51
|
Obrig TG. Escherichia coli Shiga Toxin Mechanisms of Action in Renal Disease. Toxins (Basel) 2010; 2:2769-2794. [PMID: 21297888 PMCID: PMC3032420 DOI: 10.3390/toxins2122769] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2010] [Revised: 11/13/2010] [Accepted: 11/24/2010] [Indexed: 02/06/2023] Open
Abstract
Shiga toxin-producing Escherichia coli is a contaminant of food and water that in humans causes a diarrheal prodrome followed by more severe disease of the kidneys and an array of symptoms of the central nervous system. The systemic disease is a complex referred to as diarrhea-associated hemolytic uremic syndrome (D(+)HUS). D(+)HUS is characterized by thrombocytopenia, microangiopathic hemolytic anemia, and acute renal failure. This review focuses on the renal aspects of D(+)HUS. Current knowledge of this renal disease is derived from a combination of human samples, animal models of D(+)HUS, and interaction of Shiga toxin with isolated renal cell types. Shiga toxin is a multi-subunit protein complex that binds to a glycosphingolipid receptor, Gb3, on select eukaryotic cell types. Location of Gb3 in the kidney is predictive of the sites of action of Shiga toxin. However, the toxin is cytotoxic to some, but not all cell types that express Gb3. It also can cause apoptosis or generate an inflammatory response in some cells. Together, this myriad of results is responsible for D(+)HUS disease.
Collapse
Affiliation(s)
- Tom G Obrig
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, 685 W. Baltimore St., HSF I Suite 380, Baltimore, MD 21201, USA; ; Tel.: +1-410-706-6917
| |
Collapse
|
52
|
WEN QIONG, HUANG ZHAO, ZHOU SHUFENG, LI XIAOYAN, LUO NING, YU XUEQING. Urinary proteins from patients with nephrotic syndrome alters the signalling proteins regulating epithelial-mesenchymal transition. Nephrology (Carlton) 2010; 15:63-74. [DOI: 10.1111/j.1440-1797.2009.01152.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
53
|
Bazzi C, Rizza V, Raimondi S, Casellato D, Napodano P, D'Amico G. In crescentic IgA nephropathy, fractional excretion of IgG in combination with nephron loss is the best predictor of progression and responsiveness to immunosuppression. Clin J Am Soc Nephrol 2009; 4:929-35. [PMID: 19406958 DOI: 10.2215/cjn.05711108] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
BACKGROUND AND OBJECTIVES The aim of this study was to evaluate the relationship between proteinuric markers (urinary excretion of IgG, alpha2-macroglobulin, alpha1-microglobulin) and serum creatinine (sCr), histologic lesions, progression, and immunosuppression responsiveness in crescentic IgA nephropathy. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS Fractional excretion of IgG (FEIgG) and of alpha1-microglobulin and urinary excretion of alpha2-macroglobulin were evaluated in 37 patients, 23 treated with steroids and cyclophosphamide. For assessment of the effective tubular load of proteins in surviving nephrons, new markers that take into account not only the absolute excretion value but also nephron loss were obtained dividing proteinuric markers for percentage of nonobsolescent glomeruli (surviving glomeruli [SG]). For each parameter, low- and high-risk groups were defined according to cutoffs with the highest sensitivity and specificity for progression (ESRD/doubling sCr) assessed by receiver operating characteristic analysis; follow up was 60 +/- 40 mo. RESULTS FEIgG/SG is the most powerful progression predictor: 5 versus 83% in all patients; in treated patients, 0 versus 89%, increased to 0 versus 100% by sCr and FEIgG/SG in combination (low risk: both markers or only one below cutoff (n = 15); high risk: both markers above cutoff (n = 8). The nonprogressors showed at last observation 65% proteinuria reduction and 10% sCr reduction. CONCLUSIONS In crescentic IgA nephropathy, FEIgG/SG, which evaluates altered size selectivity in relation to nephron loss, is the best progression predictor. In treated patients, progression prediction was increased by FEIgG/SG and sCr in combination. Treatment may be restricted to low-risk patients.
Collapse
Affiliation(s)
- Claudio Bazzi
- Fondazione D'Amico per Ricerca sulle Malattie Renali, Via Cherubini, 6, 20100 Milan, Italy.
| | | | | | | | | | | |
Collapse
|
54
|
Protein load impairs factor H binding promoting complement-dependent dysfunction of proximal tubular cells. Kidney Int 2009; 75:1050-9. [PMID: 19242507 DOI: 10.1038/ki.2009.8] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Intrarenal complement activation plays an important role in the progression of chronic kidney disease. A key target of the activated complement cascade is the proximal tubule, a site where abnormally filtered plasma proteins and complement factors combine to promote injury. This study determined whether protein overloading of human proximal tubular cells (HK-2) in culture enhances complement activation by impairing complement regulation. Addition of albumin or transferrin to the cells incubated with diluted human serum as a source of complement caused increased apical C3 deposition. Soluble complement receptor-1 (an inhibitor of all 3 activation pathways) blocked complement deposition while the classical and lectin pathway inhibitor, magnesium chloride-EGTA, was, ineffective. Media containing albumin as well as complement had additive proinflammatory effects as shown by increased fractalkine and transforming growth factor-beta mRNA expression. This paralleled active C3 and C5b-9 generations, effects not shared by transferrin. Factor H, one of the main natural inhibitors of the alternative pathway, binds to heparan sulfate proteoglycans. Both the density of heparan sulfate and factor H binding were reduced with protein loading, thereby enhancing the albumin- and serum-dependent complement activation potential. Thus, protein overload reduces the ability of the tubule cell to bind factor H and counteract complement activation, effects instrumental to renal disease progression.
Collapse
|
55
|
Pippin JW, Brinkkoetter PT, Cormack-Aboud FC, Durvasula RV, Hauser PV, Kowalewska J, Krofft RD, Logar CM, Marshall CB, Ohse T, Shankland SJ. Inducible rodent models of acquired podocyte diseases. Am J Physiol Renal Physiol 2009; 296:F213-29. [DOI: 10.1152/ajprenal.90421.2008] [Citation(s) in RCA: 193] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Glomerular diseases remain the leading cause of chronic and end-stage kidney disease. Significant advances in our understanding of human glomerular diseases have been enabled by the development and better characterization of animal models. Diseases of the glomerular epithelial cells (podocytes) account for the majority of proteinuric diseases. Rodents have been extensively used experimentally to better define mechanisms of disease induction and progression, as well as to identify potential targets and therapies. The development of podocyte-specific genetically modified mice has energized the research field to better understand which animal models are appropriate to study acquired podocyte diseases. In this review we discuss inducible experimental models of acquired nondiabetic podocyte diseases in rodents, namely, passive Heymann nephritis, puromycin aminonucleoside nephrosis, adriamycin nephrosis, liopolysaccharide, crescentic glomerulonephritis, and protein overload nephropathy models. Details are given on the model backgrounds, how to induce each model, the interpretations of the data, and the benefits and shortcomings of each. Genetic rodent models of podocyte injury are excluded.
Collapse
|
56
|
Macconi D, Chiabrando C, Schiarea S, Aiello S, Cassis L, Gagliardini E, Noris M, Buelli S, Zoja C, Corna D, Mele C, Fanelli R, Remuzzi G, Benigni A. Proteasomal processing of albumin by renal dendritic cells generates antigenic peptides. J Am Soc Nephrol 2008; 20:123-30. [PMID: 19092126 DOI: 10.1681/asn.2007111233] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The role of dendritic cells (DC) that accumulate in the renal parenchyma of non-immune-mediated proteinuric nephropathies is not well understood. Under certain circumstances, DC capture immunologically ignored antigens, including self-antigens, and present them within MHC class I, initiating an autoimmune response. We studied whether DC could generate antigenic peptides from the self-protein albumin. Exposure of rat proximal tubular cells to autologous albumin resulted in its proteolytic cleavage to form an N-terminal 24-amino acid peptide (ALB1-24). This peptide was further processed by the DC proteasome into antigenic peptides that had binding motifs for MHC class I and were capable of activating syngeneic CD8+ T cells. In vivo, the rat five-sixths nephrectomy model allowed the localization and activation of renal DC. Accumulation of DC in the renal parenchyma peaked 1 wk after surgery and decreased at 4 wk, concomitant with their appearance in the renal draining lymph nodes. DC from renal lymph nodes, loaded with ALB1-24, activated syngeneic CD8+ T cells in primary culture. The response of CD8+ T cells of five-sixths nephrectomized rats was amplified with secondary stimulation. In contrast, DC from renal lymph nodes of five-sixths nephrectomized rats treated with the proteasomal inhibitor bortezomib lost their capacity to stimulate CD8+ T cells in primary and secondary cultures. These data suggest that albumin can be a source of potentially antigenic peptides upon renal injury and that renal DC play a role in processing self-proteins through a proteasome-dependent pathway.
Collapse
Affiliation(s)
- Daniela Macconi
- Mario Negri Institute for Pharmacological Research, Via Gavazzeni 11, 24125 Bergamo, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
57
|
Nacar A, Karabay G, Unlükal N, Yazici C, Ozdemir H. Correlation between proteinuria level and renal morphology with special reference to electron microscopy in kidney donors. Ren Fail 2008; 30:507-11. [PMID: 18569931 DOI: 10.1080/08860220802064713] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
The aims of this study were to evaluate whether there is a correlation between protein level in urine and renal morphology in kidney transplant donors, as well as to detect the role of electron microscopy. For this purpose, kidney biopsies of 10 donors with urine protein levels were evaluated. Seven patients were female and three were male. Two had physiologic proteinuria (< 150 mg/24h), four had non-significant proteinuria (150-300 mg/24h), and three had significant (> 300 mg/24h) proteinuria. Serum creatinine levels were in normal ranges in all patients except for one who had a slight increase (1.76 mg/dL). Seven cases were reported to have normal or nonspecific light microscopic findings. Two of those seven cases had physiologic proteinuria, three had non-significant proteinuria, and two had significant proteinuria. One case had IgA nephropathy with significant proteinuria. One donor had early stage focal segmental glomerulosclerosis with non-significant proteinuria, and one donor had focal interstitial fibrosis with normal urine protein level. There was no statistically significant difference between score means of ultrastructural morphology of the six patients with same patients' light microscopic results and score means of light microscopic results with urine protein levels of all patients. However, there was a significant difference between score means of ultrastructural morphology with urine protein levels of those six patients. In conclusion, urine protein levels and light microscopic findings did not always reflect the detailed morphology alone and together. Therefore, combining with electron microscopic examination could be more beneficial in relieving problems occurring in long-term prognoses.
Collapse
Affiliation(s)
- Ahmet Nacar
- Department of Histology and Embryology, Mustafa Kemal University Medical Faculty, Hatay, Turkey.
| | | | | | | | | |
Collapse
|
58
|
Abstract
Tubulointerstitial fibrosis is an integral part of the structural changes of the kidney in chronic progressive renal failure. The accumulation of the extracellular matrix in the tubulointerstitial space is mediated mainly by myofibroblasts. These are derived from resident interstitial fibroblasts, tubular epithelial cells, periadventitial cells, and possibly also mesenchymal stem cells and endothelial cells. Fibrosis is usually preceded by tubulointerstitial infiltration of mononuclear inflammatory cells. Proteinuria is one of several mechanisms of primary glomerular or vascular disease to transmit the disease process to the interstitial space. Increased protein filtration may have direct toxic effects on tubular epithelial cells, induce chemokine and cytokine secretion and result in increased expression of adhesion molecules, all contributing to the influx of mononuclear cells. Inflammatory cells in return secrete cytokines, which stimulate resident fibroblasts and tubular epithelial cells to differentiate into matrix-producing cells. The phenotypic conversion of primary epithelial cells into mesenchymal cells, termed epithelial-mesenchymal transition (EMT), has been studied in great detail in recent years. Several signal transduction pathways of this process have been clarified and may eventually result in novel therapeutic approaches. The severity of proteinuria and the extent of EMT have both been associated with the decline in renal function in clinical studies. Limiting proteinuria results in a slower decline of renal function deterioration, whereas reducing EMT has had beneficial effects in a number of animal studies, including those indicating reversal of fibrotic lesions. However, the association between proteinuria and EMT and vice versa is far from clear and has not been carefully studied.
Collapse
Affiliation(s)
- Frank M Strutz
- Department of Nephrology and Rheumatology, Georg-August-University Medical Center, Göttingen, Germany.
| |
Collapse
|
59
|
Zanchi C, Zoja C, Morigi M, Valsecchi F, Liu XY, Rottoli D, Locatelli M, Buelli S, Pezzotta A, Mapelli P, Geelen J, Remuzzi G, Hawiger J. Fractalkine and CX3CR1 mediate leukocyte capture by endothelium in response to Shiga toxin. THE JOURNAL OF IMMUNOLOGY 2008; 181:1460-9. [PMID: 18606701 DOI: 10.4049/jimmunol.181.2.1460] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Shiga toxins (Stx) are the virulence factors of enterohemorrhagic Escherichia coli O157:H7, a worldwide emerging diarrheal pathogen, which precipitates postdiarrheal hemolytic uremic syndrome, the leading cause of acute renal failure in children. In this study, we show that Stx2 triggered expression of fractalkine (FKN), a CX3C transmembrane chemokine, acting as both adhesion counterreceptor on endothelial cells and soluble chemoattractant. Stx2 caused in HUVEC expression of FKN mRNA and protein, which promoted leukocyte capture, ablated by Abs to either endothelial FKN or leukocyte CX3CR1 receptor. Exposure of human glomerular endothelial cells to Stx2 recapitulated its FKN-inducing activity and FKN-mediated leukocyte adhesion. Both processes required phosphorylation of Src-family protein tyrosine kinase and p38 MAPK in endothelial cells. Furthermore, they depended on nuclear import of NF-kappaB and other stress-responsive transcription factors. Inhibition of their nuclear import with the cell-penetrating SN50 peptide reduced FKN mRNA levels and FKN-mediated leukocyte capture by endothelial cells. Adenoviral overexpression of IkappaBalpha inhibited FKN mRNA up-regulation. The FKN-mediated responses to Stx2 were also dependent on AP-1. In mice, both virulence factors of Stx-producing E. coli, Stx and LPS, are required to elicit hemolytic uremic syndrome. In this study, FKN was detected within glomeruli of C57BL/6 mice injected with Stx2, and further increased after Stx2 plus LPS coadministration. This was associated with recruitment of CX3CR1-positive cells. Thus, in response to Stx2, FKN is induced playing an essential role in the promotion of leukocyte-endothelial cell interaction thereby potentially contributing to the renal microvascular dysfunction and thrombotic microangiopathy that underlie hemolytic uremic syndrome due to enterohemorrhagic E. coli O157:H7 infection.
Collapse
Affiliation(s)
- Cristina Zanchi
- Mario Negri Institute for Pharmacological Research, Bergamo, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
60
|
Wu SH, Lu C, Dong L, Chen ZQ. Signal transduction involved in CTGF-induced production of chemokines in mesangial cells. Growth Factors 2008; 26:192-200. [PMID: 19378419 DOI: 10.1080/08977190802227828] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
OBJECTIVE AND DESIGN This study investigates the regulatory role of connective tissue growth factor (CTGF) on production of fractalkine, monocyte-chemoattractant protein-1 (MCP-1) and regulated on activation, normal T cell expressed and secreted (RANTES) in human mesangial cells, and explore the mechanisms of CTGF action. METHODS Cultured human mesangial cells were treated with CTGF. Expressions of mRNA and proteins of fractalkine, MCP-1 and RANTES were analyzed by real-time polymerase chain reaction (PCR) and by enzyme-linked immunosorbent assay, respectively. Expressions of phosphorylated extracellular signal-regulated kinase 1/2 (ERK1/2), phosphoinositide 3-kinase (PI3-K) and protein kinase B (PKB) were assessed by Western blotting. Activities of nuclear factor-KB (NF-KB) were determined by NF-kappaB luciferase reporter assay. RESULTS CTGF enhanced the mRNA expressions and protein release of fractalkine, MCP-1 and RANTES, and the expressions of phosphorylated ERK1/2, PI3-K and PKB, and activities of NF-KB. Blockade of ERK1/2 inhibited the CTGF-induced expression ofphosphorylated ERK1/2 and NF-kappaB, and partially decreased the expressions of the above chemokines. PI3-K blockade downregulated the CTGF-stimulated expressions of phosphorylated PI3-K, PKB and NF-kappaB but not phosphorylated ERK1/2, partially decreased the expressions of the above chemokines. NF-kappaB blockade abrogated the CTGF-activated NF-kappaB and partially decreased the expressions of the above chemokines. Soluble heparin and K252a, an inhibitor of Trk, blocked CTGF-induced production of the above chemokines and the activation of the above signaling proteins. CONCLUSION These results demonstrated that CTGF induces production of fractalkine, MCP-1 and RANTES via ERK1/2 and PI3-K/PKB/NF-kappaB-dependent signal pathway mediated by cell surface heparin sulfate proteoglycans and the tyrosine kinase receptor TrkA in human mesangial cells.
Collapse
Affiliation(s)
- Sheng-Hua Wu
- Department of Pediatrics, Central Laboratory, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People's Republic of China.
| | | | | | | |
Collapse
|
61
|
Shin GT, Kim DR, Lim JE, Yim H, Kim H. Upregulation and function of GADD45gamma in unilateral ureteral obstruction. Kidney Int 2008; 73:1251-65. [PMID: 18354378 DOI: 10.1038/ki.2008.93] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
We performed differential display analysis to determine transcriptional activity in the rat kidney, following unilateral ureteral obstruction and found a 12-fold increase in the expression of Growth Arrest and DNA Damage-45gamma (GADD45gamma), a stress-responsive molecule that interacts with cell-cycle proteins. GADD45gamma was strongly expressed in as little as 6 h following ureteric obstruction in the renal tubules, and was also found in kidney tissue of patients with chronic glomerulonephritis. Adenovirus-mediated expression of GADD45gamma in cultured renal tubular cells activated p38 along with a significant upregulation of C-C and C-X3-C chemokine ligands and fibrosis-related factors such as several matrix metalloproteinases, transforming growth factor-beta1, decorin, and bone morphogenetic protein 2. Silencing of GADD45gamma expression significantly blunted the upregulation of these inflammatory and fibrogenic mediators and monocyte infiltration in the ureteral obstructed rat kidney. Our study shows that GADD45gamma is quickly upregulated in the kidney with an obstructed ureter, enhancing the production of factors regulating the pathogenesis of kidney disease.
Collapse
Affiliation(s)
- G-T Shin
- Department of Nephrology, Ajou University School of Medicine, Suwon, Republic of Korea.
| | | | | | | | | |
Collapse
|
62
|
Kruse JJCM, Bomhoff-Wijdenes IFM, Te Poele JAM, Stewart FA. Fractalkine: an important candidate for directing periglomerular leukocyte accumulation in irradiated mouse kidneys. Acta Oncol 2008; 46:945-50. [PMID: 17917825 DOI: 10.1080/02841860701218618] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Radiation-induced impairment of renal function is preceded by capillary endothelial cell damage, which initiates a cascade of inflammatory and thrombotic events. Accumulation of leukocytes in the irradiated kidney, especially in areas surrounding the glomeruli, has been clearly demonstrated. The chemokine fractalkine has recently been identified as a key mediator of leukocyte adhesion that functions without the requirement of integrins or selectin-mediated rolling. In this study we investigate the possible involvement of fractalkine in the inflammatory response of the irradiated kidney. Mouse kidneys were irradiated with single doses of 16 or 0 Gy, and protein and mRNA levels of fractalkine and PECAM-1 were examined after 10 to 40 weeks. These changes were correlated with the progressive increase and distribution of leukocytes in the irradiated kidneys. Increased fractalkine immunoreactivity was seen at glomerular sites 30 to 40 weeks after irradiation. This fractalkine expression was strongly associated with the presence of leukocytes surrounding the Bowman's capsule of the same glomeruli. No significant changes in mRNA levels of fractalkine were seen in whole kidney extracts after irradiation, but expression levels were not determined for isolated glomeruli. PECAM-1 protein levels did not change with time after irradiation, although a significant decrease in mRNA expression was seen at 10 weeks. This study is the first demonstration of increased fractalkine after irradiation and the results suggest that fractalkine may be an important mechanism of leukocyte trafficking in the development of a radiation induced inflammatory response.
Collapse
|
63
|
Ishida Y, Gao JL, Murphy PM. Chemokine receptor CX3CR1 mediates skin wound healing by promoting macrophage and fibroblast accumulation and function. THE JOURNAL OF IMMUNOLOGY 2008; 180:569-79. [PMID: 18097059 DOI: 10.4049/jimmunol.180.1.569] [Citation(s) in RCA: 237] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Wounds heal through a highly regulated, self-limited inflammatory response, however, precise inflammatory mediators have not been fully delineated. In this study, we report that in a mouse model of excisional skin wound healing the chemokine CX3CL1 and its receptor CX3CR1 were both highly induced at wound sites; CX3CL1 colocalized with macrophages and endothelial cells, whereas CX3CR1 colocalized mainly with macrophages and fibroblasts. Loss of CX3CR1 function delayed wound closure in both CX3CR1 knockout (KO) mice and in wild-type mice infused with anti-CX3CR1-neutralizing Ab. Conversely, transfer of bone marrow from donor wild-type mice, but not from donor CX3CR1 KO mice, restored wound healing to normal in CX3CR1 KO-recipient mice. Direct effects of CX3CR1 disruption at the wound site included marked reduction of macrophages and macrophage products, such as TGF-beta1 and vascular endothelial growth factor. Consistent with this, we observed reduced alpha-smooth muscle actin (a marker for myofibroblasts) and collagen deposition in skin from wounded CX3CR1 KO mice, as well as reduced neovascularization. Together, the data support a molecular model of skin wound repair in which CX3CR1 mediates direct recruitment of bone marrow-derived monocytes/macrophages which release profibrotic and angiogenic mediators.
Collapse
Affiliation(s)
- Yuko Ishida
- Molecular Signaling Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | |
Collapse
|
64
|
Kato N, Watanabe Y, Ohno Y, Inoue T, Kanno Y, Suzuki H, Okada H. Mapping quantitative trait loci for proteinuria-induced renal collagen deposition. Kidney Int 2008; 73:1017-23. [PMID: 18305463 DOI: 10.1038/ki.2008.7] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The progression of chronic kidney disease is a complex process influenced by genetic factors. Proteinuria is a predictor of functional deterioration and an accelerator of disease progression through renal parenchymal damage and interstitial fibrosis. To determine genetic components that might mediate renal fibrosis due to proteinuria, we mapped loci influencing the phenotype of two mouse strains differing in proteinuria-induced renal type I collagen (COLI) deposition. Collagen I deposition in 129S1/svImJ and C57BL/6J mice differs significantly among tested strains. We backcrossed 120 hemi-nephrectomized (129S1/svImJ x C57BL/6J) F1 x 129S1/svImJ backcrossed mice loaded with bovine serum albumin giving rise to proteinuria and renal COLI deposition. Quantitative trait loci (QTL) mapping was performed and our analysis identified one suggestive linkage for renal COLI deposition peaking at 87 cM near D2Mit224 (logarithm of odds: 2.41) on Chr 2. In silico analysis uncovered nine candidate genes. Hence, although more studies are needed, these QTL provide an initial cue to subsequent gene discovery, which might help unravel the genetics of renal fibrosis.
Collapse
Affiliation(s)
- N Kato
- Department of Nephrology, Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan
| | | | | | | | | | | | | |
Collapse
|
65
|
Koziolek MJ, Schmid H, Cohen CD, Blaschke S, Hemmerlein B, Zapf A, Müller GA, Strutz F. Potential role of fractalkine receptor expression in human renal fibrogenesis. Kidney Int 2007; 72:599-607. [PMID: 17579663 DOI: 10.1038/sj.ki.5002368] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The inhibition of several chemokine/chemokine receptors has been shown to reduce progressive renal interstitial fibrosis. In this study, we examined the expression of the CX(3)C receptor in human renal biopsies with interstitial fibrosis and from normal kidneys by real-time polymerase chain reaction (PCR) and immunohistochemistry. The CX(3)C receptor was not only detected in mononuclear, tubular epithelial, and dendritic cells but also in alpha-smooth muscle actin and vimentin-positive interstitial myofibroblasts in fibrotic kidneys. Real-time PCR indicated a significant upregulation of CX(3)C receptor mRNA in fibrotic kidneys compared with non-fibrotic nephropathies or donor biopsies. In renal fibroblasts in vitro, hydrogen peroxide increased the expression of the CX(3)C receptor, an increase that was inhibited by N-acetylcysteine and catalase. However, neither proinflammatory nor profibrotic cytokines resulted in this upregulation. Stimulation of fibroblasts by CX(3)C ligand led to a significant enhancement of migration, which was abrogated by pre-incubation with a blocking anti-CX(3)C receptor antibody. Our studies indicate that renal fibrosis is associated with the expression of CX(3)C receptors on human renal fibroblasts. The expression is induced by reactive oxygen species suggesting a role of oxidative stress.
Collapse
Affiliation(s)
- M J Koziolek
- Department of Nephrology and Rheumatology, Georg-August-University Goettingen, Robert-Koch-Street 40, D-37075 Goettingen, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
66
|
Gaedeke J, Neumayer HH, Peters H. Pharmacological management of renal fibrotic disease. Expert Opin Pharmacother 2007; 7:377-86. [PMID: 16503810 DOI: 10.1517/14656566.7.4.377] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Chronic kidney diseases frequently advance to end-stage renal failure, and the number of patients affected is steadily increasing worldwide. At the molecular level, progression of renal insufficiency correlates closely with ongoing pathological matrix protein expansion (i.e., renal fibrosis), in a manner independent of the underlying disorder. Overactivity of the renin-angiotensin system and of the TGF-beta system have been identified as key mediators of kidney matrix accumulation, and are principal targets in the management of chronic renal disease. This review provides a recent overview of the therapeutic options that are clinically established, and of novel molecular strategies that will approach clinical practice in the near future.
Collapse
Affiliation(s)
- Jens Gaedeke
- Department of Nephrology, Charité Universitätsmedizin Berlin, Campus Charité Mitte, Humboldt University, Schumannstrasse 20/21D-10098 Berlin, Germany.
| | | | | |
Collapse
|
67
|
Wang W, Soltero L, Zhang P, Huang XR, Lan HY, Adrogue HJ. Renal inflammation is modulated by potassium in chronic kidney disease: possible role of Smad7. Am J Physiol Renal Physiol 2007; 293:F1123-30. [PMID: 17634402 DOI: 10.1152/ajprenal.00104.2007] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
High-potassium diets have been shown to be beneficial in cardiovascular disease partly because of a blood pressure-lowering effect. The effect of potassium on inflammation has not been studied. We investigated the influence of potassium supplementation on the degree of renal inflammation and the intracellular signaling mechanisms that could mediate inflammation in chronic kidney disease (CKD). CKD was created in male Sprague-Dawley rats by subtotal nephrectomy. Two groups of CKD rats were pair fed with diets containing 2.1% potassium (potassium-supplemented diet) or 0.4% potassium (basal diet). Body weight, blood pressure, and blood and urine electrolytes were measured biweekly. The animals were euthanized at week 8, and the remnant kidneys were analyzed by histology, immunohistochemistry, Western blotting, and real-time quantitative PCR. In the CKD pair-fed groups, blood potassium concentration did not differ significantly, but blood pressure was lower in the potassium-supplemented group. Compared with the basal diet, potassium supplementation decreased renal tubulointerstitial injury and suppressed renal inflammation as evidenced by decreased macrophage infiltration, lower expression of inflammatory cytokines, and decreased NF-kappaB activation. These renoprotective effects were associated with downregulation of renal transforming growth facto-beta, upregulation of renal Smad7, and lower blood pressure. Our results show that potassium supplementation can reduce renal inflammation and hence, could modulate the progression of kidney injury in CKD.
Collapse
Affiliation(s)
- Wansheng Wang
- Dept. of Medicine-Renal Section, Baylor College of Medicine, Houston, TX 77030, USA.
| | | | | | | | | | | |
Collapse
|
68
|
Benigni A, Caroli C, Longaretti L, Gagliardini E, Zoja C, Galbusera M, Moioli D, Romagnani P, Tincani A, Andreoli L, Remuzzi G. Involvement of renal tubular Toll-like receptor 9 in the development of tubulointerstitial injury in systemic lupus. ACTA ACUST UNITED AC 2007; 56:1569-78. [PMID: 17469139 DOI: 10.1002/art.22524] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
OBJECTIVE Toll-like receptor 9 (TLR-9), a receptor for CpG DNA, has been implicated in the activation of immune cells in lupus. We undertook this study to determine whether the expression of TLR-9 in resident renal cells in lupus nephritis is related to the development of tubulointerstitial injury. METHODS TLR-9 was analyzed in selectively retrieved renal tissue from (NZB x NZW)F1 mice at different stages of disease by laser capture microdissection combined with real-time quantitative reverse transcriptase-polymerase chain reaction, and in renal biopsy specimens from lupus nephritis patients by immunohistochemistry. We investigated for the molecular component responsible for TLR-9 activation by cultured proximal tubular cells in serum from patients with lupus. RESULTS Renal tissue from NZB x NZW mice displayed robust TLR-9 expression localized to proximal tubular cells. TLR-9 levels correlated with proteinuria and tubulointerstitial injury to the extent that a cyclin-dependent kinase inhibitor, while reducing proteinuria and renal structural damage, prevented tubular TLR-9 generation in lupus mice. Consistently, exaggerated TLR-9 staining was found in proximal tubular cells of lupus patients, which correlated with tubulointerstitial damage. DNA-containing immune complexes purified from sera of patients with lupus induced TLR-9 in cultured proximal tubular cells. This was prevented by CCGG-rich short oligonucleotides, specific antagonists of CpG DNA, indicating that the DNA component of immune complexes was required for TLR-9 stimulation. CONCLUSION These findings suggest that tubular TLR-9 activation has a pathogenetic role in tubulointerstitial inflammation and damage in experimental and human lupus nephritis, and they indicate a novel target for future therapies.
Collapse
MESH Headings
- Animals
- Biopsy
- Cells, Cultured
- Female
- Gene Expression Regulation/drug effects
- Humans
- Kidney Tubules, Proximal/metabolism
- Kidney Tubules, Proximal/pathology
- Lupus Erythematosus, Systemic/complications
- Lupus Erythematosus, Systemic/genetics
- Lupus Erythematosus, Systemic/metabolism
- Mice
- Mice, Inbred NZB
- Mice, Inbred Strains
- Nephritis, Interstitial/etiology
- Nephritis, Interstitial/genetics
- Nephritis, Interstitial/metabolism
- Protein Kinase Inhibitors/pharmacology
- Purines/pharmacology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Roscovitine
- Toll-Like Receptor 9/genetics
- Toll-Like Receptor 9/metabolism
Collapse
Affiliation(s)
- Ariela Benigni
- Mario Negri Institute for Pharmacological Research, Bergamo, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
69
|
Kim YH, Chung IY, Choi MY, Kim YS, Lee JH, Park CH, Kang SS, Roh GS, Choi WS, Yoo JM, Cho GJ. Triamcinolone suppresses retinal vascular pathology via a potent interruption of proinflammatory signal-regulated activation of VEGF during a relative hypoxia. Neurobiol Dis 2007; 26:569-76. [PMID: 17434742 DOI: 10.1016/j.nbd.2007.02.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2006] [Revised: 01/31/2007] [Accepted: 02/04/2007] [Indexed: 10/23/2022] Open
Abstract
We examined the effect of triamcinolone acetonide (TA), a corticosteroid, on the relationship between vascular pathophysiology and vascular endothelial growth factor (VEGF) activation in the retina of a rat model of oxygen-induced retinopathy (OIR). OIR was induced by exposure of hyperoxia (80% oxygen) to Sprague-Dawley (SD) rats from P2 to P14 and then returned to normoxic conditions. TA was intravitreal-injected once into the right eye of OIR rats at P15. Effects of TA on vascular pathophysiology or changes of various genes in response to hypoxia and/or proinflammation under hypoxic retina were assessed by the Evans-blue method, fluorescein isothiocyanate-dextran (FITC-D) infusion, immunoblotting, and ELIZA. TA not only reduced retinal neovascularization and vascular leakage in the OIR-rat retina, but also blocked the induction of hypoxia-response proinflammatory genes before it negatively controlled VEGF activation. These findings suggest a potential that TA suppresses retinal neovascular pathophysiology via proinflammation-mediated activation of VEGF during hypoxia.
Collapse
Affiliation(s)
- Y H Kim
- Department of Anatomy and Neurobiology, College of Medicine, Gyeongsang National University, Chilam-dong 92, Jinju, Gyungnam 660-751, South Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
70
|
Gerstung M, Roth T, Dienes HP, Licht C, Fries JWU. Endothelin-1 induces NF-kappaB via two independent pathways in human renal tubular epithelial cells. Am J Nephrol 2007; 27:294-300. [PMID: 17460393 DOI: 10.1159/000101999] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2007] [Accepted: 03/19/2007] [Indexed: 11/19/2022]
Abstract
BACKGROUND Endothelin-1 (ET-1) is a major transcriptional activator of renal proximal tubule cells acting in an autocrine and paracrine manner. In animal studies, ET-1 has been implicated in progressive renal interstitial fibrosis by promoting gene expression, possibly via the inflammatory NF-kappaB signal pathway. While ET-1-dependent mechanisms of signal transduction have been studied mainly in tumor cell lines, we analyzed the mechanism of ET-1-induced, NF-kappaB-mediated target gene activation in proximal tubule cells. METHODS Human renal proximal tubule cells were stimulated with ET-1 and gene expression analyzed by protein microarray, Western blot, non-radioactive electromobility shift assay, and quantitative real-time polymerase chain reaction. RESULTS Activation of NF-kappaB occurs only via an ET-1-specific type A receptor (not type B as in animals). Induction can be blocked by bosentan, and endothelin-A but not endothelin-B receptor-specific antagonists. Protein microarray screening shows activation of two independent cascades (via the endothelin-A receptor, or via diacylglycerol) leading to NF-kappaB induction. The independent induction is also reflected by target gene expression such as the vascular cell adhesion molecule-1, interleukin-6, and fractalkine at different time points. CONCLUSION Thus prohibiting ET-1-mediated gene transcription necessitates blocking of NF-kappaB and diacylglycerol signal transduction in proximal tubule cells.
Collapse
Affiliation(s)
- Melanie Gerstung
- Department of Pathology, University of Cologne, Cologne, Germany
| | | | | | | | | |
Collapse
|
71
|
Morigi M, Buelli S, Zanchi C, Longaretti L, Macconi D, Benigni A, Moioli D, Remuzzi G, Zoja C. Shigatoxin-induced endothelin-1 expression in cultured podocytes autocrinally mediates actin remodeling. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 169:1965-75. [PMID: 17148661 PMCID: PMC1762486 DOI: 10.2353/ajpath.2006.051331] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Shigatoxin (Stx) is the offending agent of post-diarrheal hemolytic uremic syndrome, characterized by glomerular ischemic changes preceding microvascular thrombosis. Because podocytes are highly sensitive to Stx cytotoxicity and represent a source of vasoactive molecules, we studied whether Stx-2 modulated the production of endothelin-1 (ET-1), taken as candidate mediator of podocyte dysfunction. Stx-2 enhanced ET-1 mRNA and protein expression via activation of nuclear factor kappaB (NF-kappaB) and activator protein-1 (Ap-1) to the extent that transfection with the dominant-negative mutant of IkappaB-kinase 2 or with Ap-1 decoy oligodeoxynucleotides reduced ET-1 mRNA levels. We propose a role for p38 and p42/44 mitogen-activated protein kinases (MAPKs) in mediating NF-kappaB-dependent gene transcription induced by Stx-2, based on data that Stx-2 phosphorylated p38 and p42/44 MAPKs and that MAPK inhibitors reduced transcription of NF-kappaB promoter/luciferase reporter gene construct induced by Stx-2. Stx-2 caused F-actin redistribution and intercellular gaps via production of ET-1 acting on ETA receptor, because cytoskeleton changes were prevented by ETA receptor blockade. Exogenous ET-1 induced cytoskeleton rearrangement and intercellular gaps via phosphatidylinositol-3 kinase and Rho-kinase pathway and increased protein permeability across the podocyte monolayer. These data suggest that the podocyte is a target of Stx, a novel stimulus for the synthesis of ET-1, which may control cytoskeleton remodeling and glomerular permeability in an autocrine fashion.
Collapse
Affiliation(s)
- Marina Morigi
- Mario Negri Institute for Pharmacological Research, Unit of Nephrology and Dialysis, Azienda Ospedaliera, Ospedali Riuniti di Bergamo, Via Gavazzeni 11, 24125 Bergamo, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
72
|
Durkan AM, Alexander RT, Liu GY, Rui M, Femia G, Robinson LA. Expression and targeting of CX3CL1 (fractalkine) in renal tubular epithelial cells. J Am Soc Nephrol 2006; 18:74-83. [PMID: 17151328 DOI: 10.1681/asn.2006080862] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The chemokine CX3CL1 plays a key role in glomerulonephritis and can act as both chemoattractant and adhesion molecule. CX3CL1 also is upregulated in tubulointerstitial injury, but little is known about the subcellular distribution and function of CX3CL1 in renal tubular epithelial cells (RTEC). Unexpectedly, it was found that CX3CL1 is expressed predominantly on the apical surface of tubular epithelium in human renal transplant biopsy specimens with acute rejection or acute tubular necrosis. For studying the targeting of CX3CL1 in polarized RTEC, MDCK cells that expressed untagged or green fluorescent protein-tagged CX3CL1 were generated. The chemokine was present on the apical membrane and in subapical vesicles. Apical targeting of CX3CL1 was not due to signals that were conferred by its intracellular domain, to associations with lipid rafts, or to O-glycosylation but, rather, depended on N-linked glycosylation of the protein. With the use of fluorescence recovery after photobleaching, it was found that CX3CL1 is immobile in the apical membrane. However, CX3CL1 partitioned with the triton-soluble rather than -insoluble cellular fraction, indicating that it is not associated directly with the actin cytoskeleton or with lipid rafts. Accordingly, disruption of rafts through cholesterol depletion did not render CX3CL1 mobile. For exploration of potential functions of apical CX3CL1, binding of CX3CR1-expressing leukocytes to polarized RTEC was examined. Leukocyte adhesion to the luminal surface was enhanced significantly when CX3CL1 was present. These data demonstrate that CX3CL1 is expressed preferentially on the apical membrane of RTEC and suggest a novel function for the chemokine in recruitment and retention of leukocytes in tubulointerstitial inflammation.
Collapse
Affiliation(s)
- Anne M Durkan
- Division of Nephrology, Hospital for Sick Children, 555 University Avenue, Toronto, ON, Canada
| | | | | | | | | | | |
Collapse
|
73
|
Zoja C, Abbate M, Remuzzi G. Progression of chronic kidney disease: insights from animal models. Curr Opin Nephrol Hypertens 2006; 15:250-7. [PMID: 16609291 DOI: 10.1097/01.mnh.0000222691.53970.83] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
PURPOSE OF REVIEW Chronic kidney diseases are emerging as a worldwide public health problem. Clarification of the mechanisms underlying progression of proteinuric nephropathies received significant input from the generation of transgenic and knockout animals and from novel approaches to block mediators of injury. Reviewed here are advances in animal models used as a tool to address some relevant questions to the pathophysiology of human chronic nephropathies. RECENT FINDINGS Gene targeting in rodents identified podocyte loss as central event in the development of glomerulosclerosis. The trigger is dysfunction or absence of podocyte molecules that stabilize the slit diaphragm or anchor foot processes to the basement membrane. Sustained injury of the glomerular barrier to proteins is transmitted to the tubulointerstitial compartment leading to inflammation and fibrosis. Blocking NF-kappaB activity and chemokine signals in the kidney effectively interrupts such process. Growth factors produced by tubular cells and inflammatory cells contribute to interstitial fibrogenesis via myofibroblast activation. SUMMARY Development of genetically engineered animals and techniques to specifically manipulate cellular mediators has highlighted the determinants of glomerulosclerosis and tubulointerstitial injury. This knowledge will provide basis for novel interventions to protect the podocyte in chronic progressive glomerulopathies and to halt renal scarring and loss of function.
Collapse
Affiliation(s)
- Carla Zoja
- Mario Negri Institute for Pharmacological Research, Bergamo, Italy.
| | | | | |
Collapse
|
74
|
Abstract
The possibility that proteinuria may accelerate kidney disease progression to end-stage renal failure has received support from the results of increasing numbers of experimental and clinical studies. Evidence indicating that this process occurs through multiple pathways, including induction of tubular chemokine expression and complement activation that lead to inflammatory cell infiltration in the interstitium and sustained fibrogenesis, is reviewed. Macrophages are prominent in the interstitial inflammatory infiltrate. This cell type mediates progression of renal injury to the extent that macrophage numbers in renal biopsy predict renal survival in patients with chronic renal disease. Chemoattractants and adhesive molecules for inflammatory cells are upregulated by excess ultrafiltered protein load of proximal tubular cells via activation of NF-kappaB-dependent and NF-kappaB-independent pathways. This mechanism is a potential target for therapeutic approaches, as shown by beneficial effects of manipulations with inhibitory molecules of NF-kappaB activation or of chemokine receptors in experimental studies. Targeting complement synthesis or activation in proximal tubule might offer novel therapeutic opportunities. Finally, proximal tubular cell receptors for uptake of plasma proteins that are under investigation may provide activation signals on excess tubular protein handling.
Collapse
Affiliation(s)
- Mauro Abbate
- Mario Negri Institute for Pharmacological Research, Bergamo, Italy
| | | | | |
Collapse
|
75
|
Halimi J, Laouad I, Buchler M, Al-Najjar A, Chatelet V, Houssaini TS, Nivet H, Lebranchu Y. Early Proteinuria Is a Strong Indicator of Donor Renal Lesions, Ischemia-Reperfusion Injury and Immunological Aggression. Transplant Proc 2006; 38:2319-20. [PMID: 16980078 DOI: 10.1016/j.transproceed.2006.06.122] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND Early proteinuria is associated with reduced long-term graft survival. However, the determinants and mechanisms of proteinuria early after transplantation have not been identified. METHODS Parameters associated with proteinuria within the first 3 months following transplantation were retrospectively assessed among 484 renal transplant recipients. RESULTS Proteinuria was more abundant in patients with a history of two or more rejection episodes (0.42 +/- 0.68 vs 0.18 +/- 0.39 g/d; P = .02). Proteinuria was greater when donor age was 60 or more (OR: 4.43; P = .003), when recipient death was due to cardiovascular causes (OR: 1.98; P = .002), or when cold (OR: 1.77; P = .006) or warm (1.21; P = .09) ischemia times were prolonged. CONCLUSIONS Proteinuria early after transplantation was related to pretransplant renal lesions, ischemia-reperfusion, and immunologic injuries.
Collapse
Affiliation(s)
- J Halimi
- François Rabelais University, Department of Nephrology and Clinical Immunology, Hôpital Bretonneau, 2 Boulevard Tonnellé, 37044 Tours cedex 9, France.
| | | | | | | | | | | | | | | |
Collapse
|
76
|
Biemesderfer D. Regulated intramembrane proteolysis of megalin: Linking urinary protein and gene regulation in proximal tubule? Kidney Int 2006; 69:1717-21. [PMID: 16557231 DOI: 10.1038/sj.ki.5000298] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Regulated intramembrane proteolysis (RIP) represents an evolutionarily conserved process linking receptor function with transcriptional regulation. Best characterized by the Notch signaling pathway, RIP involves regulated ectodomain shedding followed by gamma-secretase-mediated release of the C-terminal, cytosolic domain. The C-terminus in turn translocates to the nucleus where it interacts with other proteins to regulate expression of specific genes. Recent studies in our laboratory have shown that megalin, a scavenger receptor in proximal tubule, is subjected to RIP in a manner very similar to that of Notch. We showed that megalin in subjected to protein kinase C-regulated, metalloprotease-mediated ectodomain shedding producing a membrane-associated C-terminal fragment (MCTF). The MCTF in turn forms the substrate for gamma-secretase. These data implicate megalin as a central element of a Notch-like signaling pathway linking protein reabsorption and gene regulation in proximal tubule. The likelihood that megalin processing plays an important role in the progression of proteinuric kidney disease is discussed.
Collapse
Affiliation(s)
- D Biemesderfer
- Section of Nephrology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, Connecticut 06520, USA.
| |
Collapse
|
77
|
Wu SH, Wu XH, Lu C, Dong L, Zhou GP, Chen ZQ. Lipoxin A4 inhibits connective tissue growth factor-induced production of chemokines in rat mesangial cells. Kidney Int 2006; 69:248-56. [PMID: 16408113 DOI: 10.1038/sj.ki.5000025] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Connective tissue growth factor (CTGF) is involved in mitogenesis, matrix production, and chemotaxis in mesenchymal cells. The effects of CTGF on the production of chemokines remain unclear. The present studies investigate the regulatory role of CTGF in the production of fractalkine, monocyte chemoattractant protein-1 (MCP-1), and RANTES (regulated upon activation, normal T cell expressed and secreted) in cultured mesangial cells of rats, and the modulatory effects of lipoxin A(4) (LXA(4)) on actions of CTGF. CTGF enhanced the mRNA expression and protein release of fractalkine, MCP-1, and RANTES, the expression of phospho (P)-p42/44 mitogen-activated protein kinase (MAPK), P-phosphoinositide 3-kinase (PI3-K), P-Akt, and activity of nuclear factor-kappaB (NF-kappaB) in mesangial cells. P-p42/44 MAPK blockade inhibited the CTGF-induced expression of P-p42/44 MAPK but not NF-kappaB, and partially decreased the levels of the above chemokines in supernatants. P-PI3-K blockade downregulated the CTGF-stimulated expression of P-PI3-K, P-Akt, and NF-kappaB but not P-p42/44 MAPK, and partially decreased the release of the above chemokines. NF-kappaB blockade abrogated the CTGF-activated NF-kappaB and partially decreased the secretion of the above chemokines. LXA(4) dose-dependently inhibited the CTGF-stimulated mRNA expression and protein release of the above chemokines, and the expression of P-p42/44MAPK, P-PI3-K, P-Akt, and NF-kappaB. In conclusion, these results demonstrate that CTGF induces production of fractalkine, MCP-1, and RANTES via the p42/44 MAPK-, PI3-K/Akt-, and NF-kappaB-dependent signal pathway, and LXA(4) downregulates the above effects of CTGF on rat mesangial cells.
Collapse
Affiliation(s)
- S-H Wu
- Department of Pediatrics, Central Laboratory, The First Affiliated Hospital of Nanjing Medical University, Jiangsu, People's Republic of China.
| | | | | | | | | | | |
Collapse
|
78
|
Abstract
Albumin is the most abundant plasmaprotein serving multiple functions as a carrier of metabolites, hormones, vitamins, and drugs, as an acid/base buffer, as antioxidant and by supporting the oncotic pressure and volume of the blood. The presence of albumin in urine is considered to be the result of the balance between glomerular filtration and tubular reabsorption. Albuminuria has been accepted as an independent risk factor and a marker for renal as well as cardiovascular disease, and during the past decade, evidence has suggested that albumin itself may cause progression of renal disease. Thus, the reduction of proteinuria and, in particular, albuminuria has become a target in itself to prevent deterioration of renal function. Studies have shown albumin and its ligands to induce expression of inflammatory and fibrogenic mediators, and it has been hypothesized that increased filtration of albumin causes excessive tubular reabsorption, resulting in inflammation and fibrosis, resulting in the loss of renal function. In addition, it is known that tubular dysfunction in itself may cause albuminuria owing to decreased reabsorption of filtered albumin, and, recently, it has been suggested that significant amounts of albumin fragments are excreted in the urine as a result of tubular degradation. Thus, although both tubular and glomerular dysfunction influences renal handling of albumin, it appears that tubular reabsorption plays a central role in mediating the effects of albumin on renal function. The present paper will review the mechanisms for tubular albumin uptake and the possible implications for the development of renal disease.
Collapse
Affiliation(s)
- H Birn
- Department of Cell Biology, Institute of Anatomy, University of Aarhus, Aarhus C, Denmark
| | | |
Collapse
|
79
|
Kikuchi Y, Imakiire T, Hyodo T, Kushiyama T, Higashi K, Hyodo N, Suzuki S, Miura S. Advanced glycation end-product induces fractalkine gene upregulation in normal rat glomeruli. Nephrol Dial Transplant 2005; 20:2690-6. [PMID: 16234287 DOI: 10.1093/ndt/gfi232] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND We previously reported that fractalkine was upregulated in streptozotocin-induced diabetic kidneys. Fractalkine in diabetic kidneys was detected on glomerular capillaries and the mesangium. This upregulation was suppressed by treatment with angiotensin-converting enzyme inhibitor (ACE-I) or aminoiguanidine. We examined what factors induce fractalkine upregulation in normal rat glomeruli. METHODS Glomeruli were collected from the kidneys of normal Sprague-Dawley rats by a microdissection method. Ten glomeruli were incubated in a solution with glucose, mannitol, angiotensin II, tumour necrosis factor (TNF)-alpha and advanced glycation end-product (AGE)-bovine serum albumin (BSA) for 1, 2 and 4 h. Fractalkine mRNA expression in glomeruli was examined by reverse transcription-polymerase chain reaction. RESULTS Fractalkine mRNA levels in the 30 mM glucose solution significantly increased (121%) compared with those in the control or 30 mM mannitol solution at 1 h. Fractalkine mRNA levels in the 15 mM glucose solution showed no significant differences at 1 or 2 h, but significantly increased (106%) after 4 h incubation. Fractalkine mRNA levels in 10(-6)-10(-8) M angiotensin II solution showed no significant differences. Fractalkine mRNA levels in the 5 or 10 ng/ml TNF-alpha solution significantly increased compared with those in the control in a time- and dose-dependent manner (by 94 to 253%). Fractalkine mRNA levels in the 50-200 microg/ml AGE-BSA solution also increased compared with those in BSA solution in a time- and dose-dependent manner (by 119 to 261%). By pre-incubation with MG132, a nuclear factor-kappaB inhibitor, fractalkine upregulation by AGE-BSA or 30 mM glucose was completely suppressed. CONCLUSIONS High glucose levels, AGE formation and cytokine activation in diabetes may induce fractalkine upregulation in the kidneys and lead to progression of diabetic nephropathy.
Collapse
Affiliation(s)
- Yuichi Kikuchi
- Second Department of Internal Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513 Japan.
| | | | | | | | | | | | | | | |
Collapse
|
80
|
Halimi JM, Laouad I, Buchler M, Al-Najjar A, Chatelet V, Houssaini TS, Nivet H, Lebranchu Y. Early low-grade proteinuria: causes, short-term evolution and long-term consequences in renal transplantation. Am J Transplant 2005; 5:2281-8. [PMID: 16095510 DOI: 10.1111/j.1600-6143.2005.01020.x] [Citation(s) in RCA: 109] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Proteinuria 1 year after transplantation is associated with poor renal outcome. It is unclear whether low-grade (<1 g/24 h) proteinuria earlier after transplantation and its short-term change affect long-term graft survival. The effects of proteinuria and its change on long-term graft survival were retrospectively assessed in 484 renal transplant recipients. One- and 3-month proteinuria correlated with donor age, donor cardiovascular death, prolonged cold and warm ischemia times and acute rejection. One- and 3-month proteinuria (per 0.1 g/24 h, hazard ratio (HR): 1.07 and 1.15, p<0.0001)-especially low-grade proteinuria (HR: 1.20 and 1.26, p<0.0001)-were powerful, independent predictors of graft loss. Its short-term reduction correlated with arterial pressure (AP) (the lower the 3-month diastolic and 12-month systolic AP, the lower the risk of increasing proteinuria during 1-3 months and 3-12 months periods, respectively: Odds ratio (OR) per 10 MmHg: 0.78, p=0.01 and 0.85, respectively, p=0.02), and was associated with decreased long-term graft loss (per 0.1 g/24 h: HR: 0.88 and 0.98, respectively, p<0.0001), independently of initial proteinuria. Early low-grade proteinuria due to pre-transplant renal lesions, ischemia-reperfusion and immunologic injuries is a potent predictor of graft loss. Short-term reduction in proteinuria is associated with improved long-term graft survival.
Collapse
Affiliation(s)
- Jean-Michel Halimi
- Department of Nephrology and Clinical Immunology, Francois Rabelais University, Tours, France.
| | | | | | | | | | | | | | | |
Collapse
|
81
|
Schmieder RE, Klingbeil AU, Fleischmann EH, Veelken R, Delles C. Additional antiproteinuric effect of ultrahigh dose candesartan: a double-blind, randomized, prospective study. J Am Soc Nephrol 2005; 16:3038-45. [PMID: 16120821 DOI: 10.1681/asn.2005020138] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Proteinuria indicates future renal and cardiovascular morbidity, and, conversely, its reduction is associated with improved outcome. In a randomized, double-blind trial with parallel group design, the antiproteinuric effect of candesartan at high doses was analyzed. Patients with normal or mildly impaired renal function, protein excretion rate of 1 to 10 g/d, and treatment with an angiotensin-converting enzyme inhibitor or angiotensin receptor blocker for 3 mo were eligible. After a 4-wk treatment with 16 mg/d candesartan, patients (n = 32) were allocated to double-blind therapy with either 32 or 64 mg/d candesartan for 12 wk (including 4 wk of uptitration), followed again by 4 wk of candesartan 16 mg/d. Proteinuria at study entry was similar in both groups (geometric mean [95% confidence interval (CI)]; 32 mg/d candesartan 2.14 g/d [95% CI, 1.45 to 3.17]; 64 mg/d candesartan 2.54 g/d [95% CI, 1.91 to 3.40]; NS). After the double-blind treatment phase, proteinuria was reduced to 1.42 g/d (0.85 to 2.37) in the 64-mg/d group (P = 0.017), without any change in the 32-mg/d group (2.02 g/d [95% CI, 1.26 to 3.26]). The change in proteinuria differed between the two groups in absolute (P = 0.025) and relative terms (-29 +/- 50 versus -0 +/- 26%; P = 0.012). After downtitration to 16 mg/d candesartan, proteinuria increased again to 2.38 g/d (1.57 to 3.62) in the 64-mg/d group (P = 0.001) but remained unchanged in the 32-mg/d group (2.04 g/d [95% CI, 1.17 to 3.57]; NS). No change in BP was noticed in response to the different doses of candesartan. These data indicate an additive antiproteinuric effect of ultrahigh dose of the angiotensin receptor blocker candesartan compared with standard dose.
Collapse
Affiliation(s)
- Roland E Schmieder
- Universitätsklinikum Erlangen, Medizinische Klinik IV, Krankenhausstrasse 12, Erlangen 91054, Germany.
| | | | | | | | | |
Collapse
|
82
|
Morigi M, Buelli S, Angioletti S, Zanchi C, Longaretti L, Zoja C, Galbusera M, Gastoldi S, Mundel P, Remuzzi G, Benigni A. In response to protein load podocytes reorganize cytoskeleton and modulate endothelin-1 gene: implication for permselective dysfunction of chronic nephropathies. THE AMERICAN JOURNAL OF PATHOLOGY 2005; 166:1309-20. [PMID: 15855633 PMCID: PMC1606387 DOI: 10.1016/s0002-9440(10)62350-4] [Citation(s) in RCA: 121] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Effacement of podocyte foot processes occurs in many proteinuric nephropathies and is accompanied by rearrangement of the actin cytoskeleton. Here, we studied whether protein overload affects intracellular pathways, leading to cytoskeletal architecture changes and ultimately to podocyte dysfunction. Mouse podocytes bound and endocytosed both albumin and IgG via receptor-specific mechanisms. Protein overload caused redistribution of F-actin fibers instrumental to up-regulation of the prepro-endothelin (ET)-1 gene and production of the corresponding peptide. Increased DNA-binding activity for nuclear factor (NF)-kappaB and Ap-1 nuclear proteins was measured in nuclear extracts of podocytes exposed to excess proteins. Both Y27632, which inhibits Rho kinase-dependent stress fiber formation, and jasplakinolide, an F-actin stabilizer, decreased NF-kappaB and Ap-1 activity and reduced ET-1 expression. This suggested a role for the cytoskeleton, through activated Rho, in the regulation of the ET-1 peptide. Focal adhesion kinase (FAK), an integrin-associated nonreceptor tyrosine kinase, was phosphorylated by albumin treatment via Rho kinase-triggered actin reorganization. FAK activation led to NF-kappaB- and Ap-1-dependent ET-1 expression. These data suggest that reorganization of the actin cytoskeletal network in response to protein load is implicated in modulation of the ET-1 gene via Rho kinase-dependent FAK activation of NF-kappaB and Ap-1 in differentiated podocytes. Increased ET-1 generation might alter glomerular permselectivity and amplify the noxious effect of protein overload on dysfunctional podocytes.
Collapse
Affiliation(s)
- Marina Morigi
- Mario Negri Institute for Pharmacological Research, Via Gavazzeni 11, 24125 Bergamo, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
83
|
Hirschberg R, Wang S. Proteinuria and growth factors in the development of tubulointerstitial injury and scarring in kidney disease. Curr Opin Nephrol Hypertens 2005; 14:43-52. [PMID: 15586015 DOI: 10.1097/00041552-200501000-00008] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW There are ongoing debates as to the role and mechanisms of proteinuria in tubulointerstitial fibrogenesis. Moreover, recent experimental findings have allowed for further insights into mediators and interactions between cells in the renal interstitium during fibrogenesis. RECENT FINDINGS Proteinuria or albuminuria are likely just markers for the glomerular ultrafiltration and tubular actions of ultrafiltered, biologically active growth factors which 'activate' tubular cells causing basolateral secretion of chemokines and cytokines. Chemokines attract and activate macrophages. Tubular cell-derived platelet-derived growth factor (PDGF) and macrophage-derived transforming growth factor-beta cause fibroblast proliferation. Several growth factors contribute to their transition into extracellular matrix-producing myofibroblasts. This cascade of events provides targets for some currently available and several novel therapies. SUMMARY Albuminuria or glomerular proteinuria appear to be markers but ultrafiltered, bioactive growth factors are culprits in proteinuria-associated interstitial fibrosis. Interactions of tubular cells with macrophages and fibroblasts in the interstitium via defined growth factor/cytokines provide opportunities for therapeutic interventions.
Collapse
Affiliation(s)
- Raimund Hirschberg
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA 90502, USA.
| | | |
Collapse
|
84
|
Amiloride and the regulation of NF-kappaB: an unsung crosstalk and missing link between fluid dynamics and oxidative stress-related inflammation--controversy or pseudo-controversy? Biochem Biophys Res Commun 2005; 327:373-81. [PMID: 15629126 DOI: 10.1016/j.bbrc.2004.11.166] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2004] [Indexed: 11/17/2022]
Abstract
Understanding the biophysics of fluid dynamics within the context of transcriptional regulation, mediated by nuclear factor (NF)-kappaB, is crucial to developing a consensus on the molecular basis of fluid mechanics and imbalance. Amiloride, an antikaliuretic-diuretic agent, has recently entered the realm of NF-kappaB as a key player in regulating the molecular association of fluid dynamics with inflammation and oxidative stress. With the identification of flanking regions encoding the amiloride-sensitive channels that are NF-kappaB-responsive, a new theme emerges which underlies the significance of this association. What is the role of NF-kappaB in regulating fluid mechanics-is it a physiologic or immunologic function? Conversely, amiloride is purported as a major regulator of this transcriptional pathway. It is the mainstream of this survey, therefore, to outline current advances on the biophysics and nature of the interaction existing between amiloride, amiloride-sensitive channels, and NF-kappaB, while searching for potential molecular mechanisms.
Collapse
|
85
|
Zandi-Nejad K, Eddy AA, Glassock RJ, Brenner BM. Why is proteinuria an ominous biomarker of progressive kidney disease? Kidney Int 2005:S76-89. [PMID: 15485426 DOI: 10.1111/j.1523-1755.2004.09220.x] [Citation(s) in RCA: 122] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Progressive tubule injury and interstitial fibrosis frequently accompany glomerulopathies associated with proteinuria. Clinical experience indicates that higher levels of proteinuria prior to, as well as after initiation of treatment predict more rapid decline in renal function and more pronounced tubulointerstitial injury. It has been proposed that filtration of potentially tubulotoxic plasma proteins is responsible for the observed correlations between proteinuria and progression (i.e., proteinuria is a cause and not only a consequence of progressive renal injury). Numerous attempts have been made to identify the species of putative tubulotoxic proteins in this progressive injury process, but much uncertainty persists. These uncertainties stem from nonphysiologic exposure of apical cell surfaces to proteins in vitro, the extremely high concentrations of various proteins tested in vitro, and the nonuniformity of end points measured. Furthermore, there is often a lack of correlation between in vitro and in vivo findings, and a lack of uniformity of results even for seemingly similar in vitro experiments. Less controversy is evident in the potential pathways whereby injured tubules evoke a tubulointerstitial inflammatory and fibrotic response, with many in vivo models serving to incriminate excessive cytokine and chemokine production, infiltration of various inflammatory cells, and the balance between apoptosis and cell proliferation. Despite many years of concerted efforts, we believe it is still unclear whether proteinuria is a cause (and if so, which species of protein), or only a consequence of progressive renal injury. Nevertheless, pending the resolution of these uncertainties by more decisive and unambiguous experimentation, the strongly predictive inverse relationship between level of proteinuria and long-term renal survival currently justifies aggressive antiproteinuric treatment strategies, with a goal of reducing protein excretion rate to the lowest level possible without the induction of symptoms or undue risk.
Collapse
Affiliation(s)
- Kambiz Zandi-Nejad
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | |
Collapse
|
86
|
Lin SL, Chiang WC, Chen YM, Lai CF, Tsai TJ, Hsieh BS. The renoprotective potential of pentoxifylline in chronic kidney disease. J Chin Med Assoc 2005; 68:99-105. [PMID: 15813241 DOI: 10.1016/s1726-4901(09)70228-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Current interventions with proven efficacy, such as glycemic and blood pressure control, dietary protein restriction, and angiotensin II blockade, slow the progression of chronic kidney disease (CKD); however, whether long-term cessation of CKD progression is possible remains unclear. Because of the pathogenetic complexity of this condition, multidrug interventions with the least adverse effects should be investigated as the next step in attempts to stop CKD progression. Pentoxifylline, a non-selective phosphodiesterase inhibitor with indiscernible toxicity, exerts potent inhibitory effects against cell proliferation, inflammation, and extracellular matrix accumulation, all of which play important roles in CKD progression. Pentoxifylline monotherapy markedly reduces proteinuria in patients with membranous nephropathy. Moreover, limited human studies have proven pentoxifylline efficacy in reducing proteinuria in patients with diabetes receiving angiotensin-converting enzyme inhibitors, and in patients with nephrotic syndrome secondary to lupus nephritis despite immunosuppressive therapy. Further clinical trials are necessary to examine whether pentoxifylline can improve renal outcomes in patients receiving interventions of proven efficacy.
Collapse
Affiliation(s)
- Shuei-Liong Lin
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan, R.O.C
| | | | | | | | | | | |
Collapse
|
87
|
Erkan E, Garcia CD, Patterson LT, Mishra J, Mitsnefes MM, Kaskel FJ, Devarajan P. Induction of renal tubular cell apoptosis in focal segmental glomerulosclerosis: roles of proteinuria and Fas-dependent pathways. J Am Soc Nephrol 2004; 16:398-407. [PMID: 15601749 DOI: 10.1681/asn.2003100861] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The hypothesis that apoptosis represents a proximate mechanism by which tubule cells are damaged in FSGS was tested. Thirty kidney biopsy specimens from children with idiopathic early FSGS were studied retrospectively. Unexpected, apoptosis was evident in both proximal and distal tubule cells. There was a significant correlation between the degree of proteinuria and the number of apoptotic cells. Fas protein was detected predominantly in the tubule cells that underwent apoptosis. When compared with patients with other chronic proteinuric states, those with FSGS displayed a proliferation/apoptosis ratio in favor of proliferation in the glomerulus but dramatically in favor of apoptosis in the tubules. When both proteinuria and apoptosis were included in a stepwise logistic regression procedure, only apoptosis was found to predict independently the development of ESRD. Prolonged incubation of cultured Madin-Darby canine kidney (distal/collecting) cells with albumin also resulted in a dose- and duration-dependent induction of apoptosis and activation of the Fas pathway, lending support to the novel finding of distal tubule cell apoptosis in patients with FSGS. The results indicate that an elevated tubule cell apoptosis rate at the time of initial biopsy represents an independent predictor of progression to ESRD in patients with early FSGS.
Collapse
Affiliation(s)
- Elif Erkan
- Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA
| | | | | | | | | | | | | |
Collapse
|
88
|
Abstract
This paper reviews current concepts regarding the pathophysiology, diagnostic evaluation, and treatment of microalbuminuria and proteinuria in adults. Microalbuminuria (in diabetics) and proteinuria are early markers for potentially serious renal disease, and are associated with increased risk of atherosclerotic cardiovascular disease. Proteinuria also contributes to renal scarring, and accelerates the progression of chronic kidney disease to end-stage renal failure. Screening of diabetics for microalbuminuria, and the initial workup of proteinuria, should occur in the primary care setting. Reduction of microalbuminuria in diabetics may retard its progression to overt diabetic nephropathy. Therapy of renal diseases should aim for optimal blood pressure control and the maximum possible reduction in urinary protein excretion. Angiotensin-converting enzyme inhibitor (ACE-I) and/or angiotensin-receptor blocker (ARB) therapy is the most effective measure to achieve this. These drugs also provide protection against the cardiovascular problems that are highly prevalent in this patient population.
Collapse
Affiliation(s)
- K K Venkat
- Division of Nephrology, Department of Medicine, Henry Ford Hospital, 2799 West Grand Blvd., Detroit, MI 48202, USA.
| |
Collapse
|
89
|
Imai E, Nakajima H, Kaimori JY. Albumin turns on a vicious spiral of oxidative stress in renal proximal tubules. Kidney Int 2004; 66:2085-7. [PMID: 15496183 DOI: 10.1111/j.1523-1755.2004.00044.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
90
|
Sukkar MB, Issa R, Xie S, Oltmanns U, Newton R, Chung KF. Fractalkine/CX3CL1 production by human airway smooth muscle cells: induction by IFN-gamma and TNF-alpha and regulation by TGF-beta and corticosteroids. Am J Physiol Lung Cell Mol Physiol 2004; 287:L1230-40. [PMID: 15321787 DOI: 10.1152/ajplung.00014.2004] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Chemokine synthesis by airway smooth muscle cells (ASMC) may be an important process underlying inflammatory cell recruitment in airway inflammatory diseases such as asthma and chronic obstructive pulmonary disease (COPD). Fractalkine (FKN) is a recently described CX(3)C chemokine that has dual functions, serving as both a cell adhesion molecule and a chemoattractant for monocytes and T cells, expressing its unique receptor, CX(3)CR1. We investigated FKN expression by human ASMC in response to the proinflammatory cytokines IL-1beta, TNF-alpha, and IFN-gamma, the T helper 2-type cytokines IL-4, IL-10, and IL-13, and the fibrogenic cytokine transforming growth factor (TGF)-beta. Neither of these cytokines alone had any significant effect on ASMC FKN production. Combined stimulation with IFN-gamma and TNF-alpha induced FKN mRNA and protein expression in a time- and concentration-dependent manner. TGF-beta had a significant inhibitory effect on cytokine-induced FKN mRNA and protein expression. Dexamethasone (10(-8)-10(-6) M) significantly upregulated cytokine-induced FKN mRNA and protein expression. Finally, we used selective inhibitors of the mitogen-activated protein kinases c-Jun NH(2)-terminal kinase (JNK) (SP-610025), p38 (SB-203580), and extracellular signal-regulated kinase (PD-98095) to investigate their role in FKN production. SP-610025 (25 microM) and SB-203580 (20 microM), but not PD-98095, significantly attenuated cytokine-induced FKN protein synthesis. IFN-gamma- and TNF-alpha-induced JNK phosphorylation remained unaltered in the presence of TGF-beta but was inhibited by dexamethasone, indicating that JNK is not involved in TGF-beta- or dexamethasone-mediated regulation of FKN production. In summary, FKN production by human ASMC in vitro is regulated by inflammatory and anti-inflammatory factors.
Collapse
Affiliation(s)
- Maria B Sukkar
- Department of Thoracic Medicine, National Heart and Lung Institute, Imperial College, London, Dovehouse St., SW3 6LY, London, UK
| | | | | | | | | | | |
Collapse
|
91
|
Lin SL, Chen YM, Chiang WC, Tsai TJ, Chen WY. Review Article. Pentoxifylline: A potential therapy for chronic kidney disease. Nephrology (Carlton) 2004; 9:198-204. [PMID: 15363050 DOI: 10.1111/j.1440-1797.2004.00267.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Almost all forms of chronic kidney disease progressing to end-stage kidney failure are characterized by diffuse fibrosis, a final common pathway converging from multiple pathogenetic networks regardless of the initial injury. Four principal interventions including glycaemic and blood pressure control, dietary protein restriction, and angiotensin II blockade have been proven to slow progression of diabetic and/or non-diabetic chronic kidney disease. However, the ultimate solution to halt disease progression in the long term is still pending. Because of the pathogenetic complexity of kidney disease, multidrug intervention with the least side-effects should, without doubt, be the next step to stop kidney disease progression. Animal and cellular studies have demonstrated the rationale for pentoxifylline (i.e. its effects against cell proliferation, inflammation, and extracellular matrix accumulation) in the treatment of chronic kidney disease induced by immune- or non-immune-mediated mechanisms. Limited human studies have proven its efficacy in reducing proteinuria in patients with diabetes receiving angiotensin-converting enzyme inhibitors, and in patients with nephrotic syndrome refractory to conventional immunosuppressive therapy. Moreover, monotherapy with pentoxifylline markedly reduces proteinuria in patients with membranous nephropathy. Further studies are required to examine whether pentoxifylline can improve the renal outcome in patients receiving interventions with proven efficacy.
Collapse
Affiliation(s)
- Shuei-Liong Lin
- Department of Internal Medicine, National Taiwan University Hospital and Department of Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | | | | | | | | |
Collapse
|
92
|
Zoja C, Benigni A, Remuzzi G. Cellular responses to protein overload: key event in renal disease progression. Curr Opin Nephrol Hypertens 2004; 13:31-7. [PMID: 15090857 DOI: 10.1097/00041552-200401000-00005] [Citation(s) in RCA: 115] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Insight into the mechanisms underlying the progression of chronic proteinuric nephropathies has attracted the interest of the renal community in the last two decades. Systemic hypertension, proteinuria, cytokines and growth factors, and reactive oxygen species have all been implicated. Reviewed here are the determinants of tubulointerstitial injury; the focus is on protein ultrafiltration and reabsorption, which ultimately contribute, by activating fibrogenic mechanisms in tubular cells, to renal scarring. RECENT FINDINGS Protein overloading of proximal tubular cells--a well-documented consequence of exuberant protein ultrafiltration--differentially regulates transcription of NF-kappaB-dependent and NF-kappa-B-independent genes. This forms endothelin-1, cytokines and chemokines; all of these, being secreted toward the basolateral compartment of tubular epithelial cells, foster local recruitment of mononuclear cells. Autocrine pathways of activation of tubular epithelial cells contribute to interstitial injury and fibrosis. Albumin endocytosis in proximal tubular cells triggers events that include protein kinase C-dependent generation of reactive oxygen species, nuclear translocation of NF-kappaB, and activation of mitogen-activated protein kinase. In-vivo evidence that proteinuria activates transcription factors, including NF-kappaB, and overexpression of chemokine and fibrogenic cytokines is also available. SUMMARY Proteinuria incites a multitude of inflammatory and fibrogenic mediators, all of which contribute to renal scarring. Specific antagonism of multiple injurious pathways might help to arrest, or even reverse, the progression of renal damage.
Collapse
Affiliation(s)
- Carla Zoja
- Mario Negri Institute for Pharmacological Research, Via Gavazzeni 11, 24125 Bergamo, Italy.
| | | | | |
Collapse
|