The Myeloid-Kidney Interface in Health and Disease : Clinical Journal of the American Society of Nephrology

Journal Logo

Review

The Myeloid-Kidney Interface in Health and Disease

Vlasschaert, Caitlyn1; Moran, Sarah M.1; Rauh, Michael J.2

Author Information
CJASN 17(2):p 323-331, February 2022. | DOI: 10.2215/CJN.04120321
  • Free

Abstract

Introduction

Innate immune cells of the myeloid lineage form a kidney surveillance system that is poised to quickly react to a variety of insults, from pathogens to endogenous threats. Circulating and resident myeloid cells interact with the various kidney substructures, and deficits in this interaction are the root cause of many kidney disorders. Recent efforts in single-cell sequencing have provided clarity on many intricacies of the kidney-myeloid interface (1–4). This review will provide an overview of current knowledge regarding the interactions between myeloid cells and native kidneys and provide an updated framework for future studies investigating alterations in this interface.

Myeloid Cells in Normal Kidney Function

Myeloid cells are important to kidney health and include monocytes, macrophages, dendritic cells, and neutrophils. Neutrophils are circulating myeloid cells that are rapidly mobilized to sites of inflammation and infection. Recruited neutrophils eliminate pathogens via phagocytosis and via release of intracellular contents, including antimicrobial peptides (degranulation) and neutrophil extracellular traps (NETs), which physically trap bacteria—much like a spider’s web traps prey (Figure 1A). Neutrophils are abundant yet short lived: their survival, both in circulation and after recruitment to tissues, is typically limited to <5 days (5).

F1
Figure 1.:
Myeloid cells in kidney health: Graphical representation of cooperative myeloid cell responses to ascending urinary tract infection. (A) Neutrophils are abundant circulating myeloid cells that can be rapidly recruited to help contain perceived threats while bridging to other innate and adaptive immune system responses. Neutrophils employ three main lines of defense to neutralize invading pathogens and toxins: phagocytosis, degranulation, and release of neutrophil extracellular traps (NETs). (B) Kidney-resident macrophages are versatile phagocytes that coordinate multistaged responses to organ threat and injury. Macrophages clean up day-to-day cellular debris in the kidney via phagocytosis and, from this basal state, they can be primed to assume different roles. For example, macrophages assume a proinflammatory phenotype upon recognition of pathogen-associated molecular patterns (e.g., LPS) and damage-associated molecular patterns (e.g., uromodulin, cytokines) by different pattern-recognition receptors located on the cell surface and intracellularly. This activation enhances phagocytosis (including the phagocytosis of neutrophils that have first phagocytosed pathogens) and prompts the release of proinflammatory cytokines such as IL-6, IL-1β, and IL-18 (see Table 1). As the threat resolves, macrophages with reparative functions support tissue healing and return to homeostasis. (C) Dendritic cells play key roles in activation of adaptive immune system responses (8). Similar to macrophages, conventional dendritic cells are activated by pathogen- and damage-associated molecular patterns encountered in their tissue of residence. Activated conventional dendritic cells then present representative antigens on their cell surfaces using MHC molecules and migrate to kidney-draining lymph nodes to induce proliferation and differentiation of T cells specific to this antigen. Activated conventional dendritic cells can also produce cytokines and chemokines to recruit neutrophils.

Macrophages are comparatively much longer lived (on the order of months) and have the broadest roles in organ homeostasis, from neutralizing threats to orchestrating adaptive repair after tissue injury (Figure 1B). Although historically dichotomized into polar M1 (proinflammatory) and M2 (anti-inflammatory) states, macrophages are now recognized to exist as several subsets along a spectrum of activity (6,7). Macrophages engulf and destroy perceived threats, such as foreign material and senescent or abnormal endogenous cells. Dendritic cells similarly recognize these foreign antigens but chiefly serve to present these to T cells, thereby activating the adaptive immune system (Figure 1C) (8). Kidney macrophages largely derive from infiltrating bone marrow–derived monocytes, but there also exists a subset of embryonically derived and self-renewing resident macrophages (2). Resident macrophages and dendritic cells—collectively referred to as histiocytes—are concentrated in the medulla because the hypersaline medullary milieu favors their recruitment and enhances their defense capacity (9). This spatial zonation is thought to enable more rapid response to pathogen- and damage-associated molecular patterns. Whereas pathogen-associated molecular patterns are bacterial and viral epitopes, damage-associated molecular patterns are endogenous signals of noninfectious damage released by structural cells of the kidney, including epithelial cells, endothelial cells, and fibroblasts (2,10). For example, in acute tubular necrosis, leached uromodulin acts as a damage-associated molecular pattern to attract macrophages and dendritic cells to the site of tubular damage (11). Similarly, in response to retrograde colonization by uropathogens, epithelial cells of the kidney pelvis release chemokines to recruit neighboring phagocytes (2). These and many more coordinated immune responses to threat are a product of intricate kidney-myeloid crosstalk.

Myeloid cells communicate alerts bidirectionally with neighboring cells by way of cytokines. Important cytokines in kidney homeostasis are outlined in Table 1. Of these, IL-6 signaling has important, complex, and, at times, conflicting roles in controlling inflammation (12). IL-6 is proinflammatory in the acute response to tissue injury, setting off a cascade of immune responses, including B- and CD4+ T-cell activation, and the production of acute-phase proteins, such as C-reactive protein and fibrinogen by hepatocytes (13). Tight regulation of IL-6 signaling in the kidney microenvironment is important for mounting an appropriate inflammatory response (14). In early tubular injury, proinflammatory macrophages secrete large amounts of IL-6 to stimulate the activation of neighboring fibroblasts, epithelial cells, and progenitor cells (15). In adaptive kidney repair, this controlled burst of acute inflammation is followed by a period of wound healing and resolution of injury (16), which is coordinated by distinct subsets of macrophages (1). Profibrotic macrophages contribute to the activation of myofibroblasts at the site of injury (17), which produce the collagenous extracellular matrix that is used to provide structural support to the cellular remodeling that occurs after tissue injury (4,7). Activation of the fibrotic phase is typically effected by IL-4, IL-13, TGF-β (17), and—paradoxically—also IL-6 (18). In summary, IL-6 stimulates production of critical proteins and recruits myeloid cells involved in induction and resolution of the inflammatory response.

Table 1. - Cytokines in kidney homeostasis
Cytokine Function
Proinflammatory cytokines
 IL-6 Pleiotropic cytokine with local and systemic roles in the  inflammatory response a
 IL-1β Proinflammatory cytokines released upon activation of the  NLRP3 inflammasome, a multimeric protein complex that  can sense a variety of substrates, such as monosodium  urate, glucose, ATP, and multiple bacterial antigens (17).  This pleiotropy is possible because NLRP3 does not directly  recognize these as discrete ligands but instead detects  specific changes to cellular homeostasis resulting from their  presence (18). Once activated, NLRP3 inflammasome  components perform activating cleavage of precursor forms  of IL-1β and IL-18, which are then released locally during  programmed inflammatory cell death (pyroptosis) (17)
 IL-18
 IFN family cytokines Released upon detection of viral pathogen-associated  molecular patterns (critical role in host defense)
 TNFα Promotes neutrophil migration to sites of kidney injury and  potentiates phagocytosis
Chemokines
 CCL2 (MCP-1) Recruit monocytes to the glomerular endothelium and guide  the infiltration of monocytes-turned-macrophages to the  kidney parenchyma during injury
 CCL3 (MIP-1)
 CX3CL1 (fractalkine)
 CXCL1 Facilitate neutrophil infiltration of the kidney after injury
 CXCL8 (IL-8)
Anti-inflammatory cytokines
 IL-4 Promote alternative (M2-type) activation of macrophages
 IL-10
aSee text for further details.

Mechanisms of Myeloid Cell Dysfunction in Kidney Pathology

Myeloid cells are involved in virtually all forms of kidney dysfunction and overt kidney disease. Their contributions to pathology can be broadly divided into whether the myeloid cells become trapped in a dysfunctional inflammatory loop in response to injury, or whether the myeloid cells are themselves the primary cause of injury due to an intrinsic defect (Figure 2).

F2
Figure 2.:
Mechanisms of myeloid cell dysfunction in kidney disease. Detrimental myeloid cell interactions with the kidney can be broadly categorized into two types. The left-sided panels (A and B) depict scenarios where myeloid cells are exerting otherwise normal responses to abnormal stimuli. (A) Macrophages are situated in a hyperinflammatory milieu where the initial organ threat has not resolved, such as persistent hyperglycemia in uncontrolled diabetes or continued ingestion of medications causing acute interstitial nephritis. Macrophages contribute to the perpetuation of the inflammation and to organ fibrosis as they become nested in a dysfunctional loop. (B) Neutrophils and macrophages are responding to antigens perceived as foreign (abnormally deposited endogenous immune complexes), leading to injury to the glomerulus. This occurs in IgA nephropathy (depicted) and in anti–glomerular basement membrane disease. The right-sided panels (C and D) depict myeloid cells that are rendered dysfunctional outside of the kidney niche but contribute to injury therein. (C) The formation of dysregulated neutrophil extracellular traps (NETs), which occurs after autoactivation in ANCA-associated vasculitis and in other autoimmune glomerulonephritides. Moreover, these NETs expose double-stranded DNA (dsDNA) and nuclear proteins to the extracellular environment, which is thought to promote autoimmune complex formation in lupus nephritis. (D) Mutations in bone marrow progenitor cells can lead to the production of a spectrum of clonal and dysfunctional myeloid cells. Named disorders include acute myeloid leukemia (AML), chronic myeloid leukemia (CML), myelodsplastic syndrome (MDS), primary myelofibrosis (PMF), and clonal hematopoiesis of indeterminate potential (CHIP). As discussed in the main text, the dysregulated mutant myeloid cells in these disorders can have a plethora of downstream effects on kidney structure and function. Gd-IgA, galactose-deficient IgA.

Myeloid Cells in Chronic Kidney Inflammation and Fibrosis

Proinflammatory macrophages can perpetuate inflammation, especially when there is continued presence of damage-associated molecular patterns (Figure 2A) (7). In diabetic kidney disease (DKD), sustained hyperglycemia is associated with pervasive cytokine signaling and macrophage infiltration (19,20). In early DKD, immune cells have been shown to infiltrate the kidney at a density seven to eight times that in those without diabetes (21). Whereas activated macrophages are mostly restricted to the glomerulus in early DKD, there is subsequent progressive infiltration of the neighboring parenchyma as chronic inflammation develops (22). This persistent inflammation can lead to progressive fibrosis, the common end point in many CKDs (23).

An overwhelming acute inflammatory signal can also prompt chronic inflammation and kidney scarring. Drug-induced acute interstitial nephritis (AIN) is a local hypersensitivity reaction that represents the third most common cause of AKI in patients admitted to the hospital (24). Surveillant dendritic cells recognize the offending drug, and subsequent T-cell activation then sets off an inflammatory cascade that can recruit a broad spectrum of myeloid cells, including monocytes in transit, neutrophils, and often also eosinophils (24). The magnitude of this inflammatory cascade and the consequent degree of kidney impairment are highly variable and difficult to predict in drug-induced AIN (25,26). At one extreme, an intense inflammatory response and marked interstitial influx of various myeloid cells can promote severe AKI with pyuria and leave dysregulated residual inflammation, marked fibrosis, and chronic kidney impairment in its wake. Kidney biopsy can enable assessment of the degree of acuity, nature of myeloid cellular infiltrate, and an assessment of fibrosis and chronicity (27).

Myeloid Cells Recruited to Pathogenic Glomerular Immune Complexes

Immune complexes can form or deposit within the glomerulus as a result of autoantibody formation—a failure of the adaptive immune system. Innate immune cells of the myeloid lineage are programmed to detect and eliminate abnormal autoantigens. This type of immune response is protective against malignancy, for example, but destructive when otherwise healthy tissues are targeted, such as in GN (Figure 2B). IgA nephropathy results from deposition of immune complexes within the glomerular mesangium. Nephritogenic galactose-deficient IgA1 is the product of underglycosylation of the IgA hinge region, a process that is augmented by IL-6 signaling (28,29). Galactose-deficient IgA1 is the target antigen of autoantibody formation in IgA nephropathy, and deposition of these immune complexes in the mesangium leads to proliferative changes, including endocapillary and mesangial hypercellularity with local cytokine release, complement system activation, and macrophage infiltration (30). In IgA nephropathy, mesangial macrophage recruitment on kidney biopsy is associated with a more severe clinical course of disease (31). Within proliferative glomerular lesions in IgA nephropathy, the amount of CD163—a cell surface receptor for hemoglobin-haptoglobin complexes (32) that is specific to monocytes and monocyte-derived macrophages and that can be used as histopathologic proxy of their density—is inversely correlated with eGFR (33).

Pathologic macrophage and neutrophil responses to deposited immune complexes likewise contribute to kidney damage in anti–glomerular basement membrane disease and in lupus nephritis (27). In anti–glomerular basement membrane disease, experiments in mice have shown that an Fc receptor on the neutrophil cell surface directly binds the deposited immune complexes and promotes NET formation by the recruited neutrophil (34). Likewise, in lupus nephritis, antibodies complexed to nuclear autoantigens are deposited in the glomerulus, which activates the complement system and recruits various leukocytes (35). Macrophages exposed to autoantibody immune complexes in lupus nephritis adopt a distinct metabolic state that is skewed toward glycolysis—a phenotype that is similar, but not identical, to M1 macrophages (36). It has been proposed that targeting this unique macrophage metabolism may help treat lupus nephritis (36). In addition to their role in recognizing immune complex deposits, neutrophils have extrarenal roles in SLE and lupus nephritis. Neutrophils in SLE are primed to undergo NETosis, which externalizes immunogenic nuclear material and can promote SLE pathogenesis (37). In particular, low-density granulocytes—a subset of largely immature and proinflammatory neutrophils (38)—engage in more NET formation in SLE (39).

Cleaved Cell Surface Proteins in Kidney Injury

Maladaptive local release of myeloid intracellular content (cytokines, granules, NETs) can promote to kidney injury instead of promoting organ defense. Certain proteins cleaved from the cell surface of myeloid cells have been similarly identified as mediators of kidney injury. The urokinase-type plasminogen activator receptor (uPAR) is a membrane-bound protein found on many cells that is cleaved in the setting of inflammation (40,41). The soluble uPAR (suPAR) protein mediates pathologic activation of podocyte αvβ3-integrin signaling (40,42) and has been implicated in a broad range of kidney disease phenotypes, including all-cause CKD (43), APOL1-associated kidney disease (44), and autosomal dominant polycystic kidney disease (45). A variety of myeloid and nonmyeloid cells are known to shed suPAR (41,46), although bone marrow–derived immature myeloid lineage cells were identified as the primary source of high circulating suPAR levels in a mouse model (38). Other cleaved transmembrane proteins have been implicated in modulating inflammatory kidney damage by acting as decoy receptors that can scavenge cytokines and other inflammatory mediators. This includes soluble receptors for advanced glycemic end products in DKD (47,48) and soluble TNF receptor 1 (sTNFR-1) and sTNFR-2. Elevations in serum soluble receptors for advanced glycemic end products and sTNFRs have been associated with kidney dysfunction, all-cause mortality, and cardiovascular events (47,49,50). There is debate about whether this reflects heightened activity of the nonscavenged ligands, or instead whether the decoy receptors act as reservoirs that prolong the half-life of these inflammatory mediators (51). suPAR, sTNFR-1, sTNFR-2, and other proteins related to myeloid cell function have recently been shown to robustly correlate with histologic disease severity and clinical outcomes across a spectrum of kidney disease etiologies (52).

Kidney Sequelae of Intrinsic Myeloid Cell Defects

Thus far, this review has focused on immune cells amplifying incendiary damage in the kidney microenvironment by reacting to abnormal stimuli, such as pervasive inflammation or intruding immune complexes. Conversely, there exist scenarios where myeloid cells acquire defects outside of the kidney niche and are primary instigators of kidney disease (Figure 2). The next sections will explore how inherent myeloid cell defects, such as autoimmune activation or malignant transformation of these cells, can compromise kidney homeostasis.

Autoimmune Activation of Myeloid Cells in ANCA-Associated Vasculitis

Neutrophils, the most abundant myeloid cell in the circulation, participate in virtually all forms of autoimmune GN through varying mechanisms of injury (53). In ANCA-associated vasculitis, the contents of neutrophil granules are targeted by anti–proteinase 3 and/or anti-myeloperoxidase autoantibodies (Figure 2C). Upon autoantigen binding, neutrophils are activated: cytokines, proteases, and microparticles (54) are released via degranulation and NET formation, leading to endothelial injury and necrotizing damage of the glomerulus (55). The release of damaging neutrophil granule contents by NETosis is increasingly recognized as a common pathophysiologic mechanism in many systemic immune disorders affecting the kidney (53). The contents of these granules cooperatively disrupt glomerular endothelium integrity; activate complement; and induce the recruitment of monocytes, which are then primed to infiltrate glomeruli and amplify inflammatory and fibrotic insults therein. ANCA-stimulated neutrophils also release B cell–activating factor, which further perpetuates autoantibody formation and neutrophil autoactivation. Diverse macrophage populations amplify inflammatory insults in ANCA-associated vasculitis, and urinary biomarkers of their presence have been clinically translated into markers of disease activity. CD163 is cleaved from the macrophage and monocyte cell surface in the setting of inflammation (56), and urinary soluble CD163 is a reliable biomarker of active ANCA-associated vasculitis (57,58).

Kidney Sequelae of Malignant Myeloid Cell Disorders

Clonal and dysplastic cells of the myeloid lineage can manifest kidney sequelae as a direct consequence of disease and due to treatment-related toxicity (Figure 2D) (59,60). Acute myeloid leukemia can present with AKI due to tumor lysis syndrome or thrombotic microangiopathy (61). Less commonly, it can precipitate glomerular phenotypes, such as membranous nephropathy or FSGS (62), or cause direct injury through malignant parenchymal infiltration (63). Chronic myeloid leukemia has been associated with membranous nephropathy, minimal change disease, and membranoproliferative GN (60). Other myeloproliferative neoplasms—particularly primary myelofibrosis—have been associated with progressive kidney dysfunction and heavy proteinuria (62). Systemic autoimmune disorders are diagnosed in 10%–20% of individuals with myelodysplastic syndromes and, in a recent case series of individuals diagnosed with myelodysplastic syndrome who underwent kidney biopsy for AKI, more than half were diagnosed with rare glomerulopathies (64).

Clonal hematopoiesis of indeterminate potential (CHIP) is a newly recognized premalignant state that is associated with systemic inflammatory damage (65). With age, clonal populations of myeloid cells emerge as a result of mutations in bone marrow hematopoietic stem cells, commonly in epigenetic regulators such as TET2, DNMT3A, and ASXL1 (65,66). When the proportion of clonal cells in circulation reaches ≥4%, but there is no evidence of malignant transformation, severe cytopenias, or other World Health Organization–defined disorder, this is labeled CHIP (67). CHIP affects at least 15% of the general population aged ≥65 and it portends a 1.4-fold higher risk of all-cause mortality (65), an 11-fold higher risk of hematologic malignancy (65,66), and a two-fold higher risk of cardiovascular disease (68) that is independent of, and on par with, traditional cardiovascular risk factors (69). CHIP cells produce high levels of inflammatory cytokines (70,71), and dysregulated IL-6 signaling has been causally implicated in CHIP pathogenesis (72). Macrophages in mouse models of CHIP display an impaired ability to resolve inflammation (73) and contribute to kidney tubulointerstitial fibrosis (74,75). We recently showed that CHIP was associated with worse baseline kidney function, more kidney failure events, and higher rates of anemia in a small cohort of individuals with advanced CKD followed for up to 12 years. A cross-sectional analysis of 112 individuals with ANCA-associated vasculitis noted a higher prevalence of CHIP compared with age- and sex-matched healthy controls (30% versus 14%) (76). At present, there exist no evidence-based interventions for CHIP and, as such, no clinical guidelines pertaining to its surveillance or management (77). This is expected to change within the next few years because therapeutic strategies aimed at modifying clonal burden or controlling downstream inflammatory damage are in development (78). It is currently unknown whether these potential treatments may improve kidney or cardiovascular outcomes in patients with CHIP and kidney disease. First validating and exploring the causality and directionality of the association between CHIP and kidney disease is critical to ensure that patients affected by kidney disease are adequately considered in this ongoing effort.

Targeting Myeloid Cells in Kidney Disease

Given their broad involvement in inflammation and fibrosis, it is unsurprising that many therapies in kidney disease target myeloid cells. Among their numerous sites of action, glucocorticoids induce a type of kidney M2 macrophage polarization (78) and suppress IL-6 signaling in podocytes (79); for this reason, they are used to dampen inflammation across a broad spectrum of kidney disorders—from ANCA-associated vasculitis and lupus nephritis to nephrotic syndromes to severe cases of drug-induced AIN. Curiously, mineralocorticoids have the opposite effect: aldosterone induces a type of M1 activation of macrophages through direct actions on mineralocorticoid receptors within myeloid cell nuclei. Both hematopoietic-specific deletion of the mineralocorticoid receptor and inhibition with mineralocorticoid receptor antagonists were shown to be kidney protective in a mouse model of GN (80). The Finerenone in Reducing Kidney Failure and Disease Progression in Diabetic Kidney Disease (FIDELIO-DKD) phase 3 randomized control trial showed that finerenone (a mineralocorticoid receptor antagonist with both anti-inflammatory and antifibrotic properties [81]) delayed the progression of DKD (82). Many other components of the renin-angiotensin-aldosterone system have intricate roles in controlling the kidney inflammatory versus fibrotic balance (83). The targeted inhibition of certain myeloid cell intracellular proteins is being investigated in immune-mediated GN . Spleen tyrosine kinase inhibitors are in early stages of clinical assessment as therapies for ANCA-associated vasculitis (84) and IgA nephropathy (85), and inhibitors of diverse pathologic NETosis effector proteins have been assessed in preclinical models of ANCA-associated vasculitis (86).

Some of the most prescribed treatments in kidney failure (hemodialysis and peritoneal dialysis) have broad effects on the inflammatory network. Multiple patient- and modality-related factors contribute to inflammation, including uremic toxins and foreign bodies such as dialysis catheters and arteriovenous fistula graft material (87). Chronic elevations in IL-6, IL-1β, and other inflammatory markers are common in patients on long-term dialysis and have been associated with higher mortality, cardiovascular events, anemia, malnutrition, mineral bone disease, and cognitive decline in this population (87). Although not yet used in routine practice, proinflammatory cytokine blockade may help mitigate some of these complications, with supporting evidence for canakinumab (an IL-1β blocker) in the secondary prevention of cardiovascular events, which showed particular efficacy in patients with CKD (88), and ziltivekimab (an IL-6 blocker) for patients on hemodialysis with inflammatory anemia and erythropoietin hyporesponsiveness (89).

Conclusion

Circulating and resident myeloid cells are critical determinants of kidney health and disease. Although their role is to defend the kidney after injury, an imbalance or hyperactivation in any part of their regulatory network can, conversely, perpetuate damage. A growing spectrum of intrinsic defects are recognized to affect myeloid cell function, including the recently recognized and common clinical entity known as CHIP, wherein age-associated somatic mutations drive the emergence of hyperinflammatory myeloid cell populations. Whereas myeloid cells and their effector cytokines are the targets of several existing therapies for kidney disease, ongoing developments in our understanding will drive new therapies to emerge.

Disclosures

S.M. Moran, M.J. Rauh, and C. Vlasschaert report being employed by Queen’s University. All other authors have nothing to disclose.

Funding

This work was supported by the Government of Canada Canadian Institutes of Health Research grant 201911FBD-433120-DRA-CEDA-251523 and Physicians’ Services Incorporated Foundation grant RT8-2021.

Published online ahead of print. Publication date available at www.cjasn.org.

References

1. Conway BR, O’Sullivan ED, Cairns C, O’Sullivan J, Simpson DJ, Salzano A, Connor K, Ding P, Humphries D, Stewart K, Teenan O, Pius R, Henderson NC, Bénézech C, Ramachandran P, Ferenbach D, Hughes J, Chandra T, Denby L: Kidney single-cell atlas reveals myeloid heterogeneity in progression and regression of kidney disease. J Am Soc Nephrol 31: 2833–2854, 2020
2. Stewart BJ, Ferdinand JR, Young MD, Mitchell TJ, Loudon KW, Riding AM, Richoz N, Frazer GL, Staniforth JUL, Vieira Braga FA, Botting RA, Popescu DM, Vento-Tormo R, Stephenson E, Cagan A, Farndon SJ, Polanski K, Efremova M, Green K, Del Castillo Velasco-Herrera M, Guzzo C, Collord G, Mamanova L, Aho T, Armitage JN, Riddick ACP, Mushtaq I, Farrell S, Rampling D, Nicholson J, Filby A, Burge J, Lisgo S, Lindsay S, Bajenoff M, Warren AY, Stewart GD, Sebire N, Coleman N, Haniffa M, Teichmann SA, Behjati S, Clatworthy MR: Spatiotemporal immune zonation of the human kidney. Science 365: 1461–1466, 2019
3. Fu J, Akat KM, Sun Z, Zhang W, Schlondorff D, Liu Z, Tuschl T, Lee K, He JC: Single-cell RNA profiling of glomerular cells shows dynamic changes in experimental diabetic kidney disease. J Am Soc Nephrol 30: 533–545, 2019
4. Kuppe C, Ibrahim MM, Kranz J, Zhang X, Ziegler S, Perales-Patón J, Jansen J, Reimer KC, Smith JR, Dobie R, Wilson-Kanamori JR, Halder M, Xu Y, Kabgani N, Kaesler N, Klaus M, Gernhold L, Puelles VG, Huber TB, Boor P, Menzel S, Hoogenboezem RM, Bindels EMJ, Steffens J, Floege J, Schneider RK, Saez-Rodriguez J, Henderson NC, Kramann R: Decoding myofibroblast origins in human kidney fibrosis. Nature 589: 281–286, 2021 10.1038/s41586-020-2941-1
5. Hidalgo A, Chilvers ER, Summers C, Koenderman L: The neutrophil life cycle. Trends Immunol 40: 584–597, 2019
6. Murray PJ, Allen JE, Biswas SK, Fisher EA, Gilroy DW, Goerdt S, Gordon S, Hamilton JA, Ivashkiv LB, Lawrence T, Locati M, Mantovani A, Martinez FO, Mege JL, Mosser DM, Natoli G, Saeij JP, Schultze JL, Shirey KA, Sica A, Suttles J, Udalova I, van Ginderachter JA, Vogel SN, Wynn TA: Macrophage activation and polarization: Nomenclature and experimental guidelines. Immunity 41: 14–20, 2014
7. Tang PM-K, Nikolic-Paterson DJ, Lan H-Y: Macrophages: Versatile players in renal inflammation and fibrosis. Nat Rev Nephrol 15: 144–158, 2019
8. Kurts C, Ginhoux F, Panzer U: Kidney dendritic cells: Fundamental biology and functional roles in health and disease. Nat Rev Nephrol 16: 391–407, 2020
9. Berry MR, Mathews RJ, Ferdinand JR, Jing C, Loudon KW, Wlodek E, Dennison TW, Kuper C, Neuhofer W, Clatworthy MR: Renal sodium gradient orchestrates a dynamic antibacterial defense zone. Cell 170: 860–874.e19, 2017
10. Krausgruber T, Fortelny N, Fife-Gernedl V, Senekowitsch M, Schuster LC, Lercher A, Nemc A, Schmidl C, Rendeiro AF, Bergthaler A, Bock C: Structural cells are key regulators of organ-specific immune responses. Nature 583: 296–302, 2020
11. Anders H-J, Schaefer L: Beyond tissue injury-damage-associated molecular patterns, toll-like receptors, and inflammasomes also drive regeneration and fibrosis. J Am Soc Nephrol 25: 1387–1400, 2014
12. Fuster JJ, Walsh K: The good, the bad, and the ugly of interleukin-6 signaling. EMBO J 33: 1425–1427, 2014
13. Tanaka T, Narazaki M, Kishimoto T: IL-6 in inflammation, immunity, and disease. Cold Spring Harb Perspect Biol 6: a016295, 2014
14. Nechemia-Arbely Y, Barkan D, Pizov G, Shriki A, Rose-John S, Galun E, Axelrod JH: IL-6/IL-6R axis plays a critical role in acute kidney injury. J Am Soc Nephrol 19: 1106–1115, 2008
15. Wynn TA, Vannella KM: Macrophages in tissue repair, regeneration, and fibrosis. Immunity 44: 450–462, 2016
16. Han Y, Ma FY, Tesch GH, Manthey CL, Nikolic-Paterson DJ: Role of macrophages in the fibrotic phase of rat crescentic glomerulonephritis. Am J Physiol Renal Physiol 304: F1043–F1053, 2013
17. Wang Y, Harris DCH: Macrophages in renal disease. J Am Soc Nephrol 22: 21–27, 2011
18. Luig M, Kluger MA, Goerke B, Meyer M, Nosko A, Yan I, Scheller J, Mittrücker HW, Rose-John S, Stahl RA, Panzer U, Steinmetz OM: Inflammation-induced IL-6 functions as a natural brake on macrophages and limits GN. J Am Soc Nephrol 26: 1597–1607, 2015
19. Pavlou S, Lindsay J, Ingram R, Xu H, Chen M: Sustained high glucose exposure sensitizes macrophage responses to cytokine stimuli but reduces their phagocytic activity. BMC Immunol 19: 24, 2018
20. Chow F, Ozols E, Nikolic-Paterson DJ, Atkins RC, Tesch GH: Macrophages in mouse type 2 diabetic nephropathy: Correlation with diabetic state and progressive renal injury. Kidney Int 65: 116–128, 2004
21. Wilson PC, Wu H, Kirita Y, Uchimura K, Ledru N, Rennke HG, Welling PA, Waikar SS, Humphreys BD: The single-cell transcriptomic landscape of early human diabetic nephropathy. Proc Natl Acad Sci U S A 116: 19619–19625, 2019
22. Klessens CQF, Zandbergen M, Wolterbeek R, Bruijn JA, Rabelink TJ, Bajema IM, IJpelaar DHT: Macrophages in diabetic nephropathy in patients with type 2 diabetes. Nephrol Dial Transplant 32: 1322–1329, 2017
23. Humphreys BD: Mechanisms of renal fibrosis. Annu Rev Physiol 80: 309–326, 2018
24. Raghavan R, Shawar S: Mechanisms of drug-induced interstitial nephritis. Adv Chronic Kidney Dis 24: 64–71, 2017
25. Clarkson MR, Giblin L, O’Connell FP, O’Kelly P, Walshe JJ, Conlon P, O’Meara Y, Dormon A, Campbell E, Donohoe J: Acute interstitial nephritis: Clinical features and response to corticosteroid therapy. Nephrol Dial Transplant 19: 2778–2783, 2004
26. Muriithi AK, Leung N, Valeri AM, Cornell LD, Sethi S, Fidler ME, Nasr SH: Biopsy-proven acute interstitial nephritis, 1993–2011: A case series. Am J Kidney Dis 64: 558–566, 2014
27. Devi S, Li A, Westhorpe CL, Lo CY, Abeynaike LD, Snelgrove SL, Hall P, Ooi JD, Sobey CG, Kitching AR, Hickey MJ: Multiphoton imaging reveals a new leukocyte recruitment paradigm in the glomerulus [published correction appears in Nat Med 22: 446, 2016 10.1038/nm0416-446a]. Nat Med 19: 107–112, 2013
28. Suzuki H, Raska M, Yamada K, Moldoveanu Z, Julian BA, Wyatt RJ, Tomino Y, Gharavi AG, Novak J: Cytokines alter IgA1 O-glycosylation by dysregulating C1GalT1 and ST6GalNAc-II enzymes. J Biol Chem 289: 5330–5339, 2014
29. Makita Y, Suzuki H, Kano T, Takahata A, Julian BA, Novak J, Suzuki Y: TLR9 activation induces aberrant IgA glycosylation via APRIL- and IL-6-mediated pathways in IgA nephropathy. Kidney Int 97: 340–349, 2020
30. Peng W, Pei GQ, Tang Y, Tan L, Qin W: IgA1 deposition may induce NLRP3 expression and macrophage transdifferentiation of podocyte in IgA nephropathy. J Transl Med 17: 406, 2019
31. Silva GEB, Costa RS, Ravinal RC, Ramalho LN, Reis MA, Moyses-Neto M, Romao EA, Coimbra TM, Dantas M: Renal macrophage infiltration is associated with a poor outcome in IgA nephropathy. Clinics (São Paulo) 67: 697–703, 2012
32. Kristiansen M, Graversen JH, Jacobsen C, Sonne O, Hoffman HJ, Law SK, Moestrup SK: Identification of the haemoglobin scavenger receptor. Nature 409: 198–201, 2001
33. Hodgin JB, Berthier CC, John R, Grone E, Porubsky S, Gröne HJ, Herzenberg AM, Scholey JW, Hladunewich M, Cattran DC, Kretzler M, Reich HN: The molecular phenotype of endocapillary proliferation: Novel therapeutic targets for IgA nephropathy. PLoS One 9: e103413, 2014
34. Nishi H, Furuhashi K, Cullere X, Saggu G, Miller MJ, Chen Y, Rosetti F, Hamilton SL, Yang L, Pittman SP, Liao J, Herter JM, Berry JC, DeAngelo DJ, Zhu C, Tsokos GC, Mayadas TN: Neutrophil FcγRIIA promotes IgG-mediated glomerular neutrophil capture via Abl/Src kinases. J Clin Invest 127: 3810–3826, 2017
35. Lech M, Anders H-J: The pathogenesis of lupus nephritis. J Am Soc Nephrol 24: 1357–1366, 2013
36. Jing C, Castro-Dopico T, Richoz N, Tuong ZK, Ferdinand JR, Lok LSC, Loudon KW, Banham GD, Mathews RJ, Cader Z, Fitzpatrick S, Bashant KR, Kaplan MJ, Kaser A, Johnson RS, Murphy MP, Siegel RM, Clatworthy MR: Macrophage metabolic reprogramming presents a therapeutic target in lupus nephritis. Proc Natl Acad Sci U S A 117: 15160–15171, 2020
37. Carmona-Rivera C, Kaplan MJ: Low-density granulocytes: A distinct class of neutrophils in systemic autoimmunity. Semin Immunopathol 35: 455–463, 2013
38. Scapini P, Marini O, Tecchio C, Cassatella MA: Human neutrophils in the saga of cellular heterogeneity: Insights and open questions. Immunol Rev 273: 48–60, 2016
39. Villanueva E, Yalavarthi S, Berthier CC, Hodgin JB, Khandpur R, Lin AM, Rubin CJ, Zhao W, Olsen SH, Klinker M, Shealy D, Denny MF, Plumas J, Chaperot L, Kretzler M, Bruce AT, Kaplan MJ: Netting neutrophils induce endothelial damage, infiltrate tissues, and expose immunostimulatory molecules in systemic lupus erythematosus. J Immunol 187: 538–552, 2011
40. Hahm E, Wei C, Fernandez I, Li J, Tardi NJ, Tracy M, Wadhwani S, Cao Y, Peev V, Zloza A, Lusciks J, Hayek SS, O’Connor C, Bitzer M, Gupta V, Sever S, Sykes DB, Scadden DT, Reiser J: Bone marrow-derived immature myeloid cells are a main source of circulating suPAR contributing to proteinuric kidney disease. Nat Med 23: 100–106, 2017
41. Gussen H, Hohlstein P, Bartneck M, Warzecha KT, Buendgens L, Luedde T, Trautwein C, Koch A, Tacke F: Neutrophils are a main source of circulating suPAR predicting outcome in critical illness. J Intensive Care 7: 26, 2019
42. Wei C, El Hindi S, Li J, Fornoni A, Goes N, Sageshima J, Maiguel D, Karumanchi SA, Yap HK, Saleem M, Zhang Q, Nikolic B, Chaudhuri A, Daftarian P, Salido E, Torres A, Salifu M, Sarwal MM, Schaefer F, Morath C, Schwenger V, Zeier M, Gupta V, Roth D, Rastaldi MP, Burke G, Ruiz P, Reiser J: Circulating urokinase receptor as a cause of focal segmental glomerulosclerosis. Nat Med 17: 952–960, 2011
43. Hayek SS, Sever S, Ko YA, Trachtman H, Awad M, Wadhwani S, Altintas MM, Wei C, Hotton AL, French AL, Sperling LS, Lerakis S, Quyyumi AA, Reiser J: Soluble urokinase receptor and chronic kidney disease. N Engl J Med 373: 1916–1925, 2015
44. Hayek SS, Koh KH, Grams ME, Wei C, Ko YA, Li J, Samelko B, Lee H, Dande RR, Lee HW, Hahm E, Peev V, Tracy M, Tardi NJ, Gupta V, Altintas MM, Garborcauskas G, Stojanovic N, Winkler CA, Lipkowitz MS, Tin A, Inker LA, Levey AS, Zeier M, Freedman BI, Kopp JB, Skorecki K, Coresh J, Quyyumi AA, Sever S, Reiser J: A tripartite complex of suPAR, APOL1 risk variants and αvβ3 integrin on podocytes mediates chronic kidney disease. Nat Med 23: 945–953, 2017
45. Hayek SS, Landsittel DP, Wei C, Zeier M, Yu ASL, Torres VE, Roth S, Pao CS, Reiser J: Soluble urokinase plasminogen activator receptor and decline in kidney function in autosomal dominant polycystic kidney disease. J Am Soc Nephrol 30: 1305–1313, 2019
46. Amor C, Feucht J, Leibold J, Ho YJ, Zhu C, Alonso-Curbelo D, Mansilla-Soto J, Boyer JA, Li X, Giavridis T, Kulick A, Houlihan S, Peerschke E, Friedman SL, Ponomarev V, Piersigilli A, Sadelain M, Lowe SW: Senolytic CAR T cells reverse senescence-associated pathologies. Nature 583: 127–132, 2020
47. Thomas MC, Woodward M, Neal B, Li Q, Pickering R, Marre M, Williams B, Perkovic V, Cooper ME, Zoungas S, Chalmers J, Hillis GS; ADVANCE Collaborative Group: Relationship between levels of advanced glycation end products and their soluble receptor and adverse outcomes in adults with type 2 diabetes. Diabetes Care 38: 1891–1897, 2015
48. Wadén JM, Dahlström EH, Elonen N, Thorn LM, Wadén J, Sandholm N, Forsblom C, Groop PH; FinnDiane Study Group: Soluble receptor for AGE in diabetic nephropathy and its progression in Finnish individuals with type 1 diabetes. Diabetologia 62: 1268–1274, 2019
49. Bhatraju PK, Zelnick LR, Shlipak M, Katz R, Kestenbaum B: Association of soluble TNFR-1 concentrations with long-term decline in kidney function: The multi-ethnic study of atherosclerosis. J Am Soc Nephrol 29: 2713–2721, 2018
50. Nin JWM, Jorsal A, Ferreira I, Schalkwijk CG, Prins MH, Parving HH, Tarnow L, Rossing P, Stehouwer CD: Higher plasma soluble Receptor for Advanced Glycation End Products (sRAGE) levels are associated with incident cardiovascular disease and all-cause mortality in type 1 diabetes: A 12-year follow-up study. Diabetes 59: 2027–2032, 2010
51. Neirynck N, Glorieux G, Schepers E, Verbeke F, Vanholder R: Soluble tumor necrosis factor receptor 1 and 2 predict outcomes in advanced chronic kidney disease: A prospective cohort study. PLoS One 10: e0122073, 2015
52. Srivastava A, Schmidt IM, Palsson R, Weins A, Bonventre JV, Sabbisetti V, Stillman IE, Rennke HG, Waikar SS: The associations of plasma biomarkers of inflammation with histopathologic lesions, kidney disease progression, and mortality-The Boston Kidney Biopsy Cohort Study. Kidney Int Rep 6: 685–694, 2021
53. Gupta S, Kaplan MJ: The role of neutrophils and NETosis in autoimmune and renal diseases. Nat Rev Nephrol 12: 402–413, 2016
54. Xiao H, Heeringa P, Hu P, Liu Z, Zhao M, Aratani Y, Maeda N, Falk RJ, Jennette JC: Antineutrophil cytoplasmic autoantibodies specific for myeloperoxidase cause glomerulonephritis and vasculitis in mice. J Clin Invest 110: 955–963, 2002
55. Kitching AR, Anders HJ, Basu N, Brouwer E, Gordon J, Jayne DR, Kullman J, Lyons PA, Merkel PA, Savage COS, Specks U, Kain R: ANCA-associated vasculitis. Nat Rev Dis Primers 6: 71, 2020
56. Møller HJ, Peterslund NA, Graversen JH, Moestrup SK: Identification of the hemoglobin scavenger receptor/CD163 as a natural soluble protein in plasma. Blood 99: 378–380, 2002
57. O’Reilly VP, Wong L, Kennedy C, Elliot LA, O’Meachair S, Coughlan AM, O’Brien EC, Ryan MM, Sandoval D, Connolly E, Dekkema GJ, Lau J, Abdulahad WH, Sanders JS, Heeringa P, Buckley C, O’Brien C, Finn S, Cohen CD, Lindemeyer MT, Hickey FB, O’Hara PV, Feighery C, Moran SM, Mellotte G, Clarkson MR, Dorman AJ, Murray PT, Little MA: Urinary soluble CD163 in active renal vasculitis. J Am Soc Nephrol 27: 2906–2916, 2016
58. Moran SM, Monach PA, Zgaga L, Cuthbertson D, Carette S, Khalidi NA, Koening CL, Langford CA, McAlear CA, Moreland L, Pagnoux C, Seo P, Specks U, Sreih A, Wyse J, Ytterberg SR, Merkel PA, Little MA; Vasculitis Clinical Research Consortium: Urinary soluble CD163 and monocyte chemoattractant protein-1 in the identification of subtle renal flare in anti-neutrophil cytoplasmic antibody-associated vasculitis. Nephrol Dial Transplant 35: 283–291, 2020
59. Salahudeen AK, Bonventre JV: Onconephrology: The latest frontier in the war against kidney disease. J Am Soc Nephrol 24: 26–30, 2013
60. Jhaveri KD, Shah HH, Calderon K, Campenot ES, Radhakrishnan J: Glomerular diseases seen with cancer and chemotherapy: A narrative review. Kidney Int 84: 34–44, 2013
61. Rosner MH, Jhaveri KD, McMahon BA, Perazella MA: Onconephrology: The intersections between the kidney and cancer. CA Cancer J Clin 71: 47–77, 2021
62. Said SM, Leung N, Sethi S, Cornell LD, Fidler ME, Grande JP, Herrmann S, Tefferi A, D’Agati VD, Nasr SH: Myeloproliferative neoplasms cause glomerulopathy. Kidney Int 80: 753–759, 2011
63. Tapper EB, Luptakova K, Joyce RM, Tzachanis D: A 78-year-old man with acute myeloid leukemia (AML) and acute renal failure. Am J Case Rep 15: 364–367, 2014
64. Schwotzer N, Provot F, Ville S, Daniel L, Le Fur A, Kissling S, Jourde-Chiche N, Karras A, Moreau A, Augusto JF, Gnemmi V, Perrochia H, Bataille S, Le Quintrec M, Goujon JM, Rotman S, Fakhouri F: Spectrum of kidney involvement in patients with myelodysplastic syndromes. Kidney Int Rep 6: 746–754, 2021
65. Jaiswal S, Fontanillas P, Flannick J, Manning A, Grauman PV, Mar BG, Lindsley RC, Mermel CH, Burtt N, Chavez A, Higgins JM, Moltchanov V, Kuo FC, Kluk MJ, Henderson B, Kinnunen L, Koistinen HA, Ladenvall C, Getz G, Correa A, Banahan BF, Gabriel S, Kathiresan S, Stringham HM, McCarthy MI, Boehnke M, Tuomilehto J, Haiman C, Groop L, Atzmon G, Wilson JG, Neuberg D, Altshuler D, Ebert BL: Age-related clonal hematopoiesis associated with adverse outcomes. N Engl J Med 371: 2488–2498, 2014
66. Genovese G, Kähler AK, Handsaker RE, Lindberg J, Rose SA, Bakhoum SF, Chambert K, Mick E, Neale BM, Fromer M, Purcell SM, Svantesson O, Landén M, Höglund M, Lehmann S, Gabriel SB, Moran JL, Lander ES, Sullivan PF, Sklar P, Grönberg H, Hultman CM, McCarroll SA: Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence. N Engl J Med 371: 2477–2487, 2014
67. Steensma DP, Bejar R, Jaiswal S, Lindsley RC, Sekeres MA, Hasserjian RP, Ebert BL: Clonal hematopoiesis of indeterminate potential and its distinction from myelodysplastic syndromes. Blood 126: 9–16, 2015
68. Jaiswal S, Natarajan P, Silver AJ, Gibson CJ, Bick AG, Shvartz E, McConkey M, Gupta N, Gabriel S, Ardissino D, Baber U, Mehran R, Fuster V, Danesh J, Frossard P, Saleheen D, Melander O, Sukhova GK, Neuberg D, Libby P, Kathiresan S, Ebert BL: Clonal hematopoiesis and risk of atherosclerotic cardiovascular disease. N Engl J Med 377: 111–121, 2017
69. Jaiswal S, Libby P: Clonal haematopoiesis: Connecting ageing and inflammation in cardiovascular disease. Nat Rev Cardiol 17: 137–144, 2020
70. Bick AG, Weinstock JS, Nandakumar SK, Fulco CP, Bao EL, Zekavat SM, Szeto MD, Liao X, Leventhal MJ, Nasser J, Chang K, Laurie C, Burugula BB, Gibson CJ, Lin AE, Taub MA, Aguet F, Ardlie K, Mitchell BD, Barnes KC, Moscati A, Fornage M, Redline S, Psaty BM, Silverman EK, Weiss ST, Palmer ND, Vasan RS, Burchard EG, Kardia SLR, He J, Kaplan RC, Smith NL, Arnett DK, Schwartz DA, Correa A, de Andrade M, Guo X, Konkle BA, Custer B, Peralta JM, Gui H, Meyers DA, McGarvey ST, Chen IY, Shoemaker MB, Peyser PA, Broome JG, Gogarten SM, Wang FF, Wong Q, Montasser ME, Daya M, Kenny EE, North KE, Launer LJ, Cade BE, Bis JC, Cho MH, Lasky-Su J, Bowden DW, Cupples LA, Mak ACY, Becker LC, Smith JA, Kelly TN, Aslibekyan S, Heckbert SR, Tiwari HK, Yang IV, Heit JA, Lubitz SA, Johnsen JM, Curran JE, Wenzel SE, Weeks DE, Rao DC, Darbar D, Moon JY, Tracy RP, Buth EJ, Rafaels N, Loos RJF, Durda P, Liu Y, Hou L, Lee J, Kachroo P, Freedman BI, Levy D, Bielak LF, Hixson JE, Floyd JS, Whitsel EA, Ellinor PT, Irvin MR, Fingerlin TE, Raffield LM, Armasu SM, Wheeler MM, Sabino EC, Blangero J, Williams LK, Levy BD, Sheu WH, Roden DM, Boerwinkle E, Manson JE, Mathias RA, Desai P, Taylor KD, Johnson AD, Auer PL, Kooperberg C, Laurie CC, Blackwell TW, Smith AV, Zhao H, Lange E, Lange L, Rich SS, Rotter JI, Wilson JG, Scheet P, Kitzman JO, Lander ES, Engreitz JM, Ebert BL, Reiner AP, Jaiswal S, Abecasis G, Sankaran VG, Kathiresan S, Natarajan P; NHLBI Trans-Omics for Precision Medicine Consortium: Inherited causes of clonal haematopoiesis in 97,691 whole genomes. Nature 586: 763–768, 2020
71. Cook EK, Izukawa T, Young S, Rosen G, Jamali M, Zhang L, Johnson D, Bain E, Hilland J, Ferrone CK, Buckstein J, Francis J, Momtaz B, McNaughton AJM, Liu X, Snetsinger B, Buckstein R, Rauh MJ: Comorbid and inflammatory characteristics of genetic subtypes of clonal hematopoiesis. Blood Adv 3: 2482–2486, 2019
72. Bick AG, Pirruccello JP, Griffin GK, Gupta N, Gabriel S, Saleheen D, Libby P, Kathiresan S, Natarajan P: Genetic interleukin 6 signaling deficiency attenuates cardiovascular risk in clonal hematopoiesis. Circulation 141: 124–131, 2020
73. Cull AH, Snetsinger B, Buckstein R, Wells RA, Rauh MJ: Tet2 restrains inflammatory gene expression in macrophages. Exp Hematol 55: 56–70.e13, 2017
74. Sano S, Oshima K, Wang Y, MacLauchlan S, Katanasaka Y, Sano M, Zuriaga MA, Yoshiyama M, Goukassian D, Cooper MA, Fuster JJ, Walsh K: Tet2-mediated clonal hematopoiesis accelerates heart failure through a mechanism involving the IL-1β/NLRP3 inflammasome. J Am Coll Cardiol 71: 875–886, 2018
75. Sano S, Oshima K, Wang Y, Katanasaka Y, Sano M, Walsh K: CRISPR-mediated gene editing to assess the roles of Tet2 and Dnmt3a in clonal hematopoiesis and cardiovascular disease. Circ Res 123: 335–341, 2018
76. Arends CM, Weiss M, Christen F, Eulenberg-Gustavus C, Rousselle A, Kettritz R, Eckardt KU, Chan W, Hoyer K, Frick M, Bullinger L, Bieringer M, Schreiber A, Damm F: Clonal hematopoiesis in patients with anti-neutrophil cytoplasmic antibody-associated vasculitis. Haematologica 105: e264–e267, 2020
77. Steensma DP, Bolton KL: What to tell your patient with clonal hematopoiesis and why: Insights from 2 specialized clinics. Blood 136: 1623–1631, 2020
78. Ikezumi Y, Suzuki T, Karasawa T, Hasegawa H, Kawachi H, Nikolic-Paterson DJ, Uchiyama M: Contrasting effects of steroids and mizoribine on macrophage activation and glomerular lesions in rat thy-1 mesangial proliferative glomerulonephritis. Am J Nephrol 31: 273–282, 2010
79. Xing C-Y, Saleem MA, Coward RJ, Ni L, Witherden IR, Mathieson PW: Direct effects of dexamethasone on human podocytes. Kidney Int 70: 1038–1045, 2006
80. Huang LL, Nikolic-Paterson DJ, Han Y, Ozols E, Ma FY, Young MJ, Tesch GH: Myeloid mineralocorticoid receptor activation contributes to progressive kidney disease. J Am Soc Nephrol 25: 2231–2240, 2014
81. Grune J, Beyhoff N, Smeir E, Chudek R, Blumrich A, Ban Z, Brix S, Betz IR, Schupp M, Foryst-Ludwig A, Klopfleisch R, Stawowy P, Houtman R, Kolkhof P, Kintscher U: Selective mineralocorticoid receptor cofactor modulation as molecular basis for finerenone’s antifibrotic activity. Hypertension 71: 599–608, 2018
82. Bakris GL, Agarwal R, Anker SD, Pitt B, Ruilope LM, Rossing P, Kolkhof P, Nowack C, Schloemer P, Joseph A, Filippatos G; FIDELIO-DKD Investigators: Effect of finerenone on chronic kidney disease outcomes in type 2 diabetes. N Engl J Med 383: 2219–2229, 2020
83. Bernstein KE, Khan Z, Giani JF, Cao DY, Bernstein EA, Shen XZ: Angiotensin-converting enzyme in innate and adaptive immunity. Nat Rev Nephrol 14: 325–336, 2018
84. McAdoo SP, Prendecki M, Tanna A, Bhatt T, Bhangal G, McDaid J, Masuda ES, Cook HT, Tam FWK, Pusey CD: Spleen tyrosine kinase inhibition is an effective treatment for established vasculitis in a pre-clinical model. Kidney Int 97: 1196–1207, 2020
85. Kim MJ, McDaid JP, McAdoo SP, Barratt J, Molyneux K, Masuda ES, Pusey CD, Tam FW: Spleen tyrosine kinase is important in the production of proinflammatory cytokines and cell proliferation in human mesangial cells following stimulation with IgA1 isolated from IgA nephropathy patients. J Immunol 189: 3751–3758, 2012
86. O’Sullivan KM, Holdsworth SR: Neutrophil extracellular traps: A potential therapeutic target in MPO-ANCA associated vasculitis? Front Immunol 12: 635188, 2021
87. Nowak KL, Chonchol M: Does inflammation affect outcomes in dialysis patients? Semin Dial 31: 388–397, 2018
88. Ridker PM, MacFadyen JG, Glynn RJ, Koenig W, Libby P, Everett BM, Lefkowitz M, Thuren T, Cornel JH: Inhibition of interleukin-1β by canakinumab and cardiovascular outcomes in patients with chronic kidney disease. J Am Coll Cardiol 71: 2405–2414, 2018
89. Pergola PE, Devalaraja M, Fishbane S, Chonchol M, Mathur VS, Smith MT, Lo L, Herzog K, Kakkar R, Davidson MH: Ziltivekimab for treatment of anemia of inflammation in patients on hemodialysis: Results from a phase 1/2 multicenter, randomized, double-blind, placebo-controlled trial. J Am Soc Nephrol 32: 211–222, 2021
Keywords:

chronic kidney disease; macrophages; chronic inflammation; fibrosis

Copyright © 2022 by the American Society of Nephrology