Abstract
Activated immune cell populations contribute to hypertension in part through inciting damage to the kidney and by provoking inappropriate sodium reabsorption in the nephron. Inflammatory mediators called cytokines produced by T lymphocytes and macrophages act on specific sodium transporters in the kidney, augmenting their activity or expression, with consequent expansion of intravascular fluid volume and cardiac output. The overlapping functions of these cytokines, each of which may activate multiple receptors, present challenges in precisely targeting inflammatory signaling cascades in hypertension. Moreover, broad immune suppression could expose the hypertensive patient to disproportional risks of infection or malignancy. Nevertheless, the possibility that incisive immunomodulatory therapies could provide cardiovascular and renal protection through both blood pressure-dependent and -independent mechanisms justifies comprehensive investigation into the relevant signaling pathways and tissue sites in which inflammatory cytokines function to exaggerate blood pressure elevation and target organ damage in hypertension.
Keywords: hypertension, cytokine signaling, organ damage
hypertension impacts a billion people worldwide and leads to severe kidney and cardiovascular disease (34). Despite the availability of several classes of anti-hypertensive medications, blood pressure remains poorly controlled in up to half of patients with hypertension (17), highlighting the need to reduce barriers to compliance with existing treatments and to develop novel anti-hypertensive therapies. Blood pressure homeostasis is maintained by coordinated inputs from the kidney, heart, vasculature, nervous system, and even the immune system (9). While dysfunction in any one of these cardiovascular control centers has been shown to enhance the susceptibility to experimental hypertension, Guyton (22) suggested that sustained elevations in blood pressure reflect inappropriate reabsorption of sodium in the kidney nephron. Otherwise, in response to blood pressure elevation, the kidney should excrete salt and water in sufficient amounts to lower intravascular volume and return blood pressure back toward normal. In recent years, several laboratories including our own have sought to understand how perturbations in immune responses can provoke renal sodium retention leading to hypertension and consequent target organ damage.
Preclinical studies have implicated both the innate and adaptive immune systems in the pathogenesis of hypertension. Among myeloid cell populations of the innate immune system, LysM-expressing inflammatory monocytes, CD39-responsive neutrophils, and CD11c+ dendritic cells (DCs) bearing oxidized isoketals can all potentiate blood pressure elevation (19, 28, 66). These DCs transfer susceptibility to hypertension only in the presence of circulating lymphocytes, suggesting that DCs provoke increases in blood pressure by presenting antigens to the T cell receptor with concomitant provision of a costimulatory signal (28, 63). Although the actions of myeloid cells in the vasculature and kidney to regulate blood pressure have historically received the most attention, an emerging literature indicates that macrophages resident in the dermis can also modulate salt sensitivity through effects on nonosmotic sodium storage outside the vasculature (38, 67, 73).
Within the adaptive immune system, both B and T lymphocytes contribute to the chronic hypertensive response (6, 23). However, B cells appear to regulate blood pressure by modulating T cell function, as transfer of B cells does not recapitulate hypertension in the absence of T cells (23). We observed that animals lacking functional T cells had preserved natriuresis, blunted oxidative stress in the kidney, enhanced renal generation of nitric oxide, and blunted hypertension in response to activation of the renin-angiotensin system (RAS) (11). T cell inhibition yielded similar protective effects in other rodent models of hypertension (13, 14), suggesting that activated T cells promote hypertension by augmenting oxidative stress and sodium reabsorption in the kidney. Individual T cell subpopulations can have varying effects on renal sodium transport by modulating the function of neighboring mononuclear cells, elaborating vasoactive cytokines, and altering levels of reactive oxygen species (ROS) within the vasculature and kidney. The potential role of T lymphocytes, and those with a memory phenotype in particular, has been elegantly summarized earlier in this Perspective series (25). Below, we will consider the contribution to hypertension of several inflammatory cytokines typically produced by T and myeloid cell populations.
Inflammatory Cytokines in Hypertension
A host of inflammatory cytokines shape the hypertensive response, typically by regulating oxidant stress and/or levels of nitric oxide in the vasculature or kidney. IL-17A, a cytokine produced by Th17 cells that figure prominently in autoimmunity, provokes generation of ROS in the blood vessel wall and augments sodium transport in the proximal tubule of the nephron with consequent blood pressure elevation (26, 27, 39). IL-6 helps to sustain Th17 cell differentiation and potentiates hypertension probably by promoting renal sodium reabsorption both directly in the collecting tubule epithelium and indirectly by facilitating RAS-induced renal vasoconstriction (4, 36, 51). Moreover, IL-17A directly promotes renal sodium retention by enhancing expression and activity of sodium-hydrogen exchanger 3 in the proximal tubule and activity of the sodium chloride cotransporter in the distal nephron (43). Studies such as these that uncover the precise functions of individual IL-17 isoforms to regulate blood pressure will be critical to the successful targeting of IL-17 in hypertension. To this point, globally interrupting IL-17's downstream signaling pathways did not impact blood pressure and worsened kidney damage in more severe hypertension, and a broad IL-17 blocking antibody had no effect (31, 41). Thus, future interventions will need to selectively disrupt the prohypertensive functions of IL-17A while preserving IL-17's protective actions in the kidney.
Interferon-γ (IFN) is the prototypical inflammatory cytokine produced by cytotoxic CD8+ T cells. While CD8+ and not CD4+ T cells appear to mediate T cell-dependent hypertension during RAS activation (60, 72), and IFN facilitates sodium transport in the proximal and distal nephron (26), interpreting the actions of IFN in hypertension presents the same challenges as those of IL-17. IFN deficiency has provided either robust or no protection from RAS-dependent hypertension (49, 70), and deficiency of the receptor for IFN afforded only compartment-specific protection for the kidney through a blood pressure-independent mechanism (41). While discrepancies in the results from these studies can be ascribed to differences in study design, rodent strain, and even epigenetic factors such as gut microbiota, collectively these experiments would indicate that blood pressure is exquisitely sensitive to a cytokine's precise localization and dose.
Studies of two prototypical macrophage cytokines, tumor necrosis factor-α (TNF) and IL-1, corroborate this hypothesis. Circulating levels of TNF and IL-1 are dramatically increased in the hypotensive state of sepsis (21). Moreover, intravenous infusion of these cytokines causes natriuresis (29, 52, 53, 61). Nevertheless, genetic deficiency of TNF or the receptor for IL-1 affords significant protection from hypertension, suggesting that on balance these cytokines are prohypertensive in the intact animal (23, 58, 71). To resolve these paradoxes, others and we began to examine the actions of these cytokines in cardiovascular control centers.
Prohypertensive Actions of TNF and IL-1
TNF is a pleiotropic cytokine produced in several cardiovascular control centers with complex physiologic effects on renal function that have been comprehensively summarized in this journal (45). TNF enhances the appetite for salt, so TNF acting in the central nervous system (CNS) may contribute to hypertension (58). In our hands, TNF secreted by proinflammatory Th1 lymphocytes did not contribute to hypertension but did impact injury within the kidney glomerulus (72), consistent with the known effects of TNF to injure the glomerular endothelium (3). We also found that the presence of TNF in the blood vessel wall did not augment RAS-dependent vasoconstriction (70). By contrast, TNF suppresses nitric oxide synthase (NOS)3 expression in the thick ascending limb of the nephron such that deleting TNF selectively from the kidney via a cross-transplantation strategy attenuates the chronic hypertensive response (46, 70). These data would suggest that targeting TNF's actions within the kidney should ameliorate hypertension (Fig. 1). However, even within the kidney, activating the two receptors for TNF, TNFR1 and TNFR2, has divergent effects on blood pressure (5, 7, 57), which could confound attempts to target signaling pathways downstream of TNF in hypertensive patients.
Fig. 1.
Ligation of the IL-1R1 receptor on intrarenal myeloid cells inhibits nitric oxide synthase (NOS)2 expression, leading to reduced nitric oxide (NO) production. Binding of ANG II to the type 1 angiotensin receptor (AT1R) on the medullary thick ascending limb (mTAL) epithelial cell in the kidney enhances TNF-α production, which inhibits NOS3 expression and thereby reduces NO production. In each case, the blunted NO generation permits augmented activity of the NKCC2 sodium cotransporter and exaggerated sodium reabsorption.
The inducible form of IL-1, IL-1β, is cleaved to its active form by the NLRP3 inflammasome. Blood pressure elevation induces components of this inflammasome, whereas deficiency of these components blunts the hypertensive response (32, 55). Once activated, IL-1 could promote hypertension through actions in several tissue beds. IL-1 injected into the CNS provokes catecholamine release and blood pressure elevation (44, 54, 59). IL-1 triggers constriction of the pulmonary vasculature and aorta (16, 64), but we did not find that IL-1 receptor (IL-1R1) activation potentiates RAS-induced vasoconstriction in the systemic circulation (71). Rather, our experiments revealed that IL-1R1 expression on intrarenal myeloid cells constrains their elaboration of nitric oxide (NO), releasing the NKCC2 sodium cotransporter from tonic NO-dependent inhibition with consequent renal sodium retention during angiotensin II-dependent hypertension (71) (Fig. 1). By contrast, IL-1R1 activation on macrophages recruited to combat parasitic infection enhances NO production (37), so further studies are warranted to understand whether IL-1's actions in myeloid cells are context-specific or whether IL-1R1 activation on kidney parenchymal cells modulates the phenotype of infiltrating myeloid cells. Such an interaction in which the kidney epithelium regulates hematopoietic cell differentiation has been appreciated during the pathogenesis of acute kidney injury (35).
Prospects for Targeting the TNF or IL-1 Pathways in Human Hypertension
When considering the use of any immunomodulatory drug, weighing the benefits of cardiovascular protection against the risks of infection and compromised tumor surveillance is paramount. Broad immunosuppression appears to blunt the hypertensive response in human patients (24). However, more targeted approaches might reduce blood pressure and/or attenuate end-organ damage in hypertension without markedly enhancing infection risk. Even individual cytokine blockade is not without risk as TNF antagonism, for example, has been associated with enhanced susceptibility to tuberculosis reactivation (20).
In light of these risks, pursuing additional preclinical studies to more precisely target intracellular signaling pathways downstream of TNF and IL-1 in hypertension might be prudent. On the other hand, these intracellular mediators may receive signals via multiple receptors leading to inconsistent effects on blood pressure. For example, IL-1R1 forms a signaling complex with several IL-1 receptor-associated kinase (IRAK) isoforms and Myd88 that together liberate the p65 subunit of NF-κB to allow its nuclear translocation (65), but Myd88 deletion has been reported to have pro- and anti-hypertensive effects in the same model of RAS activation (30, 56). An alternative to blocking signaling pathways downstream of TNF or IL-1 would be to globally target TNF or IL-1 signals with a tissue-specific strategy. Such a biomedical engineering approach has proved elusive to date, but harnessing the body's endogenous “tissue address” machinery with SNARE technology, piggyback transposons, or antibody conjugates represents three creative strategies in this direction (33, 50, 68).
A promising feature of the preclinical data that encourages translation to the hypertension clinic is the congruent effect of cytokine deficiency vs. blockade on blood pressure. Accordingly, TNF blockade blunted the hypertensive response in the rat subjected to RAS activation and in a mouse model of systemic lupus erythematosus (SLE) (18, 62). Similarly, we saw that IL-1R1 blockade with anakinra afforded protection from hypertension on par with that seen in IL-1R1 deficiency (71). As noted above with pharmacologic infusion models, the therapeutic benefit of TNF or IL-1 antagonists may depend on their dose and the tissues they reach, particularly given the discrepant functions of the two TNF receptors and the divergent expression profiles and induction patterns of the two IL-1 isoforms, IL-1α and IL-1β.
Studies examining the roles of TNF and IL-1 in human hypertension are inconclusive but point to the need for mechanistic studies in larger hypertensive cohorts. Levels of both TNF and IL-1β are elevated in the monocytes and circulation of some hypertensive populations (2, 12, 15), suggesting they may play a role in blood pressure elevation, but it is conversely possible that the enhanced cytokine expression is the result rather than the cause of blood pressure elevation in these patients. TNF or IL-1 blockade was not reported to reduce blood pressure in clinical studies of these therapies for normotensive patients with rheumatologic disease, and in studies of patients with heart failure, TNF antagonism only rarely yielded a reportable hypotensive response (8, 40). Nevertheless, blood pressure reductions accruing from blockade of lymphocyte proliferation correlated with lower urinary TNF levels (24), and TNF blockade in one small study of hypertensive patients reduced continuous ambulatory blood pressure (69). Regarding blockade of the IL-1 signaling pathways, large, ongoing studies with the Il-1β inhibitor canakinumab and the IL-1R1 antagonist anakinra in patients with atherosclerosis should reveal whether targeting the IL-1 signaling pathway can ameliorate cardiovascular damage and may also lend clues to the effects of IL-1 receptor activation on human blood pressure (1, 47, 48).
Concluding Remarks
Immune activation clearly promotes renal sodium retention in experimental models of salt-sensitive hypertension. Nevertheless, the employment of small rodents to study the mechanisms that link inflammatory cytokines to enhanced sodium transport in the nephron poses considerable challenges. Although we have seen clear links between natriuresis, aquaresis, and acute reductions in total body weight (as a measure of changes in intravascular volume) with genetic manipulations that fully prevent hypertension (10, 11), typically we are not able to link natriuresis to acute changes in total body weight in our experiments with cytokine-deficient mice that are only partially protected from hypertension. Admittedly, our experimental systems may not be sufficiently sensitive to detect small changes in intravasculature volume. However, an alternative possibility is that alterations in total body sodium attributable solely to kidney function do not fully capture the full complexity of sodium homeostasis as it relates to blood pressure regulation. In this regard, as tonicity-sensing macrophages regulate the storage of sodium bound to proteoglycans in the dermis (38), targeting functions of inflammatory cells and the cytokines they produce has the capacity to reduce morbidity in hypertension both by directly limiting end-organ injury and by improving sodium homeostasis throughout the body.
Despite the compelling evidence for the role of inflammatory cytokines in the pathogenesis of hypertension, the risks of immune suppression in hypertensive patients compel us to pursue carefully targeted disruptions of the immune response and to reserve such interventions for patients who clearly have biologically resistant hypertension. Evaluating inflammatory biomarkers may further allow us to reserve testing of immunomodulatory therapies for hypertensive patients with evidence of underlying immune activation. One motivation for exploring new immunomodulatory therapies for hypertension despite their inherent risks is that these novel approaches may have the capacity to attenuate injury to the brain, heart, and kidney even when they fail to reduce blood pressure (42). We therefore submit that future studies should continue to define in vivo both the signaling pathways through which cytokines regulate sodium homeostasis and the key cardiovascular control tissues in which cytokines act to modulate blood pressure.
GRANTS
This study was funded by National Institutes of Health Grants DK087893 and HL128355; Veterans Health Administration, Office of Research and Development, Biomedical Laboratory Research and Development Grant BX000893; Edna and Fred L. Mandel Center for Hypertension and Atherosclerosis Research; and Duke O'Brien Center for Kidney Research.
DISCLOSURES
No conflicts of interest, financial or otherwise, are declared by the author(s).
AUTHOR CONTRIBUTIONS
S.D.C. conception and design of research; S.D.C. performed experiments; S.D.C. analyzed data; S.D.C. interpreted results of experiments; S.D.C. and A.D.J. prepared figures; S.D.C. drafted manuscript; S.D.C. and A.D.J. edited and revised manuscript; S.D.C. and A.D.J. approved final version of manuscript.
REFERENCES
- 1.Abbate A, Kontos MC, Grizzard JD, Biondi-Zoccai GG, Van Tassell BW, Robati R, Roach LM, Arena RA, Roberts CS, Varma A, Gelwix CC, Salloum FN, Hastillo A, Dinarello CA, Vetrovec GW, Investigators VA. Interleukin-1 blockade with anakinra to prevent adverse cardiac remodeling after acute myocardial infarction [Virginia Commonwealth University Anakinra Remodeling Trial (VCU-ART) Pilot study]. Am J Cardiol 105: 1371–1377, 2010. [DOI] [PubMed] [Google Scholar]
- 2.Bautista LE, Vera LM, Arenas IA, Gamarra G. Independent association between inflammatory markers (C-reactive protein, interleukin-6, and TNF-alpha) and essential hypertension. J Hum Hypertens 19: 149–154, 2005. [DOI] [PubMed] [Google Scholar]
- 3.Bertani T, Abbate M, Zoja C, Corna D, Perico N, Ghezzi P, Remuzzi G. Tumor necrosis factor induces glomerular damage in the rabbit. Am J Pathol 134: 419–430, 1989. [PMC free article] [PubMed] [Google Scholar]
- 4.Brands MW, Banes-Berceli AK, Inscho EW, Al-Azawi H, Allen AJ, Labazi H. Interleukin 6 knockout prevents angiotensin II hypertension: role of renal vasoconstriction and janus kinase 2/signal transducer and activator of transcription 3 activation. Hypertension 56: 879–884, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Castillo A, Islam MT, Prieto MC, Majid DSA. Tumor necrosis factor-α receptor type 1, not type 2, mediates its acute responses in the kidney. Am J Physiol Renal Physiol 302: F1650–F1657, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Chan CT, Sobey CG, Lieu M, Ferens D, Kett MM, Diep H, Kim HA, Krishnan SM, Lewis CV, Salimova E, Tipping P, Vinh A, Samuel CS, Peter K, Guzik TJ, Kyaw TS, Toh BH, Bobik A, Drummond GR. Obligatory role for B cells in the development of angiotensin II-dependent hypertension. Hypertension 66: 1023–1033, 2015. [DOI] [PubMed] [Google Scholar]
- 7.Chen CC, Pedraza PL, Hao S, Stier CT, Ferreri NR. TNFR1-deficient mice display altered blood pressure and renal responses to ANG II infusion. Am J Physiol Renal Physiol 299: F1141–F1150, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Chung ES, Packer M, Lo KH, Fasanmade AA, Willerson JT, Investigators ftA. Randomized, double-blind, placebo-controlled, pilot trial of infliximab, a chimeric monoclonal antibody to tumor necrosis factor-α, in patients with moderate-to-severe heart failure: results of the anti-TNF therapy against congestive heart failure (attach) trial. Circulation 107: 3133–3140, 2003. [DOI] [PubMed] [Google Scholar]
- 9.Coffman TM. Under pressure: the search for the essential mechanisms of hypertension. Nat Med 17: 1402–1409, 2011. [DOI] [PubMed] [Google Scholar]
- 10.Crowley SD, Gurley SB, Herrera MJ, Ruiz P, Griffiths R, Kumar AP, Kim HS, Smithies O, Le TH, Coffman TM. Angiotensin II causes hypertension and cardiac hypertrophy through its receptors in the kidney. Proc Natl Acad Sci USA 103: 17985–17990, 2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Crowley SD, Song YS, Lin EE, Griffiths R, Kim HS, Ruiz P. Lymphocyte responses exacerbate angiotensin II-dependent hypertension. Am J Physiol Regul Integr Comp Physiol 298: R1089–R1097, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Dalekos GN, Elisaf M, Bairaktari E, Tsolas O, Siamopoulos KC. Increased serum levels of interleukin-1beta in the systemic circulation of patients with essential hypertension: additional risk factor for atherogenesis in hypertensive patients? J Lab Clin Med 129: 300–308, 1997. [DOI] [PubMed] [Google Scholar]
- 13.De Miguel C, Das S, Lund H, Mattson DL. T lymphocytes mediate hypertension and kidney damage in Dahl salt-sensitive rats. Am J Physiol Regul Integr Comp Physiol 298: R1136–R1142, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.De Miguel C, Guo C, Lund H, Feng D, Mattson DL. Infiltrating T lymphocytes in the kidney increase oxidative stress and participate in the development of hypertension and renal disease. Am J Physiol Renal Physiol 300: F734–F742, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Dorffel Y, Latsch C, Stuhlmuller B, Schreiber S, Scholze S, Burmester GR, Scholze J. Preactivated peripheral blood monocytes in patients with essential hypertension. Hypertension 34: 113–117, 1999. [DOI] [PubMed] [Google Scholar]
- 16.Dorrance AM. Interleukin 1-beta (IL-1beta) enhances contractile responses in endothelium-denuded aorta from hypertensive, but not normotensive, rats. Vascul Pharmacol 47: 160–165, 2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Egan BM, Zhao Y, Axon RN. US trends in prevalence, awareness, treatment, and control of hypertension, 1988–2008. JAMA 303: 2043–2050, 2010. [DOI] [PubMed] [Google Scholar]
- 18.Elmarakby AA, Quigley JE, Pollock DM, Imig JD. Tumor necrosis factor alpha blockade increases renal Cyp2c23 expression and slows the progression of renal damage in salt-sensitive hypertension. Hypertension 47: 557–562, 2006. [DOI] [PubMed] [Google Scholar]
- 19.Fabbiano S, Menacho-Marquez M, Robles-Valero J, Pericacho M, Matesanz-Marin A, Garcia-Macias C, Sevilla MA, Montero MJ, Alarcon B, Lopez-Novoa JM, Martin P, Bustelo XR. Immunosuppression-independent role of regulatory T cells against hypertension-driven renal dysfunctions. Mol Cell Biol 35: 3528–3546, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Gardam MA, Keystone EC, Menzies R, Manners S, Skamene E, Long R, Vinh DC. Anti-tumour necrosis factor agents and tuberculosis risk: mechanisms of action and clinical management. Lancet Infect Dis 3: 148–155, 2003. [DOI] [PubMed] [Google Scholar]
- 21.Girardin E, Grau GE, Dayer JM, Roux-Lombard P, Group* tJS, Lambert PH. Tumor necrosis factor and interleukin-1 in the serum of children with severe infectious purpura. N Engl J Med 319: 397–400, 1988. [DOI] [PubMed] [Google Scholar]
- 22.Guyton AC. Blood pressure control–special role of the kidneys and body fluids. Science 252: 1813–1816, 1991. [DOI] [PubMed] [Google Scholar]
- 23.Guzik TJ, Hoch NE, Brown KA, McCann LA, Rahman A, Dikalov S, Goronzy J, Weyand C, Harrison DG. Role of the T cell in the genesis of angiotensin II induced hypertension and vascular dysfunction. J Exp Med 204: 2449–2460, 2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Herrera J, Ferrebuz A, MacGregor EG, Rodriguez-Iturbe B. Mycophenolate mofetil treatment improves hypertension in patients with psoriasis and rheumatoid arthritis. J Am Soc Nephrol 17: S218–225, 2006. [DOI] [PubMed] [Google Scholar]
- 25.Itani HA, Harrison DG. Memories that last in hypertension. Am J Physiol Renal Physiol 308: F1197–F1199, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Kamat NV, Thabet SR, Xiao L, Saleh MA, Kirabo A, Madhur MS, Delpire E, Harrison DG, McDonough AA. Renal transporter activation during angiotensin-II hypertension is blunted in interferon-γ−/− and interleukin-17A−/− mice. Hypertension 65: 569–576, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Karbach S, Croxford AL, Oelze M, Schuler R, Minwegen D, Wegner J, Koukes L, Yogev N, Nikolaev A, Reissig S, Ullmann A, Knorr M, Waldner M, Neurath MF, Li H, Wu Z, Brochhausen C, Scheller J, Rose-John S, Piotrowski C, Bechmann I, Radsak M, Wild P, Daiber A, von Stebut E, Wenzel P, Waisman A, Munzel T. Interleukin 17 drives vascular inflammation, endothelial dysfunction, and arterial hypertension in psoriasis-like skin disease. Arterioscler Thromb Vasc Biol 34: 2658–2668, 2014. [DOI] [PubMed] [Google Scholar]
- 28.Kirabo A, Fontana V, de Faria AP, Loperena R, Galindo CL, Wu J, Bikineyeva AT, Dikalov S, Xiao L, Chen W, Saleh MA, Trott DW, Itani HA, Vinh A, Amarnath V, Amarnath K, Guzik TJ, Bernstein KE, Shen XZ, Shyr Y, Chen SC, Mernaugh RL, Laffer CL, Elijovich F, Davies SS, Moreno H, Madhur MS, Roberts J 2nd, Harrison DG. DC isoketal-modified proteins activate T cells and promote hypertension. J Clin Invest 124: 4642–4656, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Kohan DE, Merli CA, Simon EE. Micropuncture localization of the natriuretic effect of interleukin 1. Am J Physiol Renal Fluid Electrolyte Physiol 256: F810–F813, 1989. [DOI] [PubMed] [Google Scholar]
- 30.Kossmann S, Schönfelder T, Finger S, Brähler M, Minwegen D, Knorr M, Daiber A, Radsak M, Münzel T, Wenzel P. Abstract 11888: MyD88 expressed by myeloid cells is essential for angiotensin II-induced vascular dysfunction, inflammation and arterial hypertension. Circulation 130: A11888, 2014. [Google Scholar]
- 31.Krebs CF, Lange S, Niemann G, Rosendahl A, Lehners A, Meyer-Schwesinger C, Stahl RA, Benndorf RA, Velden J, Paust HJ, Panzer U, Ehmke H, Wenzel UO. Deficiency of the interleukin 17/23 axis accelerates renal injury in mice with deoxycorticosterone acetate+angiotensin II-induced hypertension. Hypertension 63: 565–571, 2014. [DOI] [PubMed] [Google Scholar]
- 32.Krishnan SM, Dowling JK, Ling YH, Diep H, Chan CT, Ferens D, Kett MM, Pinar A, Samuel CS, Vinh A, Arumugam TV, Hewitson TD, Kemp-Harper BK, Robertson AA, Cooper MA, Latz E, Mansell A, Sobey CG, Drummond GR. Inflammasome activity is essential for one kidney/deoxycorticosterone acetate/salt-induced hypertension in mice. Br J Pharmacol 173: 752–765, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Kvirkvelia N, McMenamin M, Gutierrez VI, Lasareishvili B, Madaio MP. Human anti-alpha3(IV)NC1 antibody drug conjugates target glomeruli to resolve nephritis. Am J Physiol Renal Physiol 309: F680–F684, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Lawes CM, Vander Hoorn S, Rodgers A, International Society of H. Global burden of blood-pressure-related disease, 2001. Lancet 371: 1513–1518, 2008. [DOI] [PubMed] [Google Scholar]
- 35.Lee S, Huen S, Nishio H, Nishio S, Lee HK, Choi BS, Ruhrberg C, Cantley LG. Distinct macrophage phenotypes contribute to kidney injury and repair. J Am Soc Nephrol 22: 317–326, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Li K, Guo D, Zhu H, Hering-Smith KS, Hamm LL, Ouyang J, Dong Y. Interleukin-6 stimulates epithelial sodium channels in mouse cortical collecting duct cells. Am J Physiol Regul Integr Comp Physiol 299: R590–R595, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Lima-Junior DS, Costa DL, Carregaro V, Cunha LD, Silva AL, Mineo TW, Gutierrez FR, Bellio M, Bortoluci KR, Flavell RA, Bozza MT, Silva JS, Zamboni DS. Inflammasome-derived IL-1beta production induces nitric oxide-mediated resistance to Leishmania. Nat Med 19: 909–915, 2013. [DOI] [PubMed] [Google Scholar]
- 38.Machnik A, Neuhofer W, Jantsch J, Dahlmann A, Tammela T, Machura K, Park JK, Beck FX, Muller DN, Derer W, Goss J, Ziomber A, Dietsch P, Wagner H, van Rooijen N, Kurtz A, Hilgers KF, Alitalo K, Eckardt KU, Luft FC, Kerjaschki D, Titze J. Macrophages regulate salt-dependent volume and blood pressure by a vascular endothelial growth factor-C-dependent buffering mechanism. Nat Med 15: 545–552, 2009. [DOI] [PubMed] [Google Scholar]
- 39.Madhur MS, Lob HE, McCann LA, Iwakura Y, Blinder Y, Guzik TJ, Harrison DG. Interleukin 17 promotes angiotensin II-induced hypertension and vascular dysfunction. Hypertension 55: 500–507, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Mann DL, McMurray JJ, Packer M, Swedberg K, Borer JS, Colucci WS, Djian J, Drexler H, Feldman A, Kober L, Krum H, Liu P, Nieminen M, Tavazzi L, van Veldhuisen DJ, Waldenstrom A, Warren M, Westheim A, Zannad F, Fleming T. Targeted anticytokine therapy in patients with chronic heart failure: results of the Randomized Etanercept Worldwide Evaluation (RENEWAL). Circulation 109: 1594–1602, 2004. [DOI] [PubMed] [Google Scholar]
- 41.Markó L, Kvakan H, Park JK, Qadri F, Spallek B, Binger KJ, Bowman EP, Kleinewietfeld M, Fokuhl V, Dechend R, Müller DN. Interferon-γ signaling inhibition ameliorates angiotensin II-induced cardiac damage. Hypertension 60: 1430–1436, 2012. [DOI] [PubMed] [Google Scholar]
- 42.Muller DN, Shagdarsuren E, Park JK, Dechend R, Mervaala E, Hampich F, Fiebeler A, Ju X, Finckenberg P, Theuer J, Viedt C, Kreuzer J, Heidecke H, Haller H, Zenke M, Luft FC. Immunosuppressive treatment protects against angiotensin II-induced renal damage. Am J Pathol 161: 1679–1693, 2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Norlander AE, Saleh MA, Kamat NV, Ko B, Gnecco J, Zhu L, Dale BL, Iwakura Y, Hoover RS, McDonough AA, Madhur MS. Interleukin-17A regulates renal sodium transporters and renal injury in angiotensin II-induced hypertension. Hypertension 68: 167–174, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Palazzolo DL, Quadri SK. Interleukin-1 stimulates catecholamine release from the hypothalamus. Life Sci 47: 2105–2109, 1990. [DOI] [PubMed] [Google Scholar]
- 45.Ramseyer VD, Garvin JL. Tumor necrosis factor-α: regulation of renal function and blood pressure. Am J Physiol Renal Physiol 304: F1231–F1242, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Ramseyer VD, Hong NJ, Garvin JL. Tumor necrosis factor alpha decreases nitric oxide synthase type 3 expression primarily via Rho/Rho kinase in the thick ascending limb. Hypertension 59: 1145–1150, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Ridker PM, Howard CP, Walter V, Everett B, Libby P, Hensen J, Thuren T, Group CPI. Effects of interleukin-1beta inhibition with canakinumab on hemoglobin A1c, lipids, C-reactive protein, interleukin-6, and fibrinogen: a phase IIb randomized, placebo-controlled trial. Circulation 126: 2739–2748, 2012. [DOI] [PubMed] [Google Scholar]
- 48.Ridker PM, Thuren T, Zalewski A, Libby P. Interleukin-1beta inhibition and the prevention of recurrent cardiovascular events: rationale and design of the Canakinumab Anti-inflammatory Thrombosis Outcomes Study (CANTOS). Am Heart J 162: 597–605, 2011. [DOI] [PubMed] [Google Scholar]
- 49.Saleh MA, McMaster WG, Wu J, Norlander AE, Funt SA, Thabet SR, Kirabo A, Xiao L, Chen W, Itani HA, Michell D, Huan T, Zhang Y, Takaki S, Titze J, Levy D, Harrison DG, Madhur MS. Lymphocyte adaptor protein LNK deficiency exacerbates hypertension and end-organ inflammation. J Clin Invest 125: 1189–1202, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Schnitzer JE. Caveolae: from basic trafficking mechanisms to targeting transcytosis for tissue-specific drug and gene delivery in vivo. Adv Drug Deliv Rev 49: 265–280, 2001. [DOI] [PubMed] [Google Scholar]
- 51.Schrader LI, Kinzenbaw DA, Johnson AW, Faraci FM, Didion SP. IL-6 deficiency protects against angiotensin II induced endothelial dysfunction and hypertrophy. Arterioscler Thromb Vasc Biol 27: 2576–2581, 2007. [DOI] [PubMed] [Google Scholar]
- 52.Schreiner GF, Kohan DE. Regulation of renal transport processes and hemodynamics by macrophages and lymphocytes. Am J Physiol Renal Fluid Electrolyte Physiol 258: F761–F767, 1990. [DOI] [PubMed] [Google Scholar]
- 53.Shahid M, Francis J, Majid DSA. Tumor necrosis factor-α induces renal vasoconstriction as well as natriuresis in mice. Am J Physiol Renal Physiol 295: F1836–F1844, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Shi P, Diez-Freire C, Jun JY, Qi Y, Katovich MJ, Li Q, Sriramula S, Francis J, Sumners C, Raizada MK. Brain microglial cytokines in neurogenic hypertension. Hypertension 56: 297–303, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Shirasuna K, Karasawa T, Usui F, Kobayashi M, Komada T, Kimura H, Kawashima A, Ohkuchi A, Taniguchi S, Takahashi M. NLRP3 deficiency improves angiotensin II-induced hypertension but not fetal growth restriction during pregnancy. Endocrinology 156: 4281–4292, 2015. [DOI] [PubMed] [Google Scholar]
- 56.Singh MV, Cicha MZ, Meyerholz DK, Chapleau MW, Abboud FM. Dual ativation of TRIF and MyD88 adaptor proteins by angiotensin II evokes opposing effects on pressure, cardiac hypertrophy, and inflammatory gene expression. Hypertension 66: 647–656, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Singh P, Bahrami L, Castillo A, Majid DS. TNF-α type 2 receptor mediates renal inflammatory response to chronic angiotensin II administration with high salt intake in mice. Am J Physiol Renal Physiol 304: F991–F999, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Sriramula S, Haque M, Majid DSA, Francis J. Involvement of tumor necrosis factor-α in angiotensin II-mediated effects on salt appetite, hypertension, and cardiac hypertrophy. Hypertension 51: 1345–1351, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Takahashi H, Nishimura M, Sakamoto M, Ikegaki I, Nakanishi T, Yoshimura M. Effects of interleukin-1 beta on blood pressure, sympathetic nerve activity, and pituitary endocrine functions in anesthetized rats. Am J Hypertens 5: 224–229, 1992. [DOI] [PubMed] [Google Scholar]
- 60.Trott DW, Thabet SR, Kirabo A, Saleh MA, Itani H, Norlander AE, Wu J, Goldstein A, Arendshorst WJ, Madhur MS, Chen W, Li CI, Shyr Y, Harrison DG. Oligoclonal CD8+ T cells play a critical role in the development of hypertension. Hypertension 64: 1108–1115, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.van Lanschot JJ, Mealy K, Jacobs DO, Evans DA, Wilmore DW. Splenectomy attenuates the inappropriate diuresis associated with tumor necrosis factor administration. Surg Gynecol Obstet 172: 293–297, 1991. [PubMed] [Google Scholar]
- 62.Venegas-Pont M, Manigrasso MB, Grifoni SC, LaMarca BB, Maric C, Racusen LC, Glover PH, Jones AV, Drummond HA, Ryan MJ. Tumor necrosis factor-alpha antagonist etanercept decreases blood pressure and protects the kidney in a mouse model of systemic lupus erythematosus. Hypertension 56: 643–649, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Vinh A, Chen W, Blinder Y, Weiss D, Taylor WR, Goronzy JJ, Weyand CM, Harrison DG, Guzik TJ. Inhibition and genetic ablation of the B7/CD28 T-cell costimulation axis prevents experimental hypertension. Circulation 122: 2529–2537, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Voelkel NF, Tuder RM, Bridges J, Arend WP. Interleukin-1 receptor antagonist treatment reduces pulmonary hypertension generated in rats by monocrotaline. Am J Respir Cell Mol Biol 11: 664–675, 1994. [DOI] [PubMed] [Google Scholar]
- 65.Weber A, Wasiliew P, Kracht M. Interleukin-1 (IL-1) pathway. Sci Signal 3: cm1, 2010. [DOI] [PubMed] [Google Scholar]
- 66.Wenzel P, Knorr M, Kossmann S, Stratmann J, Hausding M, Schuhmacher S, Karbach SH, Schwenk M, Yogev N, Schulz E, Oelze M, Grabbe S, Jonuleit H, Becker C, Daiber A, Waisman A, Münzel T. Lysozyme M-positive monocytes mediate angiotensin II-induced arterial hypertension and vascular dysfunction/clinical perspective. Circulation 124: 1370–1381, 2011. [DOI] [PubMed] [Google Scholar]
- 67.Wiig H, Schroder A, Neuhofer W, Jantsch J, Kopp C, Karlsen TV, Boschmann M, Goss J, Bry M, Rakova N, Dahlmann A, Brenner S, Tenstad O, Nurmi H, Mervaala E, Wagner H, Beck FX, Muller DN, Kerjaschki D, Luft FC, Harrison DG, Alitalo K, Titze J. Immune cells control skin lymphatic electrolyte homeostasis and blood pressure. J Clin Invest 123: 2803–2815, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Woodard LE, Wilson MH. piggyBac-ing models and new therapeutic strategies. Trends Biotechnol 33: 525–533, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Yoshida S, Takeuchi T, Kotani T, Yamamoto N, Hata K, Nagai K, Shoda T, Takai S, Makino S, Hanafusa T. Infliximab, a TNF-α inhibitor, reduces 24-h ambulatory blood pressure in rheumatoid arthritis patients. J Hum Hypertens 28: 165–169, 2014. [DOI] [PubMed] [Google Scholar]
- 70.Zhang J, Patel MB, Griffiths R, Mao A, Song YS, Karlovich NS, Sparks MA, Jin H, Wu M, Lin EE, Crowley SD. Tumor necrosis factor-α produced in the kidney contributes to angiotensin II-dependent hypertension. Hypertension 64: 1275–1281, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Zhang J, Rudemiller NP, Patel MB, Karlovich NS, Wu M, McDonough AA, Griffiths R, Sparks MA, Jeffs AD, Crowley SD. Interleukin-1 receptor activation potentiates salt reabsorption in angiotensin II-induced hypertension via the NKCC2 co-transporter in the nephron. Cell Metab 23: 360–368, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Zhang JD, Patel MB, Song YS, Griffiths R, Burchette J, Ruiz P, Sparks MA, Yan M, Howell DN, Gomez JA, Spurney RF, Coffman TM, Crowley SD. A novel role for type 1 angiotensin receptors on T lymphocytes to limit target organ damage in hypertension. Circ Res 110: 1604–1617, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Zhang MZ, Yao B, Wang Y, Yang S, Wang S, Fan X, Harris RC. Inhibition of cyclooxygenase-2 in hematopoietic cells results in salt-sensitive hypertension. J Clin Invest 125: 4281–4294, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]