Está en la página 1de 12

Am J Physiol Renal Physiol 304: F1231–F1242, 2013.

First published March 20, 2013; doi:10.1152/ajprenal.00557.2012. Review

Tumor necrosis factor-␣: regulation of renal function and blood pressure


Vanesa D. Ramseyer1,2 and Jeffrey L. Garvin1,2,3
1
Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan;
2
Department of Physiology, School of Medicine, Wayne State University, Detroit, Michigan; and 3Department of Physiology
and Biophysics, Case Western Reserve University School of Medicine, Cleveland, Ohio
Submitted 27 September 2012; accepted in final form 15 March 2013

Ramseyer VD, Garvin JL. Tumor necrosis factor-␣: regulation of renal


function and blood pressure. Am J Physiol Renal Physiol 304: F1231–F1242, 2013.
First published March 20, 2013; doi:10.1152/ajprenal.00557.2012.—Tumor necro-
sis factor-␣ (TNF-␣) is a pleiotropic cytokine that becomes elevated in chronic
inflammatory states such as hypertension and diabetes and has been found to
mediate both increases and decreases in blood pressure. High levels of TNF-␣
decrease blood pressure, whereas moderate increases in TNF-␣ have been associ-
ated with increased NaCl retention and hypertension. The explanation for these
disparate effects is not clear but could simply be due to different concentrations of
TNF-␣ within the kidney, the physiological status of the subject, or the type of
stimulus initiating the inflammatory response. TNF-␣ alters renal hemodynamics
and nephron transport, affecting both activity and expression of transporters. It also
mediates organ damage by stimulating immune cell infiltration and cell death. Here
we will summarize the available findings and attempt to provide plausible expla-
nations for such discrepancies.
TNF-␣; hypertension; natriuresis; TNF receptor; septic shock; transport; renal
hemodynamics; blood pressure

Tumor Necrosis Factor-␣ in the Kidney: Synthesis TNF-␣ production is augmented in proximal tubule cells in
and Secretion response to IL-1␣ but not IL-2 or IFN␥ (161). This increase is
inhibited by cycloheximide, suggesting that it is not due to
TNF-␣ IS A PROINFLAMMATORY cytokine that was originally
increased TNF shedding from the membrane but rather en-
described as antitumorigenic (19, 60) and produced by immune hanced protein synthesis.
cells like macrophages and lymphocytes (15, 105, 142); how- In the thick ascending limb, CaSR activation stimulates Gi
ever, further studies revealed it is also produced by endothelial and Gq protein (3); this activates phosphatidylinositol-specific
and epithelial cells (14, 43, 61, 66, 71, 98, 99, 101, 141, 160). phospholipase C, which in turn produces inositol triphosphate
TNF-␣ is expressed as a 26-kDa plasma membrane protein that and diacylglycerol. Inositol triphosphate increases intracellular
is secreted into the extracellular space by the metalloproteinase Ca, which forms a complex with calmodulin whereas diacyl-
TNF-␣-converting enzyme (TACE, also called a disintegrin glycerol activates protein kinase C (PKC). These two conver-
and metalloproteinase 17 or ADAM 17; Refs. 16, 76, 89). In gent cascades lead to activation of calcineurin. Activated
solution, this 17-kDa protein forms homotrimers and activates calcineurin dephosphorylates nuclear factor of activated T cells
two distinct receptors (151). Transmembrane TNF-␣ also (NFAT), allowing translocation of this transcription factor to
forms trimers (136) and can activate TNF receptors. Moreover, the nucleus and transcription of TNF-␣ (1, 3, 156). In partic-
the intracellular domain of transmembrane TNF is palmitoy- ular, NFAT5 (also called Ton/EBP for tonicity element binding
lated (148) and can be phosphorylated (111), providing an protein) has been shown to partially mediate the CaSR-induced
additional step for regulation of TNF-␣ release. increase in TNF-␣ expression (53) and is itself activated by
Infiltrating inflammatory cells (142), podocytes (119), mes- changes in osmolality in renal cells (95); however, it is not
angial cells (6, 13, 14, 74), and epithelial cells from proximal known whether changes in osmolality induce TNF-␣ release by
tubules (101, 113, 163, 164), thick ascending limbs (3, 35, 90, thick ascending limbs nor whether this TNF-␣ release is
156) and collecting ducts (140) are sources of TNF-␣ in the mediated via NFAT5.
kidney. TNF-␣ increases in response to endotoxemia (26), TNF-␣ levels have been shown to increase in response to
lipopolysaccharide (LPS; Refs. 34, 40, 74, 90), angiotensin II ANG II in thick ascending limbs (35) and podocytes (119). In
(ANG II; Ref. 35), calcium-sensing receptor (CaSR) activation podocytes, activation of both AT1 and AT2 receptors was
(3, 156), hypertension (32, 79), renal failure (115), glomeru- responsible for this effect, with AT1 mediating the earlier
lonephritis (153), diabetic nephropathy (42, 100), and intersti- phase and AT2 the later phase. ANG II activates PKC in the
tial tubular nephritis (50, 94). thick ascending limb (57, 128) and thus may increase NFAT
translocation to the nucleus along with TNF-␣ gene transcrip-
Address for reprint requests and other correspondence: V. D. Ramseyer,
tion; however, it is still unclear whether PKC and NFAT
Hypertension and Vascular Research Division, Henry Ford Hospital, 2799 West mediate ANG II-induced increases in TNF-␣ expression in the
Grand Boulevard, Detroit, MI 48202-2689 (e-mail: vramsey1@hfhs.org). thick ascending limb. In addition, TNF-␣ production is known
http://www.ajprenal.org 1931-857X/13 Copyright © 2013 the American Physiological Society F1231
Downloaded from www.physiology.org/journal/ajprenal by ${individualUser.givenNames} ${individualUser.surname} (115.178.202.039) on December 25, 2017.
Copyright © 2013 American Physiological Society. All rights reserved.
Review
F1232 TNF AND RENAL FUNCTION

to increase in response to LPS in thick ascending limbs and ANG II is elevated (under conditions like low-salt diet, renal
podocytes, particularly during endotoxemia. vascular hypertension, etc.), then receptor expression would be
Finally, TNF-␣ production in the kidney can be increased by elevated as well. Such data suggest that the cellular pattern of
infiltrating immune cells, especially macrophages (105, 142, TNFR expression in the kidney could affect its response to
169). Increased innate immune activation, neutrophil and mac- TNF-␣ depending on its initial physiological/pathophysiolog-
rophage infiltration, and enhanced TNF-␣ production are ob- ical state.
served in infections (83), cisplatin nephropathy (73, 114),
diabetic nephropaty (38), antiglomerular basement membrane TNF-␣ and Blood Pressure
nephropathy (142), obstructive nephropathy (134), etc. In such
situations stimuli for TNF-␣ production include activation of The role of TNF-␣ in blood pressure regulation has been
Toll-like receptor 4 in response to LPS (85), oxidative stress studied in several different models, with sometimes conflicting
(30), antibody deposition, and complement activation (142, results. There are a number of reports showing that TNF-␣
159). In addition, dendritic cell, macrophage, and lymphocyte either mediates or supports increases in blood pressure. In
recruitment to the kidney is observed in lupus erythematosus ANG II-infused hypertensive rats fed high-salt diet, etanercept
and hypertension (27, 51, 96). The stimuli for their migration delayed progression of hypertension. Protection appeared to be
to the kidney and TNF-␣ production are likely increased due to reduced renal damage, since proteinuria and monocyte/
reactive oxygen species and enhanced expression of endothe- macrophage infiltration were diminished (33). In addition,
lial adhesion molecules and chemokines by the kidney in infusion of ANG II for 2 wk failed to increase blood pressure
response to elevations in renal-derived TNF-␣. Inhibition of in TNF-␣ knockout (KO) mice, but this was restored by replace-
AT1 receptors (7, 121) as well as reactive oxygen scavenging ment therapy with recombinant TNF-␣ (0.3 ␮g·kg⫺1·day⫺1; Ref.
(30) reduces renal-derived TNF-␣, proinflammatory cytokine 131). In addition, Guzik et al. (51) showed that TNF-␣ pro-
production, and immune cell infiltration and activation. Simi- duction by T lymphocytes from ANG II-hypertensive mice was
larly, ANG II infusion increases leukocyte infiltration and higher than in control mice and that etanercept infusion blunted
production of inflammatory cytokines, all of which are pre- ANG II-induced increases in blood pressure. These data seem
vented by blockade of TNF-␣ with etanercept (33, 96). These to indicate that TNF-␣ mediates ANG II-dependent increases
data indicate that endotoxins, ANG II, and reactive oxygen in blood pressure. Similarly, in a mouse model of lupus
species stimulate TNF-␣ expression by the kidney; this in turn erythematosus etanercept infusion for 4 wk reduced blood
induces inflammatory cell recruitment and activation, which pressure, albuminuria, glomerulosclerosis, and NF-␬B activity,
further increase TNF-␣ levels. suggesting that TNF-␣ causes inflammation and mediates the
increase in blood pressure (152).
TNF-␣ Receptors Metabolic syndrome is characterized by the simultaneous
presence of obesity, dyslipidemia, insulin resistance, and hy-
The actions of TNF-␣ are mediated by two distinct recep- pertension. It has been shown that elevated TNF-␣ levels are
tors: type 1 (TNFR1 or p55) and type 2 (TNFR2 or p75). associated with the development of hypertension (63, 170).
Kinetically, TNF-␣ binding to TNFR1 shows slow dissocia- Etanercept treatment for 6 wk prevented the increase in blood
tion, whereas it is 20 –30 times faster with TNFR2 (48). pressure observed in insulin-resistant fructose-fed rats without
Therefore, some have proposed that TNFR2 serves as a “local affecting insulin sensitivity (147). TNF-␣ neutralization also
reservoir” of TNF-␣, both providing readily available TNF-␣ restored maximum acetylcholine-induced relaxation in mesen-
for TNFR1 activation and operating as a “ligand passing” teric arteries and restored nitric oxide synthase 3 (NOS3)
receptor in which TNFR2 captures TNF-␣ and passes it on to expression (147).
TNFR1. In addition, it has been shown that TNFR1 activation TNF-␣ also appears to play a role in blood pressure regula-
requires expression of TNFR2, which can form heterocom- tion during pregnancy. In preeclampsia, plasma TNF-␣ is
plexes with TNFR1 (28, 87, 107). Although the effects of doubled (10, 25, 122), and infusing TNF-␣ for 5 days (⬍0.5
soluble TNF-␣ may be mediated by either receptor, most of the ␮g·kg⫺1·day⫺1) increases blood pressure in pregnant rats but
actions of membrane-bound TNF-␣ are mediated by TNFR2 not in virgin rats (80, 81). Blockade of TNF-␣ with etanercept
(47). TNFRs form homotrimers in the plasma membrane via reduced blood pressure. In addition, healthy endothelial cells
their preligand assembly domain (22). TNFRs do not have an incubated with serum from preeclamptic rats exhibited in-
intrinsic catalytic property; rather, ligand binding causes a creased endothelin-1 release and this was prevented by etan-
conformational change in the preassembled receptor that leads ercept (79). Similarly, infusion of a neutralizing anti-TNF
to recruitment of signaling proteins. antibody reduced blood pressure, albuminuria, and renal dam-
In healthy animals, TNFR1 and 2 have been found in the age in a model of preeclampsia achieved by adoptive transfer
cortex in proximal tubules and collecting ducts as well as of serum from preeclamptic women to pregnant mice (62).
endothelial cells of the glomeruli and renal vasculature (8, 20). Finally, TNF-␣ is increased is chronic kidney disease, which
TNFR1 has also been found in vascular smooth muscle cells of is characterized by progressive loss of renal function, renal
the renal vasculature (20). However, TNFR expression varies injury, and hypertension (130, 139, 162). This disease can be
in different diseases. In glomerulonephritic kidneys, TNFR2 mimicked by 5/6 nephrectomy, which also increases TNF-␣
expression was high in glomeruli and postcapillary venules of release. In rats with renal failure, neutralizing TNF-␣ with
the cortex, whereas in obstructive nephropathy (which primar- soluble TNFR1 prevented the increase in blood pressure,
ily affects tubules) TNFR2 staining was restricted to tubular reduced fibrosis, decreased macrophage infiltration and albu-
epithelial cells (153). ANG II also increases TNFR1 and 2 minuria, and restored the ratio of L-arginine to asymmetric
expression in podocytes (119). Thus one would expect that if dimethylarginine (139). It also restored expression of NOS3,

AJP-Renal Physiol • doi:10.1152/ajprenal.00557.2012 • www.ajprenal.org


Downloaded from www.physiology.org/journal/ajprenal by ${individualUser.givenNames} ${individualUser.surname} (115.178.202.039) on December 25, 2017.
Copyright © 2013 American Physiological Society. All rights reserved.
Review
TNF AND RENAL FUNCTION F1233
endothelin-1, transforming growth factor-␤ macrophage che- In contrast to the link between TNF-␣ and hypertension,
moattractant protein, intercellular adhesion molecules, and TNF-␣ also appears to play a role in the hypotension associated
vascular cell adhesion molecules (139). with septic shock (145). This is a complex pathological con-
However, there are also a number of reports indicating that dition that results from an exaggerated and deregulated host
TNF-␣ either plays no role or tends to reduce blood pressure. response to infection with extreme increases in TNF-␣ levels.
For example, in DOCA-salt hypertensive rats etanercept did Activation of the innate immune response with increased
not reduce blood pressure (32). Although it remains unclear neutrophil and macrophage tissue infiltration is observed.
why TNF-␣ blockade failed to afford protection in these rats, However, attempts to reduce the immune response by blocking
it could be due to the high systolic blood pressure, or it could the action of TNF or inhibiting downstream mediators have
simply be that TNF-␣ does not participate in this model of provided conflicting results. Indeed, in animals pretreatment
hypertension. Supporting this notion is the fact that the DOCA- with TNF-␣ blockers like neutralizing antibodies (37, 146) or
salt model of hypertension is based on elevated mineralocor- soluble receptor fusion constructs (150) before the onset of
ticoid levels, and the increase in blood pressure can be pre- sepsis has shown a positive outcome with a lower mortality
vented with mineralocorticoid receptor antagonists (44, 149). rate; however, in humans numerous trials have failed to dem-
In addition, Muller et al. (96) showed that in a double- onstrate a beneficial effect of anti-TNF therapy in septic
transgenic rat model of ANG II-dependent hypertension, in patients (4, 41, 112). Failure of this approach is partly attrib-
which human angiotensinogen and renin are expressed, etan- uted to the need of a competent immune system to withstand
ercept did not alter blood pressure even though it reduced the systemic infection and partly to the fact that in humans
albuminuria and infiltration by inflammatory cells. The dispar- treatment is started after sepsis has been initiated. TNF-␣
ity with the previous report by Elmarakby et al. (33) in which
decreases blood pressure in septic shock by numerous mech-
TNF-␣ blockade delayed the increase in blood pressure in
anisms, including reductions in cardiac contractility (77, 165),
ANG II-induced hypertension could be explained by the fol-
increased endothelial permeability (5, 97), exaggerated NO
lowing facts: 1) Muller et al. started anti-TNF treatment after
production (72), resistance to catecholamines (17, 158), in-
rats were hypertensive whereas Elmarakby et al. started ANG
II and etanercept at the same time; 2) the blood pressure in the creased sodium excretion (126), and decreases in vasopressin
double-transgenic rats was much higher than in those infused receptors and aquaporin 2 (58).
with ANG II; 3) Muller et al. only measured blood pressure at Why TNF-␣ is implicated as a causative agent of both
day 20 whereas Elmarakby et al. found that the blood pressure- increases and decreases in blood pressure is not clear. When
lowering effect of etanercept took place from days 6 to 12 after considered in toto, most evidence seems to support a role for
the start of the anti-TNF treatment; and 4) Muller et al. used chronic moderately elevated concentrations of TNF-␣ in hy-
regular salt diet while Elmarakby et al. fed the rats high-salt pertension, as the opposing reports have alternative explana-
diet. tions. This view is also supported by the fact that nearly all
Using a different anti-TNF treatment, Ferreri et al. (36) studies show that neutralizing TNF-␣ protects against renal
showed that in ANG II-induced hypertension a neutralizing damage, which in itself is a cause of hypertension. In contrast,
anti-TNF antibody induced an acute increase in blood pressure higher concentrations of TNF-␣ are associated with decreases
after 30 min of infusion. The explanation for this discrepancy in blood pressure and more severe inflammation. In these cases
is unclear but is likely the duration of the study. In reports the decrease in blood pressure is likely dependent on both renal
where TNF-␣ was found to mediate increases in blood and nonrenal systemic actions of TNF-␣ (Fig. 1).
pressure, animals were studied for weeks rather than hours
or days. These investigators later showed that infusion of
1.6 ␮g·kg⫺1·min⫺1 ANG II raised blood pressure higher in
TNFR1 KO mice than in wild-type (WT) at 2, 4, and 5 days of
180
ANG II infusion. Proteinuria, water intake, glomerular filtra-
re

↑ macrophage infiltration
tion rate (GFR), and urinary output were also increased
blood pressur

↑ neutrophile infiltration
whereas Na excretion was reduced, suggesting increased renal ↓ ↓ GFR
Na reabsorption. These results are in stark contrast to those ↓ ↓ RBF
p

120 ↓ Renal NaCl reabsorption


mmHg))

shown in TNF-␣ KO mice (131) where the increase in blood ↑ macrophage infiltration ↓ proteinuria
pressure induced by infusion of 1 ␮g·kg⫺1·min⫺1 ANG II was ↑ lymphocyte activation
↓ NOS3 and NO
Renal failure
(m

blunted compared with WT mice and to those shown by Guzik


ystolic b

↑ Renal NaCl reabsorption


et al. (51) where etanercept reduced the increase in blood 60
↑ renal damage
↑ proteinuria
pressure induced by infusion of ANG II at a rate of 490
ng·kg⫺1·min⫺. While the explanation is not clear, it could be
Sy

due to the different doses of ANG II used (and hence possibly


different mechanisms leading to hypertension), due to the
1 2 5 10
background of the mice (C57Bc/6J vs. B6129SF2/J), or be-
TNF in plasma (fold increase from normal levels)
cause infusion of ANG II in TNFR1 KO mice increases both
TNF-␣ and TNFR2 (24). This later point raises the possibility Fig. 1. Diagram representing the effects of increasing levels of TNF-␣ in renal
that the observed phenotype is the result of hyperactive TNFR2 function and the hypothetical effect on blood pressure. Increases of 1- to 2-fold in
TNF-␣ are present in hypertension whereas increases of TNF-␣ ⬎5 times are
as opposed to a lack of TNFR1 and TNFR2 has been shown to observed in septic shock. Dashed line represents a theoretical relationship between
mediate renal macrophage infiltration, glomerulosclerosis, and TNF-␣ levels and blood pressure. NO, nitric oxide; NOS3, nitric oxide synthase
interstitial fibrosis in ANG II-induced hypertension (129). type 3; GFR, glomerular filtration rate; RBF, renal blood flow.

AJP-Renal Physiol • doi:10.1152/ajprenal.00557.2012 • www.ajprenal.org


Downloaded from www.physiology.org/journal/ajprenal by ${individualUser.givenNames} ${individualUser.surname} (115.178.202.039) on December 25, 2017.
Copyright © 2013 American Physiological Society. All rights reserved.
Review
F1234 TNF AND RENAL FUNCTION

Renal Hemodynamics Na, Water, Urea, and Glucose Transport


At high doses (ⱖ0.6 mg/kg), infusion of TNF-␣ in healthy In spite of reductions in GFR and RBF in models of sepsis
rats lowers blood pressure. This is accompanied by lower GFR and during acute TNF-␣ infusion, urinary Na excretion in-
and renal blood flow (RBF); however, renal vascular resistance creases. This indicates that TNF-␣ directly affects Na absorp-
(RVR) increases (20, 127). The decreases in GFR and RBF can tion along the nephron transport pathway independently of
be explained by the drop in blood pressure, which was shown changes in renal hemodynamics. The effects of TNF-␣ have
to be due to an increase in nitric oxide synthase type 2 (NOS2; been studied in proximal tubular cells, thick ascending limbs,
Refs. 72, 91, 117), but this cannot be the entire explanation as distal tubular cells, and collecting ducts.
RVR increases. Elevated RVR has been related to increases in In LLCPk1 cells, a model of proximal tubule cells, TNF-␣
superoxide production (127), and it may be due to TNF-␣ increases paracellular permeability, thereby activating extra-
acting differently on renal vessels than those of the general cellular signal-regulated kinases (ERK), which results in phos-
circulation or on nephron segments that alter RVR such as the phorylation of the guanidine exchange factor GEF-H1. Phos-
macula densa or connecting tubule. Increased RVR and re- phorylated GEF-H1 enhances RhoA GTPase, which binds and
duced RBF and GFR were observed in both WT and TNFR2 activates Rho kinase and increases myosin light chain phos-
KO mice but absent in TNFR1 KO, suggesting that the re- phorylation, due in part to Rho kinase-induced inhibition of
sponse was due to activation of TNFR1. Activation of TNFR2 myosin light chain phosphatase and in part to direct activation
(TNF-␣ treatment in TNFR1 KO mice) dilated blood vessels. of myosin light chain kinase. The increase in paracellular
Thus the effect of TNF-␣ on renal hemodynamics in healthy permeability is preceded by increased cofilin phosphorylation
animals is mediated primarily by TNFR1 (20). and formation of actin stress fibers (69, 70). Whether the
increase in paracellular permeability increases or decreases net
Na and fluid absorption is unclear from these reports, as they
Renin-Angiotensin System were not measured directly and arguments can be made either
TNF-␣ decreases renin promoter activity and expression in way. However, given that TNF-␣ reduces expression of renal
primary cultures of juxtaglomerular (JG) cells and in As4.1 Na-K-ATPase (126), which ultimately drives all Na absorption in
cells (a cell line that constitutively releases renin). In As4.1 the proximal nephron, it is likely that the combined actions of
cells, TNF-␣ (0.1 to 100 ng/ml) reduces renin release after 30 TNF-␣ on proximal nephron transport are inhibitory (Fig. 2A).
h of treatment. Moreover, TNF-␣ KO mice show a threefold TNF-␣ also enhances NO formation in proximal tubular
increase in renal renin mRNA content but not changes in cells due to increased inducible NO synthase (iNOS or NOS2;
plasma renin activity. These data suggest that TNF-␣ nega- Refs. 68, 91). Although one report indicated that NO stimu-
tively regulates renin expression via a reduction in mRNA lated proximal tubule transport (157), most have shown that it
levels due to declining transcription (144). inhibits Na and fluid absorption in this segment (31, 118). Thus
Todorov et al. (144) showed that TNF-␣ actions on renin NO-induced inhibition of proximal reabsorption could account
for at least part of the increase in urinary Na excretion seen
expression were dependent on NF-␬B binding to a cAMP-
when TNF-␣ is infused. Whether the TNF-␣-induced increase
responsive element (CRE) located on the renin promoter.
in paracellular permeability in proximal tubules depends on
Binding of the proteins NF-␬B-p50, p-65, p-52, and c-Rel to
NO remains unclear.
CRE lowered transcription. The transcriptional coactivator
In the thick ascending limb, ANG II and CaSR increase
CBP, the protein that binds the transcription factor CRE
TNF-␣ release (3, 35, 156). In this nephron segment, both
binding protein (CREB), interacts with NF-␬B and CREB/ATF endogenous and exogenous TNF-␣ reduce NaCl transport via
transcription factors to initiate transcription; however, silenc- activation of cyclooxygenase (COX2)-induced increases in
ing CBP did not affect TNF-induced decreases in renin mRNA prostaglandin E (PGE) and inhibition of the Na-K-2Cl cotrans-
or promoter activity. Therefore, the authors concluded the porter (NKCC2; Refs. 2, 3, 12, 34). Increased TNF-␣ sup-
effect most likely is not due to NF-␬B competing with CREB/ presses Rb uptake, a measure of thick ascending limb ion
ATF for CBP but rather that NF-␬B represses transcription transport. In addition, activation of CaSR, which increases
after binding to CRE. This conclusion is supported by the TNF-␣ release, has also been shown to increase COX2 expres-
ability of c-Rel to act as a transcriptional repressor (92). sion and reduce ouabain-sensitive oxygen consumption in the
Although TNF-␣ decreases renin content, it does not reduce renin thick ascending limb (2, 155), which may explain how CaSR
release by primary cultures of JG cells, and plasma renin activity activation leads to natriuresis.
is not significantly increased in TNF-␣ KO mice. Thus the acute About 80% of transcellular Na reabsorption in the thick
effects of TNF-␣ on blood pressure are not likely due to changes ascending limb occurs through NKCC2. There are three iso-
in plasma renin activity (144). forms of this transporter: A, B, and F. A is found throughout
Similar to its effect on renin levels, TNF-␣ was shown to the thick ascending limb, B mainly in the cortex, and F
reduce angiotensinogen levels in human kidney 2 (HK-2) cells, primarily in the medulla. The level of expression in the kidney
a renal proximal tubular cell line (123). Since intrarenal ang- is 10B:20A:70F, and the order of ion affinity is NKCC2B ⬎
iotensinogen levels correlate with blood pressure and are in- NKCC2A ⬎ NKCC2F (21). In TNF-␣ KO mice, expression of
dependent of plasma levels (45), in theory TNF-␣ could lower the A isoform was upregulated and this was reversed by
blood pressure by reducing angiotensinogen expression. How- infusion of TNF-␣ (0.01 mg·kg⫺1·day⫺1), whereas the other
ever, to the best of our knowledge neither intrarenal nor isoforms did not change; in addition, bumetanide-sensitive O2
intratubular levels of angiotensinogen have been measured in consumption (an indicator of NKCC2 activity) was increased
TNF-␣ KO mice nor has the receptor involved been identified. in TNF-␣ KO mice and was also restored by TNF-␣ infusion

AJP-Renal Physiol • doi:10.1152/ajprenal.00557.2012 • www.ajprenal.org


Downloaded from www.physiology.org/journal/ajprenal by ${individualUser.givenNames} ${individualUser.surname} (115.178.202.039) on December 25, 2017.
Copyright © 2013 American Physiological Society. All rights reserved.
Review
TNF AND RENAL FUNCTION F1235

Fig. 2. Cartoon showing the effects of TNF-␣ on ion transport in


vitro in epithelial cells of the kidney. Effects of TNF-␣ in proximal
tubular cells (A), in thick ascending limb cells (B), and in collecting
duct cells (C). Red boxes and arrows represent pathways activated
by TNF-␣ that are likely to increase Na reabsorption. Green boxes
and arrows represent pathways activated by TNF-␣ that are likely
to inhibit Na reabsorption. TNFR, TNF-␣ receptor; NHE3, Na/H
exchanger type 3; NOS2, nitric oxide synthase type 2; ERK,
extracellular signal-regulated kinases; Pi, phosphate; pGEF-H1,
phosphorylated guanine nucleotide exchange factor-H1; ROCK,
Rho-dependent kinase; MLCK, myosin light chain kinase; pMLC,
phosphorylated myosin light chain; SF, actin stress fibers; NKCC2,
Na-K-2Cl cotransporter; COX2, cyclooxygenase type 2; PGE2,
prostaglandin type 2; CaSR, calcium-sensitive receptor; ANG II,
angiotensin II; ENaC, epithelial Na channel; PKA, cAMP-depen-
dent protein kinase; PI3K, phosphatidylinositol 3 kinase; SMase,
sphingomyelinase; PKC, protein kinase C.

AJP-Renal Physiol • doi:10.1152/ajprenal.00557.2012 • www.ajprenal.org


Downloaded from www.physiology.org/journal/ajprenal by ${individualUser.givenNames} ${individualUser.surname} (115.178.202.039) on December 25, 2017.
Copyright © 2013 American Physiological Society. All rights reserved.
Review
F1236 TNF AND RENAL FUNCTION

(12). In addition to reducing NKCC2, TNF-␣ reduces expres- but V2 receptor and aquaporin 2 expression were decreased,
sion of thick ascending limb K channels (126), which would whereas a siRNA against the TNFR p50 subunit attenuated the
also be expected to reduce Na absorption. Finally, TNF-␣ decrease in these proteins (58). Thus TNF-␣ may reduce
reduces NOS3 expression and NO production in this nephron vasopressin-dependent water retention due to both inhibition of
segment (116). NO produced in response to arginine (103, 110) water transport and reductions in the osmotic gradient developed
endothelin-1 (109, 56) and clonidine (108) reduces NaCl re- by the thick ascending limb, which drives water absorption.
absorption mainly by decreasing NKCC2 activity by the thick Maintenance of urinary osmolality depends not only on
ascending limb. These data suggest that TNF-␣ affects Na water and Na but also in large measure on urea. Blous infusion
transport regulation by the thick ascending limb by reducing of TNF-␣ at a dose of 1 mg/kg decreased fractional urea
basal Na reabsorption while blunting the ability of other excretion, inner medulla urea concentration and osmolality,
natriuretic autacoids and hormones to further decrease trans- tubular urea reabsorption, and urinary urea, whereas it
port. Given that thick ascending limbs absorb 30% of the increased plasma osmolality and urea. Such findings were
filtered Na load, it is likely that direct actions of TNF-␣ in this attributed to reduced urea transporters UTA1, UTA2, UT-A3,
segment contribute to elevated Na excretion (Fig. 2B). UT-A4, and UT-B (124).
In Madin-Darby canine kidney cells, a canine model of In addition to affecting Na and fluid absorption, TNF-␣ may
distal tubular cells, TNF-␣ increases paracellular permeability diminish glucose absorption in the proximal nephron. Injection
by activating RhoA GTPase, Rho kinase, and myosin light of TNF-␣ (1 mg/kg, bolus) decreases expression of sodium
chain kinase, similar to LLCPk1 cells (69, 70). The net effect glucose transporters SGLT2 and SGLT3 and Na-K-ATPase
of this activity on transport is unknown. within 12 h; however, it also increases Glut1 and SGLT1 in the
In mpkCCDcL4 cells, a murine collecting duct cell line, 10 kidney. The increase in Glut1 and SGLT1 may occur as a
ng/ml TNF-␣ increased protein kinase A (PKA) activity inde- compensatory response to the reduced intracellular glucose
pendently of cAMP. LPS and TNF-␣ both increased transep- levels resulting from 1) decreased glucose entry due to reduc-
ithelial Na transport, measured as an increase in amiloride-
tions in SGLT2 and 3, and 2) the lower Na gradient due to
sensitive short-circuit current, and this effect depended on
reduced Na-K-ATPase (125).
PKA activation (154). In contrast, in Xenopus distal nephron
cells (A6), 100 ng/ml TNF-␣ inhibited endothelial Na channel
(ENaC) activity by reducing the channel open probability; Renal Injury
however, this effect was only seen when phosphatidylinositol Even though inhibition of transport could be interpreted as
3-kinase (PI3K) was inhibited. TNF-␣ inhibited ENaC via a protecting the kidney from injury, the majority of the literature
mechanism that involved activation of sphingomyelinase and indicates that TNF-␣ induces renal damage. In part, this is
increased ceramide and activation of PKC, which in turn undoubtedly due to diminished renal perfusion such as seen in
induced externalization of phosphatidylserine. The presence of sepsis (84, 127). It is also due in part to recruitment of immune
phosphatidylserine in the cytosolic leaflet of the apical mem- cells into the kidney, releasing cytokines that cause inflamma-
brane is sufficient to maintain ENaC activity; thus disappear-
tion and cell death (169). Finally, it is also likely due to direct
ance of this phospholipid from the inner leaflet was likely the
actions of TNF-␣ on renal cells (65, 102).
cause of reduced ENaC activity (11). The role of PI3K in
TNFR1 contains a death domain, and thus the proapoptotic
preventing this effect was attributed to its ability to inhibit
actions of TNF-␣ have been mainly attributed to this receptor
sphingomyelinase activity (18). These disparate findings could
(138, 64). TNFR1 interacts with the adaptor protein TNF recep-
be explained by differences in the concentration of TNF-␣
tested, the experimental conditions, or the origin of the respec- tor-associated death domain (TRADD) through TNFR1-DD (59),
tive cells. For example, the A6 cells were grown with 1 ␮g leading to recruitment of TNF receptor-associated factor 2
aldosterone whereas the mpkCCD cells were not, and aldoste- (TRAF2; Ref. 133). TRAF2 then interacts with the E3 ubiquitin
rone activates PI3K (55). Whether or not other pertinent ligases “cellular inhibitor of apoptosis” (c-IAP) 1 and 2 to form
hormones are also present in the collecting duct and ultimately the membrane-bound complex I (93). c-IAP polyubiquitinates
determine whether TNF-␣ increases or decreases ENaC activ- chains of RIP1, which is essential for recruitment of transforming
ity could be the key to understanding its opposing effects on growth factor-activated kinase 1 (TAK1) and activation of inhib-
tubular transport and blood pressure. When infused in vivo, itor of kappa B (I␬B) kinase (I␬K; Ref. 29). I␬K phosphorylates
TNF-␣ reduces expression of all three subunits of ENaC (126); I␬B, which is then degraded via the ubiquitin-proteasome path-
thus its actions on the cortical collecting duct likely also way, allowing NF-␬B to translocate to the nucleus where it
contribute to the natriuresis caused by systemic infusion (Fig. initiates gene transcription targeting anti-apoptotic genes like
2C). Finally, TNF-␣ increases nitrate/nitrite formation in inner c-FLIP, c-IAP-1 and 2, and TRAF1 and 2. Additionally, associ-
medullary collecting ducts, suggesting elevated NO production ation of RIP1 and TRAF-2 with TNFR1 can lead to activation of
that was supported by increased iNOS (68, 91) and NO has mitogen-activated protein kinase kinase kinase 1 (MEKKK-1)
been reported to inhibit Na reabsorption by this segment (132). and c-Jun NH2-terminal kinases (JNK), which phosphorylate and
TNF-␣ KO mice showed increased ambient urinary osmo- activate c-Jun, a member of the transcription factor complex
lality compared with WT, but water deprivation for 24 h activator protein 1. Prolonged JNK activation mediates protea-
increased urine osmolality to the same extent in both strains somal degradation of c-FLIP and activation of caspase-8, leading
(12). LPS, which potently induces TNF-␣ release, decreases to apoptosis (23, 135). TNF-␣ also activates apoptosis signaling
arginine-vasopressin type 2 receptor levels and aquaporin 2 kinase-1 (ASK1) via TNFR1 by inducing ASK1-interacting pro-
expression in the renal inner medulla (49). In support of this, in tein-1, which promotes ASK1 dissociation from the adaptor pro-
a model of sepsis plasma arginine vasopressin did not change tein 14 –3-3, phosphorylation at the stimulatory site threonine 845

AJP-Renal Physiol • doi:10.1152/ajprenal.00557.2012 • www.ajprenal.org


Downloaded from www.physiology.org/journal/ajprenal by ${individualUser.givenNames} ${individualUser.surname} (115.178.202.039) on December 25, 2017.
Copyright © 2013 American Physiological Society. All rights reserved.
Review
TNF AND RENAL FUNCTION F1237
(ASK1pThr845), and dephosphorylation of ASK1 at the inhibi- Concluding Remarks
tory site serine 967 (86, 143, 166, 167).
In the normal kidney, strong staining for phospho-serine 967 The discrepancy between blood pressure-lowering effects and
ASK1 (ASK1pser967, the inhibitory site) colocalized with prohypertensive actions of TNF-␣ is still a matter for debate but
TNFR1 in glomerular and endothelial cells from peritubular is likely the result of many variables. For example, the TNF-␣
capillaries, whereas coexpression of TNFR1 and ASK1pThr845 levels in plasma, the type of immune response (innate vs. adap-
(the stimulatory site) was not found. In contrast, in renal tive), and the extent of the response (moderate vs. exaggerated)
allografts with acute cellular rejection, active ASK1pThr845 can determine whether blood pressure is increased or a shock
response takes place. In general, plasma TNF-␣ levels in endo-
was strong in endothelial cells from glomerular and peritubular
toxemic shock increase rapidly by 10-fold or more, whereas in
capillaries and in some tubular epithelial cells, whereas
hypertension or heart failure TNF-␣ levels are increased 1- to
ASK1pSer967 was not detected. Similarly, in renal allografts
2-fold. Similarly, animal models mimicking sepsis are infused
with acute tubular necrosis, ASK1pSer967 was diminished but
with a bolus of 0.01 to 1 mg/kg TNF-␣, whereas the doses used
some tubular epithelial cells expressed a strong signal for
to induce hypertension in pregnant rats or TNF-␣ KO mice treated
ASK1pThre845. This pattern of expression was also found in
with ANG II are 0.5 ␮g/kg or lower and are maintained for days.
normal kidney tissue treated with a TNFR1-specific activator;
This may mean that at high doses TNF-␣ has nonspecific effects
however, when a TNFR2-specific activator was used, both
or activates signaling cascades leading to hypotension that sur-
TNFR1 and ASK1pSer967 were elevated (9).
passes its hypertensive actions. Alternatively, the nonreceptor-
In contrast to TNFR1, activation of TNFR2 is associated mediated actions of TNF-␣, which are due to the presence of a
with both apoptosis and cell survival. TNFR2 interacts with lectin-like domain in human TNF-␣, could be activated at high
TRAF1 and 2, leading to RIP/c-IAP-induced activation of doses. This domain, although far removed from the receptor
NF-␬B (78, 120). Therefore, TNFR2 has been mainly associ- binding site, nevertheless recognizes oligosaccharides and medi-
ated with cell proliferation and survival (137). However, stud- ates some TNF-␣ actions like trypanolytic activity (88). In addi-
ies have shown a proapoptotic or cytotoxic function of TNFR2 tion, sudden and exaggerated activation of the innate immune
activation (54, 67), and various models including TNFR2- response, with massive increases in macrophage and neutrophil
mediated activation of TNFR1 (107), cytosolic depletion of infiltration, seems to result in a shock state and leads to hypoten-
c-IAP (39), and TNFR2-induced downregulation of NF-␬B sion. In contrast, more moderate and chronic activation of the
(46) have been proposed. A proapoptotic function for TNFR2 adaptive immune response with participation of macrophages and
has been posited based on the fact that both TNFR2 and the lymphocytes contributes to slowly developing renal damage, in-
complement molecule C3 were upregulated in glomerulonephritic creased NaCl retention, and hypertension.
kidneys and this was significantly reduced in TNFR2 KO mice, One of the problems in dissecting the prohypertensive vs.
suggesting that TNFR2 can activate complement (153). TNFR2 prohypotensive actions of TNF-␣ is that the approaches uti-
also activates endothelial/epithelial tyrosine kinase (Etk), which lized to block TNF-␣ differ in their side effects. The use of
has been linked to regulation of epithelial cell junctions (52), chimeric proteins as opposed to antibodies to block the actions
whereas in endothelial cells it is involved in TNF-induced angio- of TNF or the use of a KO mouse model could explain why
genic events (104) and mediates activation of PI3K and Akt (168). TNF-␣ seems to be protective in some studies but detrimental
TNFR2-induced activation of Etk is TRAF2 independent. Bind- in others. Depending on which portion of the TNFR is con-
ing of TNF-␣ to TNFR2 induces Etk unfolding, phosphorylation, served during anti-TNF treatment, it may or may not induce
and activation (104). In normal kidneys, TNFR2 was confined to complement deposition and a cytotoxic reaction. In addition,
cells in the glomeruli and interstitium, with a strong signal for Etk while KO models provide a promising tool to study the role of
in glomerular endothelial cells. In renal allografts with acute certain genes in disease, they may also generate misleading
cellular rejection or acute tubular necrosis, a strong signal for results. Impaired development of germinal centers and follic-
TNFR2 and Etkp was seen in most tubular epithelial cells. Similar ular dendritic cell networks in secondary lymphoid organs has
observations were made when kidney tissue cultures were treated been observed in TNF-␣ and TNFR1 KO mice (75, 82, 106).
with a TNFR2-specific activator. Incubating kidney tissue with Moreover, overexpression of TNFR1 in TNFR2 KO mice and
this activator also increased proliferation of tubular epithelial cells elevated TNFR2 in TNFR1 KO has been shown: thus a
expressing Etkp, and this effect was more pronounced than when seemingly protective role of TNFR1 that is absent in TNFR1
a TNFR1-specific activator was used (9). KO might be simply due to overexpression of TNFR2. Simi-
Finally, circulating levels of TNFR play an important role in larly, a detrimental response in TNF-␣ or TNFR1 KO could be
determining the severity of certain conditions, and therefore, attributable to their inability to handle infections, not the
TNFR levels should be reported. In this regard, both Gohda et absence of the direct effects of TNF on ion transport or blood
al. (42) and Niewczas et al. (100) reported a strong positive pressure. Finally, membrane-bound TNF effects, which have
correlation between circulating TNFR1 and TNFR2 with pro- been very poorly explored, could also confuse results.
gression to chronic kidney disease in type 1 diabetes and Overall, blockade of TNF-␣ during hypertension seems to
end-stage renal disease in type 2 diabetes. This correlation was provide a beneficial outcome, while handling the development
independent of other markers, including circulating TNF-␣, of autoimmune diseases in patients treated with TNF-neutral-
blood pressure, albuminuria, and GFR. These authors reported izing agents is not a trivial undertaking (75). On the other hand,
that plasma concentrations of TNFR1 and 2 were clinical the battle between lowering TNF-␣ levels in septic shock and
predictors of end-stage renal disease and chronic kidney dis- stimulating the immune system to fight the infection still
ease; however, they did not assess whether elevated circulating represents a clinical challenge. Therefore, the discovery of
TNFRs were the cause or the consequence of a dire outcome. specific mediators of TNF-induced increases and decreases in

AJP-Renal Physiol • doi:10.1152/ajprenal.00557.2012 • www.ajprenal.org


Downloaded from www.physiology.org/journal/ajprenal by ${individualUser.givenNames} ${individualUser.surname} (115.178.202.039) on December 25, 2017.
Copyright © 2013 American Physiological Society. All rights reserved.
Review
F1238 TNF AND RENAL FUNCTION

blood pressure would represent a great advance in the gener- 16. Black RA, Rauch CT, Kozlosky CJ, Peschon JJ, Slack JL, Wolfson
ation of effective alternate treatments for hypertension and MF, Castner BJ, Stocking KL, Reddy P, Srinivasan S, Nelson N,
Boiani N, Schooley KA, Gerhart M, Davis R, Fitzner JN, Johnson
shock-induced hypotension. RS, Paxton RJ, March CJ, Cerretti DP. A metalloproteinase disinteg-
rin that releases tumour-necrosis factor-alpha from cells. Nature 385:
GRANTS 729 –733, 1997.
This work was supported by grants from the National Institutes of Health 17. Bucher M, Kees F, Taeger K, Kurtz A. Cytokines down-regulate
(HL-028982, HL-070985, and HL-090550; to J. L. Garvin) and from the alpha1-adrenergic receptor expression during endotoxemia. Crit Care
American Heart Association (11PRE7510005; to V.D. Ramseyer). Med 31: 566 –571, 2003.
18. Burow ME, Weldon CB, Collins-Burow BM, Ramsey N, McKee A,
Klippel A, McLachlan JA, Clejan S, Beckman BS. Cross-talk between
DISCLOSURES phosphatidylinositol 3-kinase and sphingomyelinase pathways as a
No conflicts of interest, financial or otherwise, are declared by the author(s). mechanism for cell survival/death decisions. J Biol Chem 275: 9628 –
9635, 2000.
19. Carswell EA, Old LJ, Kassel RL, Green S, Fiore N, Williamson B. An
AUTHOR CONTRIBUTIONS
endotoxin-induced serum factor that causes necrosis of tumors. Proc Natl
Author contributions: V.R. prepared figures; V.R. drafted manuscript; V.R. Acad Sci USA 72: 3666 –3670, 1975.
and J.L.G. edited and revised manuscript; V.R. and J.L.G. approved final 20. Castillo A, Islam MT, Prieto MC, Majid DS. Tumor necrosis factor-
version of manuscript. alpha receptor type 1, not type 2, mediates its acute responses in the
kidney. Am J Physiol Renal Physiol 302: F1650 –F1657, 2012.
REFERENCES 21. Castrop H, Schnermann J. Isoforms of renal Na-K-2Cl cotransporter
NKCC2: expression and functional significance. Am J Physiol Renal
1. Abdullah HI, Pedraza PL, Hao S, Rodland KD, McGiff JC, Ferreri Physiol 295: F859 –F866, 2008.
NR. NFAT regulates calcium-sensing receptor-mediated TNF produc- 22. Chan FK, Chun HJ, Zheng L, Siegel RM, Bui KL, Lenardo MJ. A
tion. Am J Physiol Renal Physiol 290: F1110 –F1117, 2006. domain in TNF receptors that mediates ligand-independent receptor
2. Abdullah HI, Pedraza PL, McGiff JC, Ferreri NR. Calcium-sensing assembly and signaling. Science 288: 2351–2354, 2000.
receptor signaling pathways in medullary thick ascending limb cells 23. Chang L, Kamata H, Solinas G, Luo JL, Maeda S, Venuprasad K,
mediate COX-2-derived PGE2 production: functional significance. Am J Liu YC, Karin M. The E3 ubiquitin ligase itch couples JNK activation
Physiol Renal Physiol 295: F1082–F1089, 2008. to TNFalpha-induced cell death by inducing c-FLIP(L) turnover. Cell
3. Abdullah HI, Pedraza PL, McGiff JC, Ferreri NR. CaR activation 124: 601–613, 2006.
increases TNF production by mTAL cells via a Gi-dependent mecha- 24. Chen CC, Pedraza PL, Hao S, Stier CT, Ferreri NR. TNFR1-deficient
nism. Am J Physiol Renal Physiol 294: F345–F354, 2008. mice display altered blood pressure and renal responses to ANG II
4. Abraham E. Why immunomodulatory therapies have not worked in infusion. Am J Physiol Renal Physiol 299: F1141–F1150, 2010.
sepsis. Intensive Care Med 25: 556 –566, 1999. 25. Conrad KP, Miles TM, Benyo DF. Circulating levels of immunoreac-
5. Adembri C, Sgambati E, Vitali L, Selmi V, Margheri M, Tani A, tive cytokines in women with preeclampsia. Am J Reprod Immunol 40:
Bonaccini L, Nosi D, Caldini AL, Formigli L, De Gaudio AR. Sepsis 102–111, 1998.
induces albuminuria and alterations in the glomerular filtration barrier: a 26. Cunningham PN, Dyanov HM, Park P, Wang J, Newell KA, Quigg
morphofunctional study in the rat. Crit Care 15: R277, 2011. RJ. Acute renal failure in endotoxemia is caused by TNF acting directly
6. Affres H, Perez J, Hagege J, Fouqueray B, Kornprobst M, Ardaillou on TNF receptor-1 in kidney. J Immunol 168: 5817–5823, 2002.
R, Baud L. Desferrioxamine regulates tumor necrosis factor release in 27. De PG, Castellano G, Del PA, Sozzani S, Fiore N, Loverre A,
mesangial cells. Kidney Int 39: 822–830, 1991. Parmentier M, Gesualdo L, Grandaliano G, Schena FP. The possible
7. Aki K, Shimizu A, Masuda Y, Kuwahara N, Arai T, Ishikawa A, role of ChemR23/Chemerin axis in the recruitment of dendritic cells in
Fujita E, Mii A, Natori Y, Fukunaga Y, Fukuda Y. ANG II receptor lupus nephritis. Kidney Int 79: 1228 –1235, 2011.
blockade enhances anti-inflammatory macrophages in anti-glomerular 28. Declercq W, Denecker G, Fiers W, Vandenabeele P. Cooperation of
basement membrane glomerulonephritis. Am J Physiol Renal Physiol both TNF receptors in inducing apoptosis: involvement of the TNF
298: F870 –F882, 2010.
receptor-associated factor binding domain of the TNF receptor 75. J
8. Al-Lamki RS, Wang J, Skepper JN, Thiru S, Pober JS, Bradley JR.
Immunol 161: 390 –399, 1998.
Expression of tumor necrosis factor receptors in normal kidney and
29. Ea CK, Deng L, Xia ZP, Pineda G, Chen ZJ. Activation of IKK by
rejecting renal transplants. Lab Invest 81: 1503–1515, 2001.
TNFalpha requires site-specific ubiquitination of RIP1 and polyubiquitin
9. Al-Lamki RS, Wang J, Vandenabeele P, Bradley JA, Thiru S, Luo D,
binding by NEMO. Mol Cell 22: 245–257, 2006.
Min W, Pober JS, Bradley JR. TNFR1- and TNFR2-mediated signal-
30. Ebenezer PJ, Mariappan N, Elks CM, Haque M, Francis J. Diet-
ing pathways in human kidney are cell type-specific and differentially
contribute to renal injury. FASEB J 19: 1637–1645, 2005. induced renal changes in Zucker rats are ameliorated by the superoxide
10. Alexander BT, Cockrell KL, Massey MB, Bennett WA, Granger JP. dismutase mimetic TEMPOL. Obesity (Silver Spring) 17: 1994 –2002,
Tumor necrosis factor-alpha-induced hypertension in pregnant rats re- 2009.
sults in decreased renal neuronal nitric oxide synthase expression. Am J 31. Eitle E, Hiranyachattada S, Wang H, Harris PJ. Inhibition of proxi-
Hypertens 15: 170 –175, 2002. mal tubular fluid absorption by nitric oxide and atrial natriuretic peptide
11. Bao HF, Zhang ZR, Liang YY, Ma JJ, Eaton DC, Ma HP. Ceramide in rat kidney. Am J Physiol Cell Physiol 274: C1075–C1080, 1998.
mediates inhibition of the renal epithelial sodium channel by tumor 32. Elmarakby AA, Quigley JE, Imig JD, Pollock JS, Pollock DM.
necrosis factor-alpha through protein kinase C. Am J Physiol Renal TNF-alpha inhibition reduces renal injury in DOCA-salt hypertensive
Physiol 293: F1178 –F1186, 2007. rats. Am J Physiol Regul Integr Comp Physiol 294: R76 –R83, 2008.
12. Battula S, Hao S, Pedraza PL, Stier CT, Ferreri NR. Tumor necrosis 33. Elmarakby AA, Quigley JE, Pollock DM, Imig JD. Tumor necrosis
factor-alpha is an endogenous inhibitor of Na⫹-K⫹-2Cl⫺ cotransporter factor alpha blockade increases renal Cyp2c23 expression and slows the
(NKCC2) isoform A in the thick ascending limb. Am J Physiol Renal progression of renal damage in salt-sensitive hypertension. Hypertension
Physiol 301: F94 –F100, 2011. 47: 557–562, 2006.
13. Baud L, Fouqueray B, Philippe C, Amrani A. Tumor necrosis factor 34. Escalante BA, Ferreri NR, Dunn CE, McGiff JC. Cytokines affect ion
alpha and mesangial cells. Kidney Int 41: 600 –603, 1992. transport in primary cultured thick ascending limb of Henle’s loop cells.
14. Baud L, Oudinet JP, Bens M, Noe L, Peraldi MN, Rondeau E, Am J Physiol Cell Physiol 266: C1568 –C1576, 1994.
Etienne J, Ardaillou R. Production of tumor necrosis factor by rat 35. Ferreri NR, Escalante BA, Zhao Y, An SJ, McGiff JC. Angiotensin II
mesangial cells in response to bacterial lipopolysaccharide. Kidney Int induces TNF production by the thick ascending limb: functional impli-
35: 1111–1118, 1989. cations. Am J Physiol Renal Physiol 274: F148 –F155, 1998.
15. Beutler B, Greenwald D, Hulmes JD, Chang M, Pan YC, Mathison 36. Ferreri NR, Zhao Y, Takizawa H, McGiff JC. Tumor necrosis factor-
J, Ulevitch R, Cerami A. Identity of tumour necrosis factor and the alpha-angiotensin interactions and regulation of blood pressure. J Hy-
macrophage-secreted factor cachectin. Nature 316: 552–554, 1985. pertens 15: 1481–1484, 1997.

AJP-Renal Physiol • doi:10.1152/ajprenal.00557.2012 • www.ajprenal.org


Downloaded from www.physiology.org/journal/ajprenal by ${individualUser.givenNames} ${individualUser.surname} (115.178.202.039) on December 25, 2017.
Copyright © 2013 American Physiological Society. All rights reserved.
Review
TNF AND RENAL FUNCTION F1239
37. Fong Y, Tracey KJ, Moldawer LL, Hesse DG, Manogue KB, Kenney 58. Hocherl K, Schmidt C, Kurt B, Bucher M. Inhibition of NF-␬B
JS, Lee AT, Kuo GC, Allison AC, Lowry SF. Antibodies to cachectin/ ameliorates sepsis-induced downregulation of aquaporin-2/V2 receptor
tumor necrosis factor reduce interleukin 1 beta and interleukin 6 appear- expression and acute renal failure in vivo. Am J Physiol Renal Physiol
ance during lethal bacteremia. J Exp Med 170: 1627–1633, 1989. 298: F196 –F204, 2010.
38. Fornoni A, Ijaz A, Tejada T, Lenz O. Role of inflammation in diabetic 59. Hsu H, Xiong J, Goeddel DV. The TNF receptor 1-associated protein
nephropathy. Curr Diabetes Rev 4: 10 –17, 2008. TRADD signals cell death and NF-kappa B activation. Cell 81: 495–504,
39. Fotin-Mleczek M, Henkler F, Samel D, Reichwein M, Hausser A, 1995.
Parmryd I, Scheurich P, Schmid JA, Wajant H. Apoptotic crosstalk of 60. Ichinose Y, Tsao JY, Fidler IJ. Destruction of tumor cells by mono-
TNF receptors: TNF-R2-induces depletion of TRAF2 and IAP proteins kines released from activated human blood monocytes: evidence for
and accelerates TNF-R1-dependent activation of caspase-8. J Cell Sci parallel and additive effects of IL-1 and TNF. Cancer Immunol Immu-
115: 2757–2770, 2002. nother 27: 7–12, 1988.
40. Fouqueray B, Philippe C, Herbelin A, Perez J, Ardaillou R, Baud L. 61. Imaizumi T, Itaya H, Fujita K, Kudoh D, Kudoh S, Mori K,
Cytokine formation within rat glomeruli during experimental endotox- Fujimoto K, Matsumiya T, Yoshida H, Satoh K. Expression of tumor
emia. J Am Soc Nephrol 3: 1783–1791, 1993. necrosis factor-alpha in cultured human endothelial cells stimulated with
41. Giamarellos-Bourboulis EJ. Immunomodulatory therapies for sepsis: lipopolysaccharide or interleukin-1alpha. Arterioscler Thromb Vasc Biol
unexpected effects with macrolides. Int J Antimicrob Agents 32, Suppl 1: 20: 410 –415, 2000.
S39 –S43, 2008. 62. Irani RA, Zhang Y, Zhou CC, Blackwell SC, Hicks MJ, Ramin SM,
42. Gohda T, Niewczas MA, Ficociello LH, Walker WH, Skupien J,
Kellems RE, Xia Y. Autoantibody-mediated angiotensin receptor acti-
Rosetti F, Cullere X, Johnson AC, Crabtree G, Smiles AM, Mayadas
vation contributes to preeclampsia through tumor necrosis factor-alpha
TN, Warram JH, Krolewski AS. Circulating TNF receptors 1 and 2
signaling. Hypertension 55: 1246 –1253, 2010.
predict stage 3 CKD in type 1 diabetes. J Am Soc Nephrol 23: 516 –524,
63. Ito H, Ohshima A, Tsuzuki M, Ohto N, Takao K, Hijii C, Yanagawa
2012.
43. Gomez-Guerrero C, Lopez-Armada MJ, Gonzalez E, Egido J. Sol- M, Ogasawara M, Nishioka K. Association of serum tumour necrosis
uble IgA and IgG aggregates are catabolized by cultured rat mesangial factor-alpha with serum low-density lipoprotein-cholesterol and blood
cells and induce production of TNF-alpha and IL-6, and proliferation. J pressure in apparently healthy Japanese women. Clin Exp Pharmacol
Immunol 153: 5247–5255, 1994. Physiol 28: 188 –192, 2001.
44. Gomez-Sanchez EP, Zhou M, Gomez-Sanchez CE. Mineralocortico- 64. Itoh N, Nagata S. A novel protein domain required for apoptosis.
ids, salt and high blood pressure. Steroids 61: 184 –188, 1996. Mutational analysis of human Fas antigen. J Biol Chem 268: 10932–
45. Gonzalez-Villalobos RA, Seth DM, Satou R, Horton H, Ohashi N, 10937, 1993.
Miyata K, Katsurada A, Tran DV, Kobori H, Navar LG. Intrarenal 65. Izawa-Ishizawa Y, Ishizawa K, Sakurada T, Imanishi M, Miyamoto
angiotensin II and angiotensinogen augmentation in chronic angiotensin L, Fujii S, Taira H, Kihira Y, Ikeda Y, Hamano S, Tomita S,
II-infused mice. Am J Physiol Renal Physiol 295: F772–F779, 2008. Tsuchiya K, Tamaki T. Angiotensin II receptor blocker improves tumor
46. Grech AP, Gardam S, Chan T, Quinn R, Gonzales R, Basten A, necrosis factor-alpha-induced cytotoxicity via antioxidative effect in
Brink R. Tumor necrosis factor receptor 2 (TNFR2) signaling is nega- human glomerular endothelial cells. Pharmacology 90: 324 –331, 2012.
tively regulated by a novel, carboxyl-terminal TNFR-associated factor 2 66. Jevnikar AM, Brennan DC, Singer GG, Heng JE, Maslinski W,
(TRAF2)-binding site. J Biol Chem 280: 31572–31581, 2005. Wuthrich RP, Glimcher LH, Kelley VE. Stimulated kidney tubular
47. Grell M, Douni E, Wajant H, Lohden M, Clauss M, Maxeiner B, epithelial cells express membrane associated and secreted TNF alpha.
Georgopoulos S, Lesslauer W, Kollias G, Pfizenmaier K, Scheurich Kidney Int 40: 203–211, 1991.
P. The transmembrane form of tumor necrosis factor is the prime 67. Ji W, Li Y, Wan T, Wang J, Zhang H, Chen H, Min W. Both
activating ligand of the 80 kDa tumor necrosis factor receptor. Cell 83: internalization and aip1 association are required for tumor necrosis factor
793–802, 1995. receptor 2-mediated JNK signaling. Arterioscler Thromb Vasc Biol 32:
48. Grell M, Wajant H, Zimmermann G, Scheurich P. The type 1 receptor 2271–2279, 2012.
(CD120a) is the high-affinity receptor for soluble tumor necrosis factor. 68. Kabore AF, Denis M, Bergeron MG. Association of nitric oxide
Proc Natl Acad Sci USA 95: 570 –575, 1998. production by kidney proximal tubular cells in response to lipopolysac-
49. Grinevich V, Knepper MA, Verbalis J, Reyes I, Aguilera G. Acute charide and cytokines with cellular damage. Antimicrob Agents Che-
endotoxemia in rats induces down-regulation of V2 vasopressin receptors mother 41: 557–562, 1997.
and aquaporin-2 content in the kidney medulla. Kidney Int 65: 54 –62, 69. Kakiashvili E, Dan Q, Vandermeer M, Zhang Y, Waheed F, Pham
2004. M, Szaszi K. The epidermal growth factor receptor mediates tumor
50. Guo G, Morrissey J, McCracken R, Tolley T, Klahr S. Role of necrosis factor-alpha-induced activation of the ERK/GEF-H1/RhoA
TNFR1 and TNFR2 receptors in tubulointerstitial fibrosis of obstructive pathway in tubular epithelium. J Biol Chem 286: 9268 –9279, 2011.
nephropathy. Am J Physiol Renal Physiol 277: F766 –F772, 1999. 70. Kakiashvili E, Speight P, Waheed F, Seth R, Lodyga M, Tanimura S,
51. Guzik TJ, Hoch NE, Brown KA, McCann LA, Rahman A, Dikalov S, Kohno M, Rotstein OD, Kapus A, Szaszi K. GEF-H1 mediates tumor
Goronzy J, Weyand C, Harrison DG. Role of the T cell in the genesis necrosis factor-alpha-induced Rho activation and myosin phosphoryla-
of angiotensin II induced hypertension and vascular dysfunction. J Exp
tion: role in the regulation of tubular paracellular permeability. J Biol
Med 204: 2449 –2460, 2007.
Chem 284: 11454 –11466, 2009.
52. Hamm-Alvarez SF, Chang A, Wang Y, Jerdeva G, Lin HH, Kim KJ,
71. Karkar AM, Koshino Y, Cashman SJ, Dash AC, Bonnefoy J, Meager
Ann DK. Etk/Bmx activation modulates barrier function in epithelial
A, Rees AJ. Passive immunization against tumour necrosis factor-alpha
cells. Am J Physiol Cell Physiol 280: C1657–C1668, 2001.
53. Hao S, Zhao H, Darzynkiewicz Z, Battula S, Ferreri NR. Expression (TNF-alpha) and IL-1 beta protects from LPS enhancing glomerular
and function of NFAT5 in medullary thick ascending limb (mTAL) cells. injury in nephrotoxic nephritis in rats. Clin Exp Immunol 90: 312–318,
Am J Physiol Renal Physiol 296: F1494 –F1503, 2009. 1992.
54. Heller RA, Song K, Fan N, Chang DJ. The p70 tumor necrosis factor 72. Kilbourn RG, Gross SS, Jubran A, Adams J, Griffith OW, Levi R,
receptor mediates cytotoxicity. Cell 70: 47–56, 1992. Lodato RF. NG-methyl-L-arginine inhibits tumor necrosis factor-in-
55. Helms MN, Liu L, Liang YY, Al-Khalili O, Vandewalle A, Saxena S, duced hypotension: implications for the involvement of nitric oxide. Proc
Eaton DC, Ma HP. Phosphatidylinositol 3,4,5-trisphosphate mediates Natl Acad Sci USA 87: 3629 –3632, 1990.
aldosterone stimulation of epithelial sodium channel (ENaC) and inter- 73. Kim YK, Choi TR, Kwon CH, Kim JH, Woo JS, Jung JS. Beneficial
acts with gamma-ENaC. J Biol Chem 280: 40885–40891, 2005. effect of pentoxifylline on cisplatin-induced acute renal failure in rabbits.
56. Herrera M, Hong NJ, Ortiz PA, Garvin JL. Endothelin-1 inhibits thick Ren Fail 25: 909 –922, 2003.
ascending limb transport via Akt-stimulated nitric oxide production. J 74. Kita T, Tanaka N, Nagano T. The immunocytochemical localization of
Biol Chem 284: 1454 –1460, 2009. tumour necrosis factor and leukotriene in the rat kidney after treatment
57. Herrera M, Silva GB, Garvin JL. Angiotensin II stimulates thick with lipopolysaccharide. Int J Exp Pathol 74: 471–479, 1993.
ascending limb superoxide production via protein kinase C(alpha)-de- 75. Kollias G, Kontoyiannis D. Role of TNF/TNFR in autoimmunity:
pendent NADPH oxidase activation. J Biol Chem 285: 21323–21328, specific TNF receptor blockade may be advantageous to anti-TNF treat-
2010. ments. Cytokine Growth Factor Rev 13: 315–321, 2002.

AJP-Renal Physiol • doi:10.1152/ajprenal.00557.2012 • www.ajprenal.org


Downloaded from www.physiology.org/journal/ajprenal by ${individualUser.givenNames} ${individualUser.surname} (115.178.202.039) on December 25, 2017.
Copyright © 2013 American Physiological Society. All rights reserved.
Review
F1240 TNF AND RENAL FUNCTION

76. Kriegler M, Perez C, DeFay K, Albert I, Lu SD. A novel form of pressive treatment protects against angiotensin II-induced renal damage.
TNF/cachectin is a cell surface cytotoxic transmembrane protein: rami- Am J Pathol 161: 1679 –1693, 2002.
fications for the complex physiology of TNF. Cell 53: 45–53, 1988. 97. Naikawadi RP, Cheng N, Vogel SM, Qian F, Wu D, Malik AB, Ye
77. Kumar A, Paladugu B, Mensing J, Kumar A, Parrillo JE. Nitric RD. A critical role for phosphatidylinositol (3,4,5)-trisphosphate-depen-
oxide-dependent and -independent mechanisms are involved in TNF-␣- dent Rac exchanger 1 in endothelial junction disruption and vascular
induced depression of cardiac myocyte contractility. Am J Physiol Regul hyperpermeability. Circ Res 111: 1517–1527, 2012.
Integr Comp Physiol 292: R1900 –R1906, 2007. 98. Nakamura A, Johns EJ, Imaizumi A, Niimi R, Yanagawa Y, Koh-
78. Laegreid A, Medvedev A, Nonstad U, Bombara MP, Ranges G, saka T. Role of angiotensin II-induced cAMP in mesangial TNF-alpha
Sundan A, Espevik T. Tumor necrosis factor receptor p75 mediates production. Cytokine 19: 47–51, 2002.
cell-specific activation of nuclear factor kappa B and induction of human 99. Neale TJ, Ruger BM, Macaulay H, Dunbar PR, Hasan Q, Bourke A,
cytomegalovirus enhancer. J Biol Chem 269: 7785–7791, 1994. Murray-McIntosh RP, Kitching AR. Tumor necrosis factor-alpha is
79. LaMarca B, Speed J, Fournier L, Babcock SA, Berry H, Cockrell K, expressed by glomerular visceral epithelial cells in human membranous
Granger JP. Hypertension in response to chronic reductions in uterine nephropathy. Am J Pathol 146: 1444 –1454, 1995.
perfusion in pregnant rats: effect of tumor necrosis factor-alpha blockade. 100. Niewczas MA, Gohda T, Skupien J, Smiles AM, Walker WH, Rosetti
Hypertension 52: 1161–1167, 2008. F, Cullere X, Eckfeldt JH, Doria A, Mayadas TN, Warram JH,
80. LaMarca BB, Bennett WA, Alexander BT, Cockrell K, Granger JP. Krolewski AS. Circulating TNF receptors 1 and 2 predict ESRD in type
Hypertension produced by reductions in uterine perfusion in the pregnant 2 diabetes. J Am Soc Nephrol 23: 507–515, 2012.
rat: role of tumor necrosis factor-alpha. Hypertension 46: 1022–1025, 101. Noiri E, Kuwata S, Nosaka K, Tokunaga K, Juji T, Shibata Y,
2005. Kurokawa K. Tumor necrosis factor-alpha mRNA expression in lipopo-
81. LaMarca BB, Cockrell K, Sullivan E, Bennett W, Granger JP. Role lysaccharide-stimulated rat kidney. Chronological analysis of localiza-
of endothelin in mediating tumor necrosis factor-induced hypertension in tion. Am J Pathol 144: 1159 –1166, 1994.
pregnant rats. Hypertension 46: 82–86, 2005. 102. Ortiz A, Lorz C, Justo P, Catalan MP, Egido J. Contribution of
82. Le Hir M, Bluethmann H, Kosco-Vilbois MH, Müller M, di Padova apoptotic cell death to renal injury. J Cell Mol Med 5: 18 –32, 2001.
F, Moore M, Ryffel B, Eugster HP. Differentiation of follicular den- 103. Ortiz PA, Hong NJ, Wang D, Garvin JL. Gene transfer of eNOS to the
dritic cells and full antibody responses require tumor necrosis factor thick ascending limb of eNOS-KO mice restores the effects of L-arginine
receptor-1 signaling. J Exp Med 183: 2367–2372, 1996. on NaCl absorption. Hypertension 42: 674 –679, 2003.
83. Lee S, Kim W, Kang KP, Moon SO, Sung MJ, Kim DH, Kim HJ, 104. Pan S, An P, Zhang R, He X, Yin G, Min W. Etk/Bmx as a tumor
Park SK. Agonist of peroxisome proliferator-activated receptor-gamma, necrosis factor receptor type 2-specific kinase: role in endothelial cell
rosiglitazone, reduces renal injury and dysfunction in a murine sepsis migration and angiogenesis. Mol Cell Biol 22: 7512–7523, 2002.
model. Nephrol Dial Transplant 20: 1057–1065, 2005. 105. Parameswaran N, Patial S. Tumor necrosis factor-alpha signaling in
84. Legrand M, Bezemer R, Kandil A, Demirci C, Payen D, Ince C. The macrophages. Crit Rev Eukaryot Gene Expr 20: 87–103, 2010.
role of renal hypoperfusion in development of renal microcirculatory 106. Pfeffer K. Biological functions of tumor necrosis factor cytokines and
their receptors. Cytokine Growth Factor Rev 14: 185–191, 2003.
dysfunction in endotoxemic rats. Intensive Care Med 37: 1534 –1542,
107. Pinckard JK, Sheehan KC, Schreiber RD. Ligand-induced formation
2011.
of p55 and p75 tumor necrosis factor receptor heterocomplexes on intact
85. Leon CG, Tory R, Jia J, Sivak O, Wasan KM. Discovery and
cells. J Biol Chem 272: 10784 –10789, 1997.
development of toll-like receptor 4 (TLR4) antagonists: a new paradigm
108. Plato CF, Garvin JL. ␣2-Adrenergic-mediated tubular NO production
for treating sepsis and other diseases. Pharm Res 25: 1751–1761, 2008.
inhibits thick ascending limb chloride absorption. Am J Physiol Renal
86. Liu Y, Yin G, Surapisitchat J, Berk BC, Min W. Laminar flow inhibits
Physiol 281: F679 –F686, 2001.
TNF-induced ASK1 activation by preventing dissociation of ASK1 from
109. Plato CF, Pollock DM, Garvin JL. Endothelin inhibits thick ascending
its inhibitor 14 –3-3. J Clin Invest 107: 917–923, 2001.
limb chloride flux via ETB receptor-mediated NO release. Am J Physiol
87. Lucas R, Garcia I, Donati YR, Hribar M, Mandriota SJ, Giroud C,
Renal Physiol 279: F326 –F333, 2000.
Buurman WA, Fransen L, Suter PM, Nunez G, Pepper MS, Grau 110. Plato CF, Shesely EG, Garvin JL. eNOS mediates L-arginine-induced
GE. Both TNF receptors are required for direct TNF-mediated cytotox- inhibition of thick ascending limb chloride flux. Hypertension 35: 319 –
icity in microvascular endothelial cells. Eur J Immunol 28: 3577–3586, 323, 2000.
1998. 111. Pocsik E, Duda E, Wallach D. Phosphorylation of the 26 kDa TNF
88. Lucas R, Magez S, De LR, Fransen L, Scheerlinck JP, Rampelberg precursor in monocytic cells and in transfected HeLa cells. J Inflamm 45:
M, Sablon E, De BP. Mapping the lectin-like activity of tumor necrosis 152–160, 1995.
factor. Science 263: 814 –817, 1994. 112. Qiu P, Cui X, Barochia A, Li Y, Natanson C, Eichacker PQ. The
89. Luettig B, Decker T, Lohmann-Matthes ML. Evidence for the exis- evolving experience with therapeutic TNF inhibition in sepsis: consid-
tence of two forms of membrane tumor necrosis factor: an integral ering the potential influence of risk of death. Expert Opin Investig Drugs
protein and a molecule attached to its receptor. J Immunol 143: 4034 – 20: 1555–1564, 2011.
4038, 1989. 113. Ramesh G, Brian RW. Cisplatin increases TNF-alpha mRNA stability
90. Macica CM, Escalante BA, Conners MS, Ferreri NR. TNF production in kidney proximal tubule cells. Ren Fail 28: 583–592, 2006.
by the medullary thick ascending limb of Henle’s loop. Kidney Int 46: 114. Ramesh G, Reeves WB. TNF-alpha mediates chemokine and cytokine
113–121, 1994. expression and renal injury in cisplatin nephrotoxicity. J Clin Invest 110:
91. Markewitz BA, Michael JR, Kohan DE. Cytokine-induced expression 835–842, 2002.
of a nitric oxide synthase in rat renal tubule cells. J Clin Invest 91: 115. Ramesh G, Reeves WB. Inflammatory cytokines in acute renal failure.
2138 –2143, 1993. Kidney Int Suppl S56 –S61, 2004.
92. McDonnell PC, Kumar S, Rabson AB, Gelinas C. Transcriptional 116. Ramseyer VD, Hong NJ, Garvin JL. Tumor necrosis factor alpha
activity of rel family proteins. Oncogene 7: 163–170, 1992. decreases nitric oxide synthase type 3 expression primarily via Rho/Rho
93. Micheau O, Tschopp J. Induction of TNF receptor I-mediated apoptosis kinase in the thick ascending limb. Hypertension 59: 1145–1150, 2012.
via two sequential signaling complexes. Cell 114: 181–190, 2003. 117. Rees DD, Monkhouse JE, Cambridge D, Moncada S. Nitric oxide and
94. Misseri R, Meldrum DR, Dagher P, Hile K, Rink RC, Meldrum KK. the haemodynamic profile of endotoxin shock in the conscious mouse. Br
Unilateral ureteral obstruction induces renal tubular cell production of J Pharmacol 124: 540 –546, 1998.
tumor necrosis factor-alpha independent of inflammatory cell infiltration. 118. Roczniak A, Burns KD. Nitric oxide stimulates guanylate cyclase and
J Urol 172: 1595–1599, 2004. regulates sodium transport in rabbit proximal tubule. Am J Physiol Renal
95. Miyakawa H, Woo SK, Dahl SC, Handler JS, Kwon HM. Tonicity- Fluid Electrolyte Physiol 270: F106 –F115, 1996.
responsive enhancer binding protein, a rel-like protein that stimulates 119. Rosa AC, Rattazzi L, Miglio G, Collino M, Fantozzi R. Angiotensin II
transcription in response to hypertonicity. Proc Natl Acad Sci USA 96: induces tumor necrosis factor-alpha expression and release from cultured
2538 –2542, 1999. human podocytes. Inflamm Res 61: 311–317, 2012.
96. Muller DN, Shagdarsuren E, Park JK, Dechend R, Mervaala E, 120. Rothe M, Wong SC, Henzel WJ, Goeddel DV. A novel family of
Hampich F, Fiebeler A, Ju X, Finckenberg P, Theuer J, Viedt C, putative signal transducers associated with the cytoplasmic domain of the
Kreuzer J, Heidecke H, Haller H, Zenke M, Luft FC. Immunosup- 75 kDa tumor necrosis factor receptor. Cell 78: 681–692, 1994.

AJP-Renal Physiol • doi:10.1152/ajprenal.00557.2012 • www.ajprenal.org


Downloaded from www.physiology.org/journal/ajprenal by ${individualUser.givenNames} ${individualUser.surname} (115.178.202.039) on December 25, 2017.
Copyright © 2013 American Physiological Society. All rights reserved.
Review
TNF AND RENAL FUNCTION F1241
121. Ruiz-Ortega M, Ruperez M, Lorenzo O, Esteban V, Blanco J, 143. Tobiume K, Saitoh M, Ichijo H. Activation of apoptosis signal-regu-
Mezzano S, Egido J. Angiotensin II regulates the synthesis of proin- lating kinase 1 by the stress-induced activating phosphorylation of
flammatory cytokines and chemokines in the kidney. Kidney Int Suppl pre-formed oligomer. J Cell Physiol 191: 95–104, 2002.
S12–S22, 2002. 144. Todorov V, Muller M, Schweda F, Kurtz A. Tumor necrosis factor-␣
122. Saftlas AF, Olson DR, Franks AL, Atrash HK, Pokras R. Epidemi- inhibits renin gene expression. Am J Physiol Regul Integr Comp Physiol
ology of preeclampsia and eclampsia in the United States, 1979 –1986. 283: R1046 –R1051, 2002.
Am J Obstet Gynecol 163: 460 –465, 1990. 145. Tracey KJ, Beutler B, Lowry SF, Merryweather J, Wolpe S, Milsark
123. Satou R, Miyata K, Katsurada A, Navar LG, Kobori H. Tumor IW, Hariri RJ, Fahey TJ, III, Zentella A, Albert JD. Shock and tissue
necrosis factor-␣ suppresses angiotensinogen expression through forma- injury induced by recombinant human cachectin. Science 234: 470–474, 1986.
tion of a p50/p50 homodimer in human renal proximal tubular cells. Am 146. Tracey KJ, Fong Y, Hesse DG, Manogue KR, Lee AT, Kuo GC,
J Physiol Cell Physiol 299: C750 –C759, 2010. Lowry SF, Cerami A. Anti-cachectin/TNF monoclonal antibodies pre-
124. Schmidt C, Hocherl K, Bucher M. Cytokine-mediated regulation of vent septic shock during lethal bacteraemia. Nature 330: 662–664, 1987.
urea transporters during experimental endotoxemia. Am J Physiol Renal 147. Tran LT, MacLeod KM, McNeill JH. Chronic etanercept treatment
Physiol 292: F1479 –F1489, 2007. prevents the development of hypertension in fructose-fed rats. Mol Cell
125. Schmidt C, Hocherl K, Bucher M. Regulation of renal glucose trans- Biochem 330: 219 –228, 2009.
porters during severe inflammation. Am J Physiol Renal Physiol 292: 148. Utsumi T, Takeshige T, Tanaka K, Takami K, Kira Y, Klostergaard
F804 –F811, 2007. J, Ishisaka R. Transmembrane TNF (pro-TNF) is palmitoylated. FEBS
126. Schmidt C, Hocherl K, Schweda F, Kurtz A, Bucher M. Regulation of Lett 500: 1–6, 2001.
renal sodium transporters during severe inflammation. J Am Soc Nephrol 149. Van den Berg DT, de Kloet ER, de JW. Central effects of mineralo-
18: 1072–1083, 2007. corticoid antagonist RU-28318 on blood pressure of DOCA-salt hyper-
127. Shahid M, Francis J, Majid DS. Tumor necrosis factor-␣ induces renal tensive rats. Am J Physiol Endocrinol Metab 267: E927–E933, 1994.
vasoconstriction as well as natriuresis in mice. Am J Physiol Renal 150. Van Zee KJ, Moldawer LL, Oldenburg HS, Thompson WA,
Physiol 295: F1836 –F1844, 2008. Stackpole SA, Montegut WJ, Rogy MA, Meschter C, Gallati H,
128. Silva GB, Garvin JL. Angiotensin II-dependent hypertension increases Schiller CD, Richter WF, Loetscher H, Ashkenazi A, Chamow
Na transport-related oxygen consumption by the thick ascending limb. SM, Wurm F, Calvano SE, Lowry SF, Lesslauer W. Protection
Hypertension 52: 1091–1098, 2008. against lethal Escherichia coli bacteremia in baboons (Papio anubis)
129. Singh P, Bahrami L, Castillo A, Majid DS. TNF-␣ type 2 receptor by pretreatment with a 55-kDa TNF receptor (CD120a)-Ig fusion
mediates renal inflammatory response to chronic angiotensin II admin- protein, Ro 45–2081. J Immunol 156: 2221–2230, 1996.
istration with high salt intake in mice. Am J Physiol Renal Physiol 304: 151. Vandenabeele P, Declercq W, Beyaert R, Fiers W. Two tumour
F991–F999, 2013. necrosis factor receptors: structure and function. Trends Cell Biol 5:
130. Sonkar GK, Singh RG. Evaluation of serum tumor necrosis factor alpha 392–399, 1995.
and its correlation with histology in chronic kidney disease, stable renal 152. Venegas-Pont M, Manigrasso MB, Grifoni SC, LaMarca BB, Maric
transplant and rejection cases. Saudi J Kidney Dis Transpl 20: 1000 – C, Racusen LC, Glover PH, Jones AV, Drummond HA, Ryan MJ.
1004, 2009. Tumor necrosis factor-alpha antagonist etanercept decreases blood pres-
131. Sriramula S, Haque M, Majid DS, Francis J. Involvement of tumor sure and protects the kidney in a mouse model of systemic lupus
necrosis factor-alpha in angiotensin II-mediated effects on salt appetite, erythematosus. Hypertension 56: 643–649, 2010.
hypertension, and cardiac hypertrophy. Hypertension 51: 1345–1351, 153. Vielhauer V, Stavrakis G, Mayadas TN. Renal cell-expressed TNF
2008. receptor 2, not receptor 1, is essential for the development of glomeru-
132. Stoos BA, Garcia NH, Garvin JL. Nitric oxide inhibits sodium reab- lonephritis. J Clin Invest 115: 1199 –1209, 2005.
sorption in the isolated perfused cortical collecting duct. J Am Soc 154. Vinciguerra M, Hasler U, Mordasini D, Roussel M, Capovilla M,
Nephrol 6: 89 –94, 1995. Ogier-Denis E, Vandewalle A, Martin PY, Feraille E. Cytokines and
133. Takeuchi M, Rothe M, Goeddel Anatomy of TRAF2 DV. Distinct sodium induce protein kinase A-dependent cell-surface Na,K-ATPase
domains for nuclear factor-kappaB activation and association with tumor recruitment via dissociation of NF-kappaB/IkappaB/protein kinase A
necrosis factor signaling proteins. J Biol Chem 271: 19935–19942, 1996. catalytic subunit complex in collecting duct principal cells. J Am Soc
134. Tan X, He W, Liu Y. Combination therapy with paricalcitol and Nephrol 16: 2576 –2585, 2005.
trandolapril reduces renal fibrosis in obstructive nephropathy. Kidney Int 155. Wang D, An SJ, Wang WH, McGiff JC, Ferreri NR. CaR-mediated
76: 1248 –1257, 2009. COX-2 expression in primary cultured mTAL cells. Am J Physiol Renal
135. Tang G, Minemoto Y, Dibling B, Purcell NH, Li Z, Karin M, Lin A. Physiol 281: F658 –F664, 2001.
Inhibition of JNK activation through NF-kappaB target genes. Nature 156. Wang D, Pedraza PL, Abdullah HI, McGiff JC, Ferreri NR. Calcium-
414: 313–317, 2001. sensing receptor-mediated TNF production in medullary thick ascending
136. Tang P, Hung MC, Klostergaard J. Human pro-tumor necrosis factor limb cells. Am J Physiol Renal Physiol 283: F963–F970, 2002.
is a homotrimer. Biochemistry 35: 8216 –8225, 1996. 157. Wang T, Inglis FM, Kalb RG. Defective fluid and HCO⫺ 3 absorption in
137. Tartaglia LA, Goeddel DV, Reynolds C, Figari IS, Weber RF, Fendly proximal tubule of neuronal nitric oxide synthase-knockout mice. Am J
BM, Palladino MA Jr. Stimulation of human T-cell proliferation by Physiol Renal Physiol 279: F518 –F524, 2000.
specific activation of the 75-kDa tumor necrosis factor receptor. J 158. Wray GM, Coakley JH. Severe septic shock unresponsive to noradren-
Immunol 151: 4637–4641, 1993. aline. Lancet 346: 1604, 1995.
138. Tartaglia LA, Weber RF, Figari IS, Reynolds C, Palladino MA Jr, 159. Wu X, Wittwer AJ, Carr LS, Crippes BA, DeLarco JE, Lefkowith
Goeddel DV. The two different receptors for tumor necrosis factor JB. Cytokine-induced neutrophil chemoattractant mediates neutrophil
mediate distinct cellular responses. Proc Natl Acad Sci USA 88: 9292– influx in immune complex glomerulonephritis in rat. J Clin Invest 94:
9296, 1991. 337–344, 1994.
139. Therrien FJ, Agharazii M, Lebel M, Lariviere R. Neutralization of 160. Wuthrich RP, Glimcher LH, Yui MA, Jevnikar AM, Dumas SE,
tumor necrosis factor-alpha reduces renal fibrosis and hypertension in Kelley VE. MHC class II, antigen presentation and tumor necrosis factor
rats with renal failure. Am J Nephrol 36: 151–161, 2012. in renal tubular epithelial cells. Kidney Int 37: 783–792, 1990.
140. Tikkanen I, Uhlenius N, Tikkanen T, Miettinen A, Tornroth T, 161. Yard BA, Daha MR, Kooymans-Couthino M, Bruijn JA, Paape ME,
Fyhrquist F, Holthofer H. Increased renal expression of cytokines and Schrama E, van Es LA, van der Woude FJ. IL-1 alpha stimulated TNF
growth factors induced by DOCA-NaCl treatment in Heymann nephritis. alpha production by cultured human proximal tubular epithelial cells.
Nephrol Dial Transplant 10: 2192–2198, 1995. Kidney Int 42: 383–389, 1992.
141. Timoshanko JR, Sedgwick JD, Holdsworth SR, Tipping PG. Intrinsic 162. Yeo ES, Hwang JY, Park JE, Choi YJ, Huh KB, Kim WY. Tumor
renal cells are the major source of tumor necrosis factor contributing to necrosis factor (TNF-alpha) and C-reactive protein (CRP) are positively
renal injury in murine crescentic glomerulonephritis. J Am Soc Nephrol associated with the risk of chronic kidney disease in patients with type 2
14: 1785–1793, 2003. diabetes. Yonsei Med J 51: 519 –525, 2010.
142. Tipping PG, Leong TW, Holdsworth SR. Tumor necrosis factor 163. Zager RA, Johnson AC, Hanson SY, Lund S. Ischemic proximal
production by glomerular macrophages in anti-glomerular basement tubular injury primes mice to endotoxin-induced TNF-␣ generation and
membrane glomerulonephritis in rabbits. Lab Invest 65: 272–279, 1991. systemic release. Am J Physiol Renal Physiol 289: F289 –F297, 2005.

AJP-Renal Physiol • doi:10.1152/ajprenal.00557.2012 • www.ajprenal.org


Downloaded from www.physiology.org/journal/ajprenal by ${individualUser.givenNames} ${individualUser.surname} (115.178.202.039) on December 25, 2017.
Copyright © 2013 American Physiological Society. All rights reserved.
Review
F1242 TNF AND RENAL FUNCTION

164. Zager RA, Johnson AC, Hanson SY, Lund S. Acute nephrotoxic and 168. Zhang R, Xu Y, Ekman N, Wu Z, Wu J, Alitalo K, Min W. Etk/Bmx
obstructive injury primes the kidney to endotoxin-driven cytokine/ transactivates vascular endothelial growth factor 2 and recruits phospha-
chemokine production. Kidney Int 69: 1181–1188, 2006. tidylinositol 3-kinase to mediate the tumor necrosis factor-induced an-
165. Zanotti-Cavazzoni SL, Hollenberg SM. Cardiac dysfunction in severe giogenic pathway. J Biol Chem 278: 51267–51276, 2003.
sepsis and septic shock. Curr Opin Crit Care 15: 392–397, 2009. 169. Zhu L, Yang X, Ji Y, Chen W, Guan W, Zhou SF, Yu X. Up-regulated
166. Zhang L, Chen J, Fu H. Suppression of apoptosis signal-regulating renal expression of TNF-alpha signalling adapter proteins in lupus
kinase 1-induced cell death by 14 –3-3 proteins. Proc Natl Acad Sci USA glomerulonephritis. Lupus 18: 116 –127, 2009.
96: 8511–8515, 1999. 170. Zinman B, Hanley AJ, Harris SB, Kwan J, Fantus IG. Circulating
167. Zhang R, He X, Liu W, Lu M, Hsieh JT, Min W. AIP1 mediates tumor necrosis factor-alpha concentrations in a native Canadian popula-
TNF-alpha-induced ASK1 activation by facilitating dissociation of tion with high rates of type 2 diabetes mellitus. J Clin Endocrinol Metab
ASK1 from its inhibitor 14 –3-3. J Clin Invest 111: 1933–1943, 2003. 84: 272–278, 1999.

AJP-Renal Physiol • doi:10.1152/ajprenal.00557.2012 • www.ajprenal.org


Downloaded from www.physiology.org/journal/ajprenal by ${individualUser.givenNames} ${individualUser.surname} (115.178.202.039) on December 25, 2017.
Copyright © 2013 American Physiological Society. All rights reserved.

También podría gustarte