, synergistic suppression, and blocking of suppression. J Immunol150 : 3448-3458,1993
    OpenUrlAbstract
  37. 42.↵
    Cook DN: The role of MIP-1 alpha in inflammation and hematopoiesis. J Leukocyte Biol 59:61 -66, 1996
    OpenUrlCrossRefPubMed
  38. 43.↵
    Chen S, Xia Y, Barcia GE, Wilson CB, Feng L: Neutralization of Lymphotactin (Lptn) by antibody (Ab) worsens anti-glomerular basement membrane (GBM) glomerulonephritis (GN) in WKY rats [Abstract]. J Am Soc Nephrol 9: 452A,1998
    OpenUrl
  39. 44.↵
    Arenberg DA, Keane MP, DiGiovine B, Kunkel SL, Morris SB, Xue YY, Burdick MD, Glass MC, Iannettoni MD, Strieter RM: Epithelial-neutrophil activating peptide (ENA-78) is an important angiogenic factor in non-small cell lung cancer. J Clin Invest102 : 465-472,1998
    OpenUrlCrossRefPubMed
  40. 45.↵
    Liao F, Rabin RL, Yannelli JR, Koniaris LG, Vanguri P, Farber JM: Human Mig chemokine: Biochemical and functional characterization. J Exp Med 182:1301 -1314, 1995
    OpenUrlAbstract/FREE Full Text
  41. 46.↵
    Yasumoto K, Okamoto S, Mukaida N, Murakami S, Mai M, Matsushima K: Tumor necrosis factor alpha and interferon gamma synergistically induce interleukin 8 production in a human gastric cancer cell line through acting concurrently on AP-1 and NF-κB-like binding sites of the interleukin 8 gene. J Biol Chem 267:22506 -22511, 1992
    OpenUrlAbstract/FREE Full Text
  42. 47.↵
    Nelson PJ, Ortiz BD, Pattison JM, Krensky AM: Identification of a novel regulatory region critical for expression of the RANTES chemokine in activated T lymphocytes. J Immunol157 : 1139-1148,1996
    OpenUrlAbstract
  43. 48.↵
    Jarmin DI, Kulmburg PA, Huber NE, Baumann G, Prieschl Strassmayr EE, Baumruker T: A transcription factor with AP3-like binding specificity mediates gene regulation after an allergic triggering with IgE and Ag in mouse mast cells. J Immunol 153:5720 -5729, 1994
    OpenUrlAbstract
  44. 49.↵
    Barnes PJ: Nuclear factor kappa B. Int J Biochem Cell Biol 29: 867-870,1997
    OpenUrlCrossRefPubMed
  45. 50.↵
    Heck S, Bender K, Kullmann M, Gottlicher M, Herrlich P, Cato AC: IκB alpha-independent downregulation of NF-κB activity by glucocorticoid receptor. EMBO J16 : 4698-4707,1997
    OpenUrlAbstract
  46. 51.↵
    Rovin BH, Dickerson JA, Tan LC, Hebert CA: Activation of nuclear factor-kappa B correlates with MCP-1 expression by human mesangial cells. Kidney Int 48:1263 -1271, 1995
    OpenUrlCrossRefPubMed
  47. 52.↵
    Rovin BH, Tan LC: Role of protein kinase pathways in IL-1-induced chemoattractant expression by human mesangial cells. Kidney Int 46:1059 -1068, 1994
    OpenUrlCrossRefPubMed
  48. 53.↵
    Satriano JA, Hora K, Shan Z, Stanley ER, Mori T, Schlöndorff D: Regulation of monocyte chemoattractant protein-1 and macrophage colony-stimulating factor-1 by IFN-γ, tumor necrosis factor-α, IgG aggregates, and cAMP in mouse mesangial cells. J Immunol 150:1971 -1978, 1993
    OpenUrlAbstract
  49. 54.↵
    Rovin BH, Dickerson JA, Tan LC, Fassler J: Modulation of IL-1-induced chemokine expression in human mesangial cells through alterations in redox status. Cytokine 9:178 -186, 1997
    OpenUrlCrossRefPubMed
  50. 55.↵
    Murphy PM: The molecular biology of leukocyte chemoattractant receptors. Annu Rev Immunol 12:593 -633, 1994
    OpenUrlCrossRefPubMed
  51. 56.↵
    Murphy PM: Chemokine receptors: Structure, function and role in microbial pathogenesis. Cytokine Growth Factor Rev7 : 47-64,1996
    OpenUrlCrossRefPubMed
  52. 57.↵
    Mantovani A: The chemokine system: Redundancy for robust outputs. Immunol Today 20:254 -257, 1999
    OpenUrlCrossRefPubMed
  53. 58.↵
    Mantovani A, Allavena P, Vecchi A, Sozzani S: Chemokines and chemokine receptors during activation and deactivation of monocytes and dendritic cells and in amplification of Th1 versus Th2 responses. Int J Clin Lab Res 28:77 -82, 1998
    OpenUrlCrossRefPubMed
  54. 59.↵
    Bacon KB, Premack BA, Gardner P, Schall TJ: Activation of dual T cell signaling pathways by the chemokine RANTES. Science 269:1727 -1730, 1995
    OpenUrlAbstract/FREE Full Text
  55. 60.↵
    Neote K, DiGregorio D, Mak JY, Horuk R, Schall TJ: Molecular cloning, functional expression, and signaling characteristics of a C-C chemokine receptor. Cell 72:415 -425, 1993
    OpenUrlCrossRefPubMed
  56. 61.↵
    Cai JP, Hudson S, Ye MW, Chin YH: The intracellular signaling pathways involved in MCP-1-stimulated T cell migration across microvascular endothelium. Cell Immunol 167:269 -275, 1996
    OpenUrlCrossRefPubMed
  57. 62.↵
    Darbonne WC, Rice GC, Mohler MA, Apple T, Hebert CA, Valente AJ, Baker JB: Red blood cells are a sink for interleukin 8, a leukocyte chemotaxin. J Clin Invest 88:1362 -1369, 1991
  58. 63.↵
    Neote K, Darbonne W, Ogez J, Horuk R, Schall TJ: Identification of a promiscuous inflammatory peptide receptor on the surface of red blood cells. J Biol Chem 268:12247 -12249, 1993
    OpenUrlAbstract/FREE Full Text
  59. 64.↵
    Chaudhuri A, Zbrzezna V, Polyakova J, Pogo AO, Hesselgesser J, Horuk R: Expression of the Duffy antigen in K562 cells: Evidence that it is the human erythrocyte chemokine receptor. J Biol Chem269 : 7835-7838,1994
    OpenUrlAbstract/FREE Full Text
  60. 65.↵
    Horuk R, Chitnis CE, Darbonne WC, Colby TJ, Rybicki A, Hadley TJ, Miller LH: A receptor for the malarial parasite Plasmodium vivax: The erythrocyte chemokine receptor. Science261 : 1182-1184,1993
    OpenUrlAbstract/FREE Full Text
  61. 66.↵
    Hadley TJ, Lu ZH, Wasniowska K, Martin AW, Peiper SC, Hesselgesser J, Horuk R: Postcapillary venule endothelial cells in kidney express a multispecific chemokine receptor that is structurally and functionally identical to the erythroid isoform, which is the Duffy blood group antigen. J Clin Invest 94:985 -991, 1994
  62. 67.↵
    Peiper SC, Wang ZX, Neote K, Martin AW, Showell HJ, Conklyn MJ, Ogborne K, Hadley TJ, Lu ZH, Hesselgesser J: The Duffy antigen/receptor for chemokines (DARC) is expressed in endothelial cells of Duffy negative individuals who lack the erythrocyte receptor. J Exp Med 181:1311 -1317, 1995
    OpenUrlAbstract/FREE Full Text
  63. 68.↵
    Mosmann TR, Cherwinski H, Bond MW, Giedlin MA, Coffman RL: Two types of helper T cell clone. I. Definition according to lymphokine activities and secreted proteins. J Immunol136 : 2348-2357,1986
    OpenUrlAbstract
  64. 69.↵
    Abbas AK, Murphy KM, Sher A: Functional diversity of helper T lymphocytes. Nature 383:787 -793, 1996
    OpenUrlCrossRefPubMed
  65. 70.↵
    Cher DJ, Mosmann TR: Two types of murine helper T cell clone. II. Delayed-type hypersensitivity is mediated by TH1 clones. J Immunol 138:3688 -3694, 1987
    OpenUrlAbstract
  66. 71.↵
    Cobbold S, Waldmann H: Infectious tolerance. Curr Opin Immunol 10:518 -524, 1998
    OpenUrlCrossRefPubMed
  67. 72.↵
    Bonecchi R, Bianchi G, Bordignon PP, D'Ambrosio D, Lang R, Borsatti A, Sozzani S, Allavena P, Gray PA, Mantovani A, Sinigaglia F: Differential expression of chemokine receptors and chemotactic responsiveness of type 1 T helper cells (Th1s) and Th2s. J Exp Med187 : 129-134,1998
    OpenUrlAbstract/FREE Full Text
  68. 73.↵
    Sallusto F, Lenig D, Mackay CR, Lanzavecchia A: Flexible programs of chemokine receptor expression on human polarized T helper 1 and 2 lymphocytes. J Exp Med 187:875 -883, 1998
    OpenUrlAbstract/FREE Full Text
  69. 74.↵
    Huang XR, Tipping PG, Shuo L, Holdsworth SR: Th1 responsiveness to nephritogenic antigens determines susceptibility to crescentic glomerulonephritis in mice. Kidney Int51 : 94-103,1997
    OpenUrlCrossRefPubMed
  70. 75.↵
    Huang XR, Holdsworth SR, Tipping PG: Th2 responses induce humorally mediated injury in experimental anti-glomerular basement membrane glomerulonephritis. J Am Soc Nephrol8 : 1101-1108,1997
    OpenUrlAbstract
  71. 76.↵
    Emery P, Salmon M: The immune response. 2. Systemic mediators of inflammation. Br J Hosp Med 45:164 -168, 1991
    OpenUrlPubMed
  72. 77.↵
    Crockett Torabi E: Selectins and mechanisms of signal transduction. J Leukocyte Biol 63:1 -14, 1998
    OpenUrlPubMed
  73. 78.↵
    Butcher EC, Picker LJ: Lymphocyte homing and homeostasis. Science 272:60 -66, 1996
    OpenUrlAbstract
  74. 79.↵
    Weber C, Alon R, Moser B, Springer TA: Sequential regulation of α4 β1 and α5 β1 integrin avidity by CC chemokines in monocytes: Implications for transendothelial chemotaxis. J Cell Biol 134:1063 -1073, 1996
    OpenUrlAbstract/FREE Full Text
  75. 80.↵
    Brady HR: Leukocyte adhesion molecules and kidney disease. Kidney Int 45:1285 -1300, 1994
    OpenUrlCrossRefPubMed
  76. 81.↵
    Gröne HJ, Weber C, Weber KSC, Gröne EF, Rabelink T, Klier CM, Wells TNC, Proudfoot AE, Schlondorff D, Nelson PJ: Met-RANTES-reduces vascular and tubular damage during acute renal transplant rejection: Blocking monocyte arrest and recruitment. FASEB J13 : 1371-1383,1999
    OpenUrlPubMed
  77. 82.↵
    Weber C, Springer TA: Interaction of very late antigen-4 with VCAM-1 supports transendothelial chemotaxis of monocytes by facilitating lateral migration. J Immunol161 : 6825-6834,1998
    OpenUrlAbstract/FREE Full Text
  78. 83.↵
    Campbell JJ, Foxman EF, Butcher EC: Chemoattractant receptor cross talk as a regulatory mechanism in leukocyte adhesion and migration. Eur J Immunol 27:2571 -2578, 1997
    OpenUrlCrossRefPubMed
  79. 84.↵
    Mangan DF, Mergenhagen SE, Wahl SM: Apoptosis in human monocytes: Possible role in chronic inflammatory diseases. J Periodontol 64:461 -466, 1993
    OpenUrlPubMed
  80. 85.↵
    Bohle A, Wehrmann M, Bogenschutz O, Batz C, Vogl W, Schmitt H, Muller CA, Muller GA: The long-term prognosis of the primary glomerulonephritides: A morphological and clinical analysis of 1747 cases. Pathol Res Pract 188:908 -924, 1992
    OpenUrlCrossRefPubMed
  81. 86.↵
    Grewal IS, Rutledge BJ, Fiorillo JA, Gu L, Gladue RP, Flavell RA, Rollins BJ: Transgenic monocyte chemoattractant protein-1 (MCP-1) in pancreatic islets produces monocyte-rich insulitis without diabetes: Abrogation by a second transgene expressing systemic MCP-1, J Immunol 159:401 -408, 1997
    OpenUrlAbstract
  82. 87.↵
    Boring L, Gosling J, Cleary M, Charo IF: Decreased lesion formation in CCR2—/— mice reveals a role for chemokines in the initiation of atherosclerosis. Nature 394:894 -897, 1998
    OpenUrlCrossRefPubMed
  83. 88.↵
    Lu B, Rutledge BJ, Gu L, Fiorillo J, Lukacs NW, Kunkel SL, North R, Gerard C, Rollins BJ: Abnormalities in monocyte recruitment and cytokine expression in monocyte chemoattractant protein 1-deficient mice. J Exp Med 187:601 -608, 1998
    OpenUrlAbstract/FREE Full Text
  84. 89.↵
    Boring L, Gosling J, Chensue SW, Kunkel SL, Farese RV Jr, Broxmeyer HE, Charo IF: Impaired monocyte migration and reduced type 1 (Th1) cytokine responses in C-C chemokine receptor 2 knockout mice. J Clin Invest 100:2552 -2561, 1997
    OpenUrlCrossRefPubMed
  85. 90.↵
    Zhou Y, Kurihara T, Ryseck RP, Yang Y, Ryan C, Loy J, Warr G, Bravo R: Impaired macrophage function and enhanced T cell-dependent immune response in mice lacking CCR5, the mouse homologue of the major HIV-1 coreceptor. J Immunol 160:4018 -4025, 1998
    OpenUrlAbstract/FREE Full Text
  86. 91.↵
    Rothenberg ME, MacLean JA, Pearlman E, Luster AD, Leder P: Targeted disruption of the chemokine eotaxin partially reduces antigen-induced tissue eosinophilia. J Exp Med 185:785 -790, 1997
    OpenUrlAbstract/FREE Full Text
  87. 92.↵
    Bleul CC, Farzan M, Choe H, Parolin C, Clark Lewis I, Sodroski J, Springer TA: The lymphocyte chemoattractant SDF-1 is a ligand for LESTR/fusin and blocks HIV-1 entry. Nature382 : 829-833,1996
    OpenUrlCrossRefPubMed
  88. 93.↵
    Zou YR, Kottmann AH, Kuroda M, Taniuchi I, Littman DR: Function of the chemokine receptor CXCR4 in haematopoiesis and in cerebellar development. Nature 393:595 -599, 1998
    OpenUrlCrossRefPubMed
  89. 94.↵
    Ma Q, Jones D, Borghesani PR, Segal RA, Nagasawa T, Kishimoto T, Bronson RT, Springer TA: Impaired B-lymphopoiesis, myelopoiesis, and derailed cerebellar neuron migration in CXCR4- and SDF-1-deficient mice. Proc Natl Acad Sci USA 95:9448 -9453, 1998
    OpenUrlAbstract/FREE Full Text
  90. 95.↵
    Tachibana K, Hirota S, Iizasa H, Yoshida H, Kawabata K, Kataoka Y, Kitamura Y, Matsushima K, Yoshida N, Nishikawa S, Kishimoto T, Nagasawa T: The chemokine receptor CXCR4 is essential for vascularization of the gastrointestinal tract. Nature393 : 591-594,1998
    OpenUrlCrossRefPubMed
  91. 96.↵
    Legler DF, Loetscher M, Roos RS, Clark Lewis I, Baggiolini M, Moser B: B cell-attracting chemokine 1, a human CXC chemokine expressed in lymphoid tissues, selectively attracts B lymphocytes via BLR1/CXCR5. J Exp Med 187: 655-660,1998
    OpenUrlAbstract/FREE Full Text
  92. 97.↵
    Ahuja SK, Murphy PM: Molecular piracy of mammalian interleukin-8 receptor type B by herpesvirus saimiri. J Biol Chem268 : 20691-20694,1993
    OpenUrlAbstract/FREE Full Text
  93. 98.↵
    Murphy PM: Molecular piracy of chemokine receptors by herpesviruses. Infect Agents Dis3 : 137-154,1994
    OpenUrlPubMed
  94. 99.↵
    Kledal TN, Rosenkilde MM, Coulin F, Simmons G, Johnsen AH, Alouani S, Power CA, Luttichau HR, Gerstoft J, Clapham PR, Clark Lewis I, Wells TNC, Schwartz TW: A broad-spectrum chemokine antagonist encoded by Kaposi's sarcoma-associated herpesvirus. Science277 : 1656-1659,1997
    OpenUrlAbstract/FREE Full Text
  95. 100.↵
    Boshoff C, Endo Y, Collins PD, Takeuchi Y, Reeves JD, Schweickart VL, Siani MA, Sasaki T, Williams TJ, Gray PW, Moore PS, Chang Y, Weiss RA: Angiogenic and HIV-inhibitory functions of KSHV-encoded chemokines. Science 278:290 -294, 1997
    OpenUrlAbstract/FREE Full Text
  96. 101.↵
    Pelchen-Matthews A, Signoret N, Klasse PJ, Fraile-Ramos A, Marsh M: Chemokine receptor trafficking and viral replication. Immunol Rev 168: 33-49,1999
    OpenUrlCrossRefPubMed
  97. 102.↵
    Zoja C, Wang JM, Bettoni S, Sironi M, Renzi D, Chiaffarino F, Abboud HE, Van Damme J, Mantovani A, Remuzzi G: Interleukin-1β and tumor necrosis factor-α induce gene expression and production of leukocyte chemotactic factors, colony-stimulating factors, and interleukin-6 in human mesangial cells. Am J Pathol138 : 991-1003,1991
    OpenUrlPubMed
  98. 103.↵
    Luckow B, Schlöndorff D, Wolf G: The monocyte chemoattractant protein 1. In: Molecular Nephrology, edited by Schlöndorff D, Bonventre JV, New York, Marcel Decker, 1995, pp653 -671
  99. 104.↵
    Rovin BH, Yoshiumura T, Tan L: Cytokine-induced production of monocyte chemoattractant protein-1 by cultured human mesangial cells. J Immunol 148:2148 -2153, 1992
    OpenUrlAbstract
  100. 105.↵
    Hora K, Satriano JA, Santiago A, Mori T, Stanley ER, Shan Z, Schlondorff D: Receptors for IgG complexes activate synthesis of monocyte chemoattractant peptide 1 and colony-stimulating factor 1. Proc Natl Acad Sci USA 89:1745 -1749, 1992
    OpenUrlAbstract/FREE Full Text
  101. 106.↵
    Satriano JA, Shuldiner M, Hora K, Xing Y, Shan Z, Schlöndorff D: Oxygen radicals as second messengers for expression of the monocyte chemoattractant protein, JE/MCP-1, and the monocyte colony-stimulating factor, CSF-1, in response to tumor necrosis factor-α and immunoglobulin G: Evidence for involvement of reduced nicotinamide adenine dinucleotide phosphate (NADPH)-dependent oxidase. J Clin Invest 92:1564 -1571, 1993
  102. 107.↵
    Stahl RA, Thaiss F, Disser M, Helmchen U, Hora K, Schlöndorff D: Increased expression of monocyte chemoattractant protein-1 in anti-thymocyte antibody-induced glomerulonephritis. Kidney Int44 : 1036-1047,1993
    OpenUrlCrossRefPubMed
  103. 108.↵
    Prodjosudjadi W, Gerritsma JS, Klar Mohamad N, Gerritsen AF, Bruijn JA, Daha MR, van Es LA: Production and cytokine-mediated regulation of monocyte chemoattractant protein-1 by human proximal tubular epithelial cells. Kidney Int 48:1477 -1486, 1995
    OpenUrlCrossRefPubMed
  104. 109.↵
    Schmouder RL, Strieter RM, Kunkel SL: Interferon-gamma regulation of human renal cortical epithelial cell-derived monocyte chemotactic peptide-1. Kidney Int 44:43 -49, 1993
    OpenUrlCrossRefPubMed
  105. 110.↵
    Gerritsma JS, van Kooten C, Gerritsen AF, Mommaas AM, van Es LA, Daha MR: Production of inflammatory mediators and cytokine responsiveness of an SV40-transformed human proximal tubular epithelial cell line. Exp Nephrol 6:208 -216, 1998
    OpenUrlCrossRefPubMed
  106. 111.↵
    Deckers JG, Van Der Woude FJ, Van Der Kooij SW, Daha MR: Synergistic effect of IL-1α, IFN-γ, and TNF-α on RANTES production by human renal tubular epithelial cells in vitro. J Am Soc Nephrol 9:194 -202, 1998
    OpenUrlAbstract
  107. 112.↵
    Banas B, Luckow B, Möller M, Holthöfer H, Nelson PJ, Schadde E, Brigl M, Halevy D, Reinhart B, Schlöndorff D: Chemokine and chemokine receptor expression in a novel human mesangial cell line. J Am Soc Nephrol 10:2314 -2322, 1999
    OpenUrlAbstract/FREE Full Text
  108. 113.↵
    Kakizaki Y, Sugimoto K, Waga S, Yoloyama M: Production of monocyte chemoattractant protein-1 (MCP-1) by bovine glomerular endothelial cells is enhanced by interleukin-1 (IL-1) and phorbol myristate acetate (PMA) [Abstract]. J Am Soc Nephrol 4:604A , 1993
    OpenUrl
  109. 114.↵
    Schwarz M, Radeke HH, Resch K, Uciechowski P: Lymphocyte-derived cytokines induce sequential expression of monocyte- and T cell-specific chemokines in human mesangial cells. Kidney Int52 : 1521-1531,1997
    OpenUrlCrossRefPubMed
  110. 115.↵
    Goppelt Struebe M, Stroebel M: Synergistic induction of monocyte chemoattractant protein-1 (MCP-1) by platelet-derived growth factor and interleukin-1. FEBS Lett 374:375 -378, 1995
    OpenUrlCrossRefPubMed
  111. 116.↵
    Beck-Schimmer B, Oertli B, Pasch T, Wüthrich RP: Hyaluronan induces monocyte chemoattractant protein-1 expression in renal tubular epithelial cells. J Am Soc Nephrol 9:2283 -2290, 1998
    OpenUrlAbstract
  112. 117.↵
    Deckers JG, De Haij S, van der Woude FJ, van der Kooij SW, Daha MR, van Kooten C: IL-4 and IL-13 augment cytokine- and CD40-induced RANTES production by human renal tubular epithelial cells in vitro. J Am Soc Nephrol 9:1187 -1193, 1998
    OpenUrlAbstract
  113. 118.↵
    Wang Y, Chen J, Chen L, Tay YC, Rangan GK, Harris DC: Induction of monocyte chemoattractant protein-1 in proximal tubule cells by urinary protein. J Am Soc Nephrol 8:1537 -1545, 1997
    OpenUrlAbstract
  114. 119.↵
    Zoja C, Donadelli R, Colleoni S, Figliuzzi M, Bonazzola S, Morigi M, Remuzzi G: Protein overload stimulates RANTES production by proximal tubular cells depending on NF-κB activation. Kidney Int 53:1608 -1615, 1998
    OpenUrlCrossRefPubMed
  115. 120.↵
    Wang Y, Rangan GK, Tay Y-C, Wang Y, Harris DCH: Induction of monocyte chemoattractant protein-1 by albumin is mediated by nuclear factor κB in proximal tubule cells. J Am Soc Nephrol10 : 1204-1213,1999
    OpenUrlAbstract/FREE Full Text
  116. 121.↵
    Terkeltaub R, Banka CL, Solan J, Santoro D, Brand K, Curtiss LK: Oxidized LDL induces monocytic cell expression of interleukin-8, a chemokine with T-lymphocyte chemotactic activity. Arterioscler Thromb 14: 47-53,1994
    OpenUrlAbstract/FREE Full Text
  117. 122.↵
    Wolf G, Ziyadeh FN, Thaiss F, Tomaszewski J, Caron RJ, Wenzel U, Zahner G, Helmchen U, Stahl RA: Angiotensin II stimulates expression of the chemokine RANTES in rat glomerular endothelial cells: Role of the angiotensin type 2 receptor. J Clin Invest100 : 1047-1058,1997
    OpenUrlCrossRefPubMed
  118. 123.↵
    Wolf G, Schneider A, Helmchen U, Stahl RA: AT1-receptor antagonists abolish glomerular MCP-1 expression in a model of mesangial proliferative glomerulonephritis. Exp Nephrol6 : 112-120,1998
    OpenUrlCrossRefPubMed
  119. 124.↵
    Klahr S, Ishidoya S, Morrissey J: Role of angiotensin II in the tubulointerstitial fibrosis of obstructive nephropathy. Am J Kidney Dis 26: 141-146,1995
    OpenUrlCrossRefPubMed
  120. 125.↵
    Hisada Y, Sugaya T, Yamanouchi M, Uchida H, Fujimura H, Sakurai H, Fukamizu A, Murakami K: Angiotensin II plays a pathogenic role in immune-mediated renal injury in mice. J Clin Invest103 : 627-635,1999
    OpenUrlCrossRefPubMed
  121. 126.↵
    Natori Y, Nishimura T, Yamabe H, Iyonaga K, Takeya M, Kawakami M: Production of monocyte chemoattractant protein-1 by cultured glomerular epithelial cells: Inhibition by dexamethasone. Exp Nephrol 5:318 -322, 1997
    OpenUrlPubMed
  122. 127.↵
    Feng L, Xia Y, Kreisberg JI, Wilson CB: Interleukin-1 alpha stimulates KC synthesis in rat mesangial cells: Glucocorticoids inhibit KC induction by IL-1. Am J Physiol266 : F713-F722,1994
    OpenUrlPubMed
  123. 128.↵
    Poon M, Megyesi J, Green RS, Zhang H, Rollins BJ, Safirstein R, Taubman MB: In vivo and in vitro inhibition of JE gene expression by glucocorticoids. J Biol Chem266 : 22375-22379,1991
    OpenUrlAbstract/FREE Full Text
  124. 129.↵
    Gerritsma JS, van Kooten C, Gerritsen AF, van Es LA, Daha MR: Transforming growth factor-beta 1 regulates chemokine and complement production by human proximal tubular epithelial cells. Kidney Int 53: 609-616,1998
    OpenUrlCrossRefPubMed
  125. 130.↵
    Ohishi S, Utsunomiya I, Yamamoto T, Komuro Y, Hara Y: Effects of prostaglandins and cyclic AMP on cytokine production in rat leukocytes. Eur J Pharmacol 300:255 -259, 1996
    OpenUrlCrossRefPubMed
  126. 131.↵
    Tanabe S, Lu Z, Luo Y, Quackenbush EJ, Berman MA, Collins Racie LA, Mi S, Reilly C, Lo D, Jacobs KA, Dorf ME: Identification of a new mouse beta-chemokine, thymus-derived chemotactic agent 4, with activity on T lymphocytes and mesangial cells. J Immunol159 : 5671-5679,1997
    OpenUrlAbstract
  127. 132.↵
    Luo Y, Dorf ME: Beta-chemokine TCA3 binds to mesangial cells and induces adhesion, chemotaxis, and proliferation. J Immunol 156:742 -748, 1996
    OpenUrlAbstract
  128. 133.↵
    Holdsworth SR, Kitching AR, Tipping PG: Th1 and Th2 T helper cell subsets affect patterns of injury and outcomes in glomerulonephritis. Kidney Int 55:1198 -1216, 1999
    OpenUrlCrossRefPubMed
  129. 134.↵
    Banbury Conference on Genetic Background in Mice: Mutant mice and neuroscience: Recommendations concerning genetic background. Neuron 19:755 -759, 1997
    OpenUrlCrossRefPubMed
  130. 135.↵
    Gerlai R: Gene-targeting studies of mammalian behavior: Is it the mutation or the background genotype? Trends Neurosci19 : 177-181,1996
    OpenUrlCrossRefPubMed
  131. 136.↵
    Liu H, Chao D, Nakayama EE, Taguchi H, Goto M, Xin X, Takamatsu JK, Saito H, Ishikawa Y, Akaza T, Juji T, Takebe Y, Ohishi T, Fukutake K, Maruyama Y, Yashiki S, Sonoda S, Nakamura T, Nagai Y, Iwamoto A, Shioda T: Polymorphism in RANTES chemokine promoter affects HIV-1 disease progression. Proc Natl Acad Sci USA 96:4581 -4585, 1999
    OpenUrlAbstract/FREE Full Text
  132. 137.↵
    Rovin BH, Lu L, Saxena R: A novel polymorphism in the MCP-1 gene regulatory region that influences MCP-1 expression. Biochem Biophys Res Commun 259:344 -348, 1999
    OpenUrlCrossRefPubMed
  133. 138.↵
    Neugarten J, Feith GW, Assmann KJ, Shan Z, Stanley ER, Schlöndorff D: Role of macrophages and colony-stimulating factor-1 in murine antiglomerular basement membrane glomerulonephritis. J Am Soc Nephrol5 : 1903-1909,1995
    OpenUrlAbstract/FREE Full Text
  134. 139.↵
    Lloyd CM, Minto AW, Dorf ME, Proudfoot A, Wells TN, Salant DJ, Gutierrez Ramos JC: RANTES and monocyte chemoattractant protein-1 (MCP-1) play an important role in the inflammatory phase of crescentic nephritis, but only MCP-1 is involved in crescent formation and interstitial fibrosis. J Exp Med 185:1371 -1380, 1997
    OpenUrlAbstract/FREE Full Text
  135. 140.↵
    Tesch GH, Schwarting A, Kinoshita K, Lan HY, Rollins BJ, Kelley VR: Monocyte chemoattractant protein-1 promotes macrophage-mediated tubular injury, but not glomerular injury, in nephrotoxic serum nephritis. J Clin Invest 103:73 -80, 1999
    OpenUrlCrossRefPubMed
  136. 141.↵
    Topham P, Hines D, Csizmadia V, Gerard C, Salant DI, Hancock WW: Regulatory role of CCR1 pathway in glomerulonephritis (GN): CCR1 knockout mice show enhanced glomerular injury [Abstract]. J Am Soc Nephrol 9: 487A,1998
    OpenUrl
  137. 142.↵
    Schadde E, Kretzler M, Banas B, Luckow B, Assmann KJM, Schlöndorff D: Chemokine receptor 1 to 5 expression in mouse anti-GBM-nephritis [Abstract]. J Am Soc Nephrol 8: 485A,1997
    OpenUrl
  138. 143.↵
    Fujinaka H, Yamamoto T, Feng L, Kawasaki K, Yaoita E, Hirose S, Goto S, Wilson CB, Uchiyama M, Kihara I: Crucial role of CD8-positive lymphocytes in glomerular expression of ICAM-1 and cytokines in crescentic glomerulonephritis of WKY rats. J Immunol158 : 4978-4983,1997
    OpenUrlAbstract
  139. 144.↵
    Rovin BH, Rumancik M, Tan L, Dickerson J: Glomerular expression of monocyte chemoattractant protein-1 in experimental and human glomerulonephritis. Lab Invest71 : 536-542,1994
    OpenUrlPubMed
  140. 145.↵
    Schneider A, Harendza S, Zahner G, Jocks T, Wenzel U, Wolf G, Thaiss F, Helmchen U, Stahl RAK: Cyclooxygenase metabolites mediate glomerular monocyte chemoattractant protein-1 formation and monocyte recruitment in experimental glomerulonephritis. Kidney Int55 : 430-444,1999
    OpenUrlCrossRefPubMed
  141. 146.↵
    Fujinaka H, Yamamoto T, Takeya M, Feng L, Kawasaki K, Yaoita E, Kondo D, Wilson CB, Uchiyama M, Kihara I: Suppression of anti-glomerular basement membrane nephritis by administration of anti-monocyte chemoattractant protein-1 anti-body in WKY rats. J Am Soc Nephrol8 : 1174-1178,1997
    OpenUrlAbstract
  142. 147.↵
    Sakurai H, Hisada Y, Ueno M, Sugiura M, Kawashima K, Sugita T: Activation of transcription factor NF-kappa B in experimental glomerulonephritis in rats. Biochim Biophys Acta1316 : 132-138,1996
    OpenUrlPubMed
  143. 148.↵
    Wu X, Dolecki GJ, Sherry B, Zagorski J, Lefkowith JB: Chemokines are expressed in a myeloid cell-dependent fashion and mediate distinct functions in immune complex glomerulonephritis in rat. J Immunol 158:3917 -3924, 1997
    OpenUrlAbstract
  144. 149.↵
    Tang WW, Yin S, Wittwer AJ, Qi M: Chemokine gene expression in anti-glomerular basement membrane antibody glomerulonephritis. Am J Physiol 269:F323 -F330, 1995
    OpenUrlPubMed
  145. 150.↵
    Chen S, Bacon KB, Li L, Garcia GE, Xia Y, Lo D, Thompson DA, Siani MA, Yamamoto T, Harrison JK, Feng L: In vivo inhibition of CC and CX3C chemokine-induced leukocyte infiltration and attenuation of glomerulonephritis in Wistar-Kyoto (WKY) rats by vMIP-II. J Exp Med188 : 193-198,1998
    OpenUrlAbstract/FREE Full Text
  146. 151.↵
    Natori Y, Sekiguchi M, Ou Z, Natori Y: Gene expression of CC chemokines in experimental crescentic glomerulonephritis. Clin Exp Immunol 109:143 -148, 1997
    OpenUrlCrossRefPubMed
  147. 152.↵
    Sekiguchi M, Natori Y, Iyonaga K, Takeya M: Expression of monocyte chemoattractant protein-1 in experimental crescentic glomerulonephritis in rats. J Lab Clin Med 129:239 -244, 1997
    OpenUrlCrossRefPubMed
  148. 153.↵
    Feng L, Xia Y, Yoshimura T, Wilson CB: Modulation of neutrophil influx in glomerulonephritis in the rat with anti-macrophage inflammatory protein-2 (MIP-2) antibody. J Clin Invest95 : 1009-1017,1995
  149. 154.↵
    Feng L, Chen S, Garcia GE, Xia Y, Siani MA, Botti P, Wilson CB, Harrison JK, Bacon KB: Prevention of crescentic glomerulonephritis by immunoneutralization of the fractalkine receptor CX3CR1. Kidney Int 56: 612-620,1999
    OpenUrlCrossRefPubMed
  150. 155.↵
    Natori Y, Ou ZL, Yamamoto Shuda Y: Expression of lymphotactin mRNA in experimental crescentic glomerulonephritis. Clin Exp Immunol 113:265 -268, 1998
    OpenUrlCrossRefPubMed
  151. 156.↵
    Tang WW, Qi M, Warren JS: Monocyte chemoattractant protein 1 mediates glomerular macrophage infiltration in anti-GBM Ab GN. Kidney Int 50:665 -671, 1996
    OpenUrlCrossRefPubMed
  152. 157.↵
    Wada T, Yokoyama H, Furuichi K, Kobayashi KI, Harada K, Naruto M, Su SB, Akiyama M, Mukaida N, Matsushima K: Intervention of crescentic glomerulonephritis by antibodies to monocyte chemotactic and activating factor (MCAF/MCP-1). FASEB J 10:1418 -1425, 1996
    OpenUrlPubMed
  153. 158.↵
    Stahl RKA, Thaiss F, Kahf S, Shoeppe W, Helmchen UM: Immune-mediated mesangial cell injury: Biosynthesis and function of prostanoids. Kidney Int 38:273 -281, 1990
    OpenUrlCrossRefPubMed
  154. 159.↵
    De Heer E, Prodjosudjadi W, Davidoff A, van der Wal A, Bruijn JA, Paul LC: Control of monocyte influx in glomerulonephritis in transplanted kidneys in the rat. Lab Invest78 : 1327-1337,1998
    OpenUrlPubMed
  155. 160.↵
    Wenzel U, Schneider A, Valente AJ, Abboud HE, Thaiss F, Helmchen UM, Stahl RA: Monocyte chemoattractant protein-1 mediates monocyte/macrophage influx in anti-thymocyte anti-body-induced glomerulonephritis. Kidney Int 51:770 -776, 1997
    OpenUrlCrossRefPubMed
  156. 161.↵
    Jocks T, Zahner G, Freudenberg J, Wolf G, Thaiss F, Helmchen U, Stahl RA: Prostaglandin E1 reduces the glomerular mRNA expression of monocyte-chemoattractant protein 1 in anti-thymocyte antibody-induced glomerular injury. J Am Soc Nephrol7 : 897-905,1996
    OpenUrlAbstract
  157. 162.↵
    Panzer U, Schneider A, Wilken J, Thompson DA, Kent SBH, Stahl RAK: The chemokine receptor antagonist AOP-RANTES reduces monocyte infiltration in experimental glomerulonephritis [Abstract]. J Am Soc Nephrol 9: 465A,1998
    OpenUrl
  158. 163.↵
    Zoja C, Liu XH, Donadelli R, Abbate M, Testa D, Corna D, Taraboletti G, Vecchi A, Dong QG, Rollins BJ, Bertani T, Remuzzi G: Renal expression of monocyte chemoattractant protein-1 in lupus autoimmune mice. J Am Soc Nephrol 8:720 -729, 1997
    OpenUrlAbstract
  159. 164.↵
    Zoja C, Corna D, Benedetti G, Morigi M, Donadelli R, Guglielmotti A, Pinza M, Bertani T, Remuzzi G: Bindarit retards renal disease and prolongs survival in murine lupus autoimmune disease. Kidney Int 53: 726-734,1998
    OpenUrlCrossRefPubMed
  160. 165.↵
    Moore KJ, Wada T, Barbee SD, Kelley VR: Gene transfer of RANTES elicits autoimmune renal injury in MRL-Fas(1pr) mice. Kidney Int 53:1631 -1641, 1998
    OpenUrlCrossRefPubMed
  161. 166.↵
    Wada T, Tomosugi N, Naito T, Yokoyama H, Kobayashi K, Harada A, Mukaida N, Matsushima K: Prevention of proteinuria by the administration of anti-interleukin 8 antibody in experimental acute immune complex-induced glomerulonephritis. J Exp Med180 : 1135-1140,1994
    OpenUrlAbstract/FREE Full Text
  162. 167.↵
    Ruiz Ortega M, Bustos C, Hernandez Presa MA, Lorenzo O, Plaza JJ, Egido J: Angiotensin II participates in mononuclear cell recruitment in experimental immune complex nephritis through nuclear factor-kappa B activation and monocyte chemoattractant protein-1 synthesis. J Immunol 161:430 -439, 1998
    OpenUrlAbstract/FREE Full Text
  163. 168.↵
    Anders HJ, Vielhauer V, Segerer S, Luckow B, Weller L, Gröne HJ, Schlöndorff D: CC-Chemokine and receptor expression during immune-complex glomerulonephritis (IC-GN) in mice [Abstract]. J Am Soc Nephrol10 : 525A,1999
    OpenUrl
  164. 169.↵
    Tang WW, Qi M, Warren JS, Van GY: Chemokine expression in experimental tubulointerstitial nephritis. J Immunol159 : 870-876,1997
    OpenUrlAbstract
  165. 170.↵
    Park YS, Guijarro C, Kim Y, Massy ZA, Kasiske BL, Keane WF, O'Donnell MP: Lovastatin reduces glomerular macrophage influx and expression of monocyte chemoattractant protein-1 mRNA in nephrotic rats. Am J Kidney Dis 31:190 -194, 1998
    OpenUrlCrossRefPubMed
  166. 171.↵
    Ou ZL, Natori Y: Gene expression of CC chemokines in experimental acute tubulointerstitial nephritis. J Lab Clin Med133 : 41-47,1999
    OpenUrlCrossRefPubMed
  167. 172.↵
    Kim SY, Guijarro C, O'Donnell MP, Kasiske BL, Kim Y, Keane WF: Human mesangial cell production of monocyte chemoattractant protein-1: Modulation by lovastatin. Kidney Int48 : 363-371,1995
    OpenUrlCrossRefPubMed
  168. 173.↵
    Tang WW, Feng L, Mathison JC, Wilson CB: Cytokine expression, upregulation in intercellular adhesion molecule-1, and leukocyte infiltration in experimental tubulointerstitial nephritis. Lab Invest 70:631 -638, 1994
    OpenUrlPubMed
  169. 174.↵
    Feng L, Xia Y, Chen S, Wilson CB: Blocking macrophage infiltration in rats with tubulointerstitial nephritis (TIN) with monocyte chemoattractant protein-1 (MCP-1) receptor antagonist (RA) [Abstract]. J Am Soc Nephrol 9: 494A,1998
    OpenUrl
  170. 175.↵
    Eddy AA, Giachelli CM: Renal expression of genes that promote interstitial inflammation and fibrosis in rats with protein-over-load proteinuria. Kidney Int 47:1546 -1557, 1995
    OpenUrlCrossRefPubMed
  171. 176.↵
    Pasi A, Dendorfer U, Holthofer H, Nelson PJ, Tazzari S, Armelloni S, Fornasieri A, D' Amico G, Schlondorff D: Characterization of nephropathy induced by immunization with high molecular weight dextran. Nephrol Dial Transplant 12:1849 -1855, 1997
    OpenUrlCrossRefPubMed
  172. 177.↵
    Haberstroh U, Stilo K, Pocock J, Wolf G, Helmchen U, Wenzel U, Zahner G, Stahl RA, Thaiss F: L-arginine suppresses lipopolysaccharide-induced expression of RANTES in glomeruli. J Am Soc Nephrol9 : 203-210,1998
    OpenUrlAbstract
  173. 178.↵
    Morrissey JJ, Klahr S: Differential effects of ACE and AT1 receptor inhibition on chemoattractant and adhesion molecule synthesis. Am J Physiol 274:F580 -F586, 1998
    OpenUrlPubMed
  174. 179.↵
    Diamond JR, Kees Folts D, Ding G, Frye JE, Restrepo NC: Macrophages, monocyte chemoattractant peptide-1, and TGF-beta 1 in experimental hydronephrosis. Am J Physiol266 : F926-F933,1994
    OpenUrlPubMed
  175. 180.↵
    Safirstein R, Megyesi J, Saggi SJ, Price PM, Poon M, Rollins BJ, Taubman MB: Expression of cytokine-like genes JE and KC is increased during renal ischemia. Am J Physiol261 : F1095-F1101,1991
    OpenUrlPubMed
  176. 181.↵
    Chiao H, Kohda Y, McLeroy P, Craig L, Housini I, Star RA: Alpha-melanocyte-stimulating hormone protects against renal injury after ischemia in mice and rats. J Clin Invest99 : 1165-1172,1997
    OpenUrlCrossRefPubMed
  177. 182.↵
    Chandraker A, Takada M, Nadeau KC, Peach R, Tilney NL, Sayegh MH: CD28-b7 blockade in organ dysfunction secondary to cold ischemia/reperfusion injury. Kidney Int 52:1678 -1684, 1997
    OpenUrlCrossRefPubMed
  178. 183.↵
    Shoskes DA: Effect of bioflavonoids quercetin and curcumin on ischemic renal injury: A new class of renoprotective agents. Transplantation 66:147 -152, 1998
    OpenUrlCrossRefPubMed
  179. 184.↵
    Nadeau KC, Azuma H, Tilney NL: Sequential cytokine dynamics in chronic rejection of rat renal allografts: Roles for cytokines RANTES and MCP-1. Proc Natl Acad Sci USA92 : 8729-8733,1995
    OpenUrlAbstract/FREE Full Text
  180. 185.↵
    Nagano H, Nadeau KC, Takada M, Kusaka M, Tilney NL: Sequential cellular and molecular kinetics in acutely rejecting renal allografts in rats. Transplantation 63:1101 -1108, 1997
    OpenUrlCrossRefPubMed
  181. 186.↵
    Grandaliano G, Gesualdo L, Ranieri E, Monno R, Montinaro V, Marra F, Schena FP: Monocyte chemotactic peptide-1 expression in acute and chronic human nephritides: A pathogenetic role in interstitial monocytes recruitment. J Am Soc Nephrol 7:906 -913, 1996
    OpenUrlAbstract
  182. 187.↵
    Wada T, Yokoyama H, Tomosugi N, Hisada Y, Ohta S, Naito T, Kobayashi K, Mukaida N, Matsushima K: Detection of urinary interleukin-8 in glomerular diseases. Kidney Int46 : 455-460,1994
    OpenUrlCrossRefPubMed
  183. 188.↵
    Yokoyama H, Wada T, Furuichi K, Segawa C, Shimizu M, Kobayashi K, Su S, Mukaida N, Matsushima K: Urinary levels of chemokines (MCAF/MCP-1, IL-8) reflect distinct disease activities and phases of human IgA nephropathy. J Leukocyte Biol 63:493 -499, 1998
    OpenUrlPubMed
  184. 189.↵
    Wada T, Yokoyama H, Su SB, Mukaida N, Iwano M, Dohi K, Takahashi Y, Sasaki T, Furuichi K, Segawa C, Hisada Y, Ohta S, Takasawa K, Kobayashi K, Matsushima K: Monitoring urinary levels of monocyte chemotactic and activating factor reflects disease activity of lupus nephritis. Kidney Int 49: 761-767,1996
    OpenUrlCrossRefPubMed
  185. 190.↵
    Rovin BH, Doe N, Tan LC: Monocyte chemoattractant protein-1 levels in patients with glomerular disease. Am J Kidney Dis27 : 640-646,1996
    OpenUrlCrossRefPubMed
  186. 191.↵
    Gesualdo L, Grandaliano G, Ranieri E, Monno R, Montinaro V, Manno C, Schena FP: Monocyte recruitment in cryoglobulinemic membranoproliferative glomerulonephritis: A pathogenetic role for monocyte chemotactic peptide-1. Kidney Int 51:155 -163, 1997
    OpenUrlPubMed
  187. 192.↵
    Noris M, Bernasconi S, Casiraghi F, Sozzani S, Gotti E, Remuzzi G, Mantovani A: Monocyte chemoattractant protein-1 is excreted in excessive amounts in the urine of patients with lupus nephritis. Lab Invest 73:804 -809, 1995
    OpenUrlPubMed
  188. 193.↵
    Prodjosudjadi W, Gerritsma JS, van Es LA, Daha MR, Bruijn JA: Monocyte chemoattractant protein-1 in normal and diseased human kidneys: An immunohistochemical analysis. Clin Nephrol44 : 148-155,1995
    OpenUrlPubMed
  189. 194.↵
    Yamauchi F, Kashem A, Endoh M, Nomoto Y, Sakai H: Detection of monocyte chemoattractant protein-1 (MCP-1) and its receptors in mesangial proliferative glomerulonephritis. Nephrology2 : 93-99,1996
  190. 195.↵
    Segerer S, Mack M, Regele H, Kerjaschki D, Schlöndorff D: Expression of the C-C chemokine receptor 5 in human kidney diseases. Kidney Int56 : 52-64,1999
    OpenUrlPubMed
  191. 196.↵
    Mezzano S, Droguett M, Burgos M, Ardiles L, Caorsi I, Egido J: In situ detection of chemokines (MCP-1, RANTES, IL-8), osteopontin, fibrogenic cytokines (PDGF, TGF-beta) and apoptotic cells in human idiopathic membranous nephropathy [Abstract]. J Am Soc Nephrol9 : 463A,1998
    OpenUrl
  192. 197.↵
    Cockwell P, Howie AJ, Adu D, Savage CO: In situ analysis of C-C chemokine mRNA in human glomerulonephritis. Kidney Int54 : 827-836,1998
    OpenUrlCrossRefPubMed
  193. 198.↵
    Segerer S, Mack M, Kain R, Regele H, Cartron JP, Kerjaschki D, Schlöndorff D: Chemokine receptors CCR5 and DARC in crescentic glomerulonephritis (cGN) [Abstract]. J Am Soc Nephrol 10: 591A,1999
    OpenUrl
  194. 199.↵
    Wada T, Furuichi K, Segawa-Takaeda C, Shimizu M, Sakai N, Takeda SI, Takasawa K, Kida H, Kobayashi KI, Mukaida N, Ohmoto Y, Matsushima K, Yokoyama H: MIP-1α and MCP-1 contribute to crescents and interstitial lesions in human crescentic glomerulonephritis. Kidney Int 56: 995-1003,1999
    OpenUrlCrossRefPubMed
  195. 200.↵
    Cockwell P, Brooks CJ, Adu D, Savage COS: Interleukin-8: A pathognentic role in antineutrophil cytoplasmic autoantibody-associated glomerulonephritis. Kidney Int55 : 852-863,1999
    OpenUrlCrossRefPubMed
  196. 201.↵
    Eitner F, Cui Y, Hudkins KL, Anderson DM, Schmidt A, Morton WR, Alpers CE: Chemokine receptor (CCR5) expression in human kidneys and in the HIV infected macaque. Kidney Int54 : 1945-1954,1998
    OpenUrlCrossRefPubMed
  197. 202.↵
    Hall BM, Bishop GA, Farnsworth A, Duggin GG, Horvath JS, Sheil AG, Tiller DJ: Identification of the cellular subpopulations infiltrating rejecting cadaver renal allografts: Preponderance of the T4 subset of T cells. Transplantation 37:564 -570, 1984
    OpenUrlPubMed
  198. 203.↵
    Hall BM: Cells mediating allograft rejection. Transplantation 51:1141 -1151, 1991
    OpenUrlCrossRefPubMed
  199. 204.↵
    Grandaliano G, Gesualdo L, Ranieri E, Monno R, Stallone G, Schena FP: Monocyte chemotactic peptide-1 expression and monocyte infiltration in acute renal transplant rejection. Transplantation63 : 414-420,1997
    OpenUrlCrossRefPubMed
  200. 205.↵
    Pattison J, Nelson PJ, Huie P, von Leuttichau I, Farshid G, Sibley RK, Krensky AM: RANTES chemokine expression in cell-mediated transplant rejection of the kidney. Lancet343 : 209-211,1994
    OpenUrlCrossRefPubMed
  201. 206.↵
    Budde K, Waiser J, Ceska M, Katalinic A, Kurzdorfer M, Neumayer HH: Interleukin-8 expression in patients after renal transplantation. Am J Kidney Dis 29:871 -880, 1997
    OpenUrlPubMed
  202. 207.↵
    Schmouder RL, Streiter RM, Walz A, Kunkel SL: Epithelial-derived neutrophil-activating factor-78 production in human renal tubule epithelial cells and in renal allograft rejection. Transplantation 59:118 -124, 1995
    OpenUrlPubMed
  203. 208.↵
    Robertson H, Wheeler J, Morley AR, Booth TA, Talbot D, Kirby JA: Beta-chemokine expression and distribution in paraffin-embedded transplant renal biopsy sections: Analysis by scanning laser confocal microscopy. Histochem Cell Biol 110:207 -213, 1998
    OpenUrlCrossRefPubMed
  204. 209.↵
    Schmouder RL, Strieter RM, Wiggins RC, Chensue SW, Kunkel SL: In vitro and in vivo interleukin-8 production in human renal cortical epithelia. Kidney Int 41:191 -198, 1992
    OpenUrlCrossRefPubMed
  205. 210.↵
    von Luettichau I, Nelson PJ, Pattison JM, van de Rijn M, Huie P, Warnke R, Wiedermann CJ, Stahl RA, Sibley RK, Krensky AM:RANTES chemokine expression in diseased and normal human tissues. Cytokine 8:89 -98, 1996
    OpenUrlCrossRefPubMed
  206. 211.↵
    Strehlau J, Pavlakis M, Lipman M, Shapiro M, Vasconcellos L, Harmon W, Strom TB: Quantitative detection of immune activation transcripts as a diagnostic tool in kidney transplantation. Proc Natl Acad Sci USA 94: 695-700,1997
    OpenUrlAbstract/FREE Full Text
  207. 212.↵
    Liu XH, Hadley TJ, Xu L, Peiper SC, Ray PE: Up-regulation of Duffy antigen receptor expression in children with renal disease. Kidney Int 55:1491 -1500, 1999
    OpenUrlCrossRefPubMed
  208. 213.↵
    Segerer S, Regele H, Mack M, Cartron JP, Kerjaschki D, Schlöndorff D: The Duffy antigen receptor for chemokines is upregulated during renal transplant rejection [Abstract]. J Am Soc Nephrol 10:713A , 1999
    OpenUrl
  209. 214.↵
    van Setten PA, van Hinsbergh VW, van den Heuvel LP, Preyers F, Dijkman HB, Assmann KJ, van der Velden TJ, Monnens LA: Monocyte chemoattractant protein-1 and interleukin-8 levels in urine and serum of patents with hemolytic uremic syndrome. Pediatr Res43 : 759-767,1998
    OpenUrlCrossRefPubMed
  210. 215.↵
    Brady HR, Adler S: Cell-cell and cell-matrix interactions. In: The Kidney, Vol. 1, edited by Brenner BM, 1996, pp 193-194
    OpenUrl
  211. 216.↵
    Kakizaki Y, Waga S, Sugimoto K, Tanaka H, Nukii K, Takeya M, Yoshimura T, Yokoyama M: Production of monocyte chemoattractant protein-1 by bovine glomerular endothelial cells. Kidney Int48 : 1866-1874,1995
    OpenUrlCrossRefPubMed
  212. 217.↵
    Epstein F: Nuclear factor-κB: A pivotal transcription factor in chronic inflammatory diseases. N Engl J Med336 : 1066-1071,1997
    OpenUrlCrossRefPubMed
  213. 218.↵
    Duque N, Gomez Guerrero C, Egido J: Interaction of IgA with Fc alpha receptors of human mesangial cells activates transcription factor nuclear factor-kappa B and induces expression and synthesis of monocyte chemoattractant protein-1, IL-8, and IFN-inducible protein 10. J Immunol 159:3474 -3482, 1997
    OpenUrlAbstract
  214. 219.↵
    Schlöndorff D: Renal complications of nonsteroidal anti-inflammatory drugs. Kidney Int44 : 643-653,1993
    OpenUrlCrossRefPubMed
  215. 220.↵
    Thivierge M, Le Gouill C, Tremblay MJ, Stankova J, Rola Pleszczynski M: Prostaglandin E2 induces resistance to human immunodeficiency virus-1 infection in monocyte-derived macrophages: Downregulation of CCR5 expression by cyclic adenosine monophosphate. Blood92 : 40-45,1998
    OpenUrlAbstract/FREE Full Text
  216. 221.
    Brown Z, Strieter RM, Neild GH, Thompson RC, Kunkel SL, Westwick J: IL-1 receptor antagonist inhibits monocyte chemotactic peptide 1 generation by human mesangial cells. Kidney Int42 : 95-101,1992
    OpenUrlCrossRefPubMed
  217. 222.
    Brown Z, Strieter RM, Chensue SW, Ceska M, Lindley I, Neild G, Kunkel SL, Westwick J: Cytokine-activated human mesangial cells generate the neutrophil chemoattractant, interleukin 8. Kidney Int40 : 86-90,1991
    OpenUrlCrossRefPubMed
  218. 223.
    Uciechowski P, Schwarz M, Gessner JE, Schmidt RE, Resch K, Radeke HH: IFN-gamma induces the high-affinity Fc receptor I for IgG (CD64) on human glomerular mesangial cells. Eur J Immunol28 : 2928-2935,1998
    OpenUrlCrossRefPubMed
  219. 224.
    Lee SK, Park JY, Chung SJ, Yang WS, Kim SB, Park SK, Park JS: Chemokines, osteopontin, ICAM-1 gene expression in cultured rat mesangial cells. J Korean Med Sci 13:165 -170, 1998
    OpenUrlPubMed
  220. 225.
    Wu X, Dolecki GJ, Lefkowith JB: GRO chemokines: A transduction, integration, and amplification mechanism in acute renal inflammation. Am J Physiol 269:F248 -F256, 1995
    OpenUrlPubMed
  221. 226.
    Gerritsma JS, Hiemstra PS, Gerritsen AF, Prodjosudjadi W, Verweij CL, Van Es LA, Daha MR: Regulation and production of IL-8 by human proximal tubular epithelial cells in vitro. Clin Exp Immunol103 : 289-294,1996
    OpenUrlCrossRefPubMed
  222. 227.
    van Kooten C, Boonstra JG, Paape ME, Fossiez F, Banchereau J, Lebecque S, Bruijn JA, De Fijter JW, Van Es LA, Daha MR: Interleukin-17 activates human renal epithelial cells in vitro and is expressed during renal allograft rejection. J Am Soc Nephrol9 : 1526-1534,1998
    OpenUrlAbstract
  223. 228.
    Biancone L, Conaldi PG, Toniolo A, Camussi G: Escherichia coli porin induces proinflammatory alterations in renal tubular cells. Exp Nephrol 5:330 -336, 1997
    OpenUrlPubMed
  224. 229.
    van Kooten C, Gerritsma JS, Paape ME, van Es LA, Banchereau J, Daha MR: Possible role for CD40-CD40L in the regulation of interstitial infiltration in the kidney. Kidney Int51 : 711-721,1997
    OpenUrlCrossRefPubMed
  225. 230.
    Chen X, Liao H, Ye Y: Expression of IL-8 mRNA in glomerular endothelial cells. Chung Hua I Hsueh Tsa Chih76 : 420-422,1996
  226. 231.
    Gomez Chiarri M, Hamilton TA, Egido J, Emancipator SN: Expression of IP-10, a lipopolysaccharide- and interferon-gamma-inducible protein, in murine mesangial cells in culture. Am J Pathol142 : 433-439,1993
    OpenUrlPubMed
  227. 232.
    Gonzalez Cuadrado S, Bustos C, Ruiz Ortega M, Ortiz A, Guijarro C, Plaza JJ, Egido J: Expression of leukocyte chemoattractants by interstitial renal fibroblasts: Up-regulation by drugs associated with interstitial fibrosis. Clin Exp Immunol 106:518 -522, 1996
    OpenUrlCrossRefPubMed
  228. 233.
    Gomez Chiarri M, Ortiz A, Gonzalez Cuadrado S, Seron D, Emancipator SN, Hamilton TA, Barat A, Plaza JJ, Gonzalez E, Egido J: Interferon-inducible protein-10 is highly expressed in rats with experimental nephrosis. Am J Pathol 148:301 -311, 1996
    OpenUrlPubMed
  229. 234.
    Grandaliano G, Valente AJ, Rozek MM, Abboud HE: Gamma interferon stimulates monocyte chemotactic protein (MCP-1) in human mesangial cells. J Lab Clin Med 123:282 -289, 1994
    OpenUrlPubMed
  230. 235.
    Largen PJ, Tam FW, Rees AJ, Cattell V: Rat mesangial cells have a selective role in macrophage recruitment and activation. Exp Nephrol 3: 34-39,1995
    OpenUrlPubMed
  231. 236.
    Hartner A, Goppelt Struebe M, Hocke GM, Sterzel RB: Differential regulation of chemokines by leukemia inhibitory factor, interleukin-6 and oncostatin M. Kidney Int 51:1754 -1760, 1997
    OpenUrlCrossRefPubMed
  232. 237.
    Pai R, Ha H, Kirschenbaum MA, Kamanna VS: Role of tumor necrosis factor-alpha on mesangial cell MCP-1 expression and monocyte migration: Mechanisms mediated by signal transduction. J Am Soc Nephrol 7:914 -923, 1996
    OpenUrlAbstract
  233. 238.
    Braun M, Davis AE 3rd: Cultured human glomerular mesangial cells express the C5a receptor. Kidney Int54 : 1542-1549,1998
    OpenUrlCrossRefPubMed
  234. 239.
    Ihm CG, Park JK, Hong SP, Lee TW, Cho BS, Kim MJ, Cha DR, Ha H: A high glucose concentration stimulates the expression of monocyte chemotactic peptide 1 in human mesangial cells. Nephron79 : 33-37,1998
    OpenUrlCrossRefPubMed
  235. 240.
    Kitamura M: Identification of an inhibitor targeting macrophage production of monocyte chemoattractant protein-1 as TGF-β1. J Immunol 159:1404 -1411, 1997
    OpenUrlAbstract
  236. 241.
    Singhal PC, Shan Z, Garg P, Sharma K, Sharma P, Gibbons N: Morphine modulates migration of monocytes. Nephron73 : 526-531,1996
    OpenUrlCrossRefPubMed
  237. 242.
    Kamanna VS, Pai R, Roh DD, Kirschenbaum MA: Oxidative modification of low-density lipoprotein enhances the murine mesangial cell cytokines associated with monocyte migration, differentiation, and proliferation. Lab Invest 74:1067 -1079, 1996
    OpenUrlPubMed
  238. 243.
    Simon M, Cleary TG, Hernandez JD, Abboud HE: Shiga toxin 1 elicits diverse biologic responses in mesangial cells. Kidney Int 54:1117 -1127, 1998
    OpenUrlCrossRefPubMed
  239. 244.
    Grandaliano G, Valente AJ, Abboud HE: A novel biologic activity of thrombin: Stimulation of monocyte chemotactic protein production. J Exp Med 179:1737 -1741, 1994
    OpenUrlAbstract/FREE Full Text
  240. 245.
    Wolf G, Aberle S, Thaiss F, Nelson PJ, Krensky AM, Neilson EG, Stahl RA: TNF alpha induces expression of the chemoattractant cytokine RANTES in cultured mouse mesangial cells. Kidney Int44 : 795-804,1993
    OpenUrlCrossRefPubMed
  241. 246.
    Nelson PJ, Kim HT, Manning WC, Goralski TJ, Krensky AM: Genomic organization and transcriptional regulation of the RANTES chemokine gene. J Immunol 151:2601 -2612, 1993
    OpenUrlAbstract
  242. 247.
    Satriano JA, Banas B, Luckow B, Nelson P, Schlöndorff D: Regulation of RANTES and ICAM-1 expression in murine messangial cells. J Am Soc Nephrol 8:596 -603, 1997
    OpenUrlAbstract
  243. 248.
    Heeger P, Wolf G, Meyers C, Sun MJ, O'Farrell SC, Krensky AM, Neilson EG: Isolation and characterization of cDNA from renal tubular epithelium encoding murine RANTES. Kidney Int41 : 220-225,1992
    OpenUrlCrossRefPubMed
  244. 249.
    Radeke HH, Wahl M, Resch K, Schwarz M: T helper lymphokines induce receptor expression and responsiveness to chemokines in human glomerular mesangial cells (HMC) [Abstract]. J Am Soc Nephrol9 : 466A,1998
    OpenUrl
  245. 250.
    Tam FW, Karkar AM, Smith J, Yoshimura T, Steinkasserer A, Kurrle R, Langner K, Rees AJ: Differential expression of macrophage inflammatory protein-2 and monocyte chemoattractant protein-1 in experimental glomerulonephritis. Kidney Int49 : 715-721,1996
    OpenUrlPubMed
  246. 251.
    Eddy AA, Warren JS: Expression and function of monocyte chemoattractant protein-1 in experimental nephrotic syndrome. Clin Immunol Immunopathol 78:140 -151, 1996
    OpenUrlCrossRefPubMed
  247. 252.
    Harada A, Sekido N, Akahoshi T, Wada T, Mukaida N, Matsushima K: Essential involvement of interleukin-8 (IL-8) in acute inflammation. J Leukocyte Biol 56:559 -564, 1994
    OpenUrlPubMed
  248. 253.
    Schiller B, Moran J: Focal glomerulosclerosis in the remnant kidney model-an inflammatory disease mediated by cytokines. Nephrol Dial Transplant 12:430 -437, 1997
    OpenUrlCrossRefPubMed
  249. 254.
    Shoskes DA, Jones E, Garras N, Satyanarayana K: Effect of the lazaroid U-74389G on chemokine gene expression and apoptosis in renal ischemia-reperfusion injury. Transplant Proc30 : 974-975,1998
    OpenUrlCrossRefPubMed
  250. 255.
    Feng L, Xia Y, Wilson CB: Sequential expression of C-X-C chemokines correlates with the influx of neutrophils and mononuclear cells in autoimmune tubulointerstitial nephritis in rats [Abstract]. J Am Soc Nephrol 6: 828A,1995
    OpenUrl
PreviousNext
Back to top

In this issue

Journal of the American Society of Nephrology: 11 (1)
Journal of the American Society of Nephrology
Vol. 11, Issue 1
1 Jan 2000
  • Table of Contents
  • Index by author
View Selected Citations (0)
Print
Download PDF
Sign up for Alerts
Email Article
Thank you for your help in sharing the high-quality science in JASN.
Enter multiple addresses on separate lines or separate them with commas.
Chemokines, Chemokine Receptors, and Renal Disease: From Basic Science To Pathophysiologic and Therapeutic Studies
(Your Name) has sent you a message from American Society of Nephrology
(Your Name) thought you would like to see the American Society of Nephrology web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Chemokines, Chemokine Receptors, and Renal Disease: From Basic Science To Pathophysiologic and Therapeutic Studies
STEPHAN SEGERER, PETER J. NELSON, DETLEF SCHLÖNDORFF
JASN Jan 2000, 11 (1) 152-176;

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Request Permissions
Share
Chemokines, Chemokine Receptors, and Renal Disease: From Basic Science To Pathophysiologic and Therapeutic Studies
STEPHAN SEGERER, PETER J. NELSON, DETLEF SCHLÖNDORFF
JASN Jan 2000, 11 (1) 152-176;
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like

Jump to section

  • Article
    • Abstract
    • Chemokines: Classification and Mechanisms of Action
    • Chemokines, Chemokine Receptors, and the Kidney
    • Expression of Chemokines and Chemokine Receptors in Experimental Models of Renal Diseases
    • Other Renal Disease Models
    • Chemokines in Human Kidney Diseases
    • A Model for Chemokines in Renal Diseases
    • Conclusions, Future Directions, and Therapeutic Outlook
    • Acknowledgments
    • Footnotes
    • References
    • References
  • Figures & Data Supps
  • Info & Metrics
  • View PDF

More in this TOC Section

  • Management of Obesity in Adults with CKD
  • Sphingosine-1-Phosphate Metabolism and Signaling in Kidney Diseases
  • Leveraging the Capabilities of the FDA’s Sentinel System To Improve Kidney Care
Show more Review

Cited By...

  • Epithelial Cell TGF{beta} Signaling Induces Acute Tubular Injury and Interstitial Inflammation
  • Spleen Tyrosine Kinase Is Important in the Production of Proinflammatory Cytokines and Cell Proliferation in Human Mesangial Cells following Stimulation with IgA1 Isolated from IgA Nephropathy Patients
  • Chemokines in Renal Injury
  • Differential Regulation of Kidney and Spleen Cytokine Responses in Mice Challenged with Pathology-Standardized Doses of Candida albicans Mannosylation Mutants
  • Intrapersonal and Populational Heterogeneity of the Chemokine RANTES
  • Blockade of the Kinin B1 Receptor Ameloriates Glomerulonephritis
  • New Insights into Epithelial-Mesenchymal Transition in Kidney Fibrosis
  • N-Acetyl-Seryl-Aspartyl-Lysyl-Proline Attenuates Renal Injury and Dysfunction in Hypertensive Rats With Reduced Renal Mass: Council for High Blood Pressure Research
  • Tumor Necrosis Factor-Like Weak Inducer of Apoptosis (TWEAK) Enhances Vascular and Renal Damage Induced by Hyperlipidemic Diet in ApoE-Knockout Mice
  • A CD26-Controlled Cell Surface Cascade for Regulation of T Cell Motility and Chemokine Signals
  • Serum Concentrations of Markers of TNF{alpha} and Fas-Mediated Pathways and Renal Function in Nonproteinuric Patients with Type 1 Diabetes
  • Chemokine Receptor CCR1 Regulates Inflammatory Cell Infiltration after Renal Ischemia-Reperfusion Injury
  • Complement 5a Receptor Inhibition Improves Renal Allograft Survival
  • Paricalcitol Inhibits Renal Inflammation by Promoting Vitamin D Receptor-Mediated Sequestration of NF-{kappa}B Signaling
  • Renal Urokinase-Type Plasminogen Activator (uPA) Receptor but not uPA Deficiency Strongly Attenuates Ischemia Reperfusion Injury and Acute Kidney Allograft Rejection
  • Association of Urinary Inflammatory Markers and Renal Decline in Microalbuminuric Type 1 Diabetics
  • The Cytokine TWEAK Modulates Renal Tubulointerstitial Inflammation
  • Inhibitory ITAM Signaling by Fc{alpha}RI-FcR{gamma} Chain Controls Multiple Activating Responses and Prevents Renal Inflammation
  • Role of Renal Cortical Neovascularization in Experimental Hypercholesterolemia
  • ELR+-CXC Chemokines and Their Receptors in Early Metanephric Development
  • Selective Binding and Presentation of CCL5 by Discrete Tissue Microenvironments during Renal Inflammation
  • C3a Is Required for the Production of CXC Chemokines by Tubular Epithelial Cells after Renal Ishemia/Reperfusion
  • Advanced Oxidation Protein Products Accelerate Renal Fibrosis in a Remnant Kidney Model
  • Deficiency of P-Selectin or P-Selectin Glycoprotein Ligand-1 Leads to Accelerated Development of Glomerulonephritis and Increased Expression of CC Chemokine Ligand 2 in Lupus-Prone Mice
  • Leukocyte Recruitment and Vascular Injury in Diabetic Nephropathy
  • Proinflammatory Effects of Tweak/Fn14 Interactions in Glomerular Mesangial Cells
  • Chemokine Receptor Ccr2 Deficiency Reduces Renal Disease and Prolongs Survival in MRL/lpr Lupus-Prone Mice
  • Delayed Chemokine Receptor 1 Blockade Prolongs Survival in Collagen 4A3-Deficient Mice with Alport Disease
  • Upregulation of RANTES Gene Expression in Neuroglia by Japanese Encephalitis Virus Infection
  • Hepatocyte Growth Factor Ameliorates Renal Interstitial Inflammation in Rat Remnant Kidney by Modulating Tubular Expression of Macrophage Chemoattractant Protein-1 and RANTES
  • Is There an Epidemic of HIV Infection in the US ESRD Program?
  • Intrinsic Renal Cell Expression of CD40 Directs Th1 Effectors Inducing Experimental Crescentic Glomerulonephritis
  • Complement and the Kidney
  • CCR2 Signaling Contributes to Ischemia-Reperfusion Injury in Kidney
  • Recent Advances in the Pathophysiology of Ischemic Acute Renal Failure
  • Differential Activation of NF-{kappa}B, AP-1, and C/EBP in Endotoxin-Tolerant Rats: Mechanisms for In Vivo Regulation of Glomerular RANTES/CCL5 Expression
  • Anti-Monocyte Chemoattractant Protein-1 Gene Therapy Attenuates Renal Injury Induced by Protein-Overload Proteinuria
  • Gene Therapy Expressing Amino-Terminal Truncated Monocyte Chemoattractant Protein-1 Prevents Renal Ischemia-Reperfusion Injury
  • Bone Morphogenetic Protein-7 Inhibits Constitutive and Interleukin-1{beta}-Induced Monocyte Chemoattractant Protein-1 Expression in Human Mesangial Cells: Role for JNK/AP-1 Pathway
  • Roles of Toll-Like Receptors in C-C Chemokine Production by Renal Tubular Epithelial Cells
  • Expression of Functional CCR and CXCR Chemokine Receptors in Podocytes
  • MCP-1 Induces Inflammatory Activation of Human Tubular Epithelial Cells: Involvement of the Transcription Factors, Nuclear Factor-{kappa}B and Activating Protein-1
  • Roles of SLC/CCL21 and CCR7 in Human Kidney for Mesangial Proliferation, Migration, Apoptosis, and Tissue Homeostasis
  • Spatial and Temporally Restricted Expression of Chemokines and Chemokine Receptors in the Developing Human Kidney
  • Chemokines and Transplant Immunobiology
  • IP-10 and Mig Production by Glomerular Cells in Human Proliferative Glomerulonephritis and Regulation by Nitric Oxide
  • Molecular Determinants of Receptor Binding and Signaling by the CX3C Chemokine Fractalkine
  • Chemokine Expression Precedes Inflammatory Cell Infiltration and Chemokine Receptor and Cytokine Expression during the Initiation of Murine Lupus Nephritis
  • Obstructive Nephropathy in the Mouse: Progressive Fibrosis Correlates with Tubulointerstitial Chemokine Expression and Accumulation of CC Chemokine Receptor 2- and 5-Positive Leukocytes
  • Chemokine and Chemokine Receptor Expression during Initiation and Resolution of Immune Complex Glomerulonephritis
  • Combinatorial Model of Chemokine Involvement in Glomerular Monocyte Recruitment: Role of CXC Chemokine Receptor 2 in Infiltration During Nephrotoxic Nephritis
  • Expression of the Chemokine Monocyte Chemoattractant Protein-1 and Its Receptor Chemokine Receptor 2 in Human Crescentic Glomerulonephritis
  • Molecular Determinants of Receptor Binding and Signaling by the CX3C Chemokine Fractalkine
  • Google Scholar

Similar Articles

Related Articles

  • No related articles found.
  • PubMed
  • Google Scholar

Articles

  • Current Issue
  • Early Access
  • Subject Collections
  • Article Archive
  • ASN Annual Meeting Abstracts

Information for Authors

  • Submit a Manuscript
  • Author Resources
  • Editorial Fellowship Program
  • ASN Journal Policies
  • Reuse/Reprint Policy

About

  • JASN
  • ASN
  • ASN Journals
  • ASN Kidney News

Journal Information

  • About JASN
  • JASN Email Alerts
  • JASN Key Impact Information
  • JASN Podcasts
  • JASN RSS Feeds
  • Editorial Board

More Information

  • Advertise
  • ASN Podcasts
  • ASN Publications
  • Become an ASN Member
  • Feedback
  • Follow on Twitter
  • Password/Email Address Changes
  • Subscribe to ASN Journals

© 2021 American Society of Nephrology

Print ISSN - 1046-6673 Online ISSN - 1533-3450

Powered by HighWire