Journal Information
Vol. 28. Issue. 6.December 2008
Pages 572-666
Vol. 28. Issue. 6.December 2008
Pages 572-666
DOI:
Full text access
The facilitator in acute kidney injury: TWEAK
TWEAK, el facilitador del daño renal agudo
Visits
7766
Ana Belén Sanza, María Dolores Sánchez-Ninoa, M. Concepción Izquierdoa, Juan Antonio Morenoa, Alvaro C. Uceroa, Alberto Benito-Martína, Beatríz Santamariaa, Carolina Burgosa, Jesús Egidob, Alberto Ortízb, Marta Ruíz-Ortegaa, Luis Miguel Blanco-Colioa, Adrián Ramos, Sergio Berzal, Eliécer Cotod
a Fundacion Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, Madrid, España,
b Fundacion Jiménez Díaz, Universidad Autónoma de Madrid, Fundación Renal Íñigo Álvarez de Toledo, Madrid, Madrid, España,
d Fundación Renal Íñigo Álvarez de Toledo, Genética Molecular-Hospital Central de Asturias, Madrid, Oviedo, Madrid, Asturias, España,
This item has received
Article information
Abstract
Full Text
Bibliography
Download PDF
Statistics
RESUMEN TWEAK es una citoquina de la superfamilia de las TNF que activa el receptor Fn14. TWEAK regula la proliferación celular, la muerte celular, la angiogénesis y la inflamación. La expresión de TWEAK y de su receptor Fn14 aumenta en células tubulares en caso de lesión renal. Las citoquinas proinflamatorias incrementan la expresión del receptor Fn14 en las células tubulares y TWEAK induce apoptosis en un medio inflamatorio. Además TWEAK induce la expresión de quimiocinas y citoquinas proinflamatorias. Estos resultados en células cultivadas se han reproducido en modelos experimentales de insuficiencia renal aguda. El antagonismo de TWEAK protegió la función renal y redujo la inflamación en esos modelos. En conjunto estos datos sugieren que TWEAK promueve la inflamación y la lesión tisular inflamatoria y que podría ser una nueva diana terapéutica en el daño renal.
Palabras clave:
apoptosis
Palabras clave:
Fn14
Palabras clave:
TWEAK
Palabras clave:
fracaso renal agudo
Palabras clave:
inflamación
Palabras clave:
células tubulares
ABSTRACT TWEAK is a cytokine of the TNF superfamiliy that activates the Fn14 receptor. TWEAK may regulate cell proliferation, cell death, cell differentiation, angiogenesis and inflammation. The expression of TWEAK and Fn14 is increased in tubular cells during acute kidney injury. Inflammatory cytokines increase Fn14 receptor expression in tubular cells and, in a proinflammatory milieu, TWEAK induces tubular cell apoptosis. In addition, TWEAK itself contributes to renal inflammation by promoting the secretion of chemokines and inflammatory cytokines by tubular cells. Confirmation of its role in acute kidney injury came from functional studies in the experimental model of folic acid overdose. TWEAK antagonism preserved renal function and reduced inflammation in this model. The available evidence suggests that TWEAK might be a target for therapeutic intervention in kidney injury and its role in different forms of renal injury should be further explored.
Keywords:
Keywords:
Keywords:
Keywords:
Keywords:
Keywords:
Full Text

THE PLOT: ACUTE KIDNEY INJURY



Most of the mass of the kidneys is composed of tubular cells. Loss of renal tubular cells characterizes both acute and chronic failure of renal function1. Tubular cells may be lost by shedding, death or differentiation into fibroblasts.

In human studies tubular cell death was the best histopathological correlate of renal dysfunction during acute kidney

injury (AKI).2,3 In addition sublethally injured tubular cells secrete mediators of inflammation that further aggravate AKI. However, the factors that modulate tubular cell injury and the interplay between different factors is incompletely understood. In fact, AKI is a paradigmatic situation in which an insufficient understanding of the mechanisms of injury precludes the implementation of effective physiopathology-based approaches for prevention and treatment. Numerous factors contributing to injury have been identified and many of them have been shown to be effective targets in animal models of injury.4 However the few attempts to translate this information to the clinical situation have failed. We now review the evidence involving a novel cytokine-ligand pair, TWEAK and Fn14, in AKI.5-7 We feel that they may become important targets in the treatment of AKI because they are «team players», they behave as «facilitators» that amplify injurious responses initiated by other stimuli. Thus their targeting has the potential to dampen injury induced by a wide range of stimuli.



THE STARS: TWEAK AND Fn14



Tumor necrosis factor-like weak inducer of apoptosis (TWEAK, Apo3L, TNFSF12) is a member of the tumor necrosis factor superfamily (TNFSF) of structurally-related cytokines8 (fig. 1). The human TWEAK gene is located at

chromosome 17 and encodes a 249- amino acid (aa) type II transmembrane glycoprotein (30 kDa). The intracellular domain contains a putative serine phosphorylation site. The extracellular domain contains the receptor-binding site. TWEAK may be expressed as a membrane-bound protein (mTWEAK) and as a 156-aa, 18kDa soluble protein, (sTWEAK) that results from proteolysis of mTWEAK.9,10 mTWEAK, but not sTWEAK, may enter the cell nucleus, although the relevance of this observation is unknown.11



The TWEAK receptor was initially and erroneously reported to be the tumor necrosis factor receptor superfamily

(TNFRSF) member death receptor 3 (DR3).12 In 2001 the TWEAK receptor was cloned and shown to be the previously described human fibroblast growth factorinducible 14 (Fn14, TNFRSF12A)13-15 (fig. 1). The human Fn14 gene is located at chromosome 16 and encodes a 129-aa type I transmembrane protein (14 kDa). Fn14 is processed into a 102- aa mature protein, making it the smallest TNFRSF member to date.9,16 The Fn14 extracellular domain (53-aa) contains the TWEAK-binding site.17 The intracellular domain (29-aa) contains a

TNFR-associated factor (TRAF)-binding site with three potentially phosphorylated threonines. TRAF binding has been implicated in signal transduction.18 TWEAK trimerizes and binds to Fn14 monomers, promoting receptor trimerization and signal transduction.18-21



Although Fn14 was the only characterized TWEAK receptor, some TWEAK actions may be mediated independently

of Fn14.22 Indeed CD163 was recently shown to either act as a TWEAK scavenger in pathological conditions or serve as an alternate receptor for TWEAK in cells lacking Fn14/TweakR.23



Human and murine TWEAK are closely related, with a 93% homology in the receptor-binding domain. Furthermore,

human and murine Fn14 have a 90% homology in their overall sequences, human TWEAK can bind to murine

Fn14 and vice-versa.24 In fact this pathway is remarkably conserved throughout evolution and putative TWEAK and

Fn14 ESTs can be found in species as primitive as zebrafish.25 The phylogenetical conservation suggests an important biological role.



TWEAK is widely expressed, and can be found at high levels in pancreas, intestine, heart, brain, lung, ovary and

skeletal muscle, and at lower levels in liver and kidney.8,26 TWEAK mRNA levels were initially reported to be

reduced in mice with both acute (induced by lipopolysaccharide) or chronic (autoimmune pathologies like lupus

erythematosus or hemolytic anemia) inflammatory processes in numerous tissues and peritoneal macrophages.27 In the same line of evidence, normal human arteries release sTWEAK but sTWEAK release from atherosclerotic arteries is reduced.28 Low serum sTWEAK is a marker for subclinical atherosclerosis of potential clinical relevance.28 However, elevated TWEAK expression was found in other inflammatory conditions such as experimental autoimmune encephalitis. 29



By contrast to TWEAK, resting tissue levels of Fn14 are usually low. As an example, it is undetectable in normal

arteries.28 Fn14 initially was identified as an early response gene whose transcription was induced by FGF-1 (fibroblast growth factor-1) in fibroblasts.14 Numerous cell culture and in vivo studies have demonstrated rapid induction and increased expression of Fn14 upon activation of diverse cell types by multiple stimuli, as well as in vivo in the course of liver, artery, central nervous system or kidney injury or in tumor cells.19,16,13,30,31 It has been postulated that a reduced Fn14 expression limits the actions of TWEAK in healthy tissues and rapid and quantitatively important upregulation of this receptor sensitizes to TWEAK actions28,32 (fig. 2). This sensitization would promote TWEAK activity in the presence of only mildly increased or even reduced TWEAK expression.



In experimental models of AKI and autoimmune injury renal TWEAK and, more significantly, Fn14 expression is

upregulated.31,33 The potential sources of TWEAK in the kidney include infiltrating monocytes and T lymphocytes.34,35 In particular T cells from lupus patients express TWEAK.34 Other renal sources of TWEAK include tubular epithelial cells.31 In addition, mesangial cells also express TWEAK and Fn14.36 Defining the role of a pleiotropic cytokine such as TWEAK in AKI is a challenge that has been approached by functional studies in cell cultures and experimental models.32



THE ROLES: HEROES AND VILLAINS



Evidence suggests that TWEAK and Fn14 play a role in the pathogenesis of human diseases, including atherosclerosis, stroke, rheumatoid arthritis, autoimmune kidney injury, AKI and cancer.5-7,37,38,25 TWEAK stimulates, in a cell-type- and microenvironment-dependent fashion, cell proliferation,39-43 survival, 44 migration,41,45,46 cell growth9 and apoptosis.8,47-51 TWEAK can also promote22 or inhibit52-54 cell  differentiation. Finally, TWEAK induces the expression of pro-inflammatory molecules. 8,27,36,41,45,55-58 In vivo, TWEAK can stimulate blood vessel formation (angiogenesis) 40,59 and regulates neurovascular unit permeability.60



A CROWDED PLACE: TWEAK AND INFLAMMATION



TWEAK promotes sustained NFκB activation in murine tubular epithelial cells. In tubular cells TWEAK increased

NFκB DNA-binding and transcriptional activity, IκB-α phosphorylation and RelA nuclear translocation via Fn14.32 As a consequence, TWEAK induced the mRNA expression and secretion of MCP-1, IL-6 and RANTES, which was prevented by the RelA inhibitor parthenolide. The effect of TWEAK on chemokines (previously known as  intercrines), 61 such as MCP-1 and RANTES is particularly interesting as it allows the recruitment of further

inflammatory cells and amplification of inflammation.



TNFα, used as a control for NFκB activation, yielded a different pattern of NFκB DNA-binding complexes.32 This

suggests that TWEAK induces other biological responses also dependent on NFκB that differ from those elicited by

TNFα. Thus, TNFα and TWEAK would not be redundant cytokines in renal injury. In this regard, the persistent NFκB activation induced by TWEAK in tubular cells is consistent with the recently identified non-canonical NFκB activation, which is not activated by TNFα.62



Systemic administration of TWEAK activates kidney NFκB and has a proinflammatory effect in the renal tubulointerstitium characterized by increased MCP-1, RANTES and IL-6 mRNA.32 Immunohistochemistry showed that RelA translocated to the nucleus of tubular cells and that these cells were the main site of expression of MCP-1

and RANTES. The expression of chemokines by tubular cells was followed by increased renal expression of the

mRNA for the MCP-1 receptor, CCR2, which is expressed by macrophages, as well as by an increased number of

interstitial macrophages. This demonstrates that TWEAK induces tubulointerstitial inflammation in vivo. However, TWEAK alone did not modify serum creatinine.



TWEAK had recently been found to increase MCP-1 expression in cultured glomerular mesangial cells and to upregulate whole kidney MCP-1 mRNA.36 However, that report did not address whether there was a concomitant increase of chemokine protein, which were the cells responsible for mRNA expression and whether TWEAK elicited

renal inflammation. In fact, the context of the report (cell culture studies in mesangial cells) suggested that the site

of action of TWEAK would be the glomerulus. Our findings demonstrate that in normal kidneys TWEAK elicits

tubulointerstitial, but not glomerular inflammation.32 TWEAK contributes to glomerular injury when the Fn14 receptor has been previously upregulated locally by injurious stimuli.33



THE KILLING FIELDS



Apoptosis contributes to renal cell loss.63 In addition to nephrotoxins and other forms of cell stress,64,65 several

members of the TNF ligand superfamily induce apoptotic renal cell death, including TRAIL, FasL and TNF-α.66-68

The name TWEAK derives from its weak capacity to induce apoptosis. TWEAK frequently requires co-stimuli, such as IFNγ, to induce apoptosis. 35 However, TWEAK alone may induce apoptosis in primary neurons and mesangial cells and this effect is magnified in the presence of IFNγ.36,48 TWEAK also mediates activated CD4+T cell-induced apoptosis of monocytes and macrophages (T cells presumably release concomitantly other mediators).69 By contrast, non-stimulated tubular epithelial cells are resistant to TWEAKinduced apoptosis and the lethal effect only becomes apparent in the presence of several (not one) inflammatory mediators released during AKI, such as IFNγ and TNF-α.31,70,71 This requirement for both IFNγ and TNFα in order to sensitize to cell death in the presence of

TWEAK is novel. TNFα/IFNγ increased Fn14 expression in tubular cells. Upregulation of Fn14 expression may underlie the sensitization to apoptosis. However, the level of Fn14 expression is not the only mechanism involved,

since IFNγ or TNFα alone also increase Fn14 expression but do not sensitize to cell death. Functional inhibition studies suggested that autocrine activation of the FasL/Fas system mildly contributed to cell death.31,72



The signaling pathways leading from Fn14 to cell death remains poorly understood and there is evidence that it may differ with cell type and the cell microenvironment. The lack of a death domain (DD) suggests that direct

recruitment of DD-containing adaptor proteins is not the primary mode of inducing cell death. Evidence from caspase inhibitor studies also points to several pathways for cell death. In tubular cells, the combination of TWEAK, TNFα and IFNγ resulted in activation of caspase-8, proteolysis of Bid, release of cytochrome c to the cytosol and activation of caspase-9 and caspase-3, suggesting activation of a canonical cell death receptor pathway with recruitment of the mitochondrial pathway. No evidence supporting the participation of endoplasmic reticulum stress was found. The apoptotic endoplasmic reticulum response is preserved in tubular cells exposed to paracetamol or tunicamycin.64 Most extracellular inputs are not processed in isolation, rather, multiple inputs are perceived by cells in a proinflammatory milieu.73 Thus, it is difficult to assign the final output to a single stimulus. It is conceivable that similar apoptotic pathways engaged by both TNFα and TWEAK in the presence of IFNγ cooperate in cell death induction. Indeed, TNFα may induce a delayed apoptosis in tubular cells, but the time-course differs from that induced by TWEAK/TNFα/IFNγ, as there is a lag period of 48 h, which was confirmed in cells treated with TNFa/IFNγ. 31 In addition, the intracellular molecular mechanisms differ between death induced by TNF alone and by the cytokine combination. The pancaspase inhibitor zVAD prevented the activation of caspases, and also prevented

apoptosis induced by TWEAK/TNFα/IFNγ.31 However, zVAD transformed the mode of cell death to necrosis and even increased the rate of cell death. Although the induction of necrosis in cells exposed to members of the TNF superfamily when caspases are inhibited had been previously observed,74 zVAD did not promote TNFα-induced

necrosis in tubular epithelium.31 This response is cell type-specific and suggests that pan-caspase inhibition may not be an adequate therapeutic approach to renal injury in which inflammation is contributing the cell death. In tubular cells Fn14 and reactive oxygen species were required for the necrotic response.



THE OUTCOME: PROTECTION



As outlined above cell culture data suggest that TWEAK may be deleterious in inflamed kidney. However, potential

proliferative actions of TWEAK on tubular cells or progenitor cells or the recruitment of macrophages that participate

in tissue repair might confer a protective role for TWEAK.25 In this regard, an initial interpretation of the low serum levels of TWEAK in atherosclerosis was that they may be contributing to injury. Functional inhibition of TWEAK in vivo was required to understand the outcome of these different potential actions of TWEAK and the end-result of its interactions with different components of the microenvironment. AKI induced by a folic acid overdose is an animal model which has a human counterpart in the rare cases of accidental parental overdose of folic acid.75 In addition, it shares with human AKI processes such as tubular cell death, inflammation, and tubular cell proliferation leading to spontaneous recovery and mild fibrosis.70,76 In this model, expression of TWEAK and Fn14 increased both at the mRNA and protein level.31 There was a 13-fold increase in Fn14 mRNA and a 2.5-fold increase in tubular cell Fn14 receptor. Fn14 was localized to injured, dilated proximal and distal tubules.31 While Fn14 was already very high at 24 h, TWEAK expression progressively increased, peaking at 72 h. In addition, renal expression of IFNγ and TNFα is

increased in AKI.70,32 A neutralizing anti-TWEAK antibody decreased peak serum creatinine and did not interfere

with recovery which was achieved by day 7 in control and treated mice.32 Neutralization of TWEAK also resulted in significantly milder histological injury. TWEAK did not participate in the early (24 h) inflammatory response, which

was TWEAK-independent. However, it amplified subsequent inflammation and when TWEAK was neutralized the

tubular expression of certain inflammatory molecules (MCP-1, RANTES) and interstitial inflammation by macrophages was decreased at 72 h. The in vivo effect of TWEAK on inflammatory mediators appears to be  selective. As an example, kidney IL-6 in the course of injury was TWEAK-independent. Thus, although TWEAK may modulate IL-6 expression in cultured tubular cells, other stimuli are more important regulators of IL-6 expression in AKI.



EPILOGUE



The Oxford English dictionary defines TWEAK as «improving by making fine adjustments». This meaning closely

represents the role of TWEAK in kidney injury. TWEAK regulates a wide range of cellular processes that are

involved both in the generation and the recovery from renal injury. Interestingly the precise effect of TWEAK over kidney cells is modulated by the cell microenvironment, allowing it to fine-tune the renal response to injury (fig. 3). TWEAK interacts with the cell microenvironment to elicit cell-specific responses. In the context of kidney inflammation during AKI the overall effect of TWEAK is negative for the kidney, promoting inflammation and cell death. In this context, TWEAK antagonism improves renal function. TWEAK regulates a wide range of cellular processes that are involved both in the generation of and the recovery from renal injury. Given the pleiotropic

actions of TWEAK, that include modulating the proliferation of progenitor cells,77 agonism of TWEAK should also be explored in certain clinical contexts. In addition, the possible role of TWEAK as a biomarker should be studied as urinary TWEAK levels were found to be elevated in patients with active lupus nephritis78 and low serum TWEAK levels are a marker for subclinical atherosclerosis.28



ACKNOWLEDGMENTS



This work was supported by grants from FIS 06/0046 and ISCIII-RETICS REDINREN RD 06/0016, MEC (SAF 03/884), Sociedad Espanola de Nefrologia. JAM, AR and AS were supported by FIS, MDSN and ACU by MEC, ACU, MCI, SB and BS by Fundacion Conchita Rabago and AO by the Programa de Intensificación de la Actividad

Investigadora in the Sistema Nacional de Salud of the Instituto de Salud Carlos III and the Agencia «Pedro Lain Entralgo» of the Comunidad de Madrid and FRACAM S-BIO 0283/2006.

Bibliography
[1]
REFERENCES
[2]
A. Ortiz, P. Justo, A. Sanz, C. Lorz, J. Egido: Targeting apoptosis in acute tubular necrosis. Biochem Pharmacol 2003; 66: 1589-94. [Pubmed]
[3]
K. Solez, L. Morel-Maroger, JD. Sraer: The morphology of "acute tubular necrosis" in man: analysis of 57 renal biopsies and a comparison with the glycerol model. Medicine (Baltimore) 1979; 58: 362-76.
[4]
TS. Olsen, HS. Olsen, HE Hansen: Tubular ultrastructure in acute renal failure in man: epithelial necrosis and regeneration. Virchows Arch A Pathol Anat Histopathol 1985; 406: 75-89. [Pubmed]
[5]
Lorz C, Benito-Martin A, Justo P, Sanz AB, Sanchez Niño MD, Egido J, Ortiz A: Modulation of renal tubular cell survival: where is the evidence? Current Medicinal Chemistry 2006; 13: 763-771.
[6]
Winkles JA: The TWEAK-Fn14 cytokine-receptor axis: discovery, biology and therapeutic targeting. Nat Rev Drug Discov 2008 Apr 11; [Epub ahead of print]
[7]
Burkly LC, Michaelson JS, Hahm K, Jakubowski A, Zheng TS: TWEAKing tissue remodeling by a multifunctional cytokine: role of TWEAK/Fn14 pathway in health and disease. Cytokine 2007; 40:1-16. PMID: 17981048
[8]
Sanz AB, Moreno JA, Sanchez-Nino MD, Ucero AC, Benito A, Santamaria B, Justo P, Izquierdo MC, Egido J, Blanco-Colio LM, Ortiz A: TWEAKing renal injury. Front Biosci 2008; 13 580-589. [Pubmed]
[9]
Y. Chicheportiche, P. R. Bourdon, H. Xu, Y. Hsu, H. Scott, C. Hession, I. Garci and J. L. Browning: TWEAK, a new secreted ligand in the tumor necrosis factor family that weakly induces apoptosis. J Biol Chem 1997; 272: 32401-32410. [Pubmed]
[10]
S. R. Wiley and J. A. Winkles: TWEAK, a member of the TNF superfamily, is a multifunctional cytokine that binds the TWEAKR/Fn14 receptor. Cytokine Growth Factor Rev 2003; 14:241-249. [Pubmed]
[11]
S. Campbell, J. Michaelson, L. Burkly and C. Putterman: The role of TWEAK/Fn14 in the pathogenesis of inflammation and systemic autoimmunity. Front Biosci 2004; 9: 2273-2284. [Pubmed]
[12]
F. O. Baxter, P. J. Came, K. Abell, B. Kedjouar, M. Huth, K. Rajewsky, M. Pasparakis and C. J. Watson: IKK beta/2 induces TWEAK and apoptosis in mammary epithelial cells. Development 2006; 133: 3385-3394.
[13]
S.A. Marsters, J.P. Sheridan, R.M. Pitti, J. Brush, A. Goddard, A. Ashekenazi: Identification of a ligand for the death-domain containing receptor Apo3. Curr Biol 1998; 8: 525-8. [Pubmed]
[14]
S.R. Wiley, L. Cassiano, T. Lofton, T. Davis-Smith, J.A. Winkles, V. Lindner, H. Liu, T.O. Daniel, C.A. Smith, W.C. Fanslow: A novel TNF receptor family member binds TWEAK and is implicated in angiogenesis. Immunity 2001; 15: 837-846. [Pubmed]
[15]
RL. Meighan-Mantha, D.K. Hsu, Y. Guo, S.A. Brown, S.L. Feng, K.A. Peifley, G.F. Alberts, N.G. Copeland, D.J. Gilbert, N.A. Jenkins, C.M. Richards, J.A. Winkles: The mitogen-inducible Fn14 gene encodes a type I transmembrane protein that modulates fibroblast adhesion and migration. J Biol Chem 1999; 274: 33166-33176. [Pubmed]
[16]
S.R. Wiley, J.A. Winkles: TWEAK, a member of the TNF superfamily, is a multifunctional cytokine that binds the TWEAKR/Fn14 receptor. Cytokine Growth Factor Rev 2003; 14: 241-249. [Pubmed]
[17]
S.L. Feng, Y. Guo, V.M. Factor, S.S. Thorgeirsson, D.W. Bell, J.R. Testa, K.A. Peifley, J.A. Winkles: The Fn14 immediate-early response gene is induced during liver regeneration and highly expressed in both human and murine hepatocellular carcinomas. Am J Pathol 2000; 156: 1253-1261. [Pubmed]
[18]
S. A. N. Brown, H. N. Hanscom, H. Vu, S. A. Brew and J. A. Winkles: TWEAK binding to the Fn14 cysteine-rich domain depends on charged residues located in both the A1 and D2 modules. Biochem J 2006; 397: 297-304. [Pubmed]
[19]
S.A. Brown, C.M. Richards, H.N. Hanscom, S.L. Feng and J.A. Winkles: The Fn14 cytoplasmatic tail binds tumour-necrosis-factor-receptor-associated factors 1, 2, 3 and 5 and mediates nuclear factor-kappa B activation. Biochem J 2003; 371: 395-403. [Pubmed]
[20]
N. L, Tran, W. S. McDonough, B. A. Savitch, T. F. Sawyer, J. A. Winkles and M. E. Berens: The tumor necrosis factor-like weak inducer of apoptosis (TWEAK)-fibroblast growth factor-inducible 14 (Fn14) signaling system regulates glioma cell survival via NF-kappaB pathway activation and BCLXL/ BCL-W expression. J Biol Chem 2005; 280: 3483-3492. [Pubmed]
[21]
J. A. Winkles, N. L. Tran and M. E. Berens: TWEAK and Fn14: New molecular targets for cancer therapy? Cancer Lett 2005; 235: 11-17. [Pubmed]
[22]
T. Saitoh, M. Nakayama, H. Nakano, H. Yagita, N. Yamamoto and S. Yamaoka: TWEAK induces NF-kappaB2 p100 processing and long lasting NF-kappaB activation. J Biol Chem 2003; 278: 36005-36012. [Pubmed]
[23]
T.C. Polek, M. Talpaz, B.G. Darnay, T. Spivak-Kroizman: TWEAK mediates signal transduction and differentiation of RAW264.7 cells in the absence of Fn14/TWEAKR. Evidence for a second TWEAK receptor. J Biol Chem 2003; 278: 32317-32323. [Pubmed]
[24]
Bover LC, Cardó-Vila M, Kuniyasu A, Sun J, Rangel R, Takeya M, Aggarwal BB, Arap W, Pasqualini R: A previously unrecognized protein-protein interaction between TWEAK and CD163: potential biological implications. J Immunol 2007; 178: 8183-94. PMID: 17548657
[25]
C. Bossen, K. Ingold, A. Tardivel, J.L Bodmer, O. Gaide, S. Hertig, C. Ambrose, J. Tschopp, P. Schneider: Interactions of tumor necrosis factor (TNF) and TNF receptor family members in the mouse and human. J Biol Chem 2006; 281: 13946-13971.
[26]
Zheng TS, Burkly LC. No End In Site: TWEAK/Fn14 Activation and Autoimmunity Associated End-Organ Pathologies. J Leukocyte Biology 2008 (in press)
[27]
S.A. Marsters, J.P. Sheridan, R.M. Pitti, J. Brush, A. Goddard, A. Ashkenazi: Identification of a ligand for the death-domain-containing receptor Apo3. Curr Biol 1998; 8: 525-528. [Pubmed]
[28]
Y. Chicheportiche, R. Chicheportiche, I. Sizing, J. Thompson, CB. Benjamin, C. Ambrose, JM. Daver: Proinflammatory activity of TWEAK on human dermal fibroblasts and synoviocytes: blocking and enhancing effects of anti-TWEAK monoclonal antibodies. Arthritis Res 2002; 4: 126-33. [Pubmed]
[29]
L. M. Blanco-Colio, J. L. Martín-Ventura, B. Muñóz-García, J. Orbe, J. A. Páramo, J. B. Michel, A. Ortiz, O. Meilhac and J. Egido: Identification of Soluble Tumor Necrosis Factor-Like Weak Inducer of Apoptosis (sTWEAK) as a Possible Biomarker of Subclinical Atherosclerosis. Arterioscler Thromb Vasc Biol 2007; 27: 916-922. [Pubmed]
[30]
S. Desplat- Jego, S. Varriale, R. Creidy, R. Terra, D. Bernard, M. Khrestchatisky, S. Izui, Y. Chicheportiche and J. Boucrat: TWEAK is expressed by glial cells, induces astrocyte proliferation and increases EAE severity. J Neuroimmunol 2002; 133: 116-123. [Pubmed]
[31]
K. Tanabe, I. Bonilla, J. A. Winkles and Sm. Strittmatter: Fibroblast growth factor-inducible-14 is induced in axotomized neurons and promotes neurite outgrowth. J Neurosci 2003; 23: 9675-9686. [Pubmed]
[32]
P. Justo, A. B. Sanz, M. D. Sanchez-Niño, J. A. Winkles, C. Lorz, J. Egido and A. Ortiz: Cytokine cooperation in renal tubular cell injury: the role of TWEAK. Kidney Int 2006; 70: 1750-1758 [Pubmed]
[33]
A. B. Sanz, P. Justo, M. D. Sanchez-Niño, L. M. Blanco-Colio, J. A. Winkles, M. Kreztler, A. Jakubowski, J. Egido, M. Ruiz-Ortega and A. Ortiz: The Cytokine TWEAK Modulates Renal Tubulointerstitial Inflammation. J Am Soc Nephrol 2008; 19: 695-703. [Pubmed]
[34]
Z. Zhao, L. C. Burkly, S. Campbell, N. Schwartz, A. Molano, A. Choudhury, R. A. Eisenberg, J. S. Michaelson and C. Putterman: TWEAK/Fn14 interactions are instrumental in the pathogenesis of nephritis in the chronic graft-versus-host model of systemic lupus erythematosus. J Immunol 2007; 179: 7949-7958. [Pubmed]
[35]
M. J. Kaplan, E. E. Lewis, E. A. Shelden, E. Somers, R. Pavlic, W. J. McCune and B. C. Richardson: The apoptotic ligands TRAIL, TWEAK, and Fas ligand mediate monocyte death induced by autologous lupus T cells. J Immunol 2002; 169: 6020-6029. [Pubmed]
[36]
M. Nakayama, N. Kayagaki, N. Yamaguchi, K. Okumura and H. Yagita: Involvement of TWEAK in interferon gamma-stimulated monocyte cytotoxicity. J Exp Med 2000; 192: 1373-1380. [Pubmed]
[37]
S. Campbell, L. C. Burkly, H. X. Gao, J. W. Berman, L. Su, B. Browning, T. Zheng, L. Schiffer, J. S. Michaelson and C. Putterman: Proinflammatory effects of TWEAK/Fn14 interactions in glomerular mesangial cells. J Immunol 2006; 176: 1889-1898. [Pubmed]
[38]
J. A. Winkles, N. L. Tran, S. A. Brown, N. Stains, H. E. Cunliffe and M. E. Berens: Role of TWEAK and Fn14 in tumor biology. Front Biosci 2007; 12: 2761-2771. [Pubmed]
[39]
M. Blanco-Colio, J. L. Martín-Ventura, B. Muñoz-Garcia, J. A. Moreno, O. Meilhac, A. Ortiz and J. Egido: TWEAK and Fn14. New players in the pathogenesis of atherosclerosis. Front Biosci 2007; 12: 3648-3655. [Pubmed]
[40]
P. J. Donohue, C. M. Richards, S. A. N. Brown, H. N. Hanscom, J. Buschman, S. Thangada, T. Hla, M. S. Williams and J. A. Winkles: TWEAK is an endothelial cell growth and chemotactic factor that also potentiates FGF-2 and VEGF-A mitogenic activity. Arterioscler Thromb Vasc Biol 2003; 23: 594-600. [Pubmed]
[41]
C. N. Lynch, Y. C. Wang, J. K. Lund, Y. Chen, J. A. Leal and S. R. Wiley: TWEAK induces angiogenesis and proliferation of endothelial cells. J Biol Chem 1999; 274: 8455-8459. [Pubmed]
[42]
N. Harada, M. Nakayama, H. Nakano, Y. Fukuchi, H. Yagita and K. Okumura: Pro-inflammatory effect of TWEAK/Fn14 interaction on human umbilical vein endothelial cells. Biochem Biophys Res Commun 2002; 299: 488-493. [Pubmed]
[43]
J. S. Michaelson, S. Cho, B. Browning, T. S. Zheng, J. M. Lincecum, M. Z. Wang, Y. M. Hsu and L. C. Burkly: TWEAK induces mammary epithelial branching morphogenesis. Oncogene 2005; 24: 2613-2624. [Pubmed]
[44]
A. Jakubowski, C. Ambrose, M. Parr, J. M. Lincecum, M. Z. Wang, T. S. Zheng, B. Browning, J. S. Michaelson, M. Baetscher, B. Wang, D. M. Bissell and L. C. Burkly: TWEAK induces liver progenitor cell proliferation. J Clin Invest 2005; 115: 2330-2340. [Pubmed]
[45]
A. Jakubowski, B. Browning, M. Lukashev, I. Sizing, J. S. Thompson, C. D. Benjamin, Y. Hsu, C. Ambrose, T. S. Zheng and L. C. Burkly: Dual role for TWEAK in angiogenic regulation. J Cell Sci 2002; 115: 267-274. [Pubmed]
[46]
L. Jin, A. Nakao, M. Nakayama, N. Yamaguchi, Y. Kojima, N. Nakano, R. Tsuboi, K. Okumura, H. Yagita and H. Ogawa: Induction of RANTES by TWEAK/Fn14 interaction in human keratinocytes. J Invest Dermatol 2004; 122: 1175-1179. [Pubmed]
[47]
N. L. Tran, W. S. McDonough, P. J. Donohue, J. A. Winkles, T. J. Berens, K. R. Ross, D. B. Hoelzinger, C. Beaudry, S. W. Coons and M. E. Berens: The human Fn14 receptor gene is up-regulated in migrating glioma cells in vitro and overexpressed in advanced glial tumors. Am J Pathol 2003; 162: 1313-1321. [Pubmed]
[48]
M. Nakayama, K. Ishidoh, N. Kayagaki, Y. Kojima, N. Yamaguchi, H. Nakano, E. Kominami, K. Okumura and H. Yagita: Multiple pathways of TWEAK-induced cell death. J Immunol 2002; 168: 734-743. [Pubmed]
[49]
I. Potrovita, W. Zhang, L. Burkly, K. Hahm, J. Lincecum, M. Z. Wang, M. H. Maurer, M. Rossner, A. Schneider and M. Schwaninger: Tumor necrosis factor- like weak inducer of apoptosis-induced neurodegeneration. J Neurosci 2004; 24: 8237-8244. [Pubmed]
[50]
P. Schneider, R. Schwenzer, E. Haas, F. Muhlenbeck, G. Schubert, P. Scheurich, J. Tschopp and H. Wajant: TWEAK can induce cell death via endogenous TNF and TNF receptor 1. Eur J Immunol 1999; 29: 1785-1792. [Pubmed]
[51]
C. A. Wilson and J. L. Browning: Death of HT29 adenocarcinoma cells induced by TNF family receptor activation is caspase-independent and displays features of both apoptosis and necrosis. Cell Death Differ 2002; 9: 1321-1333. [Pubmed]
[52]
M. Nakayama, K. Ishidoh, Y. Kojima, N. Harada, E. Kominami, K. Okumura and H. Yagita: Fibroblast growth factor-inducible 14 mediates multiple pathways of TWEAK-induced cell death. J Immunol 2003; 170: 341-348. [Pubmed]
[53]
N. Felli, F. Pedini, A. Zeuner, E. Petrucci, U. Testa, C. Conticello, M. Biffoni, A. Di Cataldo, J. A. Winkles, C. Peschle and R. De Maria: Multiple members of the TNF superfamily contribute to IFN-gamma-mediated inhibition of erythropoiesis. J Inmunol 2005; 1: 1464-1472.
[54]
C. Dogra, H. Changotra, S. Mohan and A. Kumar: Tumor necrosis factor-like weak inducer of apoptosis inhibits skeletal myogenesis through sustained activation of nuclear factor-kappaB and degradation of myoD protein. J Biol Chem 2006; 281: 10327-10336. [Pubmed]
[55]
S. J. Perper, B. Browning, L. C. Burkly, S. Weng, C. Gao, K. Giza, L. Su, L. Tarilonte, T. Crowell, L. Rajman, L. Runkel, M. Scott, G. J. Atkins, D. M. Findlay, T. S. Zheng and H. Hess: TWEAK is a novel arthritogenic mediator. J Immunol 2006; 177: 2610-2620. [Pubmed]
[56]
H. Xu, A. Okamoto, J. Ichikawa, T. Ando, K. Tasaka, K. Masuyama, H. Ogawa, H. Yagita, K. Okumura and A. Nakao: TWEAK/Fn14 interaction stimulates human bronchial epithelial cells to produce IL-8 and GM-CSF. Biochem Biophys Res Commun 2004; 318: 422-427. [Pubmed]
[57]
S. H. Kim, Y. J. Kang, W. J. Kim, D. K. Woo, Y. Lee, D. I. Kim, Y. B. Park, B. S. Kwon, J. E. Park and W. H. Lee: TWEAK can induce pro-inflammatory cytokines and matrix metalloproteinase-9 in macrophages. Circ J 2004; 68: 396-399. [Pubmed]
[58]
P. Saas, J. Boucraut, PR. Walker, AL. Quiquerez, M. Billot, S. Desplat-Jego, Y. Chicheportiche, PY. Dietrich: TWEAK stimulation of astrocytes and the proinflammatory consequences. Glia 2000; 32:102-7. [Pubmed]
[59]
T. Kawakita, K. Shiraki, Y. Yamanaka, Y. Yamaguchi, Y. Saitou, N. Enokimura, N. Yamamoto, H. Okano, K. Sugimoto, K. Murata and T. Nakano: Functional expression of TWEAK in human hepatocellular carcinoma: possible implication in cell proliferation and tumor angiogenesis. Biochem Biophys Res Commun 2004; 318: 726-733. [Pubmed]
[60]
D. H. Ho, H. Vu, S. A. N. Brown, P. J. Donohue, H. N. Hanscom and J. A. Winkles: Soluble tumor necrosis factor-like weak inducer of apoptosis overexpression in HEK293 cells promotes tumor growth and angiogenesis in athymic nude mice. Cancer Res 2004; 64: 8968-8972. [Pubmed]
[61]
R. Polavarapu, M. C. Gongora, J. A. Winkles and M. Yepes: Tumor necrosis factor-like weak inducer of apoptosis increases the permeability of the neurovascular unit through nuclear factor-kappaB pathway activation. J Neurosci 2005; 25: 10094-10100. [Pubmed]
[62]
Gomez-Chiarri M, Ortiz A, Seron D, Glez E, Egido J. The intercrine superfamily and renal disease. Kidney Int 1993; supl 39:S81-S85.
[63]
Beinke S, Ley SC. Functions of NF-kappaB1 and NF-kappaB2 in immune cell biology. Biochem J 2004; 382(Pt 2):393-409. [Pubmed]
[64]
Sanz AB, Santamaria B, Ruiz Ortega M, Egido J, Ortiz A. Mechanisms of renal apoptosis in health and disease. J Am Soc Nephrol 2007; Aceptado. [Pubmed]
[65]
Lorz C, Justo P, Sanz A, Subirá D, Egido J, Ortiz A. Paracetamol nephrotoxicity: a role for ER stress. J Am Soc Nephrol 2004; 15:380-9. [Pubmed]
[66]
Justo P, Lorz C, Sanz A, Egido J, Ortiz A. Intracellular mechanisms of cyclosporine A-induced tubular cell apoptosis. J Am Soc Nephrol 2003; 14: 3072-3080 [Pubmed]
[67]
Ortiz A, Lorz C, Egido J. New kids in the block: the role of FasL and Fas in kidney damage. J Nephrol 1999;12:150-158 [Pubmed]
[68]
Ortiz A, Lorz C, Egido J. The Fas ligand/Fas system in renal injury. Nephrol Dial Transplant 1999;14:1831-1834 [Pubmed]
[69]
Ortiz A, Gonzalez-Cuadrado S, Bustos C, Alonso J, Gómez-Guerrero C, López-Armada MJ, González E, Plaza JJ, Egido J. Tumor necrosis factor and glomerular damage. J Nephrol 1995;8:27-34.
[70]
MJ. Kaplan, D. Ray, RR. Mo, RL. Yung, BC. Richardson: TRAIL (Apo2 ligand) and TWEAK (Apo3 ligand) mediate CD4+ T cell killing of antigen-presenting macrophages. J Immunol 2000; 164:2897-904. [Pubmed]
[71]
A. Ortiz, C. Lorz, MP. Catalán, TM. Danoff, Y. Yamasaki, J. Egido, EG. Neilson: Expression of apoptosis regulatory proteins in tubular epithelium stressed in culture or following acute renal failure. Kidney Int 2000; 57, 969-981. [Pubmed]
[72]
N. Goes, J. Urmson, V. Ramassar, PF. Halloran: Ischemic acute tubular necrosis induces an extensive local cytokine response. Evidence for induction of interferon-gamma, transforming growth factor-beta 1, granulocyte-macrophage colony-stimulating factor, interleukin-2, and interleukin-10. Transplantation 1995; 59: 565-72.
[73]
C. Lorz, A. Ortiz, P. Justo, S. Gonzalez-Cuadrado, N. Duque, C. Gomez-Guerrero, J. Egido: Proapoptotic Fas ligand is expressed by normal kidney tubular epithelium and injured glomeruli. J Am Soc Nephrol 2000; 11: 1266-77 [Pubmed]
[74]
KA. Janes, JG. Albeck, S. Gaudet, PK. Sorger, DA. Lauffernburger, MB Yaffe: A systems model of signaling identifies a molecular basis set for cytokine-induced apoptosis. Science 2005; 310, 1646-53. [Pubmed]
[75]
A. Khwaja, L. Tatton: Resistance to the cytotoxic effects of tumor necrosis factor alpha can be overcome by inhibition of a FADD/caspase-dependent signaling pathway. J Biol Chem 1999; 274:36817-23. [Pubmed]
[76]
U. Metz-Kurschel, E. Kurschel, K. Wagner, E. Aulbert, N. Graben, T. Philipp: Folate nephropathy occurring during cytotoxic chemotherapy with high-dose folinic acid and 5-fluorouracil. Ren Fail 1990; 12(2): 93-7. [Pubmed]
[77]
A. Ortega, D. Ramila, JA. Ardura, V. Esteban, M. Ruiz-Ortega, A. Barat, R. Gazapo, RJ. Bosch, P. Esbrit: Role of parathyroid hormone-related protein in tubulointerstitial apoptosis and fibrosis after folic Acid-induced nephrotoxicity. J Am Soc Nephrol 2006; 17: 1594-603. [Pubmed]
[78]
M. Girgenrath, S. Weng, CA. Kostek, B. Browning, M. Wang, SA. Brown, JA. Winkles, JS. Michaelson, N. Allaire, P. Schneider, ML. Scott, YM. Hsu, H. Yagita, RA. Flavell, JB. Miller, LC. Burkly, TS. Zheng: TWEAK, via its receptor Fn14, is a novel regulator of mesenchymal progenitor cells and skeletal muscle regeneration. EMBO J 2006; 25: 5826-39. [Pubmed]
[79]
N. Schwartz, L. Su, L.C. Burkly, M. Mackay, C. Aranow, M. Kollaros, J.S. Michaelson, B. Rovin, C. Putterman: Urinary TWEAK and the activity of lupus nephritis. J Autoimmun 2006; 27: 242-50. [Pubmed]
Idiomas
Nefrología (English Edition)
Article options
Tools
es en

¿Es usted profesional sanitario apto para prescribir o dispensar medicamentos?

Are you a health professional able to prescribe or dispense drugs?