Abstract
AKI is characterized by a sudden, and usually reversible, decline in kidney function. In mice, ischemia–reperfusion injury (IRI) is commonly used to model the pathophysiologic features of clinical AKI. Macrophages are a unifying feature of IRI as they regulate both the initial injury response as well as the long-term outcome following resolution of injury. Initially, macrophages in the kidney take on a proinflammatory phenotype characterized by the production of inflammatory cytokines, such as CCL2 (monocyte chemoattractant protein 1), IL-6, IL-1β, and TNF-α. Release of these proinflammatory cytokines leads to tissue damage. After resolution of the initial injury, macrophages take on a reparative role, aiding in tissue repair and restoration of kidney function. By contrast, failure to resolve the initial injury results in prolonged inflammatory macrophage accumulation and increased kidney damage, fibrosis, and the eventual development of CKD. Despite the extensive amount of literature that has ascribed these functions to M1/M2 macrophages, a recent paradigm shift in the macrophage field now defines macrophages on the basis of their ontological origin, namely monocyte-derived and tissue-resident macrophages. In this review, we focus on macrophage phenotype and function during IRI-induced injury, repair, and transition to CKD using both the classic (M1/M2) and novel (ontological origin) definition of kidney macrophages.
Keywords: acute kidney failure, AKI
Clinical Overview of AKI
AKI is characterized by the abrupt loss of kidney function resulting in decreased GFR, reduced urine output, and increased serum creatinine.1 The incidence of AKI is 10%–15% in all hospital admissions and >50% in intensive care unit patients, with AKI accounting for over two million deaths every year globally.2,3 There are multiple risk factors associated with worsened outcomes after AKI, including a history of diabetes, male sex, malnutrition, cirrhosis, and older age at presentation.4–6 Causes of AKI can be broadly grouped into three categories: prerenal, intrinsic renal, and postrenal.7 Prerenal AKI is caused by decreased renal perfusion (dehydration, heart failure, hemorrhage, etc.), whereas the cause of postrenal AKI is inadequate urinary drainage (e.g., kidney stones, abdominal or pelvic mass, prostatic disease).8,9 Intrinsic AKI is caused by direct kidney damage that is the result of acute tubular necrosis, acute interstitial nephritis, and vascular diseases such as hemolytic uremic syndrome, vasculitis, and hypertensive emergencies, among others.10,11 Clinically, different criteria are used to define AKI, including Risk, Injury, Failure, Loss of kidney function, and ESKD; AKI Network; and Kidney Disease Improving Global Outcomes (KDIGO); of the three, KDIGO is the one most commonly used in clinic. KDIGO defines AKI as the presence of any of the following.
Increase in serum creatinine by 26.5 µmol/L (0.3 mg/dl) or more within 48 hours.
Decreased urine output with urine volume during the past 6 hours being <0.5 ml/kg per hour.
Within the seven prior days, 1.5 times or more increase in serum creatinine from baseline.
Complications that result from the abrupt loss of GFR include but are not limited to volume overload, acid–base disorders (most commonly metabolic acidosis), uremic complications, and electrolyte disturbances. Presently, there are no available therapies which can prevent the damage and loss of kidney function that occurs during AKI; as a result, patients who develop AKI show an increased risk of developing CKD later in life.2 Treatment options after AKI are limited to RRT in severe cases; however, mortality still approaches 50% in these patients.12
Ischemia–Reperfusion Injury Model of AKI
The ischemia–reperfusion injury (IRI) model is used to mimic clinical ischemic injury to the kidney and involves a unilateral or bilateral clamping of the renal vessels for a defined period of time.13 IRI is characterized by a temporary loss of blood flow to the tissue (ischemia) followed by restoration of blood flow (reperfusion).13 After IRI, there is an increase in oxidative damage, endothelial dysfunction, cell death via apoptosis and necrosis, and immune cell accumulation, eventually resulting in reduced kidney function.14,15 Importantly, inflammation, which is largely driven by macrophages, is a key hallmark of the response after IRI.16
Macrophages
The first description of phagocytic cells came from Elie Metchnikoff in 1883 when he observed that starfish larvae injected with thorns were quickly surrounded by white blood cells.17 These cells would later be termed macrophages, owing to their large size (macro) and ability to engulf foreign particles (phage). In vivo, the primary function of macrophages is the recognition, engulfment, and digestion of pathogens, cellular debris, and foreign substances, contributing significantly to both innate and adaptive immune response.18,19 Macrophages also play a pivotal role in tissue repair, inflammation modulation, and antigen presentation, making them essential contributors to immune surveillance and homeostasis.18 In the kidney, macrophages are involved in both the early and sustained responses to injury,19,20 as outlined below.
Epithelial–Macrophage Crosstalk after Injury
After IRI, damaged and dying tubular epithelial cells (TECs) release damage-associated molecular patterns (DAMPs), also known as alarmins and danger signals, that bind to pattern recognition receptors (PRRs) on macrophages.21 PRRs can be expressed in intracellular (e.g., cytosolic, endosomal, and cellular membranes) or extracellular compartments (e.g., secreted forms present in interstitial fluid and bloodstream).22–24 PRRs consist of four subfamilies—the nucleotide-binding oligomerization domain, leucine-rich repeats containing receptors, the Toll-like receptors, the retinoic acid-inducible gene 1–like receptors (aka retinoic acid-inducible gene 1–like helicases), and the C-type lectin receptors.25 These various PRRs recognize DAMPs, initiating an inflammatory response that coordinates leukocyte recruitment, upregulates proinflammatory cytokine production, and shapes the overall immune response.26
In this review, we begin by summarizing previous literature implicating M1 and M2 macrophages in IRI. We then describe how the recent literature has shifted away from describing macrophages on the basis of M1/M2 polarization status and, instead, defines macrophages on the basis of ontological origin (i.e., where they come from). Finally, we finish by highlighting recent data that describe the involvement of macrophages of different ontologic origins in IRI and other mouse models of AKI.
The M1/M2 Macrophage Paradigm
It has long been accepted that macrophages are highly plastic cells that can adapt their phenotype and function to meet the ongoing challenges in the tissue. For example, in vitro studies demonstrated that macrophages can polarize into a proinflammatory (M1) or anti-inflammatory (M2) phenotype depending on the cytokine environment.27 In vitro treatment of bone marrow–derived macrophages with lipopolysaccharide and IFN-γ results in a proinflammatory M1-like macrophage phenotype, whereas glucocorticoids, immune complexes, and IL-4/IL-13 induce an anti-inflammatory, M2-like phenotype.27 Each macrophage phenotype is associated with hallmark cytokine production; TNFα, IL-1β, inducible Nitric Oxide Synthase, and IL-6 for M1 macrophages, and TGF-β, IL-10, ARG1, and CD206 for M2 macrophages.27 Although it is important to highlight that M1/M2 polarization is a byproduct of in vitro cell culture conditions and macrophages in vivo often express markers associated with both M1 and M2 macrophages,28,29 the take-home message from these studies is that M1 macrophages have a proinflammatory phenotype,30–34 while M2 macrophages have a proreparative phenotype.35 Thus, it is believed that M1 macrophages play an important role in the early phase of AKI by removing dead and damaged cells, activating other immune cells via inflammatory cytokine production, and phagocytizing cell debris. By contrast, M2 macrophages produce anti-inflammatory and reparative cytokines, such as fibroblast growth factor, TGF-β, and vascular endothelial growth factor, which can drive both epithelial repair and progression to CKD, depending on the time in tissue.36 Below, we discuss the M1 and M2 driven proinflammatory and anti-inflammatory pathways involved in IRI in vivo, as well as the functional importance of each population in response to IRI.
M1 Macrophage (Proinflammatory) Pathways
Nuclear Factor Kappa-Light-Chain-Enhancer of Activated B Cells Pathway
After IRI, proinflammatory macrophages promote kidney damage by activating a number of transcription factors, including nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB).37 In its inactive state, NF-κB is sequestered in the cytoplasm by the inhibitory κB (IκB) proteins.38 NF-κB can be activated by two main pathways: the canonical (classic) and noncanonical (alternative) pathways.39 Pathogen associated molecular patterns (such as lipopolysaccharide) and proinflammatory cytokines (TNFα, IL-1β) can initiate the canonical pathway via binding to cell surface receptors, including PRRs, resulting in phosphorylation of the IκB kinase (IKK) (Figure 1). IKK activation leads to phosphorylation and subsequent ubiquitin-mediated degradation of IκB proteins, resulting in nuclear translocation of NF-κB and associated family members. P50/NF-κB1-c-Rel, p50/NF-κB1-p65/RelA, and less commonly P65/RelA, p50/NF-κB1, and c-Rel are canonical NF-κB signaling members.40
Figure 1.
The NF-κB and JAK-STAT pathways are increased in proinflammatory macrophages. DAMP, damage-associated molecular pattern; JAK-STAT, Janus kinase/signal transducer and activator of transcription; LTβR, lymphotoxin-β receptor; PRR, pattern recognition receptor; RANK, receptor activator for nuclear factor κB.
By contrast, B-cell activating factor receptor, lymphotoxin-β receptor, fibroblast growth factor-inducible factor-14 (Fn14), and receptor activator for nuclear factor κB are the main activators of the noncanonical pathway (Figure 1).41 After receptor stimulation, NF-κB inducing kinase is activated, which phosphorylates IKKα, thus leading to phosphorylation of p100 and degradation of its IκB-like C terminal.42 This results in translocation of the noncanonical dimer complex p52/NF-κB2-RelB to the nucleus.43
After nuclear translocation, NF-κB induces transcription of a wide variety of cytokines (TNF-α, IL-1, IL-2, IL-6), chemokines (CCL2, CCL3, CXCL8, CCL5), acute phase reactants (serum amyloid A), inducible effector enzymes (cyclooxygenase-2 and inducible nitric oxide synthase), and adhesion molecules (vascular cell adhesion molecule 1, intercellular adhesion molecule 1, and E-selectin) that drive the inflammatory process.38,44,45 It is likely that this is a critical pathway during the acute phase of injury46 as the inhibition of NF-κB signaling decreases the inflammatory response and fibrosis in the kidney after IRI.47,48
Janus Kinase/Signal Transducer and Activator of Transcription Pathway
The Janus kinase/signal transducer and activator of transcription (JAK-STAT) signaling pathway is an intracellular signal transduction pathway that consists of three main components: signal transducer and activator of transcription 1–6, tyrosine kinase–associated receptors, and Janus kinases (JAK1, JAK2, TYK2, and JAK3).49 The JAK-STAT pathway can be induced by cytokines, including IL-3, IL-5, granulocyte-macrophage colony stimulating factor and reactive oxygen species.50 Upon a ligand binding to its cognate receptor, the phosphorylation of a tyrosine residue on bound transcription factors (STATs) by the receptor-associated cytosolic tyrosine kinases (JAKs) results in nuclear translocation and gene transcription augmentation (Figure 1).51,52 In IRI, the activation of the JAK-STAT signaling pathway induces transcription of multiple proinflammatory cytokines.53 The JAK/STAT signaling pathway is also responsible for the recruitment of leukocytes54 and exacerbates renal injury by stimulating macrophages to secrete cytokines, such as TNF-α, IL-6, and IL-1β.54 It may be possible to target the JAK-STAT pathway in macrophages to reduce IRI-associated kidney damage. For example, inhibition of the JAK/STAT signaling cascade has been shown by several groups to reduce kidney injury and improve phenotypic outcome after IRI.54–56
Effect of Macrophage Depletion during the Acute Phase of Injury
On the basis of their production of proinflammatory cytokines, it was predicted that genetic or pharmacologic blockade or depletion of proinflammatory M1-like macrophages would reduce IRI-induced kidney damage. In agreement with this hypothesis, depletion of macrophages before IRI with liposomal clodronate (LC), an encapsulated bisphosphonate that induces apoptosis upon phagocytic uptake,57–60 significantly reduced kidney damage.61–66 The improved phenotypic outcome after IRI was associated with reduced proinflammatory cytokine production. Importantly, reintroduction of proinflammatory macrophages to macrophage-depleted mice resulted in worsened kidney phenotype,63 providing conclusive evidence that proinflammatory macrophages, which accumulate during the acute phase of injury, promote disease progression. One interesting observation from the study of proinflammatory macrophages in IRI is data showing that the depletion of macrophages with LC, but not using CD11b-DTR or CD11c-DTR mice, significantly improved kidney damage.65,66 This occurred despite the fact that all models had a similar reduction in kidney macrophage number. Instead, the authors hypothesized that the failure to rescue the kidney phenotype in CD11b-DTR and CD11c-DTR mice compared with LC-treated mice was mainly driven by differential effects on proinflammatory cytokines, such as IL-6 and monocyte chemoattractant protein 1.66 These data suggest that additional macrophage heterogeneity is present in the kidney and that each depletion method might differentially affect macrophage subsets.
M2 Macrophage (Proreparative) Pathways
ARG1
After injury, exposure to dead cell debris leads to increased granulocyte-macrophage colony stimulating factor expression by outer medullary epithelial cells, prompting increased expression of Arginase-1 (Arg1) by adjacent macrophages.67,68 Arg1, which is responsible for conversion of arginine into ornithine and eventually polyamines, is involved in the repair process through secretion of a yet unidentified factor or factors that promote tubular cell proliferation in the kidney after injury.69,70 In mice, loss of Arg1 expression in macrophages was associated with decreased tubular cell proliferation, slowed repair, worsened BUN, increased creatinine, and significantly reduced survival after AKI.67 They also found that creatinine and BUN values were not different between groups 1 day after injury, suggesting that arginase-1 is specifically required for macrophage-dependent repair after IRI.
Wnt-β Catenin Pathway
Macrophages promote tubular repair via the production of Wnt ligands.71 In healthy kidneys, canonical Wnt-β-catenin pathway activity is restricted to the renal papilla and the transcription of downstream β-catenin genes is repressed by a destruction complex. By contrast, after injury, the Wnt-β-catenin pathway is activated in both the cortical and medullary region of the kidney.71 Extracellular Wnt ligands bind to Frizzled protein (FZD), activating low-density lipoprotein receptor-related proteins (LRP5/6), and disheveled protein.72 Disheveled protein then disrupts the destruction complex, causing β-catenin accumulation in the cytoplasm and, ultimately, nuclear translocation (Figure 2). On entering the nucleus, β-catenin activates the t-cell factor/lymphoid enhancer factor transcription complex, mediating the transcription of genes associated with cell–cycle progression (n-myc, c-jun, c-myc, cyclin D1, and Bcl-w) and epithelial regeneration.72–75 In macrophages, deletion of Wnt7b in colony-stimulating factor-1 receptor expressing macrophages resulted in an impaired reparatory response after IRI,71 suggesting this pathway plays an important role in resolving damage in the kidney after injury. In agreement with this hypothesis, data from our laboratory showed that kidney resident macrophages (KRM) isolated from mice undergoing IRI had increased the expression of Wnt-associated genes,76 once again suggesting a functional role for Wnt signaling after injury.
Figure 2.
Macrophage-derived IL-22 and Wnt drive epithelial repair after injury. DVL, disheveled protein.
IL-22
IL-22 plays an important role in promoting recovery after IRI by promoting TEC proliferation.77,78 In IRI, IL-22 is predominantly expressed by mononuclear cells, including dendritic cells and macrophages, whereas the IL-22R is expressed by multiple cell types, including tubular epithelium.79,80 Upon ligand/receptor interaction, downstream signaling is initiated through the Jak1/Stat1/3/5 signaling cascade. IL-22R engagement also induces the activation of PI3K/Akt and mitogen-activated protein kinase pathways.79 Using an in vivo system of TEC regeneration, Xu et al. injected mice with adenovirus expressing IL-22 intravenously 5 days before renal IRI. They showed that increased IL-22 resulted in accelerated epithelial cell growth and improved kidney function after IRI, as measured by higher levels of serum creatinine and BUN in the control group as compared with IL-22 administered mice.78 The authors proposed that protective effects of IL-22 were dependent on the activation of AKT and STAT3 in the proximal TECs (Figure 2).78 They also observed increased Bcl-2, a key regulator of apoptosis, after treatment with IL-22, suggesting an additional mechanism through which this cytokine could exert a reparative effect. Similarly, IL-22 depletion studies showed that reduced levels of IL-22 leads to impaired epithelial repair and functional recovery after AKI.80
Effect of Macrophage Depletion on the Reparative Phase of Injury
Macrophages play an important role in the reparative phase after IRI.81,82 To test the functional importance of macrophages during the recovery phase after IRI, Lee and colleagues subjected mice to unilateral IRI and contralateral nephrectomy followed by LC-mediated macrophage depletion 48 and 72 hours after injury.63 Importantly, this approach did not affect the initial recruitment of inflammatory monocytes to the kidney during the acute phase of injury. LC-treated mice exhibited substantial diminution of tubule regeneration with fewer epithelial cells compared with the controls.63 BUN values were indistinguishable in both groups during the initial phase; however, macrophage-depleted mice exhibited less improvement in GFR at days 5 and 7 after injury.63 In agreement with these data, data collected from several other groups showed that depletion of M2-like macrophages, which accumulate during the reparative phase, resulted in failed tubular repair, worsened kidney function, and increased interstitial fibrosis.63,83–85 While most data suggest that M2-like macrophages promote repair after injury, data from other groups showed that depletion of M2-like macrophages improved histology, reduced kidney injury, and improved fibrosis outcomes.86–88 Thus, the exact role of M2-like macrophages in IRI-induced kidney damage warrants further investigation.
Kidney Fibrosis and AKI to CKD Transition
Fibrosis is the process whereby normal renal tissue is replaced by extracellular matrix (ECM). On the basis of the published data, it is hypothesized that macrophages play a profibrotic role in the kidney as depletion of macrophages resulted in reduced renal fibrosis.89 However, as mentioned above, the ability of macrophages to promote fibrosis after IRI is still controversial. There are two proposed mechanisms through which macrophages may promote fibrosis: the persistent accumulation of proreparative macrophages that produce profibrotic cytokines, such as TGF-β1,90 and the inability of macrophages to switch from the proinflammatory to the proreparative phenotype.91 Both of these processes lead to progressive renal inflammation and excessive deposition of ECM.
TGF-β1
The TGF-β pathway is the main signaling pathway that is involved in fibrosis; TGF-β is thought to be mainly produced by macrophages92,93 and signals through two major receptors, TGF-β RII (TβRII) and TGF-βRI (TβRI).94 On binding to its receptors, TGF-β induces the downstream phosphorylation of Smad2/3, which forms a complex with Smad4, to induce transcription of TGF-β target genes, including collagen and fibronectin.95 Macrophages are the major producers of TGF-β after IRI.96 Excessive production of this cytokine over time drives the accumulation of myofibroblasts, cells that deposit large amounts of ECM proteins, including collagens (Figure 3).97,98 The exact precursor cell that gives rise to myofibroblasts is not well understood, although it has been proposed that they may arise from epithelial to mesenchymal transition, endothelial to mesenchymal transition, fibrocyte recruitment, pericyte recruitment, and resident fibroblast recruitment.99 TGF-β pathway activation also results in activation of the NLRP3, Smad3, and Caspase-1 stimulators, which further exacerbate renal fibrosis.100 As a result, an imbalance develops between ECM synthesis and degradation, promoting deposition of ECM components in the kidney, glomerular sclerosis, and reduced renal function.101 Despite a significant amount of literature pointing toward macrophage-derived TGF-β being a major driver of fibrosis, data indicate that genetic deletion of Tgfb1 in myeloid lineage cells had minimal impact on fibrosis after severe IRI.96 Thus, there are likely multiple profibrotic cytokines produced by macrophages that drive fibrosis and CKD transition after IRI.
Figure 3.
TGF-β released from macrophages drives myofibroblast accumulation and ECM production after IRI. ECM, extracellular matrix; IRI, ischemia reperfusion injury.
Kidney Macrophage Classification On the Basis of Recent Literature
Recent literature has highlighted the fact that macrophages isolated in vivo express genes associated with both M1 and M2 macrophages.76 Thus, it has become evident that the original description of macrophages as M1 or M2, which was largely driven by in vitro data, does not fully recapitulate the heterogeneity of these cells in vivo. As such, recent literature favors the identification of macrophages on the basis of their ontological origin. For the purpose of this review, we will begin by defining kidney macrophage subsets in the mouse on the basis of ontologic origin. We will then briefly comment on how data from the mouse may correlate to humans.
In the mouse, it was initially believed that all macrophages originated from bone morrow monocytes.102,103 However, several paradigm shifting studies have shown that macrophages in all tissues are derived both from bone marrow monocytes and embryonic precursors.104,105 In these studies, the authors identified two distinct macrophage populations that were present in almost all tissues: monocyte-derived macrophages and tissue-resident macrophages.104–106 Monocyte-derived infiltrating macrophages (IM) originate from adult bone marrow monocytes, express high levels of CD11b and intermediate levels of F4/80 (CD11bhi, F4/80lo), and are rapidly turned over throughout the animal's lifespan (Figure 4A).107 By contrast, tissue-resident macrophages are mainly derived from embryonic precursors, express intermediate levels of CD11b and high levels of F4/80 (CD11blo, F4/80hi), and largely maintain their population through self-proliferation (Figure 4A).108–110
Figure 4.
Kidney macrophage ontogeny during development and after ischemia reperfusion injury. (A) Macrophages originate from the yolk sac, fetal liver, and bone marrow. (B) Summary of macrophage number, grouped by ontological origin, at different time points after AKI. KRM, kidney-resident macrophages.
From the start of kidney development and throughout life, KRM derive from progenitors in waves. For example, the first wave of KRM precursors entering the kidney come from yolk sac–derived erythromyeloid (CD45+CD11bloF4/80hiLy6C− cells) progenitors (EMPs) at approximately embryonic day (E) 12.5.111 Between days E13.5 and E16.5, fetal liver monocyte-derived KRM precursors begin entering the developing kidney and increase progressively, with their numbers rapidly exceeding those of yolk sac–derived macrophages by E16.5.111,112 It is still not clear whether these fetal liver monocytes come from aorta–gonad–mesonephros hematopoietic stem cells (HSCs), EMPs, or a combination of both.107,111,112 Finally, after birth, a third wave of KRM precursors derived from bone marrow HSCs begin to enter the kidney and engraft into the KRM niche; this process continues until the mice reach adulthood (approximately 12 weeks).113 Thus, KRM found in adult mice are derived from a combination of EMP (approximately 10%), fetal liver monocytes (approximately 60%), and adult, bone marrow–derived HSCs (approximately 30%).104,113,114
Despite extensive research in mice, there have been significant limitations preventing the identification of similar macrophage subsets in humans. For example, the main marker used to identify mouse-resident macrophages, F4/80, is also expressed by human eosinophils,115 although these findings have recently been called into question.116 Thus, until recently, it has not been possible to identify evolutionarily conserved macrophage populations across species. This gap in knowledge was recently addressed in a publication by our laboratory.117 In this study, we performed single-cell RNA sequencing (scRNAseq) on macrophages isolated from the kidney of adult mice, rat, pig, and humans. Using this approach, we identified a novel list of mouse KRM specific genes, including C1qa, C1qb, C1qc, Cd74, and Cd81, and showed that several of these genes were expressed in a single cluster of immune cells isolated from rat, pig, and human kidney tissue. We went on to validate that a single cluster of CD45+ immune cells from mouse, rat, and human kidney tissue expressed several of these proteins via flow cytometry. Furthermore, we showed that we could use our new KRM markers to identify a population of cells in the rat that received minimal input from peripheral monocytes, a hallmark of resident macrophages in the mouse.118 Thus, our studies identified an evolutionarily conserved KRM signature (C1qa, C1qb, C1qc, Cd81, Cd74) that was present in KRM across multiple species, including humans. The markers identified in our studies were independently confirmed in a separate study.119 In summary, there are two major populations of macrophages present in the kidney (IM, KRM) with distinct ontologic origins. Furthermore, our scRNAseq data indicate that both populations are evolutionarily conserved across multiple mammalian species.
How Macrophages of Different Origins Respond after IRI
Recently, new studies have shed light on the behavior of KRM and IM subsets during AKI.76,120–122 To understand how macrophages of each origin respond to AKI, Yao and colleagues performed scRNAseq on CD11b and F4/80 positive cells isolated from the kidneys of mice 1 or 3 days post 45 minutes of unilateral IRI.121 Analysis of combined scRNAseq at all time points revealed four clusters of KRM, including MHC2hi, Ccl4hi, Mrc1hi, and Slc40a1hi subsets. Analysis of cluster composition revealed a significant reduction in the number of MHC2hi, Ccl4hi, Mrc1hi KRM subsets 1 day postinjury; the number of all three subsets partially rebounded toward control levels on day 3 (Figure 4B).121 By contrast, the number of Slc40a1hi KRM increased day 1 postinjury followed by their subsequent reduction 3 days postinjury (Figure 4B). In homeostasis, KRM were involved in antigen processing and presentation (MHC2hi, Ccl4hi subsets), myeloid cell migration and differentiation (Ccl4hi, Mrc1hi subsets) and response to metal ions (Slc40a1hi). One day after injury, phagocytosis (MHC2hi subset), inflammatory (Ccl4hi subset), anti-inflammatory (Mrc1hi subset), and wound repair (Mrc1hi subset) functions were enhanced in KRM. Slc40a1hi KRM mainly expressed angiogenesis and wound repair genes, including Vcam1, Igf1, and Tnfaip2 on day 3.
The authors also identified six clusters of monocyte‐derived IMs, including four clusters of Ly6chi IMs (S100a9hi, Chil3hi, Fn1hi, and Hbb-bshi), one Ly6cint IM cluster (Plac8hi), and one cluster of Ly6clo IMs (Ace hi).117,121 Analysis of cluster abundance indicates that all IM subsets increased in frequency on day 1, followed by a reduction in number on day 3 after IRI (Figure 4B). Ly6chi IMs functions were associated with clearance of apoptotic cells and maintenance of the acute inflammatory response. Ly6clo IM functions were associated with maintaining endothelial stability and regulating vasculogenesis. KRMs and IMs exhibited a similar ability to activate leukocytes and phagocytose cells after injury. Collectively, these data suggest that the ontologic origin of macrophages influences their function with KRM being involved in antigen processing and presentation, phagocytosis, inflammation, and would repair. By contrast, IMs were involved in driving the acute inflammatory response, maintaining endothelial stability, and regulating vasculogenesis.
Function of KRM and IM during IRI
Despite data indicating the IM and KRM number and subsets are altered after IRI, there are limited data on the specific role of these subsets in disease progression, mainly owing to the inability to specifically target these cells. To tease out the function of IM and KRM in IRI, Park and colleagues took advantage of IM and KRM niche filling kinetics after LC injection.123 In these studies, the authors showed that KRM numbers were unchanged 72 hours post-IRI, whereas IM numbers were substantially increased,123 similar to the scRNAseq data described above.121 The authors then performed IRI in LC treated mice 2 weeks postdepletion, a time point at which KRM numbers were reduced while IM numbers were unaffected. Using this approach, the authors found that depletion of KRM resulted in worsened tubular injury and increased terminal deoxynucleotidyl transferase–mediated digoxigenin–deoxyuridine nick-end labeling positive cells suggesting that KRM promote tubular repair.123 In agreement with these data, depletion of KRM increased fibrosis in a renal artery stenosis model of ischemic kidney injury.124 The protective effect of KRM in this model was due to the production of proangiogenic and reparative factors by KRM, including Il10, Arg2, Tgfb2, and Tgfbr3.124 Additional data using a CD169-DTR mouse to deplete KRM support the idea that KRM play a protective role in IRI.125 Loss of CD169+ KRM was associated with increased inflammation, increased expression of Icam1, and worsened kidney function.125 Data from our laboratory further support the idea that KRM protect from IRI-induced kidney damage as KRM that accumulated after IRI injury had increased expression of genes associated with repair after injury, including Wnt family genes.76 Of interest, the transcriptional signature of KRM after injury mirrored that of KRM isolated from postnatal day 7 (P7) mice suggesting that dedifferentiation of KRM may promote epithelial repair and functional recovery after IRI.76 How the dedifferentiated KRM identified by our group corresponds to the KRM subsets identified via scRNAseq is unknown. In addition, data from these studies suggest that KRM have additional functions after IRI beyond production of reparative factors; these include phagocytic capacity to remove dead and dying epithelial cells, activation of the adaptive immune system, recruitment of additional immune cells, and production of proangiogenic factors that promote vessel repair after injury. Of interest, a recent study by Cheung et al. showed that injury changes the spatial distribution of KRMs. In this study, the authors identified seven distinct KRM subpopulations using scRNAseq, flow cytometry, and spatial transcriptomics and observed that after injury, the original localization of each subpopulation was disrupted and was not restored until at least 28 days after IRI.120 Once again, how these subsets correspond to previous data remain uncertain.
Additional data supporting a protective role for KRM after IRI come from studies focused on the CSF1/CSF1R signaling axis. Importantly, CSF1 signals through the CSF1R receptor expressed on KRM to drive cell survival and proliferation126,127; thus, it may be extrapolated that studies targeting the CSF1/CSF1R signaling axis were mainly focused on KRM. Treatment of mice with CSF1 beginning 3 days postinjury reduced tubular damage and improved kidney function when analyzed day 7.128 Moreover, the improvement in kidney function after CSF1 treatment was associated with increased expression of reparative cytokines, including Wnt7b and Igf1.128 Similarly, Zhang and colleagues showed that genetic or pharmacologic inhibition of CSF1 signaling reduced macrophage proliferation, increased inflammatory gene expression, and worsened kidney function after IRI.129 While the protective effect of CSF1/CSF1R signaling after IRI likely involves macrophages, one study proposed that CSF1 could directly signal to the injury tubular epithelium to drive tissue repair.84 However, selective deletion of epithelial cell–derived CSF1 prevented accumulation of reparative macrophages after IRI, delayed functional and structural recovery, and increased tubulointerstitial fibrosis.130 Thus, it is likely that the protective effects of the CSF1/CSF1R signaling axis after IRI are dependent on KRM.
The involvement of IM in IRI is clearer due to studies using Ccr2 deficient mice, which are unable to mobilize Ly6chi monocytes out of the bone marrow and to the site of injury.131 In particular, Ccr2 knockout mice were protected from IRI-induced kidney damage.132,133 The protective effect of IM deficiency after IRI was mainly attributable to reduced production of proinflammatory cytokines in the kidney, including IL-1α, IL-6, IL-12p40, and TNF-α.133
The Role of Macrophages in Other Models of AKI
This review focuses on the role of macrophages in the renal ischemia–reperfusion model of AKI; however, macrophages are also implicated in other models of AKI. While the role of macrophages in each of these models can differ, hallmarks of their function after AKI remain the similar.16 For example, after the initial injury, damaged TECs release DAMPs that bind to PRR on innate immune cells to induce phagocytosis, maturation of phagolysosomes, antigen presentation, and production of proinflammatory cytokines. In addition, IM are quickly recruited to the kidney where they produce proinflammatory cytokines, such as IL-1β, TNFα, IL-12, IL-18, and IL-23, to drive tissue damage. After resolution of the initial injury, M2-like macrophages accumulate in the kidney and promote functional recovery. Once again, prolonged accumulation of M2-like macrophages in other models of kidney injury results in fibrosis.134–136 Despite similarities in the pathophysiological process after injury, subtle differences in macrophage involvement are appreciated. For example, loss of IM (via Ccr2 knockout) did not affect cisplatin-induced AKI, whereas LC treatment to deplete KRM improved phenotypic outcome.137 However, it should be noted that these studies used a repeated low-dose cisplatin injection to induce AKI, which may alter macrophage response and involvement when compared with mice receiving a single injury event to induce AKI. For a more comprehensive view of macrophage behavior in other models of AKI, we refer readers to several other excellent studies.138–140
Macrophages play a multifunctional role in AKI, affecting both the injury and repair process. The versatile role of macrophages is heavily influenced by the kidney microenvironment, and the growth factors, cytokines, and chemokines present therein. In this review, we highlighted how the understanding of macrophages, and their phenotypes and function in the kidney, has changed over time. In the past, there was a strong focus on the M1 and M2 phenotypes, whereas recent studies focus more on ontological origin and how macrophage origin relates to function. We also observed that macrophages play an important role in the repair process, and by further exploring this in future, it may be possible to harness their reparative powers to promote tubular repair and recovery of kidney function.
Footnotes
B.M. and M.S. share first authorship.
Disclosures
K.A. Zimmerman reports the following: Ownership Interest: Regulus Therapeutics; and Patents or Royalties: OUHSC. All remaining authors have nothing to disclose.
Funding
K.A. Zimmerman: National Institute of Diabetes and Digestive and Kidney Diseases (K01DK119375 and R01DK129255-01A1), Presbyterian Health Foundation (Seed grant, Team science grant and Bridge grant), and Polycystic Kidney Disease Research Foundation (grant 826369).
Author Contributions
Conceptualization: Bibi Maryam, Morgan E. Smith.
Writing – original draft: Bibi Maryam.
Writing – review & editing: Bibi Maryam, Sarah J. Miller, Hariharasudan Natarajan, Morgan E. Smith, Kurt A. Zimmerman.
References
- 1.Bellomo R, Kellum JA, Ronco C. Acute kidney injury. Lancet. 2012;380(9843):756–766. doi: 10.1016/S0140-6736(11)61454-2 [DOI] [PubMed] [Google Scholar]
- 2.Huen SC, Cantley LG. Macrophages in renal injury and repair. Annu Rev Physiol. 2017;79:449–469. doi: 10.1146/annurev-physiol-022516-034219 [DOI] [PubMed] [Google Scholar]
- 3.Ronco C, Bellomo R, Kellum JA. Acute kidney injury. Lancet. 2019;394(10212):1949–1964. doi: 10.1016/S0140-6736(19)32563-2 [DOI] [PubMed] [Google Scholar]
- 4.Muroya Y He X Fan L, et al. Enhanced renal ischemia-reperfusion injury in aging and diabetes. Am J Physiol Renal Physiol. 2018;315(6):F1843–F1854. doi: 10.1152/ajprenal.00184.2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Montgomerie C, Spaak J, Evans M, Jacobson SH. Acute kidney injury: clinical characteristics and short-term outcomes in 1,519 patients. Kidney Dis (Basel). 2023;9(1):39–48. doi: 10.1159/000527299 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Alshogran OY, Altawalbeh SM, Almestarihi EM. Acute kidney injury development and impact on clinical and economic outcomes in patients with cirrhosis: an observational cohort study over a 10-year period. Eur J Gastroenterol Hepatol. 2023;35(4):497–504. doi: 10.1097/MEG.0000000000002520 [DOI] [PubMed] [Google Scholar]
- 7.Koza Y. Acute kidney injury: current concepts and new insights. J Inj Violence Res. 2016;8(1):58–62. doi: 10.5249/jivr.v8i1.610 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Turgut F, Somfai GM, Heussen FM, Eberle A, de Smet MD, Becker MD. Robot-assisted epiretinal membrane peeling: a prospective assessment of pre- and intra-operative times and of surgeons' subjective perceptions. J Clin Med. 2023;12(8):2768. doi: 10.3390/jcm12082768 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Gursu M Yegenaga I Tuglular S, et al. Acute kidney injury in Turkey: epidemiological characteristics, etiology, clinical course, and prognosis. BMC Nephrol. 2022;23(1):326. doi: 10.1186/s12882-022-02933-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Jacob J, Dannenhoffer J, Rutter A. Acute kidney injury. Prim Care. 2020;47(4):571–584. doi: 10.1016/j.pop.2020.08.008 [DOI] [PubMed] [Google Scholar]
- 11.Amin AA, Alabsawy EI, Jalan R, Davenport A. Epidemiology, pathophysiology, and management of hepatorenal syndrome. Semin Nephrol. 2019;39(1):17–30. doi: 10.1016/j.semnephrol.2018.10.002 [DOI] [PubMed] [Google Scholar]
- 12.Levey AS, James MT. Acute kidney injury. Ann Intern Med. 2017;167(9):Itc66–itc80. doi: 10.7326/AITC201711070 [DOI] [PubMed] [Google Scholar]
- 13.Hesketh EE Czopek A Clay M, et al. Renal ischaemia reperfusion injury: a mouse model of injury and regeneration. J Vis Exp. 2014;88:51816. doi: 10.3791/51816 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Nieuwenhuijs-Moeke GJ Pischke SE Berger SP, et al. Ischemia and reperfusion injury in kidney transplantation: relevant mechanisms in injury and repair. J Clin Med. 2020;9(1):253. doi: 10.3390/jcm9010253 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Bonventre JV, Yang L. Cellular pathophysiology of ischemic acute kidney injury. J Clin Invest. 2011;121(11):4210–4221. doi: 10.1172/JCI45161 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Han HI, Skvarca LB, Espiritu EB, Davidson AJ, Hukriede NA. The role of macrophages during acute kidney injury: destruction and repair. Pediatr Nephrol. 2019;34(4):561–569. doi: 10.1007/s00467-017-3883-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Metchnikoff E. Untersuchungen uber die intracellulare Verdauung bei Wirbellosen Thieren. Arbeit Zoologischen Instituten, Universitat Wien; 1883:141–168. [Google Scholar]
- 18.Lendeckel U, Venz S, Wolke C. Macrophages: shapes and functions. ChemTexts. 2022;8(2):12. doi: 10.1007/s40828-022-00163-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Duffield JS. Macrophages and immunologic inflammation of the kidney. Semin Nephrol. 2010;30(3):234–254. doi: 10.1016/j.semnephrol.2010.03.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Cao Q, Harris DC, Wang Y. Macrophages in kidney injury, inflammation, and fibrosis. Physiology (Bethesda). 2015;30(3):183–194. doi: 10.1152/physiol.00046.2014 [DOI] [PubMed] [Google Scholar]
- 21.Rasaei R, Sarodaya N, Kim KS, Ramakrishna S, Hong SH. Importance of deubiquitination in macrophage-mediated viral response and inflammation. Int J Mol Sci. 2020;21(21):8090. doi: 10.3390/ijms21218090 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Creagh EM, O'Neill LA. TLRs, NLRs and RLRs: a trinity of pathogen sensors that co-operate in innate immunity. Trends Immunol. 2006;27(8):352–357. doi: 10.1016/j.it.2006.06.003 [DOI] [PubMed] [Google Scholar]
- 23.Takeuchi O, Akira S. Pattern recognition receptors and inflammation. Cell. 2010;140(6):805–820. doi: 10.1016/j.cell.2010.01.022 [DOI] [PubMed] [Google Scholar]
- 24.Medzhitov R, Janeway CA, Jr. Innate immunity: impact on the adaptive immune response. Curr Opin Immunol. 1997;9(1):4–9. doi: 10.1016/s0952-7915(97)80152-5 [DOI] [PubMed] [Google Scholar]
- 25.Walsh D, McCarthy J, O'Driscoll C, Melgar S. Pattern recognition receptors--molecular orchestrators of inflammation in inflammatory bowel disease. Cytokine Growth Factor Rev. 2013;24(2):91–104. doi: 10.1016/j.cytogfr.2012.09.003 [DOI] [PubMed] [Google Scholar]
- 26.McKay DB. Intracellular pattern recognition receptors and renal ischemia. Crit Rev Immunol. 2011;31(4):297–306. doi: 10.1615/critrevimmunol.v31.i4.20 [DOI] [PubMed] [Google Scholar]
- 27.Vogel DY Glim JE Stavenuiter AW, et al. Human macrophage polarization in vitro: maturation and activation methods compared. Immunobiology. 2014;219(9):695–703. doi: 10.1016/j.imbio.2014.05.002 [DOI] [PubMed] [Google Scholar]
- 28.Fu J Sun Z Wang X, et al. The single-cell landscape of kidney immune cells reveals transcriptional heterogeneity in early diabetic kidney disease. Kidney Int. 2022;102(6):1291–1304. doi: 10.1016/j.kint.2022.08.026 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Malone AF, Wu H, Fronick C, Fulton R, Gaut JP, Humphreys BD. Harnessing expressed single nucleotide variation and single cell RNA sequencing to define immune cell chimerism in the rejecting kidney transplant. J Am Soc Nephrol. 2020;31(9):1977–1986. doi: 10.1681/ASN.2020030326 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Zhou Y Takano T Li X, et al. β-elemene regulates M1-M2 macrophage balance through the ERK/JNK/P38 MAPK signaling pathway. Commun Biol. 2022;5(1):519. doi: 10.1038/s42003-022-03369-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Liu T, Zhang L, Joo D, Sun SC. NF-κB signaling in inflammation. Signal Transduct Target Ther. 2017;2:17023. doi: 10.1038/sigtrans.2017.23 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Wu X Wang Z Shi J, et al. Macrophage polarization toward M1 phenotype through NF-κB signaling in patients with Behçet's disease. Arthritis Res Ther. 2022;24(1):249. doi: 10.1186/s13075-022-02938-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Kaneko N, Kurata M, Yamamoto T, Morikawa S, Masumoto J. The role of interleukin-1 in general pathology. Inflamm Regen. 2019;39:12. doi: 10.1186/s41232-019-0101-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Atri C, Guerfali FZ, Laouini D. Role of human macrophage polarization in inflammation during infectious diseases. Int J Mol Sci. 2018;19(6):1801. doi: 10.3390/ijms19061801 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Raziyeva K, Kim Y, Zharkinbekov Z, Kassymbek K, Jimi S, Saparov A. Immunology of acute and chronic wound healing. Biomolecules. 2021;11(5):700. doi: 10.3390/biom11050700 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Mantovani A, Schioppa T, Biswas SK, Marchesi F, Allavena P, Sica A. Tumor-associated macrophages and dendritic cells as prototypic type II polarized myeloid populations. Tumori. 2003;89(5):459–468. doi: 10.1177/030089160308900501 [DOI] [PubMed] [Google Scholar]
- 37.White S, Lin L, Hu K. NF-κB and tPA signaling in kidney and other diseases. Cells. 2020;9(6):1348. doi: 10.3390/cells9061348 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Oeckinghaus A, Ghosh S. The NF-kappaB family of transcription factors and its regulation. Cold Spring Harb Perspect Biol. 2009;1(4):a000034. doi: 10.1101/cshperspect.a000034 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Yu H, Lin L, Zhang Z, Zhang H, Hu H. Targeting NF-κB pathway for the therapy of diseases: mechanism and clinical study. Signal Transduct Target Ther. 2020;5(1):209. doi: 10.1038/s41392-020-00312-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Sun SC. The non-canonical NF-κB pathway in immunity and inflammation. Nat Rev Immunol. 2017;17(9):545–558. doi: 10.1038/nri.2017.52 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Cieniewicz B Kirillov V Daher I, et al. Ikkα-mediated noncanonical NF-κB signaling is required to support murine gammaherpesvirus 68 latency in vivo. J Virol. 2022;96(10):e0002722. doi: 10.1128/jvi.00027-22 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Senftleben U Cao Y Xiao G, et al. Activation by IKKalpha of a second, evolutionary conserved, NF-kappa B signaling pathway. Science. 2001;293(5534):1495–1499. doi: 10.1126/science.1062677 [DOI] [PubMed] [Google Scholar]
- 43.Xiao G, Fong A, Sun SC. Induction of p100 processing by NF-kappaB-inducing kinase involves docking IkappaB kinase alpha (IKKalpha) to p100 and IKKalpha-mediated phosphorylation. J Biol Chem. 2004;279(29):30099–30105. doi: 10.1074/jbc.M401428200 [DOI] [PubMed] [Google Scholar]
- 44.Li Q, Verma IM. NF-kappaB regulation in the immune system. Nat Rev Immunol. 2002;2(10):725–734. doi: 10.1038/nri910 [DOI] [PubMed] [Google Scholar]
- 45.Zhang Q, Lenardo MJ, Baltimore D. 30 years of NF-κB: a blossoming of relevance to human pathobiology. Cell. 2017;168(1-2):37–57. doi: 10.1016/j.cell.2016.12.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Smith SF, Hosgood SA, Nicholson ML. Ischemia-reperfusion injury in renal transplantation: 3 key signaling pathways in tubular epithelial cells. Kidney Int. 2019;95(1):50–56. doi: 10.1016/j.kint.2018.10.009 [DOI] [PubMed] [Google Scholar]
- 47.Cao CC Ding XQ Ou ZL, et al. In vivo transfection of NF-kappaB decoy oligodeoxynucleotides attenuate renal ischemia/reperfusion injury in rats. Kidney Int. 2004;65(3):834–845. doi: 10.1111/j.1523-1755.2004.00463.x [DOI] [PubMed] [Google Scholar]
- 48.Kim S Lee SA Yoon H, et al. Exosome-based delivery of super-repressor IκBα ameliorates kidney ischemia-reperfusion injury. Kidney Int. 2021;100(3):570–584. doi: 10.1016/j.kint.2021.04.039 [DOI] [PubMed] [Google Scholar]
- 49.Chen D Liu Y Chen J, et al. JAK/STAT pathway promotes the progression of diabetic kidney disease via autophagy in podocytes. Eur J Pharmacol. 2021;902:174121. doi: 10.1016/j.ejphar.2021.174121 [DOI] [PubMed] [Google Scholar]
- 50.Jatiani SS, Baker SJ, Silverman LR, Reddy EP. JAK/STAT pathways in cytokine signaling and myeloproliferative disorders: approaches for targeted therapies. Genes Cancer. 2010;1(10):979–993. doi: 10.1177/1947601910397187 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Negoro S Kunisada K Tone E, et al. Activation of JAK/STAT pathway transduces cytoprotective signal in rat acute myocardial infarction. Cardiovasc Res. 2000;47(4):797–805. doi: 10.1016/s0008-6363(00)00138-3 [DOI] [PubMed] [Google Scholar]
- 52.Das A, Salloum FN, Durrant D, Ockaili R, Kukreja RC. Rapamycin protects against myocardial ischemia-reperfusion injury through JAK2-STAT3 signaling pathway. J Mol Cell Cardiol. 2012;53(6):858–869. doi: 10.1016/j.yjmcc.2012.09.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Correa-Costa M Azevedo H Amano MT, et al. Transcriptome analysis of renal ischemia/reperfusion injury and its modulation by ischemic pre-conditioning or hemin treatment. PLoS One. 2012;7(11):e49569. doi: 10.1371/journal.pone.0049569 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Zhao X Zhang E Ren X, et al. Edaravone alleviates cell apoptosis and mitochondrial injury in ischemia-reperfusion-induced kidney injury via the JAK/STAT pathway. Biol Res. 2020;53(1):28. doi: 10.1186/s40659-020-00297-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Yang N Luo M Li R, et al. Blockage of JAK/STAT signalling attenuates renal ischaemia-reperfusion injury in rat. Nephrol Dial Transplant. 2008;23(1):91–100. doi: 10.1093/ndt/gfm509 [DOI] [PubMed] [Google Scholar]
- 56.Si Y Bao H Han L, et al. Dexmedetomidine protects against renal ischemia and reperfusion injury by inhibiting the JAK/STAT signaling activation. J Transl Med. 2013;11:141. doi: 10.1186/1479-5876-11-141 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.van Rooijen N, Sanders A. Elimination, blocking, and activation of macrophages: three of a kind? J Leukoc Biol. 1997;62(6):702–709. doi: 10.1002/jlb.62.6.702 [DOI] [PubMed] [Google Scholar]
- 58.Van Rooijen N, Sanders A. Liposome mediated depletion of macrophages: mechanism of action, preparation of liposomes and applications. J Immunol Methods. 1994;174(1-2):83–93. doi: 10.1016/0022-1759(94)90012-4 [DOI] [PubMed] [Google Scholar]
- 59.Zeisberger SM, Odermatt B, Marty C, Zehnder-Fjällman AH, Ballmer-Hofer K, Schwendener RA. Clodronate-liposome-mediated depletion of tumour-associated macrophages: a new and highly effective antiangiogenic therapy approach. Br J Cancer. 2006;95(3):272–281. doi: 10.1038/sj.bjc.6603240 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Guth AM, Hafeman SD, Elmslie RE, Dow SW. Liposomal clodronate treatment for tumour macrophage depletion in dogs with soft-tissue sarcoma. Vet Comp Oncol. 2013;11(4):296–305. doi: 10.1111/j.1476-5829.2012.00319.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Day YJ, Huang L, Ye H, Linden J, Okusa MD. Renal ischemia-reperfusion injury and adenosine 2A receptor-mediated tissue protection: role of macrophages. Am J Physiol Renal Physiol. 2005;288(4):F722–F731. doi: 10.1152/ajprenal.00378.2004 [DOI] [PubMed] [Google Scholar]
- 62.Jo SK, Sung SA, Cho WY, Go KJ, Kim HK. Macrophages contribute to the initiation of ischaemic acute renal failure in rats. Nephrol Dial Transplant. 2006;21(5):1231–1239. doi: 10.1093/ndt/gfk047 [DOI] [PubMed] [Google Scholar]
- 63.Lee S Huen S Nishio H, et al. Distinct macrophage phenotypes contribute to kidney injury and repair. J Am Soc Nephrol. 2011;22(2):317–326. doi: 10.1681/ASN.2009060615 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Vinuesa E, Hotter G, Jung M, Herrero-Fresneda I, Torras J, Sola A. Macrophage involvement in the kidney repair phase after ischaemia/reperfusion injury. J Pathol. 2008;214(1):104–113. doi: 10.1002/path.2259 [DOI] [PubMed] [Google Scholar]
- 65.Ferenbach DA Sheldrake TA Dhaliwal K, et al. Macrophage/monocyte depletion by clodronate, but not diphtheria toxin, improves renal ischemia/reperfusion injury in mice. Kidney Int. 2012;82(8):928–933. doi: 10.1038/ki.2012.207 [DOI] [PubMed] [Google Scholar]
- 66.Lu L Faubel S He Z, et al. Depletion of macrophages and dendritic cells in ischemic acute kidney injury. Am J Nephrol. 2012;35(2):181–190. doi: 10.1159/000335582 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Shin NS Marlier A Xu L, et al. Arginase-1 is required for macrophage-mediated renal tubule regeneration. J Am Soc Nephrol. 2022;33(6):1077–1086. doi: 10.1681/ASN.2021121548 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Cao P Sun Z Zhang F, et al. TGF-Β enhances immunosuppression of myeloid-derived suppressor cells to induce transplant immune tolerance through affecting Arg-1 expression. Front Immunol. 2022;13:919674. doi: 10.3389/fimmu.2022.919674 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Miller-Fleming L, Olin-Sandoval V, Campbell K, Ralser M. Remaining mysteries of molecular biology: the role of polyamines in the cell. J Mol Biol. 2015;427(21):3389–3406. doi: 10.1016/j.jmb.2015.06.020 [DOI] [PubMed] [Google Scholar]
- 70.Odenlund M, Holmqvist B, Baldetorp B, Hellstrand P, Nilsson BO. Polyamine synthesis inhibition induces S phase cell cycle arrest in vascular smooth muscle cells. Amino Acids. 2009;36(2):273–282. doi: 10.1007/s00726-008-0060-7 [DOI] [PubMed] [Google Scholar]
- 71.Lin SL Li B Rao S, et al. Macrophage Wnt7b is critical for kidney repair and regeneration. Proc Natl Acad Sci U S A. 2010;107(9):4194–4199. doi: 10.1073/pnas.0912228107 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Städeli R, Hoffmans R, Basler K. Transcription under the control of nuclear Arm/beta-catenin. Curr Biol. 2006;16(10):R378–R385. doi: 10.1016/j.cub.2006.04.019 [DOI] [PubMed] [Google Scholar]
- 73.Li SS Sun Q Hua MR, et al. Targeting the Wnt/β-catenin signaling pathway as a potential therapeutic strategy in renal tubulointerstitial fibrosis. Front Pharmacol. 2021;12:719880. doi: 10.3389/fphar.2021.719880 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Saegusa M, Hashimura M, Kuwata T, Hamano M, Okayasu I. Beta-catenin simultaneously induces activation of the p53-p21WAF1 pathway and overexpression of cyclin D1 during squamous differentiation of endometrial carcinoma cells. Am J Pathol. 2004;164(5):1739–1749. doi: 10.1016/s0002-9440(10)63732-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Lapham A, Adams JE, Paterson A, Lee M, Brimmell M, Packham G. The Bcl-w promoter is activated by beta-catenin/TCF4 in human colorectal carcinoma cells. Gene. 2009;432(1-2):112–117. doi: 10.1016/j.gene.2008.12.002 [DOI] [PubMed] [Google Scholar]
- 76.Lever JM Hull TD Boddu R, et al. Resident macrophages reprogram toward a developmental state after acute kidney injury. JCI Insight. 2019;4(2):e125503. doi: 10.1172/jci.insight.125503 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Kumar S. Cellular and molecular pathways of renal repair after acute kidney injury. Kidney Int. 2018;93(1):27–40. doi: 10.1016/j.kint.2017.07.030 [DOI] [PubMed] [Google Scholar]
- 78.Xu MJ, Feng D, Wang H, Guan Y, Yan X, Gao B. IL-22 ameliorates renal ischemia-reperfusion injury by targeting proximal tubule epithelium. J Am Soc Nephrol. 2014;25(5):967–977. doi: 10.1681/ASN.2013060611 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Weidenbusch M, Rodler S, Anders HJ. Interleukin-22 in kidney injury and regeneration. Am J Physiol Renal Physiol. 2015;308(10):F1041–F1046. doi: 10.1152/ajprenal.00005.2015 [DOI] [PubMed] [Google Scholar]
- 80.Kulkarni OP Hartter I Mulay SR, et al. Toll-like receptor 4-induced IL-22 accelerates kidney regeneration. J Am Soc Nephrol. 2014;25(5):978–989. doi: 10.1681/ASN.2013050528 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Hu Z, Zhan J, Pei G, Zeng R. Depletion of macrophages with clodronate liposomes partially attenuates renal fibrosis on AKI-CKD transition. Ren Fail. 2023;45(1):2149412. doi: 10.1080/0886022X.2022.2149412 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Baek JH. The impact of versatile macrophage functions on acute kidney injury and its outcomes. Front Physiol. 2019;10:1016. doi: 10.3389/fphys.2019.01016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Clements M Gershenovich M Chaber C, et al. Differential Ly6C expression after renal ischemia-reperfusion identifies unique macrophage populations. J Am Soc Nephrol. 2016;27(1):159–170. doi: 10.1681/ASN.2014111138 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Menke J Iwata Y Rabacal WA, et al. CSF-1 signals directly to renal tubular epithelial cells to mediate repair in mice. J Clin Invest. 2009;119(8):2330–2342. doi: 10.1172/JCI39087 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Jang HS, Kim J, Park YK, Park KM. Infiltrated macrophages contribute to recovery after ischemic injury but not to ischemic preconditioning in kidneys. Transplantation. 2008;85(3):447–455. doi: 10.1097/TP.0b013e318160f0d1 [DOI] [PubMed] [Google Scholar]
- 86.Ko GJ, Boo CS, Jo SK, Cho WY, Kim HK. Macrophages contribute to the development of renal fibrosis following ischaemia/reperfusion-induced acute kidney injury. Nephrol Dial Transplant. 2008;23(3):842–852. doi: 10.1093/ndt/gfm694 [DOI] [PubMed] [Google Scholar]
- 87.Kim MG, Kim SC, Ko YS, Lee HY, Jo SK, Cho W. The role of M2 macrophages in the progression of chronic kidney disease following acute kidney injury. PLoS One. 2015;10(12):e0143961. doi: 10.1371/journal.pone.0143961 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Yang Q Wang Y Pei G, et al. Bone marrow-derived Ly6C(-) macrophages promote ischemia-induced chronic kidney disease. Cell Death Dis. 2019;10(4):291. doi: 10.1038/s41419-019-1531-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Meng XM, Nikolic-Paterson DJ, Lan HY. Inflammatory processes in renal fibrosis. Nat Rev Nephrol. 2014;10(9):493–503. doi: 10.1038/nrneph.2014.114 [DOI] [PubMed] [Google Scholar]
- 90.Ricardo SD, van Goor H, Eddy AA. Macrophage diversity in renal injury and repair. J Clin Invest. 2008;118(11):3522–3530. doi: 10.1172/JCI36150 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Lech M Gröbmayr R Ryu M, et al. Macrophage phenotype controls long-term AKI outcomes--kidney regeneration versus atrophy. J Am Soc Nephrol. 2014;25(2):292–304. doi: 10.1681/ASN.2013020152 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Fadok VA, Bratton DL, Konowal A, Freed PW, Westcott JY, Henson PM. Macrophages that have ingested apoptotic cells in vitro inhibit proinflammatory cytokine production through autocrine/paracrine mechanisms involving TGF-beta, PGE2, and PAF. J Clin Invest. 1998;101(4):890–898. doi: 10.1172/JCI1112 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Huynh ML, Fadok VA, Henson PM. Phosphatidylserine-dependent ingestion of apoptotic cells promotes TGF-beta1 secretion and the resolution of inflammation. J Clin Invest. 2002;109(1):41–50. doi: 10.1172/JCI11638 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Massague J. TGFβ signalling in context. Nat Rev Mol Cell Biol. 2012;13(10):616–630. doi: 10.1038/nrm3434 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Zimmerman KA Xing D Pallero MA, et al. Calreticulin regulates neointima formation and collagen deposition following carotid artery ligation. J Vasc Res. 2015;52(5):306–320. doi: 10.1159/000443884 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Huen SC, Moeckel GW, Cantley LG. Macrophage-specific deletion of transforming growth factor-β1 does not prevent renal fibrosis after severe ischemia-reperfusion or obstructive injury. Am J Physiol Renal Physiol. 2013;305(4):F477–F484. doi: 10.1152/ajprenal.00624.2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Lu J Cao Q Zheng D, et al. Discrete functions of M2a and M2c macrophage subsets determine their relative efficacy in treating chronic kidney disease. Kidney Int. 2013;84(4):745–755. doi: 10.1038/ki.2013.135 [DOI] [PubMed] [Google Scholar]
- 98.Dihazi GH Eltoweissy M Jahn O, et al. The secretome analysis of activated human renal fibroblasts revealed beneficial effect of the modulation of the secreted peptidyl-prolyl cis-trans isomerase A in kidney fibrosis. Cells. 2020;9(7):1724. doi: 10.3390/cells9071724 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Meran S, Steadman R. Fibroblasts and myofibroblasts in renal fibrosis. Int J Exp Pathol. 2011;92(3):158–167. doi: 10.1111/j.1365-2613.2011.00764.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Sun J, Ge X, Wang Y, Niu L, Tang L, Pan S. USF2 knockdown downregulates THBS1 to inhibit the TGF-β signaling pathway and reduce pyroptosis in sepsis-induced acute kidney injury. Pharmacol Res. 2022;176:105962. doi: 10.1016/j.phrs.2021.105962 [DOI] [PubMed] [Google Scholar]
- 101.Nogueira A, Pires MJ, Oliveira PA. Pathophysiological mechanisms of renal fibrosis: a review of animal models and therapeutic strategies. In Vivo. 2017;31(1):1–22. doi: 10.21873/invivo.11019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.van Furth R, Cohn ZA, Hirsch JG, Humphrey JH, Spector WG, Langevoort HL. The mononuclear phagocyte system: a new classification of macrophages, monocytes, and their precursor cells. Bull World Health Organ. 1972;46(6):845–852. PMID: 4538544 [PMC free article] [PubMed] [Google Scholar]
- 103.van Furth R, Cohn ZA. The origin and kinetics of mononuclear phagocytes. J Exp Med. 1968;128(3):415–435. doi: 10.1084/jem.128.3.415 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Sheng J, Ruedl C, Karjalainen K. Most tissue-resident macrophages except microglia are derived from fetal hematopoietic stem cells. Immunity. 2015;43(2):382–393. doi: 10.1016/j.immuni.2015.07.016 [DOI] [PubMed] [Google Scholar]
- 105.Jang HS, Kim JI, Jung KJ, Kim J, Han KH, Park KM. Bone marrow-derived cells play a major role in kidney fibrosis via proliferation and differentiation in the infiltrated site. Biochim Biophys Acta. 2013;1832(6):817–825. doi: 10.1016/j.bbadis.2013.02.016 [DOI] [PubMed] [Google Scholar]
- 106.de Cortie K, Russell NS, Coppes RP, Stewart FA, Scharpfenecker M. Bone marrow-derived macrophages incorporate into the endothelium and influence vascular and renal function after irradiation. Int J Radiat Biol. 2014;90(9):769–777. doi: 10.3109/09553002.2014.920967 [DOI] [PubMed] [Google Scholar]
- 107.Schulz C Gomez Perdiguero E Chorro L, et al. A lineage of myeloid cells independent of Myb and hematopoietic stem cells. Science. 2012;336(6077):86–90. doi: 10.1126/science.1219179 [DOI] [PubMed] [Google Scholar]
- 108.Gomez Perdiguero E, Geissmann F. Myb-independent macrophages: a family of cells that develops with their tissue of residence and is involved in its homeostasis. Cold Spring Harb Symp Quant Biol. 2013;78:91–100. doi: 10.1101/sqb.2013.78.020032 [DOI] [PubMed] [Google Scholar]
- 109.Ginhoux F, Guilliams M. Tissue-resident macrophage ontogeny and homeostasis. Immunity. 2016;44(3):439–449. doi: 10.1016/j.immuni.2016.02.024 [DOI] [PubMed] [Google Scholar]
- 110.Viehmann SF, Bohner AMC, Kurts C, Brahler S. The multifaceted role of the renal mononuclear phagocyte system. Cell Immunol. 2018;330:97–104. doi: 10.1016/j.cellimm.2018.04.009 [DOI] [PubMed] [Google Scholar]
- 111.Hoeffel G Chen J Lavin Y, et al. C-Myb(+) erythro-myeloid progenitor-derived fetal monocytes give rise to adult tissue-resident macrophages. Immunity. 2015;42(4):665–678. doi: 10.1016/j.immuni.2015.03.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Epelman S Lavine KJ Beaudin AE, et al. Embryonic and adult-derived resident cardiac macrophages are maintained through distinct mechanisms at steady state and during inflammation. Immunity. 2014;40(1):91–104. doi: 10.1016/j.immuni.2013.11.019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Liu Z Gu Y Chakarov S, et al. Fate mapping via Ms4a3-expression history traces monocyte-derived cells. Cell. 2019;178(6):1509–1525.e19. doi: 10.1016/j.cell.2019.08.009 [DOI] [PubMed] [Google Scholar]
- 114.Munro DAD, Hughes J. The origins and functions of tissue-resident macrophages in kidney development. Front Physiol. 2017;8:837. doi: 10.3389/fphys.2017.00837 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Hamann J Koning N Pouwels W, et al. EMR1, the human homolog of F4/80, is an eosinophil-specific receptor. Eur J Immunol. 2007;37(10):2797–2802. doi: 10.1002/eji.200737553 [DOI] [PubMed] [Google Scholar]
- 116.Waddell LA Lefevre L Bush SJ, et al. ADGRE1 (EMR1, F4/80) is a rapidly-evolving gene expressed in mammalian monocyte-macrophages. Front Immunol. 2018;9:2246. doi: 10.3389/fimmu.2018.02246 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Zimmerman KA Bentley MR Lever JM, et al. Single-cell RNA sequencing identifies candidate renal resident macrophage gene expression signatures across species. J Am Soc Nephrol. 2019;30(5):767–781. doi: 10.1681/ASN.2018090931 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Zimmerman KA Yang Z Lever JM, et al. Kidney resident macrophages in the rat have minimal turnover and replacement by blood monocytes. Am J Physiol Renal Physiol. 2021;321(2):F162–F169. doi: 10.1152/ajprenal.00129.2021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Stewart BJ Ferdinand JR Young MD, et al. Spatiotemporal immune zonation of the human kidney. Science. 2019;365(6460):1461–1466. doi: 10.1126/science.aat5031 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Cheung MD Erman EN Moore KH, et al. Resident macrophage subpopulations occupy distinct microenvironments in the kidney. JCI Insight. 2022;7(20):e161078. doi: 10.1172/jci.insight.161078 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Yao W Chen Y Li Z, et al. Single cell RNA sequencing identifies a unique inflammatory macrophage subset as a druggable target for alleviating acute kidney injury. Adv Sci (Weinh). 2022;9(12):e2103675. doi: 10.1002/advs.202103675 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Nash WT, Yee MS, Okusa MD. Myeloid response to acute kidney injury. Nephron. 2023;147(1):39–43. doi: 10.1159/000526266 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Park JG Lee CR Kim MG, et al. Kidney residency of VISTA-positive macrophages accelerates repair from ischemic injury. Kidney Int. 2020;97(5):980–994. doi: 10.1016/j.kint.2019.11.025 [DOI] [PubMed] [Google Scholar]
- 124.Puranik AS Leaf IA Jensen MA, et al. Kidney-resident macrophages promote a proangiogenic environment in the normal and chronically ischemic mouse kidney. Sci Rep. 2018;8(1):13948. doi: 10.1038/s41598-018-31887-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Karasawa K Asano K Moriyama S, et al. Vascular-resident CD169-positive monocytes and macrophages control neutrophil accumulation in the kidney with ischemia-reperfusion injury. J Am Soc Nephrol. 2015;26(4):896–906. doi: 10.1681/ASN.2014020195 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Jones CV, Ricardo SD. Macrophages and CSF-1: implications for development and beyond. Organogenesis. 2013;9(4):249–260. doi: 10.4161/org.25676 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Zimmerman KA Song CJ Li Z, et al. Tissue-resident macrophages promote renal cystic disease. J Am Soc Nephrol. 2019;30(10):1841–1856. doi: 10.1681/ASN.2018080810 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Alikhan MA Jones CV Williams TM, et al. Colony-stimulating factor-1 promotes kidney growth and repair via alteration of macrophage responses. Am J Pathol. 2011;179(3):1243–1256. doi: 10.1016/j.ajpath.2011.05.037 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Zhang MZ Yao B Yang S, et al. CSF-1 signaling mediates recovery from acute kidney injury. J Clin Invest. 2012;122(12):4519–4532. doi: 10.1172/JCI60363 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Wang Y Chang J Yao B, et al. Proximal tubule-derived colony stimulating factor-1 mediates polarization of renal macrophages and dendritic cells, and recovery in acute kidney injury. Kidney Int. 2015;88(6):1274–1282. doi: 10.1038/ki.2015.295 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Serbina NV, Pamer EG. Monocyte emigration from bone marrow during bacterial infection requires signals mediated by chemokine receptor CCR2. Nat Immunol. 2006;7(3):311–317. doi: 10.1038/ni1309 [DOI] [PubMed] [Google Scholar]
- 132.Furuichi K Wada T Iwata Y, et al. CCR2 signaling contributes to ischemia-reperfusion injury in kidney. J Am Soc Nephrol. 2003;14(10):2503–2515. doi: 10.1097/01.ASN.0000089563.63641.a8 [DOI] [PubMed] [Google Scholar]
- 133.Li L Huang L Sung SS, et al. The chemokine receptors CCR2 and CX3CR1 mediate monocyte/macrophage trafficking in kidney ischemia-reperfusion injury. Kidney Int. 2008;74(12):1526–1537. doi: 10.1038/ki.2008.500 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Wilson HM, Walbaum D, Rees AJ. Macrophages and the kidney. Curr Opin Nephrol Hypertens. 2004;13(3):285–290. doi: 10.1097/00041552-200405000-00004 [DOI] [PubMed] [Google Scholar]
- 135.Ito K Chen J El Chaar M, et al. Renal damage progresses despite improvement of renal function after relief of unilateral ureteral obstruction in adult rats. Am J Physiol Renal Physiol. 2004;287(6):F1283–F1293. doi: 10.1152/ajprenal.00441.2003 [DOI] [PubMed] [Google Scholar]
- 136.Cochrane AL Kett MM Samuel CS, et al. Renal structural and functional repair in a mouse model of reversal of ureteral obstruction. J Am Soc Nephrol. 2005;16(12):3623–3630. doi: 10.1681/ASN.2004090771 [DOI] [PubMed] [Google Scholar]
- 137.Sears SM Vega AA Kurlawala Z, et al. F4/80(hi) resident macrophages contribute to cisplatin-induced renal fibrosis. Kidney360. 2022;3(5):818–833. doi: 10.34067/KID.0006442021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Ozkok A, Edelstein CL. Pathophysiology of cisplatin-induced acute kidney injury. Biomed Res Int. 2014;2014:967826. doi: 10.1155/2014/967826 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.He J, Zhao S, Duan M. The response of macrophages in sepsis-induced acute kidney injury. J Clin Med. 2023;12(3):1101. doi: 10.3390/jcm12031101 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Meng X, Jin J, Lan HY. Driving role of macrophages in transition from acute kidney injury to chronic kidney disease. Chin Med J (Engl). 2022;135(7):757–766. doi: 10.1097/CM9.0000000000002100 [DOI] [PMC free article] [PubMed] [Google Scholar]