IN INFLAMMATION

TWEAK/Fn14 and non-canonical NF-kappaB signaling in kidney disease Jonay Poveda, Luis C Tabara, Beatriz Fernandez-Fernandez, Catalina Martin-Cleary, Ana B Sanz, Rafael Selgas, Alberto Ortiz and Maria D Sanchez-Niño

Journal Name:

Frontiers in Immunology

ISSN:

1664-3224

Article type:

Mini Review Article

Received on:

30 Sep 2013

Accepted on:

26 Nov 2013

Provisional PDF published on:

26 Nov 2013

Frontiers website link:

www.frontiersin.org

Citation:

Poveda J, Tabara LC, Fernandez-fernandez B, Martin-cleary C, Sanz AB, Selgas R, Ortiz A and Sanchez-niño MD(2013) TWEAK/Fn14 and non-canonical NF-kappaB signaling in kidney disease. 4:447. doi:10.3389/fimmu.2013.00447

Article URL:

http://www.frontiersin.org/Journal/Abstract.aspx?s=1245& name=inflammation&ART_DOI=10.3389/fimmu.2013.00447 (If clicking on the link doesn't work, try copying and pasting it into your browser.)

Copyright statement:

© 2013 Poveda, Tabara, Fernandez-fernandez, Martin-cleary, Sanz, Selgas, Ortiz and Sanchez-niño. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) or licensor are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

This Provisional PDF corresponds to the article as it appeared upon acceptance, after rigorous peer-review. Fully formatted PDF and full text (HTML) versions will be made available soon.

TWEAK/Fn14 and non-canonical NF-kappaB signaling in kidney disease

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49

Jonay Poveda1+, Luis C Tabara1+, Beatriz Fernandez-Fernandez1, Catalina MartinCleary1, Ana B Sanz1, Rafael Selgas2, Alberto Ortiz1, Maria D Sanchez-Niño2* 1

Department of Nephrology, IIS-Fundacion Jimenez Diaz, Universidad Autonoma de Madrid and IRSIN, Madrid, Spain. 2 Department of Nephrology, IDIPAZ, Madrid, Spain. + Both authors contributed equally *Corresponding Author Correspondence: Dr. Maria D Sanchez-Niño Division of Nephrology IdiPAZ Paseo de la Castellana 260 28046 Madrid, Spain E-mail: [email protected] Word count: 2790 Abstract: The incidence of acute kidney injury (AKI) and chronic kidney disease (CKD) is increasing. However, there is no effective therapy for AKI and current approaches only slow down, but do not prevent progression of CKD. TWEAK is a TNF superfamily cytokine. A solid base of preclinical data suggests a role of therapies targeting the TWEAK or its receptor Fn14 in AKI and CKD. In particular TWEAK/Fn14 targeting may preserve renal function and decrease cell death, inflammation, proteinuria and fibrosis in mouse animal models. Furthermore there is clinical evidence for a role of TWEAK in human kidney injury including increased tissue and/or urinary levels of TWEAK and parenchymal renal cell expression of the receptor Fn14. In this regard, clinical trials of TWEAK targeting are ongoing in lupus nephritis. NF-κB activation plays a key role in TWEAK-elicited inflammatory responses. Activation of the noncanonical NF-κB pathway is a critical difference between TWEAK and TNF. TWEAK activation of the non-canonical NF-κB pathways promotes inflammatory responses in tubular cells. However, there is an incomplete understanding of the role of noncanonical NF-κB activation in kidney disease and on its contribution to TWEAK actions in vivo. Keywords: acute kidney injury, fibrosis, inflammation, kidney, lupus nephritis, podocyte, proteinuria. Running title: TWEAK, NF-κB and the kidney

1

50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87 88 89 90 91 92 93 94 95 96 97 98 99

1. Unsolved issues in kidney disease Acute kidney injury (AKI) and chronic kidney disease (CKD) are the most severe forms of kidney disease (1;2). AKI is characterized by a sudden loss of renal function. AKI patients have increased short- and long-term mortality and risk of CKD progression. However, there is no therapy that accelerates recovery from AKI. CKD is a major healthcare problem, with more than 20 million aged 20 years or older affected in the United States. Diabetic kidney disease is the leading cause of end stage renal disease in the Western Countries. However, current treatments based on blockade of the reninangiotensin system are not sufficient to prevent progression of diabetic kidney disease (3). Recent evidence suggests a role for TNF superfamily member Tumor necrosis factor-like weak inducer of apoptosis (TWEAK, Apo3L or TNFSF12) in both AKI and CKD, where it has been shown to regulate cell death, inflammation and fibrosis through activation of the TWEAK receptor Fn14 and a variety of intracellular signaling pathways, including the transcription factor nuclear factor-kappa B (NF-κB) (4;5) (Figure 1). Clinical trials are testing anti-TWEAK neutralizing antibodies (6;7). One key difference between TWEAK and the best characterized member of the family, TNF, is that TWEAK activates the non-canonical NF-κB pathway. We now review current information on TWEAK, non-canonical NF-κB activation and kidney disease. 2. TWEAK TWEAK may be membrane-bound or soluble, although most functional studies have been performed with soluble TWEAK. Soluble TWEAK is thought to be generated from full-length TWEAK by furin-mediated cleavage of the extracellular domain (8). The TWEAK receptor, Fn14 (TNFRSF12a), is the smallest member of the TNF receptor superfamily. Fn14 is a type I transmembrane protein which has 102 aa in its mature isoform. The extracellular domain has 53 aa and harbors a cystein rich domain required for TWEAK binding (9). Interestingly, the Fn14 intracellular domain (29aa) lacks the characteristic death domain of TNFRSF receptors but contains TNFRassociated factor (TRAF) binding sites. Fn14 trimerization recruits TRAF2 and TRAF3 upon TWEAK binding (10). TWEAK may regulate cell proliferation, cell death, cell differentiation, and inflammation (4;8). TWEAK may trigger cell death or proliferation processes, depending on cell type and microenvironment; TWEAK promotes proliferation of numerous cell types including quiescent renal tubular cells through activation of NF-κB, MAPK and PI3K/AKT pathways (11). In addition TWEAK was described as a weak inductor of apoptosis which required special microenvironment (such as the presence of interferonγ -IFNγ-) to induce cell death (12-14). Under certain circumstances TWEAK can induce apoptosis without co-treatment with other cytokines. It has been proposed that levels of Fn14 expression may sensitize cells to TWEAK but it is also clear that this cannot be the only mechanism (11). Indeed, the signaling cascade which triggers cell death following Fn14 activation remains poorly understood as Fn14 does not contain a death

2

100 101 102 103 104 105 106 107 108 109 110 111 112 113 114 115 116 117 118 119 120 121 122 123 124 125 126 127 128 129 130 131 132 133 134 135 136 137 138 139 140 141 142 143 144 145 146 147 148 149

domain(9). Induction of TNF expression by TWEAK has been reported in certain cell types. In immortalized tumor cells, TWEAK activation of Fn14 recruits a TRAF2/cellular inhibitor of apoptosis 1 (cIAP1) complex that results in the lysosomal degradation of cIAP1-TRAF2 in a cIAP1-dependent manner (15). TWEAK depletion of cIAP1 and TRAF2 activates non-canonical NF-κB signaling. However the function of non-canonical NF-κB signaling was not explored. TWEAK is expressed in many tissues. High levels are found in the pancreas, intestine, heart, brain, lung, ovary, vasculature, and skeletal muscle, and lower levels in the liver and kidney (4). Fn14 is expressed by many cell types, including epithelial, mesenchymal, and endothelial cells. In healthy tissues Fn14 expression is low. However, cellular Fn14 levels are increased in response to stress or injury. 3. TWEAK/Fn14 in kidney disease Fn14 is expressed in kidney tubular cells, mesangial cells and podocytes (12). Fn14 expression by kidney endothelium has not been well characterized. Renal infiltrating cells such as macrophages also express Fn14 (16). Fn14-expressing cells are potentially responsive to TWEAK. In addition TWEAK and Fn14 expression is increased in kidney injury and targeting of the system is beneficial in different models of kidney injury. 3.1 TWEAK/Fn14 actions on renal cells Potential kidney sources of TWEAK include infiltrating monocytes and T lymphocytes and local cells such as mesangial and tubular cells (12;17-19). During glomerular injury both mesangial cells and podocytes may be targets of the inflammatory response. Mesangial cell injury is observed in proliferative glomerulonephritis, while podocyte injury is characteristic of proteinuric kidney diseases. TWEAK promotes the expression of chemokines, adhesion molecules and matrix metalloproteinases in human and murine mesangial cells (19;20). TWEAK also increases mesangial cells proliferation, but TWEAK combined with IFN-γ promotes mesangial cells apoptosis (19;20). In human and murine podocytes TWEAK induces the expression of proinflammatory mediators in an NF-κB-dependent manner (20;21). TWEAK also promotes nephrin expression and human podocyte proliferation (20). Expression of nephrin and proliferation are not usually associated in vivo. In fact, podocytes are terminally differentiated cells that do not divide. Podocyte proliferation is only observed under very specific pathological circumstances and is usually associated with dedifferentiation and loss of podocyte markers including nephrin. In murine and human renal tubular cells TWEAK also promotes the expression of cytokines and chemokines (22). TWEAK also increases tubular cell proliferation through recruitment of the mitogen-activated protein kinases ERK and p38, the phosphatidyl-inositol 3-kinase (PI3K)/Akt pathway and the canonical NF-κB pathway (11). Similar to observations in mesangial cells, in a proinflammatory milieu TWEAK induces apoptosis of tubular cells (12). By contrast to mesangial cells, the lethal action of TWEAK in tubular cells requires the simultaneous presence of TNFα and INFγ. Surprisingly, caspase inhibition prevented the features of apoptosis induced by the cytokine cocktail but increased overall cell death through a reactive oxygen speciesdependent necrotic pathway (12). More recently, TWEAK/TNFα/INFγ-induced cell

3

150 151 152 153 154 155 156 157 158 159 160 161 162 163 164 165 166 167 168 169 170 171 172 173 174 175 176 177 178 179 180 181 182 183 184 185 186 187 188 189 190 191 192 193 194 195 196 197 198 199

death in tubular cells was shown to have features of necroptosis (23). Necroptosis is an active form of cell death that requires the kinase activity of receptor-interacting protein 1 (RIP1) and RIP3. TWEAK also promotes murine renal fibroblasts proliferation through activation of the Ras/ERK pathway (24). The proliferative effect of TWEAK on fibroblasts overrides its negative effect on extracellular matrix production. Thus, the overall effect of TWEAK targeting in experimental renal fibrosis is decreased fibrosis (24). In addition, TWEAK also promotes the expression of inflammatory cytokines in renal fibroblasts (24). So far, the proinflammatory effect of TWEAK on mesangial cells, podocytes and fibroblasts have been shown to proceed through canonical NF-κB activation involving RelA migration to the nucleus and expression of canonical RelA targets such as MCP1, RANTES and others (20;21;24). By contrast, both canonical and non-canonical NF-κB activation by TWEAK have been observed in tubular cells (22;25). The known consequences of non-canonical NF-κB activation are discussed below. 3.2 TWEAK/Fn14 expression in kidney injury TWEAK and Fn14 expression is increased in experimental animal models of AKI, lupus nephritis, albumin overdose-induced proteinuria, kidney fibrosis induced by unilateral ureteral obstruction and anti-GBM nephritis (12;21;22;24;26;27). High levels of tubular Fn14 expression have been also observed in human ischemic AKI and in acute or chronic human tubulointerstitial inflammation (26;28) In human lupus nephritis glomerular Fn14 mRNA expression was increased and was higher in proliferative than in membranous lupus nephropathy (29;30). Urinary TWEAK has been proposed as a biomarker of lupus nephritis activity (31-34). 3.3 Therapeutic modulation of TWEAK or Fn14 in experimental kidney injury Therapeutic modulation of the TWEAK/Fn14 pathway has been successful in experimental models of AKI, kidney fibrosis, lipid-induced kidney injury, proteinuriainduced kidney injury and immune-mediated glomerular injury, including lupus nephritis. The TWEAK/Fn14 pathway was modulated in mice either by gene targeting of TWEAK/Fn14, by neutralizing anti-TWEAK antibodies or by blocking anti-Fn14 antibodies. Mice with experimental ischemic or folic acid induced AKI displayed a variety of benefits from TWEAK targeting that included better histological parameters and renal function, and reduction of chemokine expression, tubular cell apoptosis and renal fibrosis, while the anti-inflammatory and anti-aging hormone klotho was increased (4;11;22;25;26;28;35). TWEAK downregulates Klotho in normal kidneys (35). Fn14-deficient mice show decreased kidney damage, inflammation and fibrosis in models of lupus nephritis (5;36). Anti-TWEAK neutralizing antibodies reduced inflammatory gene expression and renal damage in lupus nephritis (36). Reduced residual fibrosis was observed in mice which had been protected from the acute phase of ischemia reperfusion by anti-Fn14 blocking antibodies (26). Protection from fibrosis

4

200 201 202 203 204 205 206 207 208 209 210 211 212 213 214 215 216 217 218 219 220 221 222 223 224 225 226 227 228 229 230 231 232 233 234 235 236 237 238 239 240 241 242 243 244 245 246 247 248 249

by interfering with TWEAK/Fn14 is not limited to residual fibrosis following amelioration of the initial injury. TWEAK knockout mice were protected from fibrosis in the unilateral ureteral obstruction of model of persistent kidney insult while overexpression of TWEAK causes renal fibrosis in normal previously normal kidneys (24). Fn14 deficient mice were protected from anti-GBM induced glomerulonephritis (27). In addition, neutralizing anti-TWEAK antibodies improved nephritis in wild type mice without altering the adaptive immune response, indicating that TWEAK/Fn14 directly regulates the inflammatory response (27). In this regard, anti-TWEAK antibodies decreased hyperlipidemia-induced kidney inflammation and injury (37). Experimental kidney diseases in which TWEAK/Fn14 targeting has been successful share the presence of diverse degrees of local inflammation. Thus, the kidney milieu to some extent reproduces the cell culture conditions under which TWEAK promotes kidney cell death. However, the environment also influences TWEAK actions in the kidney in vivo. The TWEAK/Fn14 pathway may contribute to tissue regeneration (11;38;39). In experimental, inflammation-free unilateral nephrectomy TWEAK promotes remnant kidney growth and tubular cell proliferation (11). However, TWEAK knockout mice have decreased remnant kidney size and tubular cell proliferation (11). This information may be useful in the context of regenerative medicine. However, the regenerative potential of TWEAK was not apparent in animal models of inflammatory kidney injury, where the injurious effect was observed in all models studied so far. 4. Non-canonical NF-κB signaling The NF-κB transcription factor binds to the κB enhancer in DNA to control transcription of over 400 genes. NF-κB controls immune and inflammatory responses, developmental processes, cellular growth, and apoptosis. Dysregulation of NF-κB has been linked to cancer, inflammatory and autoimmune diseases (11;25;40). The mammalian NF-κB family has five members, RelA/p65, RelB, c-Rel, NFκB1 p50, and NF-κB2 p52 (41;42). All share a highly conserved DNAbinding/dimerization domain called the Rel homology domain (RHD), through which they form homo or heterodimers. RelA, c-Rel and RelB contain a C-terminal transactivation domain (TAD) with multiple ankyrin repeats. In order to activate transcription, they form dimers with either p50 or p52. NF-κB activation does not require the novo synthesis of NF-κB proteins. In most cells, NF-κB proteins are present as an inactive complex in the cytoplasm. The activity of NF-κB is regulated by its interaction with inhibitory IκB proteins. The IκB proteins include p105, p100, IkBα, IκBβ, IκBγ, IκBε, IκBz and Bcl-3 (43-45), NFκB1 and NFκB2 are synthesized as precursors, p105 and p100, respectively. These precursors contain an IκB-like C-terminal portion and function as NF-κB inhibitors. Ubiquitin/proteasome processing results in selective degradation of the C-terminal ankyrin repeats, disrupts the IκB-like function and generates the active NF-κB subunits p50 and p52 (46;47). NF-κB activation in response to extracellular signals can proceed through classical/canonical, alternative/non-canonical or hybrid pathways (4;40;48-51).

5

250 251 252 253 254 255 256 257 258 259 260 261 262 263 264 265 266 267 268 269 270 271 272 273 274 275 276 277 278 279 280 281 282 283 284 285 286 287 288 289 290 291 292 293 294 295 296 297 298

Classical NF-κB activation is a rapid and transient response to a wide range of stimuli, while the alternative pathway involves slow activation of the p100/RelB heterodimer leading to the generation of p52/RelB and prolonged activation of NF-κB target genes in response to a more limited set of stimuli (47;52). There is interplay between both pathways. Thus, classical NF-κB activation-induced transcription of NF-κB2 and RelB favors activation of the non-canonical pathway. Both pathways converge on the activation of a complex that contains a serine-specific IκB kinase (IKK). IKK contains at least, three distinct subunits: the catalytic kinase subunits IKKα (IKK1) and IKKβ (IKK2) and the regulatory subunit, IKKγ (NEMO). NF-κB inducing kinase (NIK, MAP3K14) is the apical kinase triggering noncanonical NF-κB activation. NIK belongs to the family of MAP3Ks that are known to be activated through T-loop phosphorylation. Upon activation, NIK activates IKKα and serves as a docking molecule that recruits IKKα to p100, facilitating ubiquitination by the β-TrCP ubiquitin ligase and subsequent proteasomal processing into the mature p52 subunit in a manner dependent on IKKα-dependent p100 phosphorylation (52-54). This allows the RelB/p52 heterodimer to translocate to the nucleus and to activate transcription of target genes (55). p100 processing is regulated by a short list of activators known to signal through NIK (55-59). This list includes TWEAK (60). A variety of functions have been described for NIK including generation and/or maintenance of memory T cells (61), the formation of Th17 cells (62), promotion of glucagon responses (63) and the pathogenesis of chronic inflammation and insulin resistance in type 2 diabetes (64). Some of these functions may be independent from activation of IKKα and the non-canonical NF-κB pathway (65) and for others the relationship to non-canonical NF-κB was not explored. Thus, NIK modulates melanoma survival and growth through a β-catenin-mediated transcription way (66), is recruited to the promoters of pro-inflammatory genes to induce H3K9 histone acetylation in response to TNFα (67) and may favor or repress Smac-mimetic induced death depending on the cell context. NIK upregulation in response to Smac mimetics/TNF repressed apoptosis induced by this combination, likely by maintaining FLICE inhibitory protein (c-FLIP) levels to suppress caspase-8 activation. Thus, resistant cells were sensitized to cell death by NIK depletion. NIK was required for activation of both canonical and non-canonical NF-κB pathways but their relative contribution to the protective effect was not explored (68). 5. Non-canonical NF-κB activation and kidney disease There is little information on the occurrence and role of non-canonical NF-κB activation in kidney disease. Few studies have addressed the overall regulation of the pathway. However, a few reports have explored individual molecules participating in non-canonical NF-κB activation, frequently without exploring function. In diabetic mice kidney cortex NIK and RelB are upregulated several fold and phosphorylation of IKK alpha was increased (69). Non-canonical NF-κB components were predominantly located in tubular epithelial cells (69). NIK overexpression in cultured human proximal tubular cells increased RelB/p52 nuclear levels and DNA binding activity and expression of inflammatory cytokines such as IL-6, IL-8, and MCP-1 (70). TRAF3 silencing also increased nuclear RelB/p52 and transcription of

6

299 300 301 302 303 304 305 306 307 308 309 310 311 312 313 314 315 316 317 318 319 320 321 322 323 324 325 326 327 328 329 330 331 332 333 334 335 336 337 338 339 340 341 342 343 344 345 346 347 348

proinflammatory cytokines. AGEs increased NIK and nuclear RelB/p52 in cultured proximal tubular cells (70). In human kidney graft biopsies with delayed graft function NIK was increased in proximal tubular, interstitial, and mesangial cells and was observed in nuclei. In pig ischemia-reperfusion tubular and glomerular NIK phosporylation was increased as observed by immunohistochemistry. In cultured proximal tubular cells thrombin induced NIK phosporylation (71). However, no functional study addressed the consequences of NIK phosphorylation. RelB targeting by siRNA may protect mice against lethal kidney ischemia (72). Mice injected with RelB siRNA had lower serum creatinine, histological tissue injury and TNF expression as compared to controls. Furthermore, RelB targeting increased survival (72). In cultured proximal tubular cells, lentiviral small hairpin RNA (shRNA)mediated knockdown of RelB, abrogated the excess apoptosis induced by TNF in combination with cisplatin. Thus, cells with targeted RelB exposed to TNF/cisplatin have the same apoptosis rate as cells treated only with cisplatin. RelB targeting protection from apoptosis was associated with phenotypic markers of epithelial-tomesenchymal transition. A transcriptomics analysis disclosed that knockdown of RelB was associated with upregulation of Snai2 and Rho GTPases. Targeting Rho kinase prevented the protective action of RelB knockdown (73). The uremic toxins p-cresylsulphate and indoxylsulfate increased NF-κB2 expression by 50-80% in cultured proximal tubular cells (74). However, whether this was associated with increased protein levels or the functional consequences of this observation for the tubular cell cytotoxicity or inflammatory response elicited by these toxins (75) were not explored. 6. TWEAK and non-canonical NF-κB activation in kidney disease A sustained NF-κB activation, persistent for up to 24 hours, was observed in tubular cells exposed to TWEAK, consistent with activation of the non-canonical pathway in addition to the already characterized canonical NF-κB activation (22) (Table 1). In this regard, in cultured renal tubular cells TWEAK increases nuclear RelB/p52 accumulation, RelB and p52 DNA binding activity and NIK- and RelBdependent CCL21 and CCL19 expression (25). Nuclear RelB/p52 migration and CCL21/CCL19 expression peaked at 24h and, thus, were delayed as compared to RelA nuclear migration and expression of canonical RelA-dependent genes such as MCP1 and RANTES that peak at 3h and 6h, respectively. By contrast, parthenolide, which inhibits the degradation of IκBα and RelA nuclear translocation, did not prevent CCL21 upregulation (22;25;76). Furthermore, TWEAK administration in vivo to healthy mice resulted in nuclear translocation of RelB and p52 in tubular cells and in increased renal CCL21 expression. Conversely, neutralizing anti-TWEAK antibodies prevented both RelB/p52 accumulation and increased expression of CCL21 in mice with folic acidinduced AKI (22). CCL21 expression had been previously shown to be dependent on non-canonical NF-κB activation in non-renal cells (55). CCL21 is T-cell and fibrocyte chemotactic factor that plays a role in renal tubulointerstitial fibrosis (77;78).

7

349 350 351 352 353 354 355 356 357 358 359 360 361 362 363 364 365 366 367 368 369 370 371 372 373 374 375 376 377 378 379 380 381 382 383 384 385 386 387 388 389 390 391 392 393 394 395 396 397 398

In summary, TWEAK is the only cytokine known to activate the non-canonical NF-κB pathway in tubular cells, both in cell culture and in vivo. Activation of the noncanonical NF-κB pathway is a key difference with TNF. However, whether TWEAK activates the non-canonical NF-κB pathway in mesangial cells, podocytes or kidney fibroblasts and the functional in these cells remains unexplored. 7. Conclusions Accumulating evidence suggests a role for TWEAK in the pathogenesis of diverse forms of kidney injury, thus making TWEAK an attractive therapeutic target. Indeed, ongoing clinical trials are targeting TWEAK in kidney disease. Recently, a phase I clinical trial of anti-TWEAK neutralizing antibodies in rheumatoid arthritis was completed (6). Intravenous administration of anti-TWEAK resulted in undetectable serum-TWEAK for a month and in decreased levels of several inflammatory biomarkers. An ongoing phase II trial in lupus nephritis patients is testing the nephroprotective effect of BIIB023 anti-TWEAK antibody (7). TWEAK is one of a handful of cytokines that activate the non-canonical NF-κB pathway and the only one to have been explored with respect to non-canonical NF-κB pathway activation in kidney cells. Functional studies suggest that non-canonical NF-κB activation is a relevant action for TWEAK-induced kidney inflammation. Potential therapeutic approaches include both the simultaneous inhibition of both NF-κB pathways when targeting TWEAK as well as the eventual independent regulation of canonical and non-canonical NF-κB responses by designing differential inhibitors. While these non-canonical NF-κB inhibitors are not yet ready for human use, progress is being made on the design of NIK inhibitors (79). However, there is little functional information on the overall role of NIK and non-canonical NF-κB activation in kidney disease and on the consequences of differential therapeutically manipulation of canonical and non-canonical NF-κB responses. Clearly, more research is needed in this area. Conflict of Interest Statement There are no conflicts of interest. Author contributions MDSN and AO devised the structure and overviewed and directed the effort. LCT, JP, BFF and CMC reviewed the TWEAK and the non-canonical NF-κB literature, respectively. ABS and RS contributed to the final form. Acknowledgments Grant support: ISCII and FEDER funds FIS PS09/00447, ISCIII-RETIC REDinREN/RD06/0016, RD12/0021, Comunidad de Madrid/CIFRA/ S2010/BMD2378. Salary support: FIS to MDSN, Programa Intensificación Actividad Investigadora (ISCIII/Agencia Laín-Entralgo/CM) to AO, FPU (Ministerio de Educación, Cultura y Deporte) to JP, Fundacion Conchita Rabago to LCT.

Table 1. TWEAK actions on kidney cells involving NF-κB activation and evidence for the role of canonical- or non-canonical pathways.

8

399 400

Cell type

Effect

Mesangial cells Inflammation Podocytes Inflammation Tubular cells Inflammation Proliferation Inflammation: CCL21, CCL19 Renal Inflammation fibroblasts 401 402 403 404 405 406 407 408 409 410 411 412 413 414 415 416 417 418 419 420 421 422 423 424 425 426 427 428 429 430 431 432 433 434 435 436 437 438 439 440

Functional modulation

NF-κB pathway involved Canonical Canonical Canonical

Reference

BAY11-7082 Parthenolide Parthenolide Parthenolide NIK siRNA, Non-canonical RelB siRNA

(20) (21) (22) (11) (25)

Parthenolide

Canonical

(24)

Legend to figures Figure 1. Key intracellular pathways activated by TWEAK engagement of Fn14 in kidney tubular cells. TWEAK signaling in kidney cells has been characterized most in detail in tubular cells. TWEAK engages both the canonical and the non-canonical NFκB pathways and kinase signaling mechanisms. Reference List 1. Kidney Disease: Improving Global Outcomes (KDIGO) Acute Kidney Injury Work Group. KDIGO Clinical Practice Guideline for Acute Kidney Injury. Kidney inter., Suppl. 2012; 2: 1138 2. Kidney Disease: Improving Global Outcomes (KDIGO) CKD Work Group. KDIGO 2012 Clinical Practice Guideline for the Evaluation and Management of Chronic Kidney Disease. Kidney inter., Suppl. 2013; 3: 1-150 3. Fernandez, F. B., Elewa, U., Sanchez-Nino, M. D., Rojas-Rivera, J. E., Martin-Cleary, C., Egido, J., Ortiz, A. (2012) 2012 update on diabetic kidney disease: the expanding spectrum, novel pathogenic insights and recent clinical trials. Minerva Med 103, 219-234. doi:R10123407 [pii]. 4. Sanz, A. B., Sanchez-Nino, M. D., Ortiz, A. (2011) TWEAK, a multifunctional cytokine in kidney injury. Kidney Int 80, 708-718. doi:ki2011180 [pii];10.1038/ki.2011.180 [doi]. 5. Sanz, A. B., Sanchez-Nino, M. D., Martin-Cleary, C., Ortiz, A., Ramos, A. M. (2013) Progress in the development of animal models of acute kidney injury and its impact on drug discovery. Expert.Opin Drug Discov 8, 879-895 6. http://clinicaltrials.gov/ct2/show/NCT00771329, accessed May 5, 2013 7. http://clinicaltrials.gov/ct2/show/NCT01499355, accessed April 14, 2013 8. Chicheportiche, Y., Bourdon, P. R., Xu, H., Hsu, Y. M., Scott, H., Hession, C., Garcia, I., Browning, J. L. (1997) TWEAK, a new secreted ligand in the tumor necrosis factor family that weakly induces apoptosis. J Biol Chem 272, 32401-32410

9

441 442 443 444 445 446 447 448 449 450 451 452 453 454 455 456 457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481 482 483 484 485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500

9. He, F., Dang, W., Saito, K., Watanabe, S., Kobayashi, N., Guntert, P., Kigawa, T., Tanaka, A., Muto, Y., Yokoyama, S. (2009) Solution structure of the cysteine-rich domain in Fn14, a member of the tumor necrosis factor receptor superfamily. Protein Sci 18, 650-656. doi:10.1002/pro.49 [doi] 10. Brown, S. A., Richards, C. M., Hanscom, H. N., Feng, S. L., Winkles, J. A. (2003) The Fn14 cytoplasmic tail binds tumour-necrosis-factor-receptor-associated factors 1, 2, 3 and 5 and mediates nuclear factor-kappaB activation. Biochem J 371, 395-403. [doi];BJ20021730 [pii]. 11. Sanz, A. B., Sanchez-Nino, M. D., Izquierdo, M. C., Jakubowski, A., Justo, P., Blanco-Colio, L. M., Ruiz-Ortega, M., Egido, J., Ortiz, A. (2009) Tweak induces proliferation in renal tubular epithelium: a role in uninephrectomy induced renal hyperplasia. J Cell Mol Med 13, 3329-3342. doi:JCMM766 [pii];10.1111/j.1582-4934.2009.00766.x [doi]. 12. Justo, P., Sanz, A. B., Sanchez-Nino, M. D., Winkles, J. A., Lorz, C., Egido, J., Ortiz, A. (2006) Cytokine cooperation in renal tubular cell injury: the role of TWEAK. Kidney Int 70, 1750-1758. doi:5001866 [pii];10.1038/sj.ki.5001866 [doi]. 13. Ortiz, A., Lorz, C., Catalan, M. P., Danoff, T. M., Yamasaki, Y., Egido, J., Neilson, E. G. (2000) Expression of apoptosis regulatory proteins in tubular epithelium stressed in culture or following acute renal failure. Kidney Int 57, 969-981. doi:kid925 [pii];10.1046/j.1523-1755.2000.00925.x [doi]. 14. Nakayama, M., Ishidoh, K., Kojima, Y., Harada, N., Kominami, E., Okumura, K., Yagita, H. (2003) Fibroblast growth factor-inducible 14 mediates multiple pathways of TWEAK-induced cell death. J Immunol 170, 341-348 15. Vince, J. E., Chau, D., Callus, B., Wong, W. W., Hawkins, C. J., Schneider, P., McKinlay, M., Benetatos, C. A., Condon, S. M., Chunduru, S. K., Yeoh, G., Brink, R., Vaux, D. L., Silke, J. (2008) TWEAK-FN14 signaling induces lysosomal degradation of a cIAP1-TRAF2 complex to sensitize tumor cells to TNFalpha. J Cell Biol 182, 171-184. doi:jcb.200801010 [pii];10.1083/jcb.200801010 [doi]. 16. Martin, C. C., Moreno, J. A., Fernandez, B., Ortiz, A., Parra, E. G., Gracia, C., Blanco-Colio, L. M., Barat, A., Egido, J. (2010) Glomerular haematuria, renal interstitial haemorrhage and acute kidney injury. Nephrol Dial Transplant 25, 4103-4106. doi:gfq493 [pii];10.1093/ndt/gfq493 [doi]. 17. Kaplan, M. J., Lewis, E. E., Shelden, E. A., Somers, E., Pavlic, R., McCune, W. J., Richardson, B. C. (2002) The apoptotic ligands TRAIL, TWEAK, and Fas ligand mediate monocyte death induced by autologous lupus T cells. J Immunol 169, 6020-6029 18. Nakayama, M., Kayagaki, N., Yamaguchi, N., Okumura, K., Yagita, H. (2000) Involvement of TWEAK in interferon gamma-stimulated monocyte cytotoxicity. J Exp Med 192, 1373-1380 19. Campbell, S., Burkly, L. C., Gao, H. X., Berman, J. W., Su, L., Browning, B., Zheng, T., Schiffer, L., Michaelson, J. S., Putterman, C. (2006) Proinflammatory effects of TWEAK/Fn14 interactions in glomerular mesangial cells. J Immunol 176, 1889-1898. doi:176/3/1889 [pii]. 20. Gao, H. X., Campbell, S. R., Burkly, L. C., Jakubowski, A., Jarchum, I., Banas, B., Saleem, M. A., Mathieson, P. W., Berman, J. W., Michaelson, J. S., Putterman, C. (2009) TNF-like weak inducer of apoptosis (TWEAK) induces inflammatory and proliferative effects in human kidney cells. Cytokine 46, 24-35. doi:S1043-4666(08)00799-0 [pii];10.1016/j.cyto.2008.12.001 [doi]. 21. Sanchez-Nino, M. D., Poveda, J., Sanz, A. B., Mezzano, S., Carrasco, S., Fernandez-Fernandez, B., Burkly, L. C., Nair, V., Kretzler, M., Hodgin, J. B., Ruiz-Ortega, M., Selgas, R., Egido, J., Ortiz, A. (2013) Fn14 in podocytes and proteinuric kidney disease. Biochim.Biophys Acta 1832, 2232-2243. doi:S0925-4439(13)00275-5 [pii];10.1016/j.bbadis.2013.08.010 [doi] 22. Sanz, A. B., Justo, P., Sanchez-Nino, M. D., Blanco-Colio, L. M., Winkles, J. A., Kreztler, M., Jakubowski, A., Blanco, J., Egido, J., Ruiz-Ortega, M., Ortiz, A. (2008) The cytokine TWEAK

10

501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526 527 528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554 555 556 557 558 559 560

modulates renal tubulointerstitial inflammation. J Am doi:ASN.2007050577 [pii];10.1681/ASN.2007050577 [doi].

Soc

Nephrol

19,

695-703.

23. Linkermann, A., Brasen, J. H., Darding, M., Jin, M. K., Sanz, A. B., Heller, J. O., De, Z. F., Weinlich, R., Ortiz, A., Walczak, H., Weinberg, J. M., Green, D. R., Kunzendorf, U., Krautwald, S. (2013) Two independent pathways of regulated necrosis mediate ischemia-reperfusion injury. Proc Natl Acad Sci U S A 110, 12024-12029. doi:1305538110 [pii];10.1073/pnas.1305538110 [doi]. 24. Ucero, A. C., Benito-Martin, A., Fuentes-Calvo, I., Santamaria, B., Blanco, J., Lopez-Novoa, J. M., Ruiz-Ortega, M., Egido, J., Burkly, L. C., Martinez-Salgado, C., Ortiz, A. (2013) TNFrelated weak inducer of apoptosis (TWEAK) promotes kidney fibrosis and Ras-dependent proliferation of cultured renal fibroblast. Biochim.Biophys Acta 1832, 1744-1755. doi:S09254439(13)00203-2 [pii];10.1016/j.bbadis.2013.05.032 [doi]. 25. Sanz, A. B., Sanchez-Nino, M. D., Izquierdo, M. C., Jakubowski, A., Justo, P., Blanco-Colio, L. M., Ruiz-Ortega, M., Selgas, R., Egido, J., Ortiz, A. (2010) TWEAK activates the non-canonical NFkappaB pathway in murine renal tubular cells: modulation of CCL21. PLoS One 5, e8955. doi:10.1371/journal.pone.0008955 [doi]. 26. Hotta, K., Sho, M., Yamato, I., Shimada, K., Harada, H., Akahori, T., Nakamura, S., Konishi, N., Yagita, H., Nonomura, K., Nakajima, Y. (2011) Direct targeting of fibroblast growth factorinducible 14 protein protects against renal ischemia reperfusion injury. Kidney Int 79, 179-188. doi:ki2010379 [pii];10.1038/ki.2010.379 [doi]. 27. Xia, Y., Campbell, S. R., Broder, A., Herlitz, L., Abadi, M., Wu, P., Michaelson, J. S., Burkly, L. C., Putterman, C. (2012) Inhibition of the TWEAK/Fn14 pathway attenuates renal disease in nephrotoxic serum nephritis. Clin Immunol 145, 108-121. doi:S1521-6616(12)00211-2 [pii];10.1016/j.clim.2012.08.008 [doi]. 28. Izquierdo, M. C., Sanz, A. B., Mezzano, S., Blanco, J., Carrasco, S., Sanchez-Nino, M. D., Benito-Martin, A., Ruiz-Ortega, M., Egido, J., Ortiz, A. (2012) TWEAK (tumor necrosis factorlike weak inducer of apoptosis) activates CXCL16 expression during renal tubulointerstitial inflammation. Kidney Int 81, 1098-1107. doi:ki2011475 [pii];10.1038/ki.2011.475 [doi]. 29. Lu, J., Szeto, C. C., Tam, L. S., Lai, F. M., Li, E. K., Chow, K. M., Li, P. K., Kwan, B. C. (2012) Relationship of intrarenal gene expression and the histological class of lupus nephritis -- a study on repeat renal biopsy. J Rheumatol. 39, 1942-1947. doi:jrheum.120177 [pii];10.3899/jrheum.120177 [doi]. 30. Lu, J., Kwan, B. C., Lai, F. M., Choi, P. C., Tam, L. S., Li, E. K., Chow, K. M., Wang, G., Li, P. K., Szeto, C. C. (2011) Gene expression of TWEAK/Fn14 and IP-10/CXCR3 in glomerulus and tubulointerstitium of patients with lupus nephritis. Nephrology.(Carlton.) 16, 426-432. doi:10.1111/j.1440-1797.2011.01449.x [doi]. 31. Schwartz, N., Su, L., Burkly, L. C., Mackay, M., Aranow, C., Kollaros, M., Michaelson, J. S., Rovin, B., Putterman, C. (2006) Urinary TWEAK and the activity of lupus nephritis. J Autoimmun. 27, 242-250. doi:S0896-8411(06)00116-8 [pii];10.1016/j.jaut.2006.12.003 [doi] 32. Schwartz, N., Rubinstein, T., Burkly, L. C., Collins, C. E., Blanco, I., Su, L., Hojaili, B., Mackay, M., Aranow, C., Stohl, W., Rovin, B. H., Michaelson, J. S., Putterman, C. (2009) Urinary TWEAK as a biomarker of lupus nephritis: a multicenter cohort study. Arthritis Res Ther. 11, R143. doi:ar2816 [pii];10.1186/ar2816 [doi]. 33. Xuejing, Z., Jiazhen, T., Jun, L., Xiangqing, X., Shuguang, Y., Fuyou, L. (2012) Urinary TWEAK level as a marker of lupus nephritis activity in 46 cases. J Biomed.Biotechnol. 2012, 359647. doi:10.1155/2012/359647 [doi] 34. El-Shehaby, A., Darweesh, H., El-Khatib, M., Momtaz, M., Marzouk, S., El-Shaarawy, N., Emad, Y. (2011) Correlations of urinary biomarkers, TNF-like weak inducer of apoptosis

11

561 562 563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582 583 584 585 586 587 588 589 590 591 592 593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 617 618 619

(TWEAK), osteoprotegerin (OPG), monocyte chemoattractant protein-1 (MCP-1), and IL-8 with lupus nephritis. J Clin Immunol 31, 848-856. doi:10.1007/s10875-011-9555-1 [doi]. 35. Moreno, J. A., Izquierdo, M. C., Sanchez-Nino, M. D., Suarez-Alvarez, B., Lopez-Larrea, C., Jakubowski, A., Blanco, J., Ramirez, R., Selgas, R., Ruiz-Ortega, M., Egido, J., Ortiz, A., Sanz, A. B. (2011) The inflammatory cytokines TWEAK and TNFalpha reduce renal klotho expression through NFkappaB. J Am Soc Nephrol 22, 1315-1325. doi:ASN.2010101073 [pii];10.1681/ASN.2010101073 [doi]. 36. Zhao, Z., Burkly, L. C., Campbell, S., Schwartz, N., Molano, A., Choudhury, A., Eisenberg, R. A., Michaelson, J. S., Putterman, C. (2007) TWEAK/Fn14 interactions are instrumental in the pathogenesis of nephritis in the chronic graft-versus-host model of systemic lupus erythematosus. J Immunol 179, 7949-7958. doi:179/11/7949 [pii]. 37. Munoz-Garcia, B., Moreno, J. A., Lopez-Franco, O., Sanz, A. B., Martin-Ventura, J. L., Blanco, J., Jakubowski, A., Burkly, L. C., Ortiz, A., Egido, J., Blanco-Colio, L. M. (2009) Tumor necrosis factor-like weak inducer of apoptosis (TWEAK) enhances vascular and renal damage induced by hyperlipidemic diet in ApoE-knockout mice. Arterioscler.Thromb.Vasc.Biol 29, 2061-2068. doi:ATVBAHA.109.194852 [pii];10.1161/ATVBAHA.109.194852 [doi]. 38. Jakubowski, A., Ambrose, C., Parr, M., Lincecum, J. M., Wang, M. Z., Zheng, T. S., Browning, B., Michaelson, J. S., Baetscher, M., Wang, B., Bissell, D. M., Burkly, L. C. (2005) TWEAK induces liver progenitor cell proliferation. J Clin Invest 115, 2330-2340. doi:10.1172/JCI23486 [doi]. 39. Girgenrath, M., Weng, S., Kostek, C. A., Browning, B., Wang, M., Brown, S. A., Winkles, J. A., Michaelson, J. S., Allaire, N., Schneider, P., Scott, M. L., Hsu, Y. M., Yagita, H., Flavell, R. A., Miller, J. B., Burkly, L. C., Zheng, T. S. (2006) TWEAK, via its receptor Fn14, is a novel regulator of mesenchymal progenitor cells and skeletal muscle regeneration. EMBO J 25, 58265839. doi:7601441 [pii];10.1038/sj.emboj.7601441 [doi]. 40. Sanz, A. B., Sanchez-Nino, M. D., Ramos, A. M., Moreno, J. A., Santamaria, B., Ruiz-Ortega, M., Egido, J., Ortiz, A. (2010) NF-kappaB in renal inflammation. J Am Soc Nephrol 21, 12541262. doi:ASN.2010020218 [pii];10.1681/ASN.2010020218 [doi]. 41. Moynagh, P. N. (2005) The NF-kappaB pathway. J Cell Sci 118, 4589-4592. doi:118/20/4589 [pii];10.1242/jcs.02579 [doi]. 42. Hoffmann, A., Natoli, G., Ghosh, G. (2006) Transcriptional regulation via the NF-kappaB signaling module. Oncogene 25, 6706-6716. doi:1209933 [pii];10.1038/sj.onc.1209933 [doi]. 43. Hayden, M. S., Ghosh, S. (2008) Shared principles in NF-kappaB signaling. Cell 132, 344-362. doi:10.1101/gad.1228704 [doi];18/18/2195 [pii]. 44. Perkins, N. D. (2007) Integrating cell-signalling pathways with NF-kappaB and IKK function. Nat Rev Mol Cell Biol 8, 49-62. doi:nrm2083 [pii];10.1038/nrm2083 [doi]. 45. Hayden, M. S., Ghosh, S. (2004) Signaling to NF-kappaB. Genes Dev. 18, 2195-2224. doi:10.1101/gad.1228704 [doi];18/18/2195 [pii]. 46. Karin, M., Ben-Neriah, Y. (2000) Phosphorylation meets ubiquitination: the control of NF[kappa]B activity. Annu.Rev Immunol 18, 621-663. doi:18/1/621 [pii];10.1146/annurev.immunol.18.1.621 [doi]. 47. Senftleben, U., Cao, Y., Xiao, G., Greten, F. R., Krahn, G., Bonizzi, G., Chen, Y., Hu, Y., Fong, A., Sun, S. C., Karin, M. (2001) Activation by IKKalpha of a second, evolutionary conserved, NF-kappa B signaling pathway. Science 293, 1495-1499. doi:10.1126/science.1062677 [doi];293/5534/1495 [pii].

12

620 621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637 638 639 640 641 642 643 644 645 646 647 648 649 650 651 652 653 654 655 656 657 658 659 660 661 662 663 664 665 666 667 668 669 670 671 672 673 674 675 676 677 678 679

48. Karin, M. (1999) How NF-kappaB is activated: the role of the IkappaB kinase (IKK) complex. Oncogene 18, 6867-6874. doi:10.1038/sj.onc.1203219 [doi]. 49. Tergaonkar, V. (2006) NFkappaB pathway: a good signaling paradigm and therapeutic target. Int J Biochem Cell Biol 38, 1647-1653. doi:S1357-2725(06)00124-5 [pii];10.1016/j.biocel.2006.03.023 [doi]. 50. Gilmore, T. D. (2006) Introduction to NF-kappaB: players, pathways, perspectives. Oncogene 25, 6680-6684. doi:1209954 [pii];10.1038/sj.onc.1209954 [doi]. 51. Scheidereit, C. (2006) IkappaB kinase complexes: gateways to NF-kappaB activation and transcription. Oncogene 25, 6685-6705. doi:1209934 [pii];10.1038/sj.onc.1209934 [doi] 52. Xiao, G., Harhaj, E. W., Sun, S. C. (2001) NF-kappaB-inducing kinase regulates the processing of NF-kappaB2 p100. Mol Cell 7, 401-409. doi:S1097-2765(01)00187-3 [pii]. 53. Xiao, G., Fong, A., Sun, S. C. (2004) Induction of p100 processing by NF-kappaB-inducing kinase involves docking IkappaB kinase alpha (IKKalpha) to p100 and IKKalpha-mediated phosphorylation. J Biol Chem 279, 30099-30105. doi:10.1074/jbc.M401428200 [doi];M401428200 [pii]. 54. Dejardin, E. (2006) The alternative NF-kappaB pathway from biochemistry to biology: pitfalls and promises for future drug development. Biochem Pharmacol 72, 1161-1179. doi:S00062952(06)00500-4 [pii];10.1016/j.bcp.2006.08.007 [doi]. 55. Dejardin, E., Droin, N. M., Delhase, M., Haas, E., Cao, Y., Makris, C., Li, Z. W., Karin, M., Ware, C. F., Green, D. R. (2002) The lymphotoxin-beta receptor induces different patterns of gene expression via two NF-kappaB pathways. Immunity. 17, 525-535. doi:S1074761302004235 [pii]. 56. Coope, H. J., Atkinson, P. G., Huhse, B., Belich, M., Janzen, J., Holman, M. J., Klaus, G. G., Johnston, L. H., Ley, S. C. (2002) CD40 regulates the processing of NF-kappaB2 p100 to p52. EMBO J 21, 5375-5385 57. Claudio, E., Brown, K., Park, S., Wang, H., Siebenlist, U. (2002) BAFF-induced NEMOindependent processing of NF-kappa B2 in maturing B cells. Nat Immunol 3, 958-965. doi:10.1038/ni842 [doi];ni842 [pii]. 58. Kayagaki, N., Yan, M., Seshasayee, D., Wang, H., Lee, W., French, D. M., Grewal, I. S., Cochran, A. G., Gordon, N. C., Yin, J., Starovasnik, M. A., Dixit, V. M. (2002) BAFF/BLyS receptor 3 binds the B cell survival factor BAFF ligand through a discrete surface loop and promotes processing of NF-kappaB2. Immunity. 17, 515-524. doi:S1074761302004259 [pii]. 59. Novack, D. V., Yin, L., Hagen-Stapleton, A., Schreiber, R. D., Goeddel, D. V., Ross, F. P., Teitelbaum, S. L. (2003) The IkappaB function of NF-kappaB2 p100 controls stimulated osteoclastogenesis. J Exp Med 198, 771-781. doi:10.1084/jem.20030116 [doi];jem.20030116 [pii]. 60. Saitoh, T., Nakayama, M., Nakano, H., Yagita, H., Yamamoto, N., Yamaoka, S. (2003) TWEAK induces NF-kappaB2 p100 processing and long lasting NF-kappaB activation. J Biol Chem 278, 36005-36012. doi:10.1074/jbc.M304266200 [doi];M304266200 [pii]. 61. Rowe, A. M., Murray, S. E., Raue, H. P., Koguchi, Y., Slifka, M. K., Parker, D. C. (2013) A Cell-Intrinsic Requirement for NF-kappaB-Inducing Kinase in CD4 and CD8 T Cell Memory. J Immunol 191, 3663-3672. doi:jimmunol.1301328 [pii];10.4049/jimmunol.1301328 [doi]. 62. Hofmann, J., Mair, F., Greter, M., Schmidt-Supprian, M., Becher, B. (2011) NIK signaling in dendritic cells but not in T cells is required for the development of effector T cells and cellmediated immune responses. J Exp Med 208, 1917-1929. doi:jem.20110128 [pii];10.1084/jem.20110128 [doi].

13

680 681 682 683 684 685 686 687 688 689 690 691 692 693 694 695 696 697 698 699 700 701 702 703 704 705 706 707 708 709 710 711 712 713 714 715 716 717 718 719 720 721 722 723 724 725 726 727 728 729 730 731 732 733 734 735 736 737 738 739

63. Sheng, L., Zhou, Y., Chen, Z., Ren, D., Cho, K. W., Jiang, L., Shen, H., Sasaki, Y., Rui, L. (2012) NF-kappaB-inducing kinase (NIK) promotes hyperglycemia and glucose intolerance in obesity by augmenting glucagon action. Nat Med 18, 943-949. doi:nm.2756 [pii];10.1038/nm.2756 [doi]. 64. Choudhary, S., Sinha, S., Zhao, Y., Banerjee, S., Sathyanarayana, P., Shahani, S., Sherman, V., Tilton, R. G., Bajaj, M. (2011) NF-kappaB-inducing kinase (NIK) mediates skeletal muscle insulin resistance: blockade by adiponectin. Endocrinology 152, 3622-3627. doi:en.2011-1343 [pii];10.1210/en.2011-1343 [doi]. 65. Hacker, H., Chi, L., Rehg, J. E., Redecke, V. (2012) NIK prevents the development of hypereosinophilic syndrome-like disease in mice independent of IKKalpha activation. J Immunol 188, 4602-4610. doi:jimmunol.1200021 [pii];10.4049/jimmunol.1200021 [doi]. 66. Thu, Y. M., Su, Y., Yang, J., Splittgerber, R., Na, S., Boyd, A., Mosse, C., Simons, C., Richmond, A. (2012) NF-kappaB inducing kinase (NIK) modulates melanoma tumorigenesis by regulating expression of pro-survival factors through the beta-catenin pathway. Oncogene 31, 2580-2592. doi:onc2011427 [pii];10.1038/onc.2011.427 [doi]. 67. Chung, S., Sundar, I. K., Hwang, J. W., Yull, F. E., Blackwell, T. S., Kinnula, V. L., Bulger, M., Yao, H., Rahman, I. (2011) NF-kappaB inducing kinase, NIK mediates cigarette smoke/TNFalpha-induced histone acetylation and inflammation through differential activation of IKKs. PLoS One 6, e23488. doi:10.1371/journal.pone.0023488 [doi];PONE-D-11-06056 [pii]. 68. Cheung, H. H., St, J. M., Beug, S. T., Lejmi-Mrad, R., LaCasse, E., Baird, S. D., Stojdl, D. F., Screaton, R. A., Korneluk, R. G. (2011) SMG1 and NIK regulate apoptosis induced by Smac mimetic compounds. Cell Death.Dis 2, e146. doi:cddis201125 [pii];10.1038/cddis.2011.25 [doi]. 69. Starkey, J. M., Haidacher, S. J., LeJeune, W. S., Zhang, X., Tieu, B. C., Choudhary, S., Brasier, A. R., Denner, L. A., Tilton, R. G. (2006) Diabetes-induced activation of canonical and noncanonical nuclear factor-kappaB pathways in renal cortex. Diabetes 55, 1252-1259. doi:55/5/1252 [pii]. 70. Zhao, Y., Banerjee, S., LeJeune, W. S., Choudhary, S., Tilton, R. G. (2011) NF-kappaBinducing kinase increases renal tubule epithelial inflammation associated with diabetes. Exp Diabetes Res 2011, 192564. doi:10.1155/2011/192564 [doi]. 71. Loverre, A., Ditonno, P., Crovace, A., Gesualdo, L., Ranieri, E., Pontrelli, P., Stallone, G., Infante, B., Schena, A., Di, P. S., Capobianco, C., Ursi, M., Palazzo, S., Battaglia, M., Selvaggi, F. P., Schena, F. P., Grandaliano, G. (2004) Ischemia-reperfusion induces glomerular and tubular activation of proinflammatory and antiapoptotic pathways: differential modulation by rapamycin. J Am Soc Nephrol 15, 2675-2686. doi:15/10/2675 [pii];10.1097/01.ASN.0000139932.00971.E4 [doi]. 72. Feng, B., Chen, G., Zheng, X., Sun, H., Zhang, X., Zhang, Z. X., Xiang, Y., Ichim, T. E., Garcia, B., Luke, P., Jevnikar, A. M., Min, W. P. (2009) Small interfering RNA targeting RelB protects against renal ischemia-reperfusion injury. Transplantation 87, 1283-1289. doi:10.1097/TP.0b013e3181a1905e [doi];00007890-200905150-00005 [pii]. 73. Benedetti, G., Fokkelman, M., Yan, K., Fredriksson, L., Herpers, B., Meerman, J., van de Water, B., de, G. M. (2013) The nuclear factor kappaB family member RelB facilitates apoptosis of renal epithelial cells caused by cisplatin/tumor necrosis factor alpha synergy by suppressing an epithelial to mesenchymal transition-like phenotypic switch. Mol Pharmacol 84, 128-138. doi:mol.112.084053 [pii];10.1124/mol.112.084053 [doi]. 74. Sun, C. Y., Hsu, H. H., Wu, M. S. (2013) p-Cresol sulfate and indoxyl sulfate induce similar cellular inflammatory gene expressions in cultured proximal renal tubular cells. Nephrol Dial Transplant 28, 70-78. doi:gfs133 [pii];10.1093/ndt/gfs133 [doi].

14

740 741 742 743 744 745 746 747 748 749 750 751 752 753 754 755 756 757 758 759 760 761 762

75. Poveda, J., Sanchez-Nino, M. D., Glorieux, G., Sanz, A. B., Egido, J., Vanholder, R., Ortiz, A. (2013) p-Cresyl sulphate has pro-inflammatory and cytotoxic actions on human proximal tubular epithelial cells. Nephrol.Dial.Transplant. 76. Hehner, S. P., Hofmann, T. G., Droge, W., Schmitz, M. L. (1999) The antiinflammatory sesquiterpene lactone parthenolide inhibits NF-kappa B by targeting the I kappa B kinase complex. J Immunol 163, 5617-5623. doi:ji_v163n10p5617 [pii] 77. Sakai, N., Wada, T., Yokoyama, H., Lipp, M., Ueha, S., Matsushima, K., Kaneko, S. (2006) Secondary lymphoid tissue chemokine (SLC/CCL21)/CCR7 signaling regulates fibrocytes in renal fibrosis. Proc Natl Acad Sci U S A 103, 14098-14103. doi:0511200103 [pii];10.1073/pnas.0511200103 [doi] 78. Carragher, D., Johal, R., Button, A., White, A., Eliopoulos, A., Jenkinson, E., Anderson, G., Caamano, J. (2004) A stroma-derived defect in NF-kappaB2-/- mice causes impaired lymph node development and lymphocyte recruitment. J Immunol 173, 2271-2279. doi:173/4/2271 [pii] 79. Li, K., McGee, L. R., Fisher, B., Sudom, A., Liu, J., Rubenstein, S. M., Anwer, M. K., Cushing, T. D., Shin, Y., Ayres, M., Lee, F., Eksterowicz, J., Faulder, P., Waszkowycz, B., Plotnikova, O., Farrelly, E., Xiao, S. H., Chen, G., Wang, Z. (2013) Inhibiting NF-kappaB-inducing kinase (NIK): discovery, structure-based design, synthesis, structure-activity relationship, and cocrystal structures. Bioorg.Med.Chem.Lett. 23, 1238-1244. doi: 10.1016/j.bmcl.2013.01.012

15

Figure 1.TIFF

Copyright of Frontiers in Immunology is the property of Frontiers Media S.A. and its content may not be copied or emailed to multiple sites or posted to a listserv without the copyright holder's express written permission. However, users may print, download, or email articles for individual use.

Fn14 and Non-Canonical NF-kappaB Signaling in Kidney Disease.

The incidence of acute kidney injury (AKI) and chronic kidney disease (CKD) is increasing. However, there is no effective therapy for AKI and current ...
447KB Sizes 0 Downloads 0 Views