Open Access

Macrophages polarization in renal inflammation and fibrosis animal models (Review)

  • Authors:
    • Ji Zeng
    • Yuan Zhang
    • Cheng Huang
  • View Affiliations

  • Published online on: December 20, 2023     https://doi.org/10.3892/mmr.2023.13152
  • Article Number: 29
  • Copyright: © Zeng et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Chronic kidney disease (CKD) is a significant public health concern. Renal fibrosis is the final common pathway in the progression of kidney diseases, irrespective of the initial injury. Substantial evidence underscores the pivotal role of renal inflammation in the genesis of renal fibrosis. The presence of macrophages within normal renal tissue is significantly increased within diseased renal tissue, indicative of their crucial regulatory function in inflammation and fibrosis. Macrophages manifest a high degree of heterogeneity, exhibiting distinct phenotypic and functional traits in response to diverse stimuli within the local microenvironment in various types of kidney diseases. Broadly, macrophages are categorized into two principal groups: Classically activated, designated as M1 macrophages and alternatively activated, designated as M2 macrophages. A number of experimental models are widely used to study the underlying mechanisms driving renal inflammation and fibrosis progression. The present review delineated the phenotypic and functional attributes of macrophages present in diverse induced models, analyzing their disposition in relation to M1 and M2 polarization states.

Introduction

Chronic kidney disease (CKD) stands as a major public health concern (1), significantly affecting the quality of life of patients (2). Renal fibrosis is the final shared route of progressive kidney diseases, irrespective of the initial injury (3). Compelling evidence highlights the pivotal role of renal inflammation in the initiation and advancement of renal fibrosis, emphasizing the contemporary focus on CKD prevention in clinical practice.

The first recognized function of macrophages is infection clearance. They are highly heterogeneous cells essential to the body's immune system (46). Macrophages are ubiquitously distributed across nearly all tissues and are established during embryonic development (5,7). In mature individuals, macrophages originate from bone marrow-derived monocytes, circulating in the blood stream and continually homing to tissues, where they ultimately differentiate into macrophages. Macrophage infiltration constitutes a common feature of CKD, with the degree of invasion and the extent of kidney injury (8).

Macrophages play a critical role in the progressive trajectory of renal fibrosis, with previous studies emphasizing their role in the clearance of dead cells and debris (characteristic function of M1 macrophages). In addition to M1 macrophages, M2 macrophages contribute to humoral immunity and tissue repair (79). Mounting evidence suggests that the ultimate outcome of renal disease is substantially influenced by the functional diversity stemming from alterations in macrophage phenotype. Studies have underscored that the polarization of macrophages holds the key to understanding kidney injury, including renal inflammation and fibrosis (79).

Macrophage polarity

Macrophages are broadly categorized into two primary groups: The classically activated, or pro-inflammatory (M1) and the alternatively activated, or anti-inflammatory (M2) macrophages (10). M1 and M2 macrophages are distinguished by their surface markers, functions and secreted factors. The M1 phenotype is induced by signaling from TNF-α and IFN-γ and is characterized by surface markers including cluster of differentiation (CD)40, CD80, CD83, and CD86. By contrast, the M2 phenotype is driven by IL-4 and marked by surface markers including CD2-6, CD163 and CD206. Functionally, M1 macrophages secrete an array of pro-inflammatory cytokines such as TNF-α, IL-1α, IL-1β, IL-6, IL-12, IL-23 and cyclooxygenase-2 (COX-2), collectively fostering an inflammatory milieu (11). By contrast, M2 macrophages release anti-inflammatory cytokines such as IL-4, IL-13, IL-12, IL-10 and TGF-β, all of which exert anti-inflammatory effects (12).

The secretions of M1 macrophage exhibit the capacity to eliminate bacteria, viruses and malignant tumor cells. However, excessive immune responses may lead to chronic inflammation and inflammatory diseases (13), necessitating the precise regulation of M1 macrophage functionality (14,15). Conversely, M2 macrophages are involved in tissue repair and immune tolerance (16).

Macrophage polarity in chronic inflammation and renal fibrosis

Most instances of acute renal inflammation are concomitant with an infiltration of M1 macrophages. Although M1 macrophages are a key feature of chronic kidney inflammation, the infiltration of these macrophages' transitions to the M2 phenotype occurs during the chronic developmental stage of the disease. The emergence of renal fibrosis is the most reliable predictor of renal disease progression and macrophages are a common feature of active fiber lesions in the kidney; therefore, an increased number of macrophages is also a marker of the progression of renal fibrosis (7). Several recent studies have similarly emphasized the pivotal role of macrophage polarization driving the renal inflammation and fibrosis (9,17,18). A number of experimental models have been extensively employed to dissect the intricacies of the mechanisms underpinning the transition from renal disease to renal fibrosis (19,20). Given this perspective, the present review endeavors to scrutinize the role of macrophage polarization in different animal models of renal fibrosis.

Unilateral ureteral obstruction (UUO)

The UUO model is the most widely employed interstitial fibrosis model due to its rapid development of tubular atrophy, interstitial fibrosis and matrix deposition (21). The UUO animal model and human obstructive nephropathies share similar etiological factors (22,23). Following complete UUO in the obstructed kidney, there is epithelial tubular cell damage (24), leading to a significant increase in renal fibrosis indicators such as inflammation mediated by macrophages (25,26) and myofibroblasts (24). M1 and M2 macrophages fulfill contrasting yet complementary roles in tissue fibrosis (27,28). Indeed, tubular epithelial cells are targeted for macrophage-induced apoptosis in the obstructed kidney (29). The process of apoptotic cell recognition and phagocytosis by macrophages facilitates their differentiation into M2 macrophages (27,30). M2 macrophages are prominent producers of TGF-β, capable of inducing myofibroblast proliferation (30,31). By contrast, M1 macrophages produce MMPs and can also induce myofibroblasts to produce MMPs that promote extracellular matrix (ECM) degradation and facilitate the resolution of fibrosis (9,31). Some data suggest that transfusion of legumain-overexpressing macrophages can alleviate UUO-induced renal fibrosis (32). Notably, macrophages of differing phenotypes can coexist, and they may revert to a resting state (M0 macrophage) or be repolarized due to the influence of the kidney microenvironment (32). During the early stages of UUO, the interstitium is dominated by the M1 phenotype, whereas M2 macrophages accumulate at later stages (33). Additionally, studies have shown that mice lacking in IRAK-M, an inhibitor of pro-inflammatory cytokines expression in internal macrophages (34), are protected in experimental models of UUO-induced kidney injury, leading to reduced renal fibrosis and a lower number of M2 macrophages. Figueiredo et al (35) found that relaxin can suppress the typical M-cytokine IL-1β expression in macrophages in response to inflammatory stimuli, thereby promoting the acquisition of an immunosuppressive M2 phenotype in macrophages. Consistent with earlier findings, relaxin can promote the acquisition of an immunosuppressive M2 phenotype. M2 macrophages mitigate kidney inflammation by releasing anti-inflammatory mediators, such as IL-10, which inhibit renal fibrosis (9,3537). Relaxin, acting as a swift yet safe antifibrotic agent, inhibits renal fibrosis in UUO (3840). Studies have demonstrated the potential of relaxin to shift macrophage polarization towards the M2 phenotype (40,41). The microenvironment may induce macrophage polarization (42). Studies have suggested that complement proteins may influence macrophage phenotypes (43,44). Although the complement proteins affect macrophage polarization (43,44), whether complement component 3 (C3) can induce macrophage polarization and regulate renal fibrosis remains uncertain. Recent data demonstrate a close correlation between interstitial infiltrating macrophages and C3-expressing renal tubules in UUO mice (44). C3 deficiency facilitates the shift of infiltrating macrophage toward the M2 phenotype during the early stage of UUO (44) (Fig. 1). In summary, while these have limitations, they offer new insights for future therapeutic approaches to renal fibrosis treatment (44).

Aldosterone and mineralocorticoid receptor (MR)

Studies have unequivocally established the pivotal role of macrophages in renal inflammation, fibrosis and the remodeling induced by MR (45,46). Although heightened macrophage infiltration has been reported previously in a rat model administered aldosterone and salt (46), the precise macrophage phenotype responsible remains somewhat elusive. The present study identified that in the MR model, it mainly mediated the M1 phenotype (5,42). Preceding the onset of fibrosis, aldosterone triggers the infiltration of monocytes and macrophages, alongside an elevated expression of inflammatory markers such as COX-2, monocyte chemoattractant protein1 and intercellular adhesion molecule 1 in the heart, vasculature and kidney (46). Studies have demonstrated the significance of aldosterone in the inflammatory and fibrotic processes in kidney diseases, wherein the aldosterone-induced enhancement of renal lesions frequently coincides with a marked inflammatory response, primarily involving macrophage-released inflammatory factors during renal fibrosis (47,48). In recent years, the influence of complement proteins on macrophage phenotypes has emerged. For example, DOCA-salt hypertensive mice that received bone marrow from knockout mice exhibited lower M1 and higher M2 macrophage count within perivascular adipose tissue, thereby concomitantly ameliorating vascular injury in contrast to mice receiving bone marrow from wild-type mice (44). Nevertheless, macrophage phenotypes should not be simplistically confined to M1 and M2 classifications, as substantial heterogeneity exists within the kidney macrophage population both in healthy and injured kidneys (12). Furthermore, there exists evidence supporting the potential of MR aldosterone antagonist MRA to diminish renal inflammation and fibrosis (49). The use of specific aldosterone synthase inhibitors or aldosterone synthase-deficient mice has demonstrated efficacy in mitigating the progression of inflammation and fibrosis (5052). Plasminogen activator inhibitor1 (PAI-1), a typical mediator of fibrosis, directly inhibits both tissue plasminogen activator and urokinase. The anti-inflammatory and anti-fibrotic effects of aldosterone could potentially arise through MR activation, fostering the expression of PAI-1 in multiple tissues, including the kidney (45). Within the kidney, aldosterone triggers PAI-1 expression via MR activation in podocytes, macrophages, mesangial cells and tubular epithelial cells (53,54). However, the clinical viability of PAI-1 inhibitors remains challenging to attain due to their limited selectivity and notable toxicity (55). In addition to PAI-1, aldosterone may also instigate the activation of cytokines such as TGF-β, NF-κB and IL-6 (54,56), which in turn might foster fibrosis development by recruiting inflammatory cells or promoting M1 macrophage polarization (53,57). Thus, MR targeting holds potential for modulating the balance between M1 and M2 phenotypes, directly influencing macrophages, or altering epithelial intercommunication, thereby delaying the onset of renal fibrosis and the development of CKD (58). Research indicates that after three weeks of aldosterone and a high-sodium diet, the expression of M1 surface markers in the kidney increased, a response that is counteracted by the addition of spironolactone. Conversely, other experiments have demonstrated that treatment with chlorophosphite solely diminishes M1 macrophage count without affecting M2 macrophage (Fig. 2). Therefore, the pro-inflammatory and pro-fibrotic consequences initiated by aldosterone can be mitigated by incorporating MR antagonists into macrophages (59). Nevertheless, the precise mechanism by which aldosterone influences macrophages and fosters macrophage polarization remains incompletely elucidated in vivo (59).

Cisplatin (CDDP)-induced renal fibrosis

CDDP is a potent anticancer drug, yet it bears significant renal adverse effects (6063). Various models of renal fibrosis induced by CDDP have been used (6466). Macrophages constitute pivotal actors within several aspects of these processes, encompassing immune responses, wound healing and tissue remodeling. M1 macrophages swiftly emerge in the kidney following injury and are recognized as proinflammatory entities. Conversely, M2 macrophages manifest later in kidney injury and exhibit a phenotype conducive to tissue repair and anti-inflammatory functions. Although M1 macrophages may persist even in chronic injury, the transition from the M1 to the M2 phenotype is associated with the development of renal fibrosis (7,67). Sears et al (68) revealed that clodrosome diminished the number of F4/80hi renal resident and M2 macrophages following small doses of cisplatin treatment but had no effect on F4/80lo infiltrating macrophages in the kidney. This depletion was accompanied by reduced renal fibrosis following small doses of cisplatin treatment. By contrast, Ccr2−/− mice exhibit a reduced number of F4/80lo infiltrating macrophages in the kidney following small doses of cisplatin treatment, with no effect on the accumulation of F4/80hi renal resident or M2 macrophages. Moreover, the genetic knockout of Ccr2 has no bearing on the development of fibrosis following small doses of cisplatin treatment. In this study, clodrosome exhibited protective effects against the reduction of the number of F4/80hi resident and M2 macrophages (68). Although recent studies undeniably emphasize the significant role played by macrophage polarization in the development of fibrotic diseases, uncertainties persist regarding whether macrophage polarization carries equivalent importance within an experimental model of renal fibrosis induced by CDDP (68,69). When endothelial cells from CDDP-treated entities were co-cultured with Raw264.7, mRNA expression levels of Arg-1 and CD206 were observed to be significantly increased. In contrast to other models, this outcome suggests that cisplatin might induce M2 macrophage polarization through the inflammatory milieu triggered by endothelial cells (69). Yu et al (69) demonstrated that cisplatin-induced an incomplete epithelial mesenchymal transition (EMT) within tubular epithelial cells (ECs), yet co-culturing cisplatin-treated ECs with M2 macrophages, rather than M1, resulted in complete EMT. Furthermore, they found that co-culturing with cisplatin-treated tubular ECs spurred fibroblast activation and promoted M2 macrophage polarization. Generally, the M1 and M2 macrophages are liable to transformation, albeit this alteration does not uniformly affect the functional transition of macrophages. For in vivo experiments, immunohistochemical methods employed for discerning M1 and M2 macrophages assume pivotal significance. One particular study used immunohistology techniques employing antibodies targeting cell surface molecules of macrophages within a CDDP-induced renal fibrosis model (70). In the CDDP-induced renal fibrosis model, the differently polarized macrophages may enact distinct roles at different stages of the disease course. M1 macrophages appearing primarily at mid-stage may participate in the progressive renal injury caused initially by CDDP. Notably, the number of M2 macrophages began to increase on day 5 and continued to increase until day 20. Similar to M2 macrophages, the levels of TGF-β1 also increased from day 7 to day 20. Therefore, the findings of this study (71) indicate that the intermediate and late progressive renal fibrosis from day 9 to day 20 may be associated with the increase in the number of M2 macrophages and the levels of TGF-β1 (7072). Cisplatin-elicited reactive oxygen species (ROS) stress within compromised epithelial cells, coupled with the secretion of pro-inflammatory cytokines by neighboring immune cells, is a plausible etiology for EMT (Fig. 3). Macrophages emerge as pivotal players in the immunological milieu during both the inflammatory and recovery phases (7375). However, the exact roles that macrophages assume in cisplatin induced EMT remain to be elucidated. Antecedently, it has been reported that phenotypes of M1 and M2 macrophages can fluidly transition contingent upon the microenvironment (42,76,77). The intricate orchestration of progressive fibrosis might stem from the intricate interplay between M1 and M2 macrophages interacting with T cells encompassing CD4 (for helper functions) and CD8 (for cytotoxicity) (78). Taken together, it may be concluded that macrophage polarization has a significant impact on CDDP-induced renal fibrosis models.

Macrophages in Adriamycin (ADR)-induced nephropathy

ADR (doxorubicin) belongs to the anthracycline class of anti-tumor drugs, known for their broad spectrum of activity against human cancers (79). ADR nephropathy (AN) serves as a rodent model for CKD and has been extensively studied, enhancing our comprehension of the mechanisms underlying the progression of chronic proteinuric renal disease (79). AN is characterized by podocyte injury, subsequent glomerulosclerosis, tubulointerstitial inflammation and fibrosis (79). In rats, ADR-induced nephropathy results in substantial proteinuria and tubular basement membrane lesions, potentially provoking an inflammatory response and interstitial fibrosis (80). AN boasts several strengths as an experimental model of kidney disease. Its renal injury induction is highly reproducible and exhibits robustness with severe tissue injury yet acceptable mortality (<5%) and morbidity (weight loss). As renal injury occurs within a few days of drug administration, the temporal occurrence of injury is consistent and predictable. Nonetheless, the model also has some limitations, as different AN batches may induce varying degrees of kidney injury (81). Research has highlighted the efficacy of M2 macrophages induced by IL-4 or IL-4/IL-13 administration against renal injury resulting from ADR-induced nephropathy in mice (67,82). These studies suggest that regulation of macrophage phenotypes in the kidney could play a pivotal role in reducing renal inflammation and proteinuria. Recent clinical studies have underscored the critical influence of proteinuria on the progression of CKD and the onset of cardiovascular disease. Inflammation and macrophage infiltration into renal tissue have been implicated as contributing factors to proteinuria. α1-acid glycoprotein (AGP), an acute-phase plasma protein, leaks into the urine in patients with proteinuria (81,82). One study found that AGP potentiates the shift of macrophage phenotype towards CD163-expressing macrophages with anti-inflammatory functions in vivo, thereby mitigating proteinuria, inflammation and renal damage (83).

In general, M2-type macrophages can be further divided into four subtypes: M2a, M2b, M2c and M2d. Previous studies have elucidated the transfer the transfer of macrophages polarized by exposure to IL-4 and IL-13, thereby attaining an M2a phenotype, as an efficacious intervention for countering ADR-induced nephropathy in severe combined immunodeficient mice (8284). Furthermore, the infusion of macrophages that have been ex vivo modified into M2c phenotypes through exposure to IL-10 and TGF-β has been shown to furnish protection against renal injury in AN (82). These findings collectively propose the ex vivo manipulation of macrophages to adopt M2 phenotypes as a promising therapeutic avenue for managing chronic inflammatory renal disease (82). However, the specific distinctions, if any, between the protective capabilities of M2a and M2c phenotypes against renal injury, remain unexplored, as do potential variations in the underlying mechanisms that substantiate the protective effects of these two distinct types of M2 macrophages. Cao et al (85,86) has extensively investigated the ramifications of introducing in vitro-generated M2 macrophages into rodent models, encompassing scenarios of acute kidney failure and CKD (82). Using a mouse model of AN, the investigation of Cao et al (85,86) showed that a solitary intravenous infusion of 1×106 macrophages generated from splenic CD116+ cells and exposed to M2c macrophage polarization, exhibited heightened protective efficacy against kidney injury compared to M2a macrophages (85). These M2c macrophages are capable of exhibiting elevated expression levels of B7-H4, a member of the B7 family (87). This protein is recognized for its capacity to curtail T cell proliferation and induce regulatory T cells, as substantiated both in vivo and in vitro. It should be noted, however, that despite undergoing some alteration in phenotype during the disease course, these M2c macrophages did not undergo a complete transformation towards the M1 phenotype (85,88). Furthermore, an alternative facet of the research demonstrated that M2 macrophages originating from the spleen (SP-M2) were associated with enhanced kidney injury mitigation within the AN model of severe combined immunodeficiency (SCID) mice (82). By contrast, some other findings (86) indicate that the utilization of bone marrow-derived M2 macrophages (BM-M2) does not confer protection to renal tissue or function within the same model. Therefore, these findings collectively suggest that in the AN experimental model involving SCID mice, the inhibitory function of BM-M2 may wane in vivo due to its proliferative effects, while SP-M2 proves protective as a result of its non-proliferative nature. The transfer of SP-M2 phenotypes appears to promote renal fibrosis (86,89). Similar to various tissues, recent investigations have also elucidated that an experimental model of doxorubicin-induced cardiotoxicity (Dox-induced cardiotoxicity) may entail the infiltration of M1 macrophages, thereby inciting inflammation and fibrosis (90,91). Exosomes (cell-derived vesicles) enhance M2 macrophages in Dox-induced cardiotoxicity model (90,92,93), a trend congruent with prior literature (9497). Immune cells are capable of internalizing exosomal miRNAs, thereby orchestrating the regulation of inflammatory responses, as previously reported (98). A mounting body of evidence further underscores the pivotal role of miRNAs in the progression of both kidney disease and inflammatory disease (99,100). This exosome-mediated inflammatory pathway introduces a novel mechanism underpinning the development of renal inflammation by facilitating communication between Tubular Epithelia Cells and macrophages. Indeed, a study demonstrated the mediation of crosstalk between TECs and macrophages through exosomal miR-19b-3p, which contributes to M1 macrophage activation (101) (Fig. 4). The findings suggest that exosomal miR-19b-3p could potentially emerge as a promising therapeutic target for the advancement of kidney disease mitigation (101). In light of the aforementioned, the precise regulation of macrophage phenotypes within the renal context emerges as a pivotal strategy for curbing renal inflammation and fibrosis in AN-induced renal fibrosis models.

Ischemia reperfusion injury (IRI) renal fibrosis

Renal IRI refers to the damage that arises from the interruption and subsequent restoration of blood flow to the kidney. The hypoxia that ensues upon IRI leads to irreversible damage to tubular cells in the S3 segment of nephrons and an exacerbated innate immune response, commonly referred to as necroinflammation (102). IRI stands out as one of the most crucial pathological process contributing to acute kidney injury (AKI) (103). The pathogenesis of renal fibrosis in IRI has been attributed to a number of factors, including inflammatory cell infiltration, the production of proinflammatory chemokines and cytokines, recruitment of bone marrow-derived fibroblasts and accumulation of M2 macrophages (104,105). Interferon regulatory factor 4 (IRF4) functions as a suppressor of innate immune signaling, by inhibiting Toll-like receptor/myeloid differentiation primary response 88 signaling (TLR/MyD88 signaling) (106). In the acute phase of IRI, IRF4 serves as an endogenous regulator of myeloid cell activation such as dendritic cells, restraining the release of TNF-α release from intrarenal myeloid cells and thereby limiting tubular cell necrosis, tissue inflammation and acute renal failure 24 h and 5 days post-IRI (107). Several studies have shown that M1 macrophage-driven chronic renal inflammation accelerates renal fibrosis formation and the progression of CKD in IRI models (107,108). The essential role of IL-4 and IL-13 as determinants for M2 macrophage activation is well-established (76). Therefore, certain findings indicate that IL-18 could inhibit the expression of macrophage M2 surface marker. As M2 macrophages have long been associated with the pathogenesis of renal fibrosis, inhibiting IL-18 in IRI-induced renal fibrosis, may impede macrophage M2 polarization, thereby accelerating the development of renal fibrosis (109). Emerging evidence underscores that macrophages can directly contribute to a fibrotic response through transitioning into myofibroblasts, a phenomenon termed macrophage-myofibroblast transition (MMT) (89,110). The MMT process introduces a novel mechanism for myofibroblast formation and the development of renal fibrosis (75). In a specific study, t IL-18 treatment significantly decreases the number of CD206+ and α-SMA+ cells in the kidneys of mice following IRI. These findings suggest that the inhibition of IL-8 reduces the progression of renal fibrosis by suppressing the transition of M2 macrophages to myofibroblasts (109).

Previous studies have shown that STAT1 activation is increased in primary cardiac myocytes following IRI (111). Other studies have shown that STAT1 modulates autophagy and regulates both cardioprotective and harmful effects of STAT1 in heart injury (111,112). Various studies have emphasized the role of alternatively activated M2-like macrophages in renal fibrosis (104,113). Although these so-called M2-like macrophages are capable of generating numerous profibrotic mediators, their ability to support host defense mechanisms is limited (114). Some investigations have showed that renal fibrosis is not only a result of persistent inflammation, but also activated macrophages and genetic factors that determine long-term outcomes of IRI (115). The IFN-γ/STAT1 pathway has been suggested to plays a crucial role in the development of classically activated macrophages and that this is regulated via different mechanisms. In conclusion, STAT1 deficiency in mice enhances renal fibrosis following renal IRI. Studies in vitro on macrophages differentiation suggest that STAT1-insufficiency dependent predominance of alternatively activated macrophages may be responsible for renal tissue remodeling (111,112,116).

Previous reports indicate that P144 (TGF-β1 inhibitory peptide) can impede the progression of liver fibrosis (117,118) and myocardial fibrosis (119). TGF-β1 has been shown to reduce fibroblast activation and collage deposition in vivo in animal models of kidney disease (120). In IRI, macrophage is the primary infiltrating immune cells in the kidney tissue. Thus, it is reasonable to hypothesize that macrophages may operate downstream of TGF-β1 in a model of renal fibrosis induced by IRI (121). Moreover, the progression of renal fibrosis after IRI has been strongly associated with an increased number of M2 macrophages (7). In addition, TGF-β1 has the capacity to induce macrophages polarization toward the M2 phenotype (104). The introduction of P144 significantly prevents the upregulation of CD206, indicating reduced M2 polarization (121). P144 not only prevents M2 macrophage polarization but also curtails the phosphorylation of Smad3 at both the transcriptional and translational levels. Thus, P144 has a substantial inhibitory effect on M2 macrophage infiltration in renal tissue (121).

Studies have shown that prior to the occurrence of AKI, macrophages are depleted, resulting in a protective effect on the kidney. The phenotypic alterations of macrophages are dynamic (122,123). However, other studies have presented evidence of the presence of M2 macrophages in injured kidneys, which could potentially lead to the secretion of fibrotic growth factors and promote extracellular matrix production (76,68,123). One study indicates that IKKα could contribute to macrophage M2 polarization. Depletion of IKKα in macrophages resulted in reduce M2 polarization and kidney fibrosis after IRI (123). Given that macrophage phenotype determines their function as well as the development of CKD, the researchers examined macrophage polarization following inflammatory stimulation. It was concluded that IKKα-dependent noncanonical NF-κB pathway activation facilitates macrophage M2 polarization, which in turn triggers cytokine-mediated activation of the Wnt/β-catenin pathway, ultimately leading to fibrosis in the injured kidney (123). The role of M2 macrophages encompasses not only anti-inflammatory functions but also pro-fibrotic attributes, notably within the context of kidney IRI. Notably, certain AKI biomarkers, such as neutrophil gelatinase-associated lipocalin and monocyte chemotactic protein-1, exhibit associations with fibrosis following AKI, within the rat bilateral IRI model. Notably, M2 macrophage, rather than M1, infiltration into the kidney demonstrated a significant association with the degree of fibrosis. Alternatively, the study proposed that suppressing M2 macrophage infiltration during the chronic phase after AKI might hold potential for preventing fibrosis progression, warranting further investigation (124).

In kidney tissue, different subsets of macrophages may exist, and macrophages may switch between phenotypes depending on the microenvironment (67). Thus, M1 macrophages further exacerbate AKI caused by IRI by releasing pro-inflammatory cytokines (67,125). Alternatively, other studies indicate that M1 macrophages also contribute to recruiting neutrophils to induce epithelial cell apoptosis (126). In addition to exerting strong pro-inflammatory effects, M1 macrophages can either promote or amplify Th1 polarization in CD4+T cells by releasing IL-12 (127). However, M2 macrophages reduce kidney inflammation by secreting anti-inflammatory cytokines (127). Furthermore, M2 macrophages may also release galectin 3, which induces renal fibrosis (128). Several studies have highlighted that MMT cells that directly lead to fibrogenesis may predominantly have a M2 phenotype (91,105,129). Altogether, M1 and M2 macrophages may promote renal fibrosis through both direct and indirect pathways. In 2009, Weis et al (130) first described the involvement of the hemeoxygenase-1 (HO-1) cytoprotective pathway in the polarization of macrophages toward the M2 phenotype. Studies have also shown that HO-1also regulates macrophage polarization and prevents damage induced by hepatic IRI through the polarization of the M2 phenotype (130132). HO-1 can facilitate a microenvironment that favors the transition from macrophages to M2 phenotype, effectively alleviating AKI damage, preventing the transition to CKD and promoting kidney tissue repair following injury (132). Several studies have reported that MCPIP1 inhibits M1 polarization by blocking NF-κB. The findings suggest that MCPIP1 deficiency can promote M2 macrophage polarization through IRF4 (133,134).

Inflammation plays a pivotal role in the pathogenesis of renal IRI (135,136). During the recovery phase of IRI, macrophages of the M2 phenotype constitute the prevailing subset. Despite M2 macrophages being crucial for short-term recovery, they are also implicated in the development of renal fibrosis subsequent to IRI. To conclude, M2 macrophages play a significant role in fibrosis progression during the transition, from AKI to CKD, with their actions at least partially contingent upon TGF-β (Fig. 5). Thus, when macrophages are regarded as therapeutic targets or tools in the progression from AKI-to-CKD, a more nuanced understanding of achieving an optimal balance between M1 and M2 macrophages to promote recovery while reducing fibrosis is essential for the formulation of clinically viable strategies to impede CKD progression (104).

Folic acid (FA)-mediated renal fibrosis

The progression of CKD following AKI is characterized by robust inflammation and fibrogenesis (137). The rodent model of FA-induced nephropathy simulates essential aspects of the AKI-CKD transition. In the early phase, the pathogenesis of FA nephropathy is characterized by acute tubular cell necrosis and an inflammation response. Persistent inflammatory cell infiltration and fibrosis are two main pathological changes in the chronic phase of AKI. The effectiveness and feasibility of inducing kidney injury by FA treatment have been experimentally validated. Therefore, FA-induced renal fibrosis is considered an ideal model for studying the transition of AKI to CKD (138). A few studies have highlighted the substantial contribution of myeloid myofibroblasts and the transition of M2 macrophages to myofibroblasts in kidney fibrosis observed in FA nephropathy and chronic renal allograft injury (110,138). Previous findings have revealed that IL-4/STAT6 signaling exerts a crucial effect in FA-induced renal fibrosis by modulating bone marrow-derived fibroblasts activation and macrophage M2 polarization (111,112). Robust evidence supports IRF-4 as a downstream effector of the IL-4/STAT6 signaling pathway, driving the differentiation of bone marrow-derived monocytes into the M2 phenotype (139141). Moreover, IRF-4 has been identified as a critical player in myeloid cells differentiation (142), macrophage activation (143) and inflammatory diseases (108). IRF-4 deficiency has been shown to suppress the inflammatory response, activation of the bone marrow-derived fibroblasts and the transition of macrophages to myofibroblasts in FA nephropathy (144).

MicroRNAs (miRs) regulate gene expression during various developmental stages of macrophages, including myelopoiesis, polarization and functional effects. In addition, miRs can also play a role in regulating the signaling and metabolic functions of macrophage polarization (145,146). In co-cultures of SOCS1 with human kidney 2 (HK-2 cells) and macrophages, the antagonism of miR-150 by locked nucleic acid (LNA)-anti-miR-150 resulted in the reversal of the JAK/STAT pathway. These findings further support the notion that LNA-anti-miR-150 reduces FA-induced renal fibrosis (147,148). In an experimental model of FA-induced renal fibrosis, macrophage numbers were significantly elevated (149). Dysregulated macrophage polarization may contribute to a crucial mechanism in the progression of chronic renal inflammation and fibrosis. M1 macrophages might not only contribute to AKI in the early stages of the disease, but also may potentially drive renal fibrosis in later stages due to the sustained detrimental effects of M1 macrophages (149151). One study (141) investigated the effects of a selective STAT6 inhibitor, AS1517499, on myeloid fibroblast activation, macrophage polarization and the development of renal fibrosis in a murine mouse model of FA-induced nephropathy. The findings revealed that within the injured kidney tissue, STAT6 signaling becomes activated, leading to the aggregation and activation of myeloid fibroblasts as well as the polarization of M2 macrophages, ultimately culminating in the progression of renal fibrotic disease. Pharmacological inhibition of STAT6 with AS1517499 resulted in the suppression of myeloid fibroblast accumulation and activation, reduction in M2 macrophage polarization, decreased production of ECM proteins and attenuation of kidney fibrosis and dysfunction (141).

SET domain-containing lysine methyltransferase 7 (also known as SETD7, SET9, SET7/9 or KMT7) is a member of the evolutionarily conserved Su (var) enhancer of zeste and trithorax domain family, initially characterized as a monomethyl transferase of lysine 4 on histone H3. Evidence indicates the involvement of SETD7 in the pathogenesis of fibrotic disorders. A study (151) demonstrates that PFI-2, a specific and potent SETD7 inhibitor, impairs the transition of M2 macrophages-to-myofibroblasts, reduces the accumulation, of bone marrow-derived myofibroblasts, dampens the inflammatory response and mitigates the development of renal fibrosis. In this study, SETD7 inhibition was shown to significantly decrease the number of NF-κB p65+ cells in FA-treated kidneys. These findings suggest that SETD7 contributes to the inflammation response in FA nephropathy through the modulation of NF-κB signaling (151) (Fig. 6). In summary, renal fibrosis represents a crucial pathological aspect of renal diseases. While macrophages play a pivotal role in kidney injury and repair, the functional behaviors of M1 and M2 macrophage cells differ within the context of renal fibrosis induced by FA.

Aristolochic acid (AA)-induced renal fibrosis

In recent years, increased attention has been directed towards the renal toxicity of Chinese herbs. AA is commonly found in various types of Chinese medicinal materials and can also be present in foods contaminated by the environment, leading to kidney injury. The hallmark of the AA-induced renal fibrosis model is the infiltration of activated monocytes/macrophages, which is also a marker of CKD development in humans (152). AA nephropathy (AAN) is characterized by tubular atrophy and interstitial fibrosis, reflecting advanced kidney disease. It is well established that sustained or recurrent episodes of AKI contribute to the progression of CKD. Indeed, in AA-induced renal fibrosis models, an early phase of acute tubular necrosis is rapidly followed by extensive interstitial recruitment of activated monocytes/macrophages and cytotoxic T lymphocytes, leading to a transient AKI episode. Subsequently, a later chronic phase is observed with progressive tubular atrophy resulting from dedifferentiation and necrosis of tubular epithelial cells. The presence of vimentin and α-SMA positive cells expressing TGF-β in interstitial regions indicates an increase in resident fibroblasts and their transformation into myofibroblasts, culminating in collagen deposition and CKD (153).

During AAN, there is a significant increase in CD11b+/F4/80+ expression, with the expression of both CD86+ and CD206+ significantly increasing at days 7, 14 and 28 following AA injection. The immunohistochemistry results for anti-F4/80, anti-CD86 and anti-CD206 in kidney tissues are consistent with the aforementioned findings, indicating the accumulation of M1 and M2 macrophages in AAN. Multiple transcription factors are involved in mediating macrophage polarization, including STATs, peroxisome proliferator-activated receptor (PPAR), Krϋppel-like factors (KLFs) and C/EBP β (154). Some findings demonstrate that 14 days following the addition of STA-21 (a STAT3 inhibitor), the M2 polarization of AA stimulated macrophages is significantly reduced, while the M1 polarization remained unchanged. Thus, STAT3 activation may contribute to AA-induced macrophage M2 polarization as well as the progression of renal fibrosis (155).

Previous studies have validated that acute AA-I exposure can modulate oxidative stress processes by enhancing ROS production in vitro, while simultaneously reducing the ability of macrophages to phagocytose apoptotic neutrophils (156,157). This observation is intriguing and suggests that acute AA-I exposure drives a pro-inflammatory macrophage phenotype with diminished phagocytic capabilities compared to alternatively activated macrophages (158,159). The generation of ROS by macrophages holds significance in a number of physiological processes, particularly those associated with the host's innate immunity responses (160,161). However, ROS release induces not only nitric oxide but also superoxide-anions, hydrogen peroxide and hydroxyl radical, among others, which are associated with mitochondrial respiration (162). Zhang et al (161) describe the importance of ROS production in activated M2-differentiated macrophages, but not in M1-like macrophages. Although the nephrotoxicity of AA is well understood, the precise mechanism by which AA contributes to CKD remains largely unknown (162). It has been reported that miR-382, a small endogenous non-coding RNA, functions as a tumor inhibitor by regulating cell apoptosis, or EMT (163). Studies showed that the upregulation of miR-382 promotes EMT in renal tubular epithelial transcription factors such as IRF4, C/EBP-β, KLF4, STAT3 and 6, PPARγ that have been shown to promote M2 polarization (163,164). Additionally, it is proposed that AA-induced M2 macrophage polarization and subsequent STAT3 phosphorylation may be mediated by the enhancement of Signal-regulatory protein α (SIRPα) targeting miR-382, a key mediator of STAT3 phosphorylation in S727 and Y705 fibrosis (165,166). Thus, renal fibrosis may be triggered by both M1 and M2 macrophages (42). However, the precise molecular mechanism underlying macrophage polarization remains to be elucidated. Research suggests that NF-κB is a key regulator of inflammation and its resolution by regulating macrophage polarization (166) (Fig. 7). In summary, the significance of macrophage infiltration, activation and polarization is increasingly recognized as a pivotal driver of meta-inflammation.

Macrophage polarization-based therapeutic prospects for renal inflammation and fibrosis

Controlling renal inflammation is arguably one of the most effective strategies for targeting renal fibrosis and maintaining renal function. Understanding the pathways of inflammation is a complex endeavor, with its resolution often dependent on the state of macrophage involvement. However, targeting macrophage-mediated renal fibrosis presents a promising avenue for targeted therapeutics, albeit a challenging one. On the one hand, inhibiting the generation of M1 macrophages and curtailing the progression of renal fibrosis is imperative. On the other hand, maintaining an adequate number of M2 macrophages is essential to facilitate tissue repair and immune regulation. The long-standing association between inflammation and macrophages suggests potential avenues for intervention by focusing on ways to shift macrophage polarization from a pro-inflammatory to an anti-inflammatory state. Thus, further exploration of macrophage polarization is paramount to addressing significant gaps in our understanding. The precise mechanisms underlying macrophage polarization across various experimental models of renal fibrosis and the effects of different macrophage phenotypes remain incompletely elucidated. Therefore, additional investigations are warranted.

Acknowledgements

The authors would like to thank the Center for Scientific Research of Anhui Medical University for valuable help in the present review and Professor Cheng Huang(Anhui Medical University) for their guidance on topic selection, ideas, opinions and arguments of this paper and their suggestions for revision when reviewing the paper.

Funding

The present study was supported by funding from the Natural Science Foundation of Anhui Province (grant no 2008085MH273), the Anhui Fund for Distinguished Young Scholars (grant no. 2022AH020050), the Scientific Research Platform Improvement Project of Anhui Medical University (grant no. 2022×kjT045) and the Research Fund of Anhui Institute of translational medicine (grant no. 2021zhyx-B06 and 2022zhyx-B07).

Availability of data and materials

Not applicable.

Authors' contributions

JZ wrote the original draft and analyzed the data. CH designed, supervised and edited the manuscript. YZ edited and revised the manuscript and completed the design and production of the figures. Data authentication is not applicable. All authors read and approved the final version of the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Bommer J: Prevalence and socio-economic aspects of chronic kidney disease. Nephrol Dial Transpl. 11:8–12. 2002. View Article : Google Scholar

2 

Decleves AE and Sharma K: Novel targets of antifibrotic and anti-inflammatory treatment in CKD. Nat Rev Nephrol. 10:257–267. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Lv W, Booz GW, Wang Y, Fan F and Roman RJ: Inflammation and renal fibrosis: Recent developments on key signaling molecules as potential therapeutic targets. Eur J Pharmacol. 820:65–76. 2018. View Article : Google Scholar : PubMed/NCBI

4 

Viehmann SF, Bohner AMC, Kurts C and Brahler S: The multifaceted role of the renal mononuclear phagocyte system. Cell Immunol. 330:97–104. 2018. View Article : Google Scholar : PubMed/NCBI

5 

Nelson PJ, Rees AJ, Griffin MD, Hughes J, Kurts C and Duffield J: The renal mononuclear phagocytic system. J Am Soc Nephrol. 23:194–203. 2012. View Article : Google Scholar : PubMed/NCBI

6 

Cao Q, Wang Y, Wang XM, Lu J, Lee VW, Ye Q, Nguyen H, Zheng G, Zhao Y, Alexander SI, et al: Renal F4/80+ CD11c+ mononuclear phagocytes display phenotypic and functional characteristics of macrophages in health and in adriamycin nephropathy. J Am Soc Nephrol. 26:349–363. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Tang PM, Nikolic-Paterson DJ and Lan HY: Macrophages: Versatile players in renal inflammation and fibrosis. Nat Rev Nephrol. 15:144–158. 2019. View Article : Google Scholar : PubMed/NCBI

8 

Wang Y and Harris DC: Macrophages in renal disease. J Am Soc Nephrol. 22:21–27. 2011. View Article : Google Scholar : PubMed/NCBI

9 

Pan B, Liu G, Jiang Z and Zheng D: Regulation of renal fibrosis by macrophage polarization. Cell Physiol Biochem. 35:1062–1069. 2015. View Article : Google Scholar : PubMed/NCBI

10 

Locati M, Curtale G and Mantovani A: Diversity, mechanisms, and significance of macrophage plasticity. Annu Rev Pathol. 15:123–147. 2020. View Article : Google Scholar : PubMed/NCBI

11 

Viola A, Munari F, Sanchez-Rodriguez R, Scolaro T and Castegna A: The metabolic signature of macrophage responses. Front Immunol. 10:14622019. View Article : Google Scholar : PubMed/NCBI

12 

Kim Y, Nurakhayev S, Nurkesh A, Zharkinbekov Z and Saparov A: Macrophage polarization in cardiac tissue repair following myocardial infarction. Int J Mol Sci. 22:27152021. View Article : Google Scholar : PubMed/NCBI

13 

Qiu P, Liu Y and Zhang J: Review: The role and mechanisms of macrophage autophagy in sepsis. Inflammation. 42:6–19. 2018. View Article : Google Scholar

14 

Kadomoto S, Izumi K and Mizokami A: Macrophage polarity and disease control. Int J Mol Sci. 23:1442021. View Article : Google Scholar : PubMed/NCBI

15 

Li C, Xu MM, Wang K, Adler AJ, Vella AT and Zhou B: Macrophage polarization and meta-inflammation. Transl Res. 191:29–44. 2018. View Article : Google Scholar : PubMed/NCBI

16 

Bashir S, Sharma Y, Elahi A and Khan F: Macrophage polarization: The link between inflammation and related diseases. Inflamm Res. 65:1–11. 2016. View Article : Google Scholar : PubMed/NCBI

17 

Wu Q, Sun S, Wei L, Liu M, Liu H, Liu T, Zhou Y, Jia Q, Wang D, Yang Z, et al: Twist1 regulates macrophage plasticity to promote renal fibrosis through galectin-3. Cell Mol Life Sci. 79:1372022. View Article : Google Scholar : PubMed/NCBI

18 

Ko GJ, Boo CS, Jo SK, Cho WY and Kim HK: Macrophages contribute to the development of renal fibrosis following ischaemia/reperfusion-induced acute kidney injury. Nephrol Dial Transplant. 23:842–852. 2008. View Article : Google Scholar : PubMed/NCBI

19 

Fogo AB: Progression and potential regression of glomerulosclerosis. Kidney Int. 59:804–819. 2001. View Article : Google Scholar : PubMed/NCBI

20 

Rodrignez-Pena A, Prieto M, Duwel A, Rivas JV, Eleno N, Pérez-Barriocanal F, Arévalo M, Smith JD, Vary CP, Bernabeu C and López-Novoa JM: Up-regulation of endoglin, a TGF-beta-binding protein, in rats with experimental renal fibrosis induced by renal mass reduction. Nwphrol Dial Transplant. 16 (Suppl 1):34–39. 2001. View Article : Google Scholar

21 

Tan X, Li Y and Liu Y: Therapeutic role and potential mechanisms of active Vitamin D in renal interstitial fibrosis. J Steroid Biochem Mol Biol. 103:491–496. 2007. View Article : Google Scholar : PubMed/NCBI

22 

Lopez-Novoa JM, Martinez-Salgado C, Rodriguez-Pena AB and Lopez-Hernandez FJ: Common pathophysiological mechanisms of chronic kidney disease: Therapeutic perspectives. Pharmacol Ther. 128:61–81. 2010. View Article : Google Scholar : PubMed/NCBI

23 

Chevalier RL, Forbes MS and Thornhill BA: Ureteral obstruction as a model of renal interstitial fibrosis and obstructive nephropathy. Kidney Int. 75:1145–1152. 2009. View Article : Google Scholar : PubMed/NCBI

24 

Grande MT, Perez-Barriocanal F and Lopez-Novoa JM: Role of inflammation in tubulo-interstitial damage associated to obstructive nephropathy. J Inflamm (Lond). 7:192010. View Article : Google Scholar : PubMed/NCBI

25 

Wang PH, Cenedeze MA, Campanholle G, Malheiros DM, Torres HA, Pesquero JB, Pacheco-Silva A and Camara NO: Deletion of bradykinin B1 receptor reduces renal fibrosis. Int Immunopharmacol. 9:653–657. 2009. View Article : Google Scholar : PubMed/NCBI

26 

Nishida M and Hamaoka K: Macrophage phenotype and renal fibrosis in obstructive nephropathy. Nephron Exp Nephrol. 110:e31–e36. 2008. View Article : Google Scholar : PubMed/NCBI

27 

Duffield JS, Ware CF, Ryffel B and Savill J: Suppression by apoptotic cells defines tumor necrosis factor-mediated induction of glomerular mesangial cell apoptosis by activated macrophages. Am J Pathol. 159:1397–1404. 2001. View Article : Google Scholar : PubMed/NCBI

28 

Kipari T and Hughes J: Macrophage-mediated renal cell death. Kidney Int. 61:760–761. 2002. View Article : Google Scholar : PubMed/NCBI

29 

Lange-Sperandio B, Cachat F, Thornhill BA and Chevalier RL: Selectins mediate macrophage infiltration in obstructive nephropathy in newborn mice. Kidney Int. 61:516–524. 2002. View Article : Google Scholar : PubMed/NCBI

30 

Fadok VA, Bratton DL, Konowal A, Freed PW, Westcott JY and 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 Investigation. 101:890–898. 1998. View Article : Google Scholar : PubMed/NCBI

31 

Song E, Ouyang N, Horbelt M, Antus B, Wang M and Exton MS: Influence of alternatively and classically activated macrophages on fibrogenic activities of human fibroblasts. Cell Immunol. 204:19–28. 2000. View Article : Google Scholar : PubMed/NCBI

32 

Wang D, Xiong M, Chen C, Du L, Liu Z, Shi Y, Zhang M, Gong J, Song X, Xiang R, et al: Legumain, an asparaginyl endopeptidase, mediates the effect of M2 macrophages on attenuating renal interstitial fibrosis in obstructive nephropathy. Kidney Int. 94:91–101. 2018. View Article : Google Scholar : PubMed/NCBI

33 

Nishida M, Okumura Y, Fujimoto S, Shiraishi I, Itoi T and Hamaoka K: Adoptive transfer of macrophages ameliorates renal fibrosis in mice. Biochem Biophys Res Commun. 332:11–16. 2005. View Article : Google Scholar : PubMed/NCBI

34 

Steiger S, Kumar SV, Honarpisheh M, Lorenz G, Gunthner R, Romoli S, Grobmayr R, Susanti HE, Potempa J, Koziel J, et al: Immunomodulatory molecule IRAK-M balances macrophage polarization and determines macrophage responses during renal fibrosis. J Immunol. 199:1440–1452. 2017. View Article : Google Scholar : PubMed/NCBI

35 

Figueiredo KA, Rossi G and Cox ME: Relaxin promotes clustering, migration, and activation states of mononuclear myelocytic cells. Ann N Y Acad Sci. 1160:353–360. 2009. View Article : Google Scholar : PubMed/NCBI

36 

Lan HY: Role of macrophage migration inhibition factor in kidney disease. Nephron Exp Nephrol. 109:e79–e83. 2008. View Article : Google Scholar : PubMed/NCBI

37 

Li MO and Flavell RA: Contextual regulation of inflammation: A duet by transforming growth factor-beta and interleukin-10. Immunity. 28:468–476. 2008. View Article : Google Scholar : PubMed/NCBI

38 

Tregear GW, Bathgate RA, Hossain MA, Lin F, Zhang S, Shabanpoor F, Scott DJ, Ma S, Gundlach AL, Samuel CS and Wade JD: Structure and activity in the relaxin family of peptides. Ann N Y Acad Sci. 1160:5–10. 2009. View Article : Google Scholar : PubMed/NCBI

39 

Samuel CS, Hewitson TD, Unemori EN and Tang ML: Drugs of the future: The hormone relaxin. Cell Mol Life Sci. 64:1539–1557. 2007. View Article : Google Scholar : PubMed/NCBI

40 

Samuel CS and Hewitson TD: Relaxin and the progression of kidney disease. Curr Opin Nephrol Hypertens. 18:9–14. 2009. View Article : Google Scholar : PubMed/NCBI

41 

Chen L, Sha ML, Li D, Zhu YP, Wang XJ, Jiang CY, Xia SJ and Shao Y: Relaxin abrogates renal interstitial fibrosis by regulating macrophage polarization via inhibition of Toll-like receptor 4 signaling. Oncotarget. 8:21044–21053. 2017. View Article : Google Scholar : PubMed/NCBI

42 

Anders HJ and Ryu M: Renal microenvironments and macrophage phenotypes determine progression or resolution of renal inflammation and fibrosis. Kidney Int. 80:915–925. 2011. View Article : Google Scholar : PubMed/NCBI

43 

Bohlson SS, O'Conner SD, Hulsebus HJ, Ho MM and Fraser DA: Complement, c1q, and c1q-related molecules regulate macrophage polarization. Front Immunol. 5:4022014. View Article : Google Scholar : PubMed/NCBI

44 

Cui J, Wu X, Song Y, Chen Y and Wan J: Complement C3 exacerbates renal interstitial fibrosis by facilitating the M1 macrophage phenotype in a mouse model of unilateral ureteral obstruction. Am J Physiol Renal Physiol. 317:F1171–F1182. 2019. View Article : Google Scholar : PubMed/NCBI

45 

Brown NJ: Contribution of aldosterone to cardiovascular and renal inflammation and fibrosis. Nat Rev Nephrol. 9:459–469. 2013. View Article : Google Scholar : PubMed/NCBI

46 

Blasi ER, Rocha R, Rudolph AE, Blomme EA, Polly ML and McMahon EG: Aldosterone/salt induces renal inflammation and fibrosis in hypertensive rats. Kidney Int. 63:1791–1800. 2003. View Article : Google Scholar : PubMed/NCBI

47 

Kang YS, Ko GJ, Lee MH, Song HK, Han SY, Han KH, Kim HK, Han JY and Cha DR: Effect of eplerenone, enalapril and their combination treatment on diabetic nephropathy in type II diabetic rats. Nephrology Dialysis Transplantation. 24:73–84. 2008. View Article : Google Scholar

48 

Yuan X, Wang X, Li Y, Li X, Zhang S and Hao L: Aldosterone promotes renal interstitial fibrosis via the AIF-1/AKT/mTOR signaling pathway. Mol Med Rep. 20:4033–4044. 2019.PubMed/NCBI

49 

Nakamura T, Girerd S, Jaisser F and Barrera-Chimal J: Nonepithelial mineralocorticoid receptor activation as a determinant of kidney disease. Kidney Int Suppl (2011). 12:12–18. 2022. View Article : Google Scholar : PubMed/NCBI

50 

Fiebeler A, Nussberger J, Shagdarsuren E, Rong S, Hilfenhaus G, Al-Saadi N, Dechend R, Wellner M, Meiners S, Maser-Gluth C, et al: Aldosterone synthase inhibitor ameliorates angiotensin II-induced organ damage. Circulation. 111:3087–3094. 2005. View Article : Google Scholar : PubMed/NCBI

51 

Lea WB, Kwak ES, Luther JM, Fowler SM, Wang Z, Ma J, Fogo AB and Brown NJ: Aldosterone antagonism or synthase inhibition reduces end-organ damage induced by treatment with angiotensin and high salt. Kidney Int. 75:936–944. 2009. View Article : Google Scholar : PubMed/NCBI

52 

Luther JM, Luo P, Wang Z, Cohen SE, Kim HS, Fogo AB and Brown NJ: Aldosterone deficiency and mineralocorticoid receptor antagonism prevent angiotensin II-induced cardiac, renal, and vascular injury. Kidney Int. 82:643–651. 2012. View Article : Google Scholar : PubMed/NCBI

53 

Ma J, Weisberg A, Griffin JP, Vaughan DE, Fogo AB and Brown NJ: Plasminogen activator inhibitor-1 deficiency protects against aldosterone-induced glomerular injury. Kidney Int. 69:1064–1072. 2006. View Article : Google Scholar : PubMed/NCBI

54 

Huang W, Xu C, Kahng KW, Noble NA, Border WA and Huang Y: Aldosterone and TGF-β1synergistically increase PAI-1 and decrease matrix degradation in rat renal mesangial and fibroblast cells. Am J Physiol Renal Physiol. 294:F1287–F1295. 2008. View Article : Google Scholar : PubMed/NCBI

55 

Sillen M and Declerck PJ: Targeting PAI-1 in cardiovascular disease: Structural insights Into PAI-1 functionality and inhibition. Front Cardiovasc Med. 7:6224732020. View Article : Google Scholar : PubMed/NCBI

56 

Leroy V, De Seigneux S, Agassiz V, Hasler U, Rafestin-Oblin ME, Vinciguerra M, Martin PY and Féraille E: Aldosterone activates NF-kappaB in the collecting duct. J Am Soc Nephrol. 20:131–144. 2009. View Article : Google Scholar : PubMed/NCBI

57 

Lee H, Fessler MB, Qu P, Heymann J and Kopp JB: Macrophage polarization in innate immune responses contributing to pathogenesis of chronic kidney disease. BMC Nephrol. 21:2702020. View Article : Google Scholar : PubMed/NCBI

58 

Patel V, Joharapurkar A and Jain M: Role of mineralocorticoid receptor antagonists in kidney diseases. Drug Dev Res. 82:341–363. 2021. View Article : Google Scholar : PubMed/NCBI

59 

Martin-Fernandez B, Rubio-Navarro A, Cortegano I, Ballesteros S, Alia M, Cannata-Ortiz P, Olivares-Alvaro E, Egido J, de Andres B, Gaspar ML, et al: Aldosterone induces renal fibrosis and inflammatory M1-macrophage subtype via mineralocorticoid receptor in rats. PLoS One. 11:e01459462016. View Article : Google Scholar : PubMed/NCBI

60 

Malik S, Suchal K, Gamad N, Dinda AK, Arya DS and Bhatia J: Telmisartan ameliorates cisplatin-induced nephrotoxicity by inhibiting MAPK mediated inflammation and apoptosis. Eur J Pharmacol. 748:54–60. 2015. View Article : Google Scholar : PubMed/NCBI

61 

Sindhu G, Nishanthi E and Sharmila R: Nephroprotective effect of vanillic acid against cisplatin induced nephrotoxicity in wistar rats: A biochemical and molecular study. Environ Toxicol Pharmacol. 39:392–404. 2015. View Article : Google Scholar : PubMed/NCBI

62 

Ozkok A and Edelstein CL: Pathophysiology of cisplatin-induced acute kidney injury. Biomed Res Int. 2014:9678262014. View Article : Google Scholar : PubMed/NCBI

63 

Jaiman S, Sharma AK, Singh K and Khanna D: Signalling mechanisms involved in renal pathological changes during cisplatin-induced nephropathy. Eur J Clin Pharmacol. 69:1863–1874. 2013. View Article : Google Scholar : PubMed/NCBI

64 

Huen SC and Cantley LG: Macrophages in renal injury and repair. Annu Rev Physiol. 79:449–469. 2017. View Article : Google Scholar : PubMed/NCBI

65 

Yuasa T, Juniantito V, Ichikawa C, Yano R, Izawa T, Kuwamura M and Yamate J: Thy-1 expression, a possible marker of early myofibroblast development, in renal tubulointerstitial fibrosis induced in rats by cisplatin. Exp Toxicol Pathol. 65:651–659. 2013. View Article : Google Scholar : PubMed/NCBI

66 

Terada N, Karim MR, Izawa T, Kuwamura M and Yamate J: Expression of beta-catenin in regenerating renal tubules of cisplatin-induced kidney failure in rats. Clin Exp Nephrol. 22:1240–1250. 2018. View Article : Google Scholar : PubMed/NCBI

67 

Lee S, Huen S, Nishio H, Nishio S, Lee HK, Choi BS, Ruhrberg C and Cantley LG: Distinct macrophage phenotypes contribute to kidney injury and repair. J Am Soc Nephrol. 22:317–326. 2011. View Article : Google Scholar : PubMed/NCBI

68 

Sears SM, Vega AA, Kurlawala Z, Oropilla GB, Krueger A, Shah PP, Doll MA, Miller R, Beverly LJ and Siskind LJ: F4/80hi resident macrophages contribute to cisplatin-induced renal fibrosis. Kidney. 360:3818–833. 2022.

69 

Yu CC, Chien CT and Chang TC: M2 macrophage polarization modulates epithelial-mesenchymal transition in cisplatin-induced tubulointerstitial fibrosis. Biomedicine (Taipei). 6:52016. View Article : Google Scholar : PubMed/NCBI

70 

Kim SH, Yu MA, Ryu ES, Jang YH and Kang DH: Indoxyl sulfate-induced epithelial-to-mesenchymal transition and apoptosis of renal tubular cells as novel mechanisms of progression of renal disease. Lab Invest. 92:488–498. 2012. View Article : Google Scholar : PubMed/NCBI

71 

Benedetti G, Fokkelman M, Yan K, Fredriksson L, Herpers B, Meerman J, van de Water B and de Graauw M: The nuclear factor κB family member RelB facilitates apoptosis of renal epithelial cells caused by cisplatin/tumor necrosis factor α synergy by suppressing an epithelial to mesenchymal transition-like phenotypic switch. Mol Pharmacol. 84:128–138. 2013. View Article : Google Scholar : PubMed/NCBI

72 

Yamamoto E, Izawa T, Juniantito V, Kuwamura M, Sugiura K, Takeuchi T and Yamate J: Involvement of endogenous prostaglandin E2 in tubular epithelial regeneration through inhibition of apoptosis and epithelial-mesenchymal transition in cisplatin-induced rat renal lesions. Histol Histopathol. 25:995–1007. 2010.PubMed/NCBI

73 

Meng XM, Nikolic-Paterson DJ and Lan HY: Inflammatory processes in renal fibrosis. Nat Rev Nephrol. 10:493–503. 2014. View Article : Google Scholar : PubMed/NCBI

74 

Lech M and Anders HJ: Macrophages and fibrosis: How resident and infiltrating mononuclear phagocytes orchestrate all phases of tissue injury and repair. Biochim Biophys Acta. 1832:989–997. 2013. View Article : Google Scholar : PubMed/NCBI

75 

Nikolic-Paterson DJ, Wang S and Lan HY: Macrophages promote renal fibrosis through direct and indirect mechanisms. Kidney Int Suppl (2011). 4:34–38. 2014. View Article : Google Scholar : PubMed/NCBI

76 

Sica A and Mantovani A: Macrophage plasticity and polarization: In vivo veritas. J Clin Invest. 122:787–795. 2012. View Article : Google Scholar : PubMed/NCBI

77 

Wijesundera KK, Izawa T, Murakami H, Tennakoon AH, Golbar HM, Kato-Ichikawa C, Tanaka M, Kuwamura M and Yamate J: M1- and M2-macrophage polarization in thioacetamide (TAA)-induced rat liver lesions; a possible analysis for hepato-pathology. Histol Histopathol. 29:497–511. 2014.PubMed/NCBI

78 

Nakagawa M, Karim MR, Izawa T, Kuwamura M and Yamate J: Immunophenotypical characterization of M1/M2 macrophages and lymphocytes in cisplatin-induced rat progressive renal fibrosis. Cells. 10:2572021. View Article : Google Scholar : PubMed/NCBI

79 

Lee VW and Harris DC: Adriamycin nephropathy: A model of focal segmental glomerulosclerosis. Nephrology (Carlton). 16:30–38. 2011. View Article : Google Scholar : PubMed/NCBI

80 

Nogueira A, Pires MJ and Oliveira PA: Pathophysiological mechanisms of renal fibrosis: A review of animal models and therapeutic strategies. In Vivo. 31:1–22. 2017. View Article : Google Scholar : PubMed/NCBI

81 

Pippin JW, Brinkkoetter PT, Cormack-Aboud FC, Durvasula RV, Hauser PV, Kowalewska J, Krofft RD, Logar CM, Marshall CB, Ohse T, et al: Inducible rodent models of acquired podocyte diseases. Am J Physiol Renal Physiol. 296:F213–F229. 2009. View Article : Google Scholar : PubMed/NCBI

82 

Wang Y, Wang YP, Zheng G, Lee VW, Ouyang L, Chang DH, Mahajan D, Coombs J, Wang YM, Alexander SI and Harris DC: Ex vivo programmed macrophages ameliorate experimental chronic inflammatory renal disease. Kidney Int. 72:290–299. 2007. View Article : Google Scholar : PubMed/NCBI

83 

Fujimura R, Watanabe H, Nishida K, Fujiwara Y, Koga T, Bi J, Imafuku T, Kobayashi K, Komori H, Miyahisa M, et al: α1-Acid glycoprotein attenuates adriamycin-induced nephropathy via CD163 expressing macrophage induction. Kidney. 360(1): 343–353. 2020. View Article : Google Scholar

84 

Lu J, Cao Q, Zheng D, Sun Y, Wang C, Yu X, Wang Y, Lee VW, Zheng G, Tan TK, et al: Discrete functions of M2a and M2c macrophage subsets determine their relative efficacy in treating chronic kidney disease. Kidney Int. 84:745–755. 2013. View Article : Google Scholar : PubMed/NCBI

85 

Cao Q, Wang Y, Zheng D, Sun Y, Wang Y, Lee VWS, Zheng G, Tan TK, Ince J, Alexander SI and Harris DC: IL-10/TGF-beta-modified macrophages induce regulatory T cells and protect against adriamycin nephrosis. J Am Soc Nephrol. 21:933–942. 2010. View Article : Google Scholar : PubMed/NCBI

86 

Cao Q, Wang Y, Zheng D, Sun Y, Wang C, Wang XM, Lee VWS, Wang Y, Zheng G, Tan TK, et al: Failed renoprotection by alternatively activated bone marrow macrophages is due to a proliferation-dependent phenotype switch in vivo. Kidney Int. 85:794–806. 2014. View Article : Google Scholar : PubMed/NCBI

87 

Wang JY and Wang WP: B7-H4, a promising target for immunotherapy. Cell Immunol. 347:1040082020. View Article : Google Scholar : PubMed/NCBI

88 

Umemura N, Saio M, Suwa T, Kitoh Y, Bai J, Nonaka K, Ouyang GF, Okada M, Balazs M, Adany R, et al: Tumor-infiltrating myeloid-derived suppressor cells are pleiotropic-inflamed monocytes/macrophages that bear M1- and M2-type characteristics. J Leukoc Biol. 83:1136–1144. 2008. View Article : Google Scholar : PubMed/NCBI

89 

Wang S, Meng XM, Ng YY, Ma FY, Zhou S, Zhang Y, Yang C, Huang XR, Xiao J, Wang YY, et al: TGF-β/Smad3 signalling regulates the transition of bone marrow-derived macrophages into myofibroblasts during tissue fibrosis. Oncotarget. 7:8809–8822. 2016. View Article : Google Scholar : PubMed/NCBI

90 

Tavakoli Dargani Z and Singla DK: Embryonic stem cell-derived exosomes inhibit doxorubicin-induced TLR4-NLRP3-mediated cell death-pyroptosis. Am J Physiol Heart Circ Physiol. 317:H460–H471. 2019. View Article : Google Scholar : PubMed/NCBI

91 

Johnson TA and Singla DK: PTEN inhibitor VO-OHpic attenuates inflammatory M1 macrophages and cardiac remodeling in doxorubicin-induced cardiomyopathy. Am J Physiol Heart Circ Physiol. 315:H1236–H1249. 2018. View Article : Google Scholar : PubMed/NCBI

92 

Zhang Y, Liu Y, Liu H and Tang WH: Exosomes: Biogenesis, biologic function and clinical potential. Cell Biosci. 9:192019. View Article : Google Scholar : PubMed/NCBI

93 

Jella KK, Nasti TH, Li Z, Malla SR, Buchwald ZS and Khan MK: Exosomes, their biogenesis and role in inter-cellular communication, tumor microenvironment and cancer immunotherapy. Vaccines (Basel). 6:E692018. View Article : Google Scholar

94 

Deng S, Zhou X, Ge Z, Song Y, Wang H, Liu X and Zhang D: Exosomes from adipose-derived mesenchymal stem cells ameliorate cardiac damage after myocardial infarction by activating S1P/SK1/S1PR1 signaling and promoting macrophage M2 polarization. Int J Biochem Cell Biol. 114:1055642019. View Article : Google Scholar : PubMed/NCBI

95 

Cai J, Qiao B, Gao N, Lin N and He W: Oral squamous cell carcinoma-derived exosomes promote M2 subtype macrophage polarization mediated by exosome-enclosed miR-29a-3p. Am J Physiol Cell Physiol. 316:C731–C740. 2019. View Article : Google Scholar : PubMed/NCBI

96 

Bardi GT, Smith MA and Hood JL: Melanoma exosomes promote mixed M1 and M2 macrophage polarization. Cytokine. 105:63–72. 2018. View Article : Google Scholar : PubMed/NCBI

97 

Singla D, Johnson T and Tavakoli Dargani Z: Exosome treatment enhances Anti-Inflammatory M2 macrophages and reduces inflammation-induced pyroptosis in doxorubicin-induced cardiomyopathy. Cells. 8:12242019. View Article : Google Scholar : PubMed/NCBI

98 

Alexander M, Hu R, Runtsch MC, Kagele DA, Mosbruger TL, Tolmachova T, Seabra MC, Round JL, Ward DM and O'Connell RM: Exosome-delivered microRNAs modulate the inflammatory response to endotoxin. Nat Commun. 6:73212015. View Article : Google Scholar : PubMed/NCBI

99 

Lorenzen JM, Kaucsar T, Schauerte C, Schmitt R, Rong S, Hübner A, Scherf K, Fiedler J, Martino F, Kumarswamy R, et al: MicroRNA-24 antagonism prevents renal ischemia reperfusion injury. J Am Soc Nephrol. 25:2717–2729. 2014. View Article : Google Scholar : PubMed/NCBI

100 

Trionfini P and Benigni A: MicroRNAs as master regulators of glomerular function in health and disease. J Am Soc Nephrol. 28:1686–1696. 2017. View Article : Google Scholar : PubMed/NCBI

101 

Lv LL, Feng Y, Wu M, Wang B, Li ZL, Zhong X, Wu WJ, Chen J, Ni HF, Tang TT, et al: Exosomal miRNA-19b-3p of tubular epithelial cells promotes M1 macrophage activation in kidney injury. Cell Death Differ. 27:210–226. 2020. View Article : Google Scholar : PubMed/NCBI

102 

Mulay SR, Linkermann A and Anders HJ: Necroinflammation in Kidney disease. J Am Soc Nephrol. 27:27–39. 2016. View Article : Google Scholar : PubMed/NCBI

103 

O'Neal JB, Shaw AD and Billings FT: Acute kidney injury following cardiac surgery: Current understanding and future directions. Crit Care. 20:1872016. View Article : Google Scholar : PubMed/NCBI

104 

Kim MG, Kim SC, Ko YS, Lee HY, Jo SK and Cho W: The role of M2 macrophages in the progression of chronic kidney disease following acute kidney injury. PLoS One. 10:e01439612015. View Article : Google Scholar : PubMed/NCBI

105 

Yang J, Lin SC, Chen G, He L, Hu Z, Chan L, Trial J, Entman ML and Wang Y: Adiponectin promotes monocyte-to-fibroblast transition in renal fibrosis. J Am Soc Nephrol. 24:1644–1659. 2013. View Article : Google Scholar : PubMed/NCBI

106 

Negishi H, Ohba Y, Yanai H, Takaoka A, Honma K, Yui K, Matsuyama T, Taniguchi T and Honda K: Negative regulation of Toll-like-receptor signaling by IRF-4. Proc Natl Acad Sci USA. 102:15989–15994. 2005. View Article : Google Scholar : PubMed/NCBI

107 

Lassen S, Lech M, Rommele C, Mittruecker HW, Mak TW and Anders HJ: Ischemia reperfusion induces IFN regulatory factor 4 in renal dendritic cells, which suppresses postischemic inflammation and prevents acute renal failure. J Immunol. 185:1976–1983. 2010. View Article : Google Scholar : PubMed/NCBI

108 

Lorenz G, Moschovaki-Filippidou F, Wurf V, Metzger P, Steiger S, Batz F, Carbajo-Lozoya J, Koziel J, Schnurr M, Cohen CD, et al: IFN regulatory Factor 4 controls post-ischemic inflammation and prevents chronic kidney disease. Front Immunol. 10:21622019. View Article : Google Scholar : PubMed/NCBI

109 

Liang H, Xu F, Zhang T, Huang J, Guan Q, Wang H and Huang Q: Inhibition of IL-18 reduces renal fibrosis after ischemia-reperfusion. Biomed Pharmacother. 106:879–889. 2018. View Article : Google Scholar : PubMed/NCBI

110 

Wang YY, Jiang H, Pan J, Huang XR, Wang YC, Huang HF, To KF, Nikolic-Paterson DJ, Lan HY and Chen JH: Macrophage-to-Myofibroblast transition contributes to interstitial fibrosis in chronic renal allograft injury. J Am Soc Nephrol. 28:2053–2067. 2017. View Article : Google Scholar : PubMed/NCBI

111 

Stephanou A, Brar BK, Scarabelli TM, Jonassen AK, Yellon DM, Marber MS, Knight RA and Latchman DS: Ischemia-induced STAT-1 expression and activation play a critical role in cardiomyocyte apoptosis. J Biol Chem. 275:10002–10008. 2000. View Article : Google Scholar : PubMed/NCBI

112 

McCormick J, Suleman N, Scarabelli TM, Knight RA, Latchman DS and Stephanou A: STAT1 deficiency in the heart protects against myocardial infarction by enhancing autophagy. J Cell Mol Med. 16:386–393. 2012. View Article : Google Scholar : PubMed/NCBI

113 

Braga TT, Correa-Costa M, Guise YF, Castoldi A, de Oliveira CD, Hyane MI, Cenedeze MA, Teixeira SA, Muscara MN, Perez KR, et al: MyD88 signaling pathway is involved in renal fibrosis by favoring a TH2 immune response and activating alternative M2 macrophages. Mol Med. 18:1231–1239. 2012. View Article : Google Scholar : PubMed/NCBI

114 

Buchacher T, Ohradanova-Repic A, Stockinger H, Fischer MB and Weber V: M2 polarization of human macrophages favors survival of the intracellular pathogen chlamydia pneumoniae. PLoS One. 10:e01435932015. View Article : Google Scholar : PubMed/NCBI

115 

Lech M, Gröbmayr R, Ryu M, Lorenz G, Hartter I, Mulay SR, Susanti HE, Kobayashi KS, Flavell RA and Anders HJ: Macrophage phenotype controls long-term AKI outcomes-kidney regeneration versus atrophy. J Am Soc Nephrol. 25:292–304. 2014. View Article : Google Scholar : PubMed/NCBI

116 

Kemmner S, Bachmann Q, Steiger S, Lorenz G, Honarpisheh M, Foresto-Neto O, Wang S, Carbajo-Lozoya J, Alt V, Schulte C, et al: STAT1 regulates macrophage number and phenotype and prevents renal fibrosis after ischemia-reperfusion injury. Am J Physiol Renal Physiol. 316:F277–F291. 2019. View Article : Google Scholar : PubMed/NCBI

117 

Dotor J, Lopez-Vazquez AB, Lasarte JJ, Sarobe P, Garcia-Granero M, Riezu-Boj JI, Martinez A, Feijoo E, Lopez-Sagaseta J, Hermida J, et al: Identification of peptide inhibitors of transforming growth factor beta 1 using a phage-displayed peptide library. Cytokine. 39:106–115. 2007. View Article : Google Scholar : PubMed/NCBI

118 

Diaz-Valdes N, Basagoiti M, Dotor J, Aranda F, Monreal I, Riezu-Boj JI, Borras-Cuesta F, Sarobe P and Feijoo E: Induction of monocyte chemoattractant protein-1 and interleukin-10 by TGFbeta1 in melanoma enhances tumor infiltration and immunosuppression. Cancer Res. 71:812–821. 2011. View Article : Google Scholar : PubMed/NCBI

119 

Qiu SS, Dotor J and Hontanilla B: Effect of P144® (Anti-TGF-β) in an ‘In Vivo’ human hypertrophic scar model in nude mice. PLoS One. 10:e01444892015. View Article : Google Scholar : PubMed/NCBI

120 

Baltanás A, Miguel-Carrasco JL, San José G, Cebrián C, Moreno MU, Dotor J, Borrás-Cuesta F, López B, González A, Díez J, et al: A synthetic peptide from transforming growth Factor-β1 type III receptor inhibits NADPH oxidase and prevents oxidative stress in the kidney of spontaneously hypertensive rats. Antioxid Redox Signal. 19:1607–1618. 2013. View Article : Google Scholar : PubMed/NCBI

121 

Li D, Zhang J, Yuan S, Wang C, Chang J, Tong Y, Liu R, Sang T, Li L, Li J, et al: TGF-β1 peptide-based inhibitor P144 ameliorates renal fibrosis after ischemia-reperfusion injury by modulating alternatively activated macrophages. Cell Prolif. 55:e132992022. View Article : Google Scholar : PubMed/NCBI

122 

Cao Q, Wang Y and Harris DCH: Macrophage heterogeneity, phenotypes, and roles in renal fibrosis. Kidney Int Suppl (2011). 4:16–19. 2014. View Article : Google Scholar : PubMed/NCBI

123 

Zhang F, Fan L, Zhang H, Huang WJ, Sun D, Pan BB, Wan X and Cao CC: Deficiency of IKK α in macrophages mitigates fibrosis progression in the kidney after renal ischemia-reperfusion injury. J Immunol Res. 2021:55210512021. View Article : Google Scholar : PubMed/NCBI

124 

Saito H, Tanaka T, Tanaka S, Higashijima Y, Yamaguchi J, Sugahara M, Ito M, Uchida L, Hasegawa S, Wakashima T, et al: Persistent expression of neutrophil gelatinase-associated lipocalin and M2 macrophage markers and chronic fibrosis after acute kidney injury. Physiol Rep. 6:e137072018. View Article : Google Scholar : PubMed/NCBI

125 

Sepe V, Libetta C, Gregorini M and Rampino T: The innate immune system in human kidney inflammaging. J Nephrol. 35:381–395. 2022. View Article : Google Scholar : PubMed/NCBI

126 

Wen Y, Yan HR, Wang B and Liu BC: Macrophage heterogeneity in kidney injury and fibrosis. Front Immunol. 12:6817482021. View Article : Google Scholar : PubMed/NCBI

127 

Muraille E, Leo O and Moser M: TH1/TH2 paradigm extended: Macrophage polarization as an unappreciated pathogen-driven escape mechanism? Front Immunol. 5:6032014. View Article : Google Scholar : PubMed/NCBI

128 

Henderson NC, Mackinnon AC, Farnworth SL, Kipari T, Haslett C, Iredale JP, Liu F-T, Hughes J and Sethi T: Galectin-3 expression and secretion links macrophages to the promotion of renal fibrosis. Am J Pathol. 172:288–298. 2008. View Article : Google Scholar : PubMed/NCBI

129 

Meng XM, Wang S, Huang XR, Yang C, Xiao J, Zhang Y, To KF, Nikolic-Paterson DJ and Lan HY: Inflammatory macrophages can transdifferentiate into myofibroblasts during renal fibrosis. Cell Death Dis. 7:e24952016. View Article : Google Scholar : PubMed/NCBI

130 

Weis N, Weigert A, von Knethen A and Brüne B: Heme oxygenase-1 contributes to an alternative macrophage activation profile induced by apoptotic cell supernatants. Mol Biol Cell. 20:1280–1288. 2009. View Article : Google Scholar : PubMed/NCBI

131 

Zhang M, Nakamura K, Kageyama S, Lawal AO, Gong KW, Bhetraratana M, Fujii T, Sulaiman D, Hirao H, Bolisetty S, et al: Myeloid HO-1 modulates macrophage polarization and protects against ischemia-reperfusion injury. JCI Insight. 3:1205962018. View Article : Google Scholar : PubMed/NCBI

132 

Naito Y, Takagi T and Higashimura Y: Heme oxygenase-1 and anti-inflammatory M2 macrophages. Arch Biochem Biophysics. 564:83–88. 2014. View Article : Google Scholar : PubMed/NCBI

133 

Ribeiro A, Dobosz E, Krill M, Kohler P, Wadowska M, Steiger S, Schmaderer C, Koziel J and Lech M: Macrophage-specific MCPIP1/Regnase-1 attenuates kidney ischemia-reperfusion injury by shaping the local inflammatory response and tissue regeneration. Cells. 11:3972022. View Article : Google Scholar : PubMed/NCBI

134 

Satoh T, Takeuchi O, Vandenbon A, Yasuda K, Tanaka Y, Kumagai Y, Miyake T, Matsushita K, Okazaki T, Saitoh T, et al: The Jmjd3-Irf4 axis regulates M2 macrophage polarization and host responses against helminth infection. Nat Immunol. 11:936–944. 2010. View Article : Google Scholar : PubMed/NCBI

135 

Fu Y, Xiang Y, Li H, Chen A and Dong Z: Inflammation in kidney repair: Mechanism and therapeutic potential. Pharmacol Ther. 237:1082402022. View Article : Google Scholar : PubMed/NCBI

136 

Bonventre JV and Zuk A: Ischemic acute renal failure: An inflammatory disease? Kidney Int. 66:480–485. 2004. View Article : Google Scholar : PubMed/NCBI

137 

Ferenbach DA and Bonventre JV: Mechanisms of maladaptive repair after AKI leading to accelerated kidney ageing and CKD. Nat Rev Nephrol. 11:264–276. 2015. View Article : Google Scholar : PubMed/NCBI

138 

Jiao B, An C, Tran M, Du H, Wang P, Zhou D and Wang Y: Pharmacological inhibition of STAT6 ameliorates myeloid fibroblast activation and alternative macrophage polarization in renal fibrosis. Front Immunol. 12:7350142021. View Article : Google Scholar : PubMed/NCBI

139 

Liang H, Zhang Z, Yan J and Wang Y, Hu Z, Mitch WE and Wang Y: The IL-4 receptor α has a critical role in bone marrow-derived fibroblast activation and renal fibrosis. Kidney Int. 92:1433–1443. 2017. View Article : Google Scholar : PubMed/NCBI

140 

Lehtonen A, Veckman V, Nikula T, Lahesmaa R, Kinnunen L, Matikainen S and Julkunen I: Differential expression of IFN regulatory factor 4 gene in human monocyte-derived dendritic cells and macrophages. J Immunol. 175:6570–6579. 2005. View Article : Google Scholar : PubMed/NCBI

141 

Hsu AT, Lupancu TJ, Lee M-C, Fleetwood AJ, Cook AD, Hamilton JA and Achuthan A: Epigenetic and transcriptional regulation of IL4-induced CCL17 production in human monocytes and murine macrophages. J Biol Chemistry. 293:11415–11423. 2018. View Article : Google Scholar : PubMed/NCBI

142 

Yamamoto M, Kato T, Hotta C, Nishiyama A, Kurotaki D, Yoshinari M, Takami M, Ichino M, Nakazawa M, Matsuyama T, et al: Shared and distinct functions of the transcription factors IRF4 and IRF8 in myeloid cell development. PLoS One. 6:e258122011. View Article : Google Scholar : PubMed/NCBI

143 

Sasaki K, Terker AS, Pan Y, Li Z, Cao S, Wang Y, Niu A, Wang S, Fan X, Zhang MZ, et al: Deletion of myeloid interferon regulatory factor 4 (Irf4) in mouse model protects against kidney fibrosis after ischemic injury by decreased macrophage recruitment and activation. J Am Soc Nephrol. 32:1037–1052. 2021. View Article : Google Scholar : PubMed/NCBI

144 

Chen M, Wen X, Gao Y, Liu B, Zhong C, Nie J and Liang H: IRF-4 deficiency reduces inflammation and kidney fibrosis after folic acid-induced acute kidney injury. Int Immunopharmacol. 100:1081422021. View Article : Google Scholar : PubMed/NCBI

145 

Nelson MC and O'Connell RM: MicroRNAs: At the interface of metabolic pathways and inflammatory responses by macrophages. Front Immunol. 11:17972020. View Article : Google Scholar : PubMed/NCBI

146 

Niu X and Schulert GS: Functional regulation of macrophage phenotypes by MicroRNAs in inflammatory arthritis. Front Immunol. 10:22172019. View Article : Google Scholar : PubMed/NCBI

147 

Luan J, Fu J, Wang D, Jiao C, Cui X, Chen C, Liu D, Zhang Y, Wang Y, Yuen PST, et al: miR-150-based RNA interference attenuates tubulointerstitial fibrosis through the SOCS1/JAK/STAT pathway in vivo and in vitro. Mol Ther Nucleic Acids. 22:871–884. 2020. View Article : Google Scholar : PubMed/NCBI

148 

Hao X, Luan J, Jiao C, Ma C, Feng Z, Zhu L, Zhang Y, Fu J, Lai E, Zhang B, et al: LNA-anti-miR-150 alleviates renal interstitial fibrosis by reducing pro-inflammatory M1/M2 macrophage polarization. Front Immunol. 13:9130072022. View Article : Google Scholar : PubMed/NCBI

149 

Li C, Ding XY, Xiang DM, Xu J, Huang XL, Hou FF and Zhou QG: Enhanced M1 and Impaired M2 macrophage polarization and reduced mitochondrial biogenesis via inhibition of AMP kinase in chronic kidney disease. Cell Physiol Biochem. 36:358–372. 2015. View Article : Google Scholar : PubMed/NCBI

150 

Wen Y, Lu X, Ren J, Privratsky JR, Yang B, Rudemiller NP, Zhang J, Griffiths R, Jain MK, Nedospasov SA, et al: KLF4 in macrophages attenuates TNF α-mediated kidney injury and fibrosis. J Am Soc Nephrol. 30:1925–1938. 2019. View Article : Google Scholar : PubMed/NCBI

151 

Liu B, Nie J, Liang H, Liang Z, Huang J, Yu W and Wen S: Pharmacological inhibition of SETD7 by PFI-2 attenuates renal fibrosis following folic acid and obstruction injury. Eur J Pharmacol. 901:1740972021. View Article : Google Scholar : PubMed/NCBI

152 

Debelle FD, Vanherweghem JL and Nortier JL: Aristolochic acid nephropathy: A worldwide problem. Kidney Int. 74:158–169. 2008. View Article : Google Scholar : PubMed/NCBI

153 

Baudoux T, Jadot I, Declèves A-E, Antoine M-H, Colet J-M, Botton O, De Prez E, Pozdzik A, Husson C, Caron N and Nortier JL: Experimental aristolochic acid nephropathy: A relevant model to study AKI-to-CKD transition. Front Med (Lausanne). 9:8228702022. View Article : Google Scholar : PubMed/NCBI

154 

Feng Y, Ren J, Gui Y, Wei W, Shu B, Lu Q, Xue X, Sun X, He W, Yang J and Dai C: Wnt/β-catenin-promoted macrophage alternative activation contributes to kidney fibrosis. J Am Soc Nephrol. 29:182–193. 2018. View Article : Google Scholar : PubMed/NCBI

155 

Wang X, Jia P, Ren T, Zou Z, Xu S, Zhang Y, Shi Y, Bao S, Li Y, Fang Y and Ding X: MicroRNA-382 promotes M2-like macrophage via the SIRP-α/STAT3 signaling pathway in aristolochic acid-induced renal fibrosis. Front Immunol. 13:8649842022. View Article : Google Scholar : PubMed/NCBI

156 

Martinez FO, Helming L, Milde R, Varin A, Melgert BN, Draijer C, Thomas B, Fabbri M, Crawshaw A, Ho LP, et al: Genetic programs expressed in resting and IL-4 alternatively activated mouse and human macrophages: Similarities and differences. Blood. 121:e57–69. 2013. View Article : Google Scholar : PubMed/NCBI

157 

Chalayer E, Gramont B, Zekre F, Goguyer-Deschaumes R, Waeckel L, Grange L, Paul S, Chung AW and Killian M: Fc receptors gone wrong: A comprehensive review of their roles in autoimmune and inflammatory diseases. Autoimmun Rev. 21:1030162022. View Article : Google Scholar : PubMed/NCBI

158 

Yadav N and Chandra H: Modulation of alveolar macrophage innate response in proinflammatory-, pro-oxidant-, and infection-models by mint extract and chemical constituents: Role of MAPKs. Immunobiology. 223:49–56. 2018. View Article : Google Scholar : PubMed/NCBI

159 

Formentini L, Santacatterina F, Nunez de Arenas C, Stamatakis K, Lopez-Martinez D, Logan A, Fresno M, Smits R, Murphy MP and Cuezva JM: Mitochondrial ROS production protects the intestine from inflammation through functional M2 macrophage polarization. Cell Rep. 19:1202–1213. 2017. View Article : Google Scholar : PubMed/NCBI

160 

Phaniendra A, Jestadi DB and Periyasamy L: Free radicals: Properties, sources, targets, and their implication in various diseases. Indian J Clin Biochem. 30:11–26. 2015. View Article : Google Scholar : PubMed/NCBI

161 

Zhang Y, Choksi S, Chen K, Pobezinskaya Y, Linnoila I and Liu ZG: ROS play a critical role in the differentiation of alternatively activated macrophages and the occurrence of tumor-associated macrophages. Cell Res. 23:898–914. 2013. View Article : Google Scholar : PubMed/NCBI

162 

Lebeau C, Debelle FD, Arlt VM, Pozdzik A, De Prez EG, Phillips DH, Deschodt-Lanckman MM, Vanherweghem JL and Nortier JL: Early proximal tubule injury in experimental aristolochic acid nephropathy: Functional and histological studies. Nephrol Dial Transplant. 20:2321–2332. 2005. View Article : Google Scholar : PubMed/NCBI

163 

Seok JK, Lee SH, Kim MJ and Lee YM: MicroRNA-382 induced by HIF-1α is an angiogenic miR targeting the tumor suppressor phosphatase and tensin homolog. Nucleic Acids Res. 42:8062–8072. 2014. View Article : Google Scholar : PubMed/NCBI

164 

Wang X, Xue N, Zhao S, Shi Y, Ding X and Fang Y: Upregulation of miR-382 contributes to renal fibrosis secondary to aristolochic acid-induced kidney injury via PTEN signaling pathway. Cell Death Dis. 11:6202020. View Article : Google Scholar : PubMed/NCBI

165 

Pan YF, Tan YX, Wang M, Zhang J, Zhang B, Yang C, Ding ZW, Dong LW and Wang HY: Signal regulatory protein alpha is associated with tumor-polarized macrophages phenotype switch and plays a pivotal role in tumor progression. Hepatology. 58:680–691. 2013. View Article : Google Scholar : PubMed/NCBI

166 

Sanz AB, Sanchez-Nino MD, Ramos AM, Moreno JA, Santamaria B, Ruiz-Ortega M, Egido J and Ortiz A: NF-kappaB in renal inflammation. J Am Soc Nephrol. 21:1254–1262. 2010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2024
Volume 29 Issue 2

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zeng J, Zhang Y and Huang C: Macrophages polarization in renal inflammation and fibrosis animal models (Review). Mol Med Rep 29: 29, 2024
APA
Zeng, J., Zhang, Y., & Huang, C. (2024). Macrophages polarization in renal inflammation and fibrosis animal models (Review). Molecular Medicine Reports, 29, 29. https://doi.org/10.3892/mmr.2023.13152
MLA
Zeng, J., Zhang, Y., Huang, C."Macrophages polarization in renal inflammation and fibrosis animal models (Review)". Molecular Medicine Reports 29.2 (2024): 29.
Chicago
Zeng, J., Zhang, Y., Huang, C."Macrophages polarization in renal inflammation and fibrosis animal models (Review)". Molecular Medicine Reports 29, no. 2 (2024): 29. https://doi.org/10.3892/mmr.2023.13152