Next Article in Journal
6-n-Butoxy-10-nitro-12,13-dioxa-11-azatricyclo[7.3.1.02,7]trideca-2,4,6,10-tetraene Improves the X-ray Sensitivity on Inhibiting Proliferation and Promoting Oxidative Stress and Apoptosis of Oral Cancer Cells
Next Article in Special Issue
Swollen Feet: Considering the Paradoxical Roles of Interleukins in Nephrotic Syndrome
Previous Article in Journal
Characterization of Novel RHD Allele Variants and Their Implications for Routine Blood Group Diagnostics
Previous Article in Special Issue
Recent Advances in Proteinuric Kidney Disease/Nephrotic Syndrome: Lessons from Knockout/Transgenic Mouse Models
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Complement Activation in Nephrotic Glomerular Diseases

by
Dominik Nell
,
Robert Wolf
,
Przemyslaw Marek Podgorny
,
Tobias Kuschnereit
,
Rieke Kuschnereit
,
Thomas Dabers
,
Sylvia Stracke
and
Tilman Schmidt
*,†
Section of Nephrology, Clinic and Policlinic of Internal Medicine A, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, 17475 Greifswald, Germany
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Biomedicines 2024, 12(2), 455; https://doi.org/10.3390/biomedicines12020455
Submission received: 30 December 2023 / Revised: 23 January 2024 / Accepted: 9 February 2024 / Published: 18 February 2024

Abstract

:
The nephrotic syndrome holds significant clinical importance and is characterized by a substantial protein loss in the urine. Damage to the glomerular basement membrane or podocytes frequently underlies renal protein loss. There is an increasing belief in the involvement of the complement system, a part of the innate immune system, in these conditions. Understanding the interactions between the complement system and glomerular structures continually evolves, challenging the traditional view of the blood–urine barrier as a passive filter. Clinical studies suggest that a precise inhibition of the complement system at various points may soon become feasible. However, a thorough understanding of current knowledge is imperative for planning future therapies in nephrotic glomerular diseases such as membranous glomerulopathy, membranoproliferative glomerulonephritis, lupus nephritis, focal segmental glomerulosclerosis, and minimal change disease. This review provides an overview of the complement system, its interactions with glomerular structures, and insights into specific glomerular diseases exhibiting a nephrotic course. Additionally, we explore new diagnostic tools and future therapeutic approaches.

1. Introduction

Nephrotic syndrome (NS) is a glomerular disease of clinical significance and is characterized by proteinuria, hypoalbuminemia, edema, and dyslipidemia [1]. Under physiological conditions, the unique structure of the glomeruli ensures the control of glomerular capillary pressure and a stable glomerular filtration rate (GFR). The glomerular filtration barrier (GFB), comprising specialized endothelial cells, the glomerular basement membrane (GBM), and the podocytes, allows for the filtration of water and small solutes, such as sodium and urea. At the same time, it prevents the passing of large molecules or cellular blood components [2]. Healthy individuals typically exhibit only small quantities of detectable urinary albumin, and even proteins with low molecular weights undergo complete reabsorption in the proximal tubules.
However, nephrotic syndrome is characterized by massive proteinuria, usually exceeding 3500 mg daily. Proteinuria occurs due to GFB dysfunction, typically localized within the GBM or podocytes. While albumin also reaches the primary urine under physiological conditions, the reabsorption mechanisms of proximal tubular cells become insufficient in the presence of a GFB dysfunction, resulting in albuminuria and tubular damage. The loss of albumin through the GFB and impaired tubular reabsorption lead to hypoalbuminemia. Additionally, there is an increased capillary permeability, resulting in the escape of albumin into the interstitial space, further driving hypoalbuminemia [1]. The clinical presentation of NS also includes the development of edema. The assumption that the loss of albumin leads to a loss of oncotic pressure and subsequent edema has mainly been discarded. Instead, there is an increased reabsorption of NaCl. Due to the presence of proteases, which end up in the urine due to proteinuria, there appears to be a proteolytic activation of the epithelial sodium channel (ENaC). Ultimately, this leads to an expansion of plasma volume and the development of edema [3,4]. The tubular resistance to atrial natriuretic peptide might further amplify the plasma volume [5]. Increased hepatic synthesis of lipoproteins and enzymes and the urinary loss of liporegulators lead to hypercholesterolemia and hypertriglyceridemia [1]. In addition, a defect of anticoagulant proteins, an increase in procoagulant factors, platelet hyperreactivity, impaired fibrinolysis, increased plasma viscosity, and urinary loss of plasminogen activator inhibitor-1 lead to significantly increased risk of thrombosis, which makes nephrotic syndrome clinically relevant [6]. Although a decrease in GFR is not necessarily the main symptom of nephrotic syndrome, the development of chronic kidney disease (CKD) is a relevant problem.
The GFB is no longer considered a simple anatomical barrier. Instead, it is now recognized as a site of signaling pathways. It is involved in the immunological diseases of the glomeruli and, for example, interact with the adaptive and innate immune system [7,8,9]. Studies further indicate that the GFB also interacts with the complement system [10,11,12]. The complement system is part of the innate immune system, consisting of numerous proteins that interact with each other, with other local cells, and immune cells. In numerous glomerulonephritides, the complement system is activated by different mechanisms [13]. Based on current clinical studies, new complement system inhibitors are expected to be approved soon.
In this review, we provide an overview of the currently known complement-mediated pathomechanisms that can lead to damage to the basement membrane and podocytes. In detail, we summarize the current knowledge on complement activation in membranous glomerulopathy, membranoproliferative glomerulonephritis, lupus nephritis, focal segmental glomerulosclerosis, and minimal change disease. We also discuss the current state of research and provide an outlook on future research findings and treatment options.

2. Complement

The complement system consists of more than 100 soluble and membrane-bound proteins, activating and deactivating each other. Although the complement system is a part of the innate immune system, it interacts with the adaptive immune system and blood coagulation [14,15,16]. During activation of the complement system, cleavage of the essential complement factor 3 (C3) by a C3 convertase occurs. Here, as at other sites of complement activation, complement cleavage products, which have independent functions, are formed. For example, C3a and C3d mediate pro-inflammatory effects through their action as anaphylotoxins and of binding to leukocytes [17]. An important cleavage product of C3 is C3b, as it is directly involved in forming another enzyme, the C5 convertase. Cleavage of complement factor 5 (C5) produces C5b, which initiates the so-called Membrane Attack Complex (MAC; C5b-9) by attaching complement factors 6, 7, 8, and 9. The MAC ultimately forms pores in lipid bilayers, leading to the lysis of pathogens by a “multi-hit” mechanism [18]. Several studies have elucidated new functions of the MAC beyond its “classical” cytolytic (pore-forming) function. Cells exposed to sublytic MAC can instigate intracellular Ca2+-dependent or Ca2+-independent signaling pathways that affect cell proliferation, induction of apoptosis, cell motility, inflammasome activation, and pro-inflammatory cytokine signaling [19]. At the same time, C5a, another cleavage product of C5, again represents a pro-inflammatory molecule [17]. Even though these processes are uniform, the initiation of complement activation varies (Figure 1).
In principle, there are three different pathways of complement activation: the classical pathway, the alternative pathway, and the lectin pathway. The easiest to understand is activation via antibodies, particularly by immunoglobulin G and M, which correspond to the classical activation pathway. C1q binds to antibodies in classical activation and conforms to an activated protease [19]. Cleavage of C4 and C2 forms the C3 convertase of the classical pathway (C4bC2b). In addition, pattern recognition molecules, such as mannose-binding lectin (MBL), ficolins, and collectins, mediate the cleavage of C2 and C4. Such activation occurs physiologically on bacterial surfaces and corresponds to the lectin pathway [20]. As in classical activation, the C3 convertase of the lectin pathway forms of complement factors 4 and 2 (C4bC2b). The C3 convertase then forms the origin of the common terminal pathway. As previously indicated, the generation of the complement factor C3b occurs concomitantly with the cleavage of C3. Together with cleaved complement factor B (Bb), complement activation is amplified in that C3b and Bb themselves act as C3 convertase of the alternative complement pathway (C3bBb) [21]. In addition, hydrolytic cleavage of C3b from C3 occurs continuously, so the alternative complement pathway is always activated [22].
This system necessitates regulatory mechanisms to counteract persistent activation, ensuring homeostasis between excessive and insufficient activation and adapting to the respective situation [23]. A critical regulator here is complement Factor H (CFH). CFH is a soluble protein that regulates the alternative C3 convertase and the local inhibition of complement in the glomeruli [24]. Accordingly, genetic and functional alterations of CFH were described in both atypical hemolytic uremic syndrome (aHUS) and C3 glomerulopathy (C3G) [25,26]. Regulators for the classical pathway (C4-binding protein (C4bp)) and the lectin pathway (MAP-1) also exist [27,28]. In addition to these soluble inhibitors, locally expressed complement inhibitors, such as CD46, CD59, or CD55 (DAF), are described [29,30,31].
Given that kidney cells can express complement factors themselves, this raises the question of whether the cellular structures of the kidney interact with the complement system [32].

3. Renal Complement Activation

Various authors postulate a role for the complement system in numerous glomerular diseases [13]. In particular, complement activation is crucial in atypical hemolytic uremic syndrome (aHUS). An increased activation of the alternative and the terminal complement pathway in this disease leads to endothelial damage. Clinically, aHUS usually presents as a thrombotic microangiopathy and acute renal failure [33].
In nephrotic syndrome, the primary damage site is the GBM, the podocyte, or both, so involvement of the complement system in glomerular injury must have other potential pathomechanisms. The GBM, produced by endothelial cells and podocytes, comprises α345(IV)collagen, laminin, nidogens, and heparan sulfate proteoglycans. The podocyte is a morphologically highly complex cell. Podocytes branch rapidly with numerous foot processes. The so-called slit diaphragm forms between the foot processes, which is a cell–cell junction harboring essential proteins such as nephrin, podocin, or synaptopodin. With the help of integrins, the foot processes also connect to the GBM [34,35,36]. The podocyte thus prevents the passage of large molecules across the GFB more than any other structure [37]. Due to its high specialization, the podocyte is susceptible to stressors, and complex biological changes occur in the podocyte, including loss of integrity and changes in cellular metabolism. Effacement of the foot processes is the final route of stressed podocytes.
However, the podocyte is no longer recognized as a simple barrier that prevents the passage of proteins into the urine. Instead, there is increasing evidence that the podocyte actively generates and regulates immune-mediated glomerular diseases, for example, by expressing Toll-like receptors and releasing chemokines and cytokines [38,39,40]. The podocyte thus actively participates in shaping a pro-inflammatory environment. The podocyte also appears to influence the course of glomerular diseases by interacting with immune cells. Podocyte expression of MHC molecules and antigen presentation with consecutive podocyte damage have already been demonstrated [8,41]. On the other hand, podocytes also appear to be able to protect themselves against immunological stress [42].
The glomerular activation of complement also represents an immunological stressor on the podocytes. However, only a strong activation of C5b-9 leads to the lysis of the podocytes. The binding of C5b-9 in sublytic doses already induces changes within the metabolism of the podocytes. The activation of the complement system on the podocyte thus potentially leads to an influence on kinases, lipid metabolism, stress on the endoplasmic reticulum, and changes in the ubiquitinin–proteasome system [43,44,45,46]. In contrast, the podocyte appears to be able to protect itself from sublytic doses of MAC. In vitro data suggest that autophagy protects the podocyte from such attacks. Lv et al. reported increased changes in disease injury-related morphology when autophagy was inhibited with 3-methyladenine, while treatment with rapamycin resulted in better podocyte survival [47]. The importance of autophagy and lysosomal degradation was confirmed in a further study [48]. In vitro studies with podocytes from patients with membranous glomerulopathy showed that although there was an increase in the number of autophagosomes in the podocytes of the patients, there was no increase in lysosomal degradation. The authors conclude from this that C5b-9 inhibits degradation in general. By examining the expression profile of complement in podocytes, it became clear that numerous complement factors can be detected using rtPCR [8]. The authors found the expression of complement factors and their receptors. After the role of pro-inflammatory complement cleavage products (e.g., C5a) had become clear, Abe et al. investigated the expression of the C5a receptor (C5aR) in various human glomerulonephritides. Using immunohistochemistry and in situ hybridization, a significant increase in C5aR was observed in the cases of IgA nephropathy and membranous glomerulopathy, including podocytes [49]. Surgically resected kidneys and minimal change disease served as controls, where the C5aR was predominantly detected tubularly by immunohistochemistry. An elegant paper elucidated the connection between complement activation on podocytes and nephrotic syndrome. Angeletti et al. investigated the role of the decay acceleration factor (DAF/CD55), a regulator of C3 convertase. The influence of DAF on podocytes was demonstrated in various mouse models. The authors assume an interaction of C3a with the podocyte, which indirectly prevents the regulation of C3 convertase [31]. Zoshima et al. found complement Factor H expressed in podocytes [10]. This work provided evidence of the functional significance of CFH production from podocytes. Using a podocyte-specific mouse model, the authors showed that increased expression of CFH is associated with improved clearance of subendothelial deposited immunoglobulin G. The mechanism by which CFH is involved in removing immunoglobulins could not be clarified in this study. The podocyte upregulates the production of VEGF under different conditions. An increased production of vascular endothelial growth factor (VEGF) also leads to an increased production of CFH. This observation was made not only in podocytes, but also in retinal cells. Blocking VEGF increased the deposition of complement factors of the alternative pathway [50]. In addition to the production of CFH, the podocyte might also be an activator of complement. In vitro data suggest the production of C3 and the cleavage into the pro-inflammatory cleavage product C3a by the podocyte [11].
Even the basement membrane regulates complement activation due to its composition. Heparan sulfate proteoglycans (HSPGs), an essential component of the GBM, appear to play a role here. It has long been assumed that HSPGs are involved in the charge selectivity of GFB. Accordingly, a loss of HSPGs has been observed in several nephrotic diseases (lupus nephritis, membranous glomerulopathy, minimal change disease). However, CFH also binds to HSPGs of the GBM. In this way, the GBM protects itself from attacks by the complement system [12].
A further potential mechanism of injury is the renal excretion of proteins and proteases in urine, which include complement factors. Urinary activation products of the complement system (C5b-9) correlate with the progression of diabetic nephropathy. Additionally, more severe tubular damage has been observed in patients with high activation products in urine [51]. In a study by Woern et al., the presence of complement factors in patients with various nephrotic conditions (including membranous nephropathy, minimal change glomerulopathy/focal segmental glomerulosclerosis, and immune complex glomerulonephritis) was demonstrated in urine. Furthermore, an increased activity of proteases from the clotting cascade and the complement cascade was observed. Examples of the latter include complement factors B and D. Therefore, a change in the composition of the complement system due to proteinuria is conceivable. However, speculation also arises about tubular kidney damage through the increased activity of proteases, leading to the activation of the complement system in urine [52].
Recently, Medica et al. published a new concept of complement regulation that goes beyond the known mechanisms. The authors showed in an in vitro model that endothelial progenitor cells could also achieve local complement regulation. These cells were able to influence endothelial cells and podocytes via vesicle-transported RNA. In a pro-inflammatory environment and in the presence of C5a, transferred RNA could maintain the functions of endothelial cells and podocytes [53].
The diverse mechanisms of activation and regulation of the complement system make a therapeutic approach in various nephrotic diseases interesting. However, a more precise understanding of the different nephrotic glomerular diseases is necessary for planning future therapies.

4. Nephrotic Glomerular Diseases

4.1. Membranous Nephropathy (MN)

MN is the most common nephrotic glomerulonephritis in adults [54]. Specific autoantibodies against podocyte-expressed phospholipase A2 receptor (PLA2R) and thrombospondin type 1 domain-containing 7 A (THSD7A) can explain about 75% of cases [55,56]. Renal biopsies are characterized by depositions of subepithelial IgG and C3, suggesting an activation of the classical complement pathway. Autoantibodies against PLA2R and THSD7A are predominantly of the IgG4 isotype. This isotype has the lowest binding capacity for the complement, so complement activation via the classical pathway is unlikely to be the primary mechanism [57]. Nevertheless, C1q and C4d deposits in the vast majority of MN support the assumption of classical activation [58,59]. On the other hand, other authors have also described the glomerular deposition of MBL in the MN. Since activation of the lectin pathway can also lead to deposits of C4d, one can interpret such findings as the primary activation of the lectin pathway [59]. However, correlations between MBL in the sera of patients and the clinical course were not meaningful. In contrast, correlations between the autoantibody titers and the clinical course are possible [60]. In addition, Bally et al. described cases of MN in patients with a genetic MBL deficiency [61]. There is also discussion about alternative complement activation in MN. Seifert et al. recently addressed the question of primary complement activation [62]. In human kidney biopsies, the authors detected deposits of C1q. As in other studies, IgG4 was the dominant isotype antibody deposition. However, Seifert et al. showed that in addition to IgG4, at least one other IgG isotype that can activate complement is deposited in the kidney. In addition, a proximity ligation assay could demonstrate a dominance of classical activation. The dominance of the classical complement activation contrasts with a study by Manral et al. Based on in vitro studies, an alternative pathway activation is primarily assumed here. In this study, the authors analyzed patients with a THSD7A-associated MN and hypothesized an alternative complement pathway activation by binding to IgG4. The observation that the binding of C3b was completely absent in factor B-depleted sera supports the assumption of an alternative activation pathway [63].

4.2. Membranoproliferative Glomerulonephritis (MPGN)

MPGN is a morphological pattern of glomerular changes characterized by a thickening of the glomerular basement membrane. The thickening of the basement membrane is caused by deposits of complement factors and, in some cases, immunoglobulins. MPGN can occur in numerous diseases [64]. Depending on the location of the deposits, MPGN type I shows subendothelial and mesangial deposits, type II MPGN intramembranous deposits, and type III MPGN additional subepithelial deposits. Since the composition and the localization of deposits vary, the clinical course of MPGN is very heterogeneous. A nephrotic syndrome occurs in 30–50% of cases [64,65]. The Consensus Report 2013 advocated restructuring the classification of MPGN. The objective was to pivot toward categorization based on immunohistochemical and immunofluorescence characteristics, departing from solely morphological distinctions [66,67]. A novel classification termed C3 glomerulopathy (C3G) was proposed for instances where C3 deposition surpassed that of immunoglobulins. This redefinition of C3G encompassed the patterns noted in MPGN types I and III, along with intramembranous glomerulonephritis/dense deposit disease (MPGN type II). Furthermore, the scope of diagnosing C3G was expanded beyond membranoproliferative patterns to encompass other manifestations of glomerulonephritis, such as mesangioproliferative patterns. In scenarios where immunoglobulin deposition was predominant, the nomenclature transitioned to immune complex-mediated MPGN (IC-MPGN). The evolution of this revised definition stemmed from advancements in comprehending complement-mediated kidney diseases, with C3 glomerulopathy (C3G) serving as a prototype [68]. Within C3G, an excessive activation of the complement system has been associated with genetic mutations in various complement genes, notably Factor H, C3, and the genes encoding FHR1, FHR2, FHR3, FHR4, and FHR5. A subclass of antibodies known as nephritic factors has been identified, playing a role in stabilizing complement activation by binding to elements such as the alternative C3 convertase, C5 convertase, Factor H, C3, C3b, C3d, or Factor B. These antibodies disrupt the alternative pathway, resulting in its hyperactivation. Numerous additional mutations, predominantly affecting the alternative complement pathway, have also been identified. These mutations involve genes associated with FHR1, FHR2, FHR3, FHR4, FHR5, and C3, which encode components forming C3 or C5 convertases or regulators that govern the timing and site of C3 convertase activity [69]. However, a strict distinction between C3G and IC-MPGN is not always feasible. Patients with IC-MPGN also exhibit genetic and acquired disruptions in the alternative pathway. A retrospective study of 140 patients suffering from idiopathic IC-MPGN or C3G revealed a prevalence of genetic disorders not only in C3G, but also in IC-MPGN. The finding of mutations in the alternative C3 convertase components linked both diseases to alternative complement activation. Antibodies stabilizing the C3 convertase were also detectable in IC-MPGN, not only in C3G [70,71].

4.3. Lupus Nephritis (LN)

LN occurs in approximately 50–75% of cases of systemic lupus erythematosus [72]. It significantly determines the morbidity and mortality of the disease. The presence of autoantibodies, such as those against dsDNA, leads to the formation of immune complexes that can deposit in the kidneys. LN exhibits diverse clinical and morphological manifestations. Morphologically, there are six distinct types, with LN Type VI characterized by advanced sclerosis and, consequently, unresponsiveness to therapy. The focal LN (Type III) and diffuse LN (Type VI) often present a nephritic syndrome with a rapid decline in GFR. Type V LN morphologically shows a membranous pattern and frequently presents clinically as a nephrotic syndrome. Overall, LN demonstrates deposits of immunoglobulins and complement factors, often referred to as a “full-house pattern” [73]. Reduced serum levels of C3 and C4 serve as activity markers of LN [74]. Accordingly, the serum has elevated corresponding cleavage products such as iC3d, C4d, and C5b-9 [75,76]. The deposition of immunoglobulins and complement factors immediately suggests activation of the classical complement pathway in LN. This is supported by C4d deposits, which are frequently diffuse in Type III/IV LN but are subepithelial in Type V LN [77,78]. Thus, the location of complement activation and various dysregulations may lead to different types of LN [79]. Activation of the lectin pathway via increased MBL levels has also been described [80]. Another indicator of lectin pathway activation is the alteration of MASP1 and MASP2 in LN, which was observed in LN type III and IV compared to the membranous form [81]. Consequently, activation of the lectin pathway might contribute to the manifestation of different types of LN.

4.4. Focal Segmental Glomerulosclerosis (FSGS)

FSGS represents a histopathological description of morphological changes within glomeruli, predominantly characterized by segmental sclerosis. The challenge lies in recognizing that this histopathological pattern can emerge from many glomerulopathies. Often, FSGS represents the common endpoint of vastly different conditions mediated by adaptation (a mismatch between glomerular load and capacity), genetics, viral association (such as HIV), or medication-induced factors (e.g., lithium). In the primary form, akin to minimal change disease (MCD), there is an assumption of a circulating factor responsible for the disease [82]. Deposits of immunoglobulins like IgM and C3 are generally perceived as nonspecific. However, evidence is mounting regarding the involvement of the complement system in FSGS [83,84,85,86]. Serum studies in patients have revealed indications of complement activation, where complement cleavage products such as C3a, C5a, sC5b-9, C4a, and C4d were elevated [84,85,86]. The local regulation of complement becomes particularly intriguing. Angeletti et al. identified CD55/DAF as a crucial regulator of local complement activation in a mouse model of adriamycin-induced FSGS. A podocyte-specific knockout of DAF resulted in an increased C3b deposition in the glomeruli. The authors argue that C3a signaling in podocytes reduced nephrin expression [31]. Indeed, mutations in nephrin have been described in humans with FSGS [87]. Another study demonstrates a reduced DAF expression in FSGS patients, establishing a potential link to the human condition [88]. Even in the frequently described deposits of IgM, they might serve as effectors of the disease rather than nonspecific deposits. Trachtmann et al. describe IgM deposits in FSGS patients, correlating with deposits of complement products from the classical complement pathway. Furthermore, the authors speculate that IgM antibodies are not deposited nonspecifically but bind to glomerular antigens. It remains unclear whether antibody binding and complement activation contribute to disease onset or if these occur secondarily to glomerular damage. However, complement activation potentially contributes to disease progression.

4.5. Minimal Change Disease (MCD)

MCD is the most common cause of nephrotic syndrome in childhood. The disease was explicitly named due to the absence of morphological changes in light microscopy, including the absence of complement factors [89]. It is important to note that diagnosing MCD in childhood often relies not on biopsy, but on clinical criteria and response to cortisone. Biopsy for MCD is commonly performed in cases of atypical presentations or during adolescence or adulthood. This fact makes scientific analysis challenging. However, due to an immunologically mediated pathophysiology, interaction with the complement system is at least conceivable [90]. Nevertheless, current data regarding complement activation do not extend beyond measuring complement cleavage products in patients’ blood. Elevated levels of C5b-9 have been observed, while serum levels of C3 and C2 were reduced [88,91]. Even measurements of serum levels of C4a were inconclusive in different studies [92]. Therefore, a clear hypothesis regarding the involvement of complement in MCD cannot be conclusively generated.

5. Discussion

With the approval of Eculizumab, the therapy for aHUS has been revolutionized, shifting the focus toward the complement system in numerous glomerulonephritides [93]. Despite the successful MAC blockade in aHUS, complement-mediated kidney diseases remained elusive. Similar to aHUS, an overactivation of the alternative complement pathway leads to C3G. The inhibition of the terminal complement pathway is rarely effective in C3G; currently, no approved therapy exists [33,94]. It remains uncertain how to distinguish G3G from IC-MPGN. Despite the dominant immunoglobulin deposits in IC-MPGN, it frequently shares a histopathological pattern with C3G. Patients with IC-MPGN also exhibit alterations in the alternative complement pathway, which suggests that they might not be distinct diseases, and the immunoglobulin deposits in IC-MPGN could merely trigger complement activation [70,71]. Similarly, this scenario might apply to atypical postinfectious glomerulonephritis, often presenting as NS [95]. Typically, postinfectious glomerulonephritis follows a self-resolving course. However, in some cases, hematuria and proteinuria persist. Even in “atypical postinfectious glomerulonephritis”, underlying disturbances in the alternative pathway have been observed, suggesting that infection and immune complex deposition could serve as triggers for complement activation in this scenario. Overall, C3G is not primarily mediated by the terminal complement pathway but by mediators released from the proximal complement cascade [68]. Inhibition of the alternative complement pathway thus represents a potential therapeutic option. Currently, numerous drugs are in clinical trials, aiming to inhibit complement activation at various points (Table 1). However, these potential agents exhibit significant physiological differences. Iptacopan, an orally available small molecule inhibiting the activity of Factor B, has made the most progress. It aims to inhibit the alternative C3 convertase (C3bBb) and the alternative C5 convertase (C3bBbC3b), thereby inhibiting alternative complement activation at multiple points and reducing pro-inflammatory cleavage products. Iptacopan’s efficacy has been demonstrated by normalizing C3 levels in patient sera. Additionally, treatment resulted in a reduction of C3 deposits in the examined kidneys and was associated with a significant decrease in proteinuria [96]. An alternative approach involves the inhibition of Factor D, which is involved in the enzymatic cleavage of Factor B; however, this approach is not currently further pursued in C3G. Other investigated substances show less specificity toward the alternative complement pathway. Approaches include inhibiting the activity of the C3 convertase by binding to C3 or downregulating hepatic C3 production through siRNA. Avacopan, an inhibitor of the C5aR already approved for ANCA-associated vasculitis therapy, is currently undergoing clinical evaluation. With Avacopan, the goal is to reduce the pro-inflammatory effect of C5a while keeping the rest of the complement cascade intact [97].
In MN, there are varying perspectives on the primary complement activation. Seifert et al. investigated human biopsies associated with PLA2R1 and THSD7A-positive MN. As previously known, this study confirmed a dominant deposition of IgG4. However, IgG4 has a limited ability to activate complement. Yet, it was demonstrated that 100% of the 39 examined cases also exhibited another IgG isotype capable of complement activation (IgG1/IgG2/IgG3). In the case of MN, activation appears to occur primarily through immunoglobulins, specifically the classical pathway [62]. Conversely, other publications highlighting the central role of the alternative and lectin pathway exist [59,63]. Currently, when specific therapy is deemed necessary, the treatment of membranous nephropathy (MN) increasingly focuses on B-cell depletion. This approach aims to inhibit the production of autoantibodies [98]. Indirectly, this would also address activation through the classical pathway. Clinical trials investigating the specific inhibition of the classical and lectin pathways are in the early phases (Table 1). Utilizing the C3 inhibitor, pegcetacoplan, leads to the comprehensive inhibition of the complement system, thereby potentially achieving heightened efficacy. However, this approach comes with the trade-off of potential adverse events. To mitigate the latter, a more precise targeting of the complement system is appealing, with a consideration for specific modulation of the alternative pathway. The alternative complement pathway serves as an amplification mechanism for complement activation, operating independently or being triggered through the classical or lectin pathway [99].
Consequently, inhibiting the alternative pathway could be a therapeutic option for diseases primarily activated through the classical pathway. Lupus nephritis is characterized by deposits of antigen–antibody complexes (immune complexes), suggesting complement system activation through the classical pathway. Paradoxically, classical pathway complement factor mutations are considered risk factors for systemic lupus [100]. Classical complement activation appears to have a protective role by eliminating apoptotic cells. Therefore, inhibiting the classical complement pathway may not necessarily be beneficial. However, a potential approach could involve the specific complement blockade while preserving classical activation. Thus, along with inhibiting the alternative complement pathway, targeting specific complement blockade could also be considered in LN.
Diverse approaches to complement inhibition are emerging for all glomerular diseases. The current state of research does not allow for clear recommendations either in favor or against a specific inhibitor. Particularly in the case of IgA nephropathy (IgAN), uncertainties regarding complement inhibition as a therapeutic option are evident. IgAN is the most common primary glomerulonephritis globally [101]. It is an autoimmune disease characterized by the production of galactose-deficient IgA1 and antibodies against these IgA molecules. The mesangial deposition of these immune complexes leads to proliferation and glomerular infiltration [102]. The deposition of immune complexes can trigger complement system activation, with the exact mechanisms remaining poorly understood. Various complement activation pathways are discussed in the context of IgAN, focusing on the lectin and the terminal complement pathways. However, amplification via the alternative complement pathway is also considered, leading to ongoing clinical trials involving different complement inhibitors (Table 1) [103]. Promising results have been observed with the inhibition of the alternative complement pathway. Iptacopan, for instance, demonstrated a reduction in proteinuria compared to placebo in a Phase 2 study [104]. Data on C5 inhibition using Eculizumab were inconsistent, necessitating further study results. Surprisingly, despite a plausible pathophysiology for lectin pathway activation, the results of the investigation of Narsoplimab were disappointing. The expected reduction in proteinuria could not be achieved, leading to the termination of the study in 2023 [103]. The current understanding of the complement system is insufficient to predict therapeutic success, as indicated by the various studies and unexpected results of clinical studies.
For that, precise therapeutic planning is challenging because current diagnostics do not definitively determine complement activation at the time of diagnosis, nor can they identify the primary complement activation pathway (alternative, classical, or lectin). Urine would be an easily accessible biomaterial to measure complement activation. Indeed, there are data on complement products in the urine for the diseases discussed above [84,88,105]. However, these data are often derived from small case series, limiting normalization. Thus, it is conceivable that the levels of complement products in urine might not correlate with disease activity but with proteinuria or impaired reabsorption. A study from 2021 provided a large dataset of more than 16,000 patients. This publication normalized urinary complement products against proteinuria and GFR, and the analysis included a control cohort and numerous glomerular diseases. Analysis like this will help interpret data and identify potential biomarkers. For example, this work showed increased urinary excretion of complement factors after normalization to proteinuria in LN compared to systemic lupus. Even more intriguing was that some complement factors, such as Factor B, correlated less with GFR but were associated with specific entities (IgAN). As Factor B participates in the complement system’s alternative activation, this could reflect the primary activation (alternative pathway). Such data could be a diagnostic tool for complement activation and activity [92]. Efforts are also being made from kidney biopsies to quantify activity and identify the primary activation [62,106]. A proximity ligation assay identifies the primary complement activation pathway by distinguishing the different C3 convertase (C3bBb vs. C4bC2b) in renal biopsies. In the study by Person et al., a clear dominance of classical complement activation was evident in LN, while aHUS showed more activation of the alternative pathway [107]. In principle, with the expansion of such studies, an assignment to the primary complement activation in individual disease entities could be achieved. Additionally, the intensity of deposits might provide insight into the activity of the complement system at the time of examination. However, a definitive assignment to the primary activation of the complement system is not yet adequately possible.
Testing specific complement inhibition requires careful consideration in light of the myriad unresolved queries and uncertainties. Given the intricate nature of complement activation and the regulatory proteins involved, a thorough examination of activation patterns in both responders and non-responders from clinical studies becomes essential. This comprehensive assessment should encompass the analysis of plasma, kidney, and urine, potentially enabling the identification of distinct activation patterns. Such patterns could, in turn, facilitate the anticipation of treatment response in prospective scenarios. When contemplating the design of clinical trials, meticulous planning becomes paramount. Patient selection may necessitate a basis in their complement patterns, as discerned through a kidney biopsy. Moreover, the incorporation of surrogate endpoints is crucial, with a focus on measures of complement activation. This may encompass assessing blood and urinary levels of complement or employing staining techniques for complement products in repeated biopsies. Such considerations are particularly pertinent in the context of proof-of-concept studies.
In summary, the complement system plays a crucial role in various glomerular diseases. However, the exact mechanisms of its functioning are still not fully understood. The clinical manifestations of different diseases can be attributed to different patterns of complement dysfunction. A more detailed analysis can help to identify pathological subtypes, which can offer insights into the prognosis and activation of the disease. Therefore, a thorough understanding of the activation and control of the complement system within specific entities is necessary. This can facilitate personalized therapeutic decisions in the future.

Author Contributions

Conceptualization: D.N., S.S. and T.S.; validation: D.N., R.W., P.M.P., T.K., R.K., T.D., S.S. and T.S.; investigation: D.N., R.W., P.M.P., T.K., R.K., T.D., S.S. and T.S.; writing—original draft preparation: D.N., R.W., P.M.P., T.K., R.K. and T.D.; writing—review and editing: D.N., S.S. and T.S.; visualization: R.W., P.M.P., T.K., R.K. and T.D.; supervision: T.S.; project administration: S.S. and T.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

T.S. has received honors from Alexion Pharma GmbH and Novartis Pharma GmbH. T.S. acts as a consultant to Eleva GmbH. The activities did not influence this work. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

  1. Claudio:, P.; Gabriella, M. Nephrotic Syndrome: Pathophysiology and Consequences. J. Nephrol. 2023, 36, 2179–2190. [Google Scholar] [CrossRef]
  2. Benzing, T.; Salant, D. Insights into Glomerular Filtration and Albuminuria. N. Engl. J. Med. 2021, 384, 1437–1446. [Google Scholar] [CrossRef]
  3. Hinrichs, G.R.; Jensen, B.L.; Svenningsen, P. Mechanisms of Sodium Retention in Nephrotic Syndrome. Curr. Opin. Nephrol. Hypertens. 2020, 29, 207–212. [Google Scholar] [CrossRef]
  4. Larionov, A.; Dahlke, E.; Kunke, M.; Zanon Rodriguez, L.; Schiessl, I.M.; Magnin, J.; Kern, U.; Alli, A.A.; Mollet, G.; Schilling, O.; et al. Cathepsin B Increases ENaC Activity Leading to Hypertension Early in Nephrotic Syndrome. J. Cell. Mol. Medi 2019, 23, 6543–6553. [Google Scholar] [CrossRef]
  5. Siddall, E.C.; Radhakrishnan, J. The Pathophysiology of Edema Formation in the Nephrotic Syndrome. Kidney Int. 2012, 82, 635–642. [Google Scholar] [CrossRef] [PubMed]
  6. Gyamlani, G.; Molnar, M.Z.; Lu, J.L.; Sumida, K.; Kalantar-Zadeh, K.; Kovesdy, C.P. Association of Serum Albumin Level and Venous Thromboembolic Events in a Large Cohort of Patients with Nephrotic Syndrome. Nephrol. Dial. Transplant. 2017, 32, 157–164. [Google Scholar] [CrossRef] [PubMed]
  7. Xia, H.; Bao, W.; Shi, S. Innate Immune Activity in Glomerular Podocytes. Front. Immunol. 2017, 8, 122. [Google Scholar] [CrossRef]
  8. Li, S.; Liu, Y.; He, Y.; Rong, W.; Zhang, M.; Li, L.; Liu, Z.; Zen, K. Podocytes Present Antigen to Activate Specific T Cell Immune Responses in Inflammatory Renal Disease. J. Pathol. 2020, 252, 165–177. [Google Scholar] [CrossRef] [PubMed]
  9. Bhargava, R.; Tsokos, G.C. The Immune Podocyte. Curr. Opin. Rheumatol. 2019, 31, 167–174. [Google Scholar] [CrossRef] [PubMed]
  10. Zoshima, T.; Hara, S.; Yamagishi, M.; Pastan, I.; Matsusaka, T.; Kawano, M.; Nagata, M. Possible Role of Complement Factor H in Podocytes in Clearing Glomerular Subendothelial Immune Complex Deposits. Sci. Rep. 2019, 9, 7857. [Google Scholar] [CrossRef] [PubMed]
  11. Mühlig, A.K.; Keir, L.S.; Abt, J.C.; Heidelbach, H.S.; Horton, R.; Welsh, G.I.; Meyer-Schwesinger, C.; Licht, C.; Coward, R.J.; Fester, L.; et al. Podocytes Produce and Secrete Functional Complement C3 and Complement Factor H. Front. Immunol. 2020, 11, 1833. [Google Scholar] [CrossRef]
  12. Borza, D.-B. Glomerular Basement Membrane Heparan Sulfate in Health and Disease: A Regulator of Local Complement Activation. Matrix Biol. 2017, 57–58, 299–310. [Google Scholar] [CrossRef] [PubMed]
  13. Petr, V.; Thurman, J.M. The Role of Complement in Kidney Disease. Nat. Rev. Nephrol. 2023, 19, 771–787. [Google Scholar] [CrossRef] [PubMed]
  14. Zhang, X.; Kimura, Y.; Fang, C.; Zhou, L.; Sfyroera, G.; Lambris, J.D.; Wetsel, R.A.; Miwa, T.; Song, W.-C. Regulation of Toll-like Receptor–Mediated Inflammatory Response by Complement in Vivo. Blood 2007, 110, 228–236. [Google Scholar] [CrossRef] [PubMed]
  15. Arbore, G.; West, E.E.; Spolski, R.; Robertson, A.A.B.; Klos, A.; Rheinheimer, C.; Dutow, P.; Woodruff, T.M.; Yu, Z.X.; O’Neill, L.A.; et al. T Helper 1 Immunity Requires Complement-Driven NLRP3 Inflammasome Activity in CD4 + T Cells. Science 2016, 352, aad1210. [Google Scholar] [CrossRef] [PubMed]
  16. Huber-Lang, M.; Sarma, J.V.; Zetoune, F.S.; Rittirsch, D.; Neff, T.A.; McGuire, S.R.; Lambris, J.D.; Warner, R.L.; Flierl, M.A.; Hoesel, L.M.; et al. Generation of C5a in the Absence of C3: A New Complement Activation Pathway. Nat. Med. 2006, 12, 682–687. [Google Scholar] [CrossRef] [PubMed]
  17. Vandendriessche, S.; Cambier, S.; Proost, P.; Marques, P.E. Complement Receptors and Their Role in Leukocyte Recruitment and Phagocytosis. Front. Cell Dev. Biol. 2021, 9, 624025. [Google Scholar] [CrossRef]
  18. Serna, M.; Giles, J.L.; Morgan, B.P.; Bubeck, D. Structural Basis of Complement Membrane Attack Complex Formation. Nat. Commun. 2016, 7, 10587. [Google Scholar] [CrossRef]
  19. Mastellos, D.C.; Hajishengallis, G.; Lambris, J.D. A Guide to Complement Biology, Pathology and Therapeutic Opportunity. Nat. Rev. Immunol. 2023, 24, 118–141. [Google Scholar] [CrossRef]
  20. Garred, P.; Genster, N.; Pilely, K.; Bayarri-Olmos, R.; Rosbjerg, A.; Ma, Y.J.; Skjoedt, M. A Journey through the Lectin Pathway of Complement—MBL and Beyond. Immunol. Rev. 2016, 274, 74–97. [Google Scholar] [CrossRef]
  21. De Boer, E.C.; Thielen, A.J.; Langereis, J.D.; Kamp, A.; Brouwer, M.C.; Oskam, N.; Jongsma, M.L.; Baral, A.J.; Spaapen, R.M.; Zeerleder, S.; et al. The Contribution of the Alternative Pathway in Complement Activation on Cell Surfaces Depends on the Strength of Classical Pathway Initiation. Clin. Trans. Immunol. 2023, 12, e1436. [Google Scholar] [CrossRef]
  22. Lachmann, P.J.; Lay, E.; Seilly, D.J. Experimental Confirmation of the C3 Tickover Hypothesis by Studies with an Ab (S77) That Inhibits Tickover in Whole Serum. FASEB J. 2018, 32, 123–129. [Google Scholar] [CrossRef]
  23. Zipfel, P.F.; Skerka, C. Complement Regulators and Inhibitory Proteins. Nat. Rev. Immunol. 2009, 9, 729–740. [Google Scholar] [CrossRef] [PubMed]
  24. Lucientes-Continente, L.; Márquez-Tirado, B.; Goicoechea De Jorge, E. The Factor H Protein Family: The Switchers of the Complement Alternative Pathway. Immunol. Rev. 2023, 313, 25–45. [Google Scholar] [CrossRef]
  25. Servais, A.; Noël, L.-H.; Roumenina, L.T.; Le Quintrec, M.; Ngo, S.; Dragon-Durey, M.-A.; Macher, M.-A.; Zuber, J.; Karras, A.; Provot, F.; et al. Acquired and Genetic Complement Abnormalities Play a Critical Role in Dense Deposit Disease and Other C3 Glomerulopathies. Kidney Int. 2012, 82, 454–464. [Google Scholar] [CrossRef]
  26. Martín Merinero, H.; Zhang, Y.; Arjona, E.; Del Angel, G.; Goodfellow, R.; Gomez-Rubio, E.; Ji, R.-R.; Michelena, M.; Smith, R.J.H.; Rodríguez De Córdoba, S. Functional Characterization of 105 Factor H Variants Associated with aHUS: Lessons for Variant Classification. Blood 2021, 138, 2185–2201. [Google Scholar] [CrossRef]
  27. Blom, A.M. A Cluster of Positively Charged Amino Acids in the Alpha-Chain of C4b-Binding Protein (C4BP) Is Pivotal for the Regulation of the Complement System and the Interaction with Bacteria. Scand. J. Clin. Lab. Investig. Suppl. 2000, 233, 37–49. [Google Scholar]
  28. Degn, S.E.; Hansen, A.G.; Steffensen, R.; Jacobsen, C.; Jensenius, J.C.; Thiel, S. MAp44, a Human Protein Associated with Pattern Recognition Molecules of the Complement System and Regulating the Lectin Pathway of Complement Activation. J. Immunol. 2009, 183, 7371–7378. [Google Scholar] [CrossRef]
  29. Post, T.W.; Liszewski, M.K.; Adams, E.M.; Tedja, I.; Miller, E.A.; Atkinson, J.P. Membrane Cofactor Protein of the Complement System: Alternative Splicing of Serine/Threonine/Proline-Rich Exons and Cytoplasmic Tails Produces Multiple Isoforms That Correlate with Protein Phenotype. J. Exp. Med. 1991, 174, 93–102. [Google Scholar] [CrossRef]
  30. Schaefer, F.; Ardissino, G.; Ariceta, G.; Fakhouri, F.; Scully, M.; Isbel, N.; Lommelé, Å.; Kupelian, V.; Gasteyger, C.; Greenbaum, L.A.; et al. Clinical and Genetic Predictors of Atypical Hemolytic Uremic Syndrome Phenotype and Outcome. Kidney Int. 2018, 94, 408–418. [Google Scholar] [CrossRef]
  31. Angeletti, A.; Cantarelli, C.; Petrosyan, A.; Andrighetto, S.; Budge, K.; D’Agati, V.D.; Hartzell, S.; Malvi, D.; Donadei, C.; Thurman, J.M.; et al. Loss of Decay-Accelerating Factor Triggers Podocyte Injury and Glomerulosclerosis. J. Exp. Med. 2020, 217, e20191699. [Google Scholar] [CrossRef]
  32. Lake, B.B.; Menon, R.; Winfree, S.; Hu, Q.; Melo Ferreira, R.; Kalhor, K.; Barwinska, D.; Otto, E.A.; Ferkowicz, M.; Diep, D.; et al. An Atlas of Healthy and Injured Cell States and Niches in the Human Kidney. Nature 2023, 619, 585–594. [Google Scholar] [CrossRef]
  33. Goodship, T.H.J.; Cook, H.T.; Fakhouri, F.; Fervenza, F.C.; Frémeaux-Bacchi, V.; Kavanagh, D.; Nester, C.M.; Noris, M.; Pickering, M.C.; Rodríguez de Córdoba, S.; et al. Atypical Hemolytic Uremic Syndrome and C3 Glomerulopathy: Conclusions from a “Kidney Disease: Improving Global Outcomes” (KDIGO) Controversies Conference. Kidney Int. 2017, 91, 539–551. [Google Scholar] [CrossRef] [PubMed]
  34. Kawachi, H.; Fukusumi, Y. New Insight into Podocyte Slit Diaphragm, a Therapeutic Target of Proteinuria. Clin. Exp. Nephrol. 2020, 24, 193–204. [Google Scholar] [CrossRef] [PubMed]
  35. Schell, C.; Huber, T.B. The Evolving Complexity of the Podocyte Cytoskeleton. J. Am. Soc. Nephrol. JASN 2017, 28, 3166–3174. [Google Scholar] [CrossRef] [PubMed]
  36. Blaine, J.; Dylewski, J. Regulation of the Actin Cytoskeleton in Podocytes. Cells 2020, 9, 1700. [Google Scholar] [CrossRef] [PubMed]
  37. Butt, L.; Unnersjö-Jess, D.; Höhne, M.; Edwards, A.; Binz-Lotter, J.; Reilly, D.; Hahnfeldt, R.; Ziegler, V.; Fremter, K.; Rinschen, M.M.; et al. A Molecular Mechanism Explaining Albuminuria in Kidney Disease. Nat. Metab. 2020, 2, 461–474. [Google Scholar] [CrossRef] [PubMed]
  38. Banas, M.C.; Banas, B.; Hudkins, K.L.; Wietecha, T.A.; Iyoda, M.; Bock, E.; Hauser, P.; Pippin, J.W.; Shankland, S.J.; Smith, K.D.; et al. TLR4 Links Podocytes with the Innate Immune System to Mediate Glomerular Injury. J. Am. Soc. Nephrol. 2008, 19, 704–713. [Google Scholar] [CrossRef] [PubMed]
  39. Srivastava, T.; Sharma, M.; Yew, K.-H.; Sharma, R.; Duncan, R.S.; Saleem, M.A.; McCarthy, E.T.; Kats, A.; Cudmore, P.A.; Alon, U.S.; et al. LPS and PAN-Induced Podocyte Injury in an in Vitro Model of Minimal Change Disease: Changes in TLR Profile. J. Cell Commun. Signal. 2013, 7, 49–60. [Google Scholar] [CrossRef]
  40. Wright, R.D.; Beresford, M.W. Podocytes Contribute, and Respond, to the Inflammatory Environment in Lupus Nephritis. Am. J. Physiol. Ren. Physiol. 2018, 315, F1683–F1694. [Google Scholar] [CrossRef]
  41. Coers, W.; Brouwer, L.; Vos, J.T.W.M.; Chand, A.; Huitema, S.; Heeringa, P.; Kallenberg, C.G.M.; Weening, J.J. Podocyte Expression of MHC Class I and II and Intercellular Adhesion Molecule-1 (ICAM-1) in Experimental Pauci-Immune Crescentic Glomerulonephritis. Clin. Exp. Immunol. 2008, 98, 279–286. [Google Scholar] [CrossRef]
  42. Dylewski, J.; Dobrinskikh, E.; Lewis, L.; Tonsawan, P.; Miyazaki, M.; Jat, P.S.; Blaine, J. Differential Trafficking of Albumin and IgG Facilitated by the Neonatal Fc Receptor in Podocytes in Vitro and in Vivo. PLoS ONE 2019, 14, e0209732. [Google Scholar] [CrossRef]
  43. Nagata, M. Podocyte Injury and Its Consequences. Kidney Int. 2016, 89, 1221–1230. [Google Scholar] [CrossRef]
  44. Takano, T.; Elimam, H.; Cybulsky, A.V. Complement-Mediated Cellular Injury. Semin. Nephrol. 2013, 33, 586–601. [Google Scholar] [CrossRef] [PubMed]
  45. Cybulsky, A.V. The Intersecting Roles of Endoplasmic Reticulum Stress, Ubiquitin–Proteasome System, and Autophagy in the Pathogenesis of Proteinuric Kidney Disease. Kidney Int. 2013, 84, 25–33. [Google Scholar] [CrossRef]
  46. Greka, A.; Mundel, P. Balancing Calcium Signals through TRPC5 and TRPC6 in Podocytes. J. Am. Soc. Nephrol. 2011, 22, 1969–1980. [Google Scholar] [CrossRef] [PubMed]
  47. Lv, Q.; Yang, F.; Chen, K.; Zhang, Y. Autophagy Protects Podocytes from Sublytic Complement Induced Injury. Exp. Cell Res. 2016, 341, 132–138. [Google Scholar] [CrossRef] [PubMed]
  48. Liu, W.J.; Li, Z.; Chen, X.; Zhao, X.; Zhong, Z.; Yang, C.; Wu, H.; An, N.; Li, W.; Liu, H. Blockage of the Lysosome-Dependent Autophagic Pathway Contributes to Complement Membrane Attack Complex-Induced Podocyte Injury in Idiopathic Membranous Nephropathy. Sci. Rep. 2017, 7, 8643. [Google Scholar] [CrossRef] [PubMed]
  49. Abe, K.; Miyazaki, M.; Koji, T.; Furusu, A.; Nakamura-Kurashige, T.; Nishino, T.; Ozono, Y.; Harada, T.; Sakai, H.; Kohno, S. Enhanced Expression of Complement C5a Receptor mRNA in Human Diseased Kidney Assessed by in Situ Hybridization. Kidney Int. 2001, 60, 137–146. [Google Scholar] [CrossRef]
  50. Keir, L.S.; Firth, R.; Aponik, L.; Feitelberg, D.; Sakimoto, S.; Aguilar, E.; Welsh, G.I.; Richards, A.; Usui, Y.; Satchell, S.C.; et al. VEGF Regulates Local Inhibitory Complement Proteins in the Eye and Kidney. J. Clin. Investig. 2016, 127, 199–214. [Google Scholar] [CrossRef]
  51. Zheng, J.; Jiang, Z.; Chen, D.; Wang, S.; Zhao, W.; Li, L. Pathological Significance of Urinary Complement Activation in Diabetic Nephropathy: A Full View from the Development of the Disease. J. Diabetes Investig. 2019, 10, 738–744. [Google Scholar] [CrossRef]
  52. Wörn, M.; Bohnert, B.N.; Alenazi, F.; Boldt, K.; Klose, F.; Junger, K.; Ueffing, M.; Birkenfeld, A.L.; Kalbacher, H.; Artunc, F. Proteasuria in Nephrotic Syndrome–Quantification and Proteomic Profiling. J. Proteom. 2021, 230, 103981. [Google Scholar] [CrossRef]
  53. Medica, D.; Franzin, R.; Stasi, A.; Castellano, G.; Migliori, M.; Panichi, V.; Figliolini, F.; Gesualdo, L.; Camussi, G.; Cantaluppi, V. Extracellular Vesicles Derived from Endothelial Progenitor Cells Protect Human Glomerular Endothelial Cells and Podocytes from Complement- and Cytokine-Mediated Injury. Cells 2021, 10, 1675. [Google Scholar] [CrossRef]
  54. Maisonneuve, P.; Agodoa, L.; Gellert, R.; Stewart, J.H.; Buccianti, G.; Lowenfels, A.B.; Wolfe, R.A.; Jones, E.; Disney, A.P.S.; Briggs, D.; et al. Distribution of Primary Renal Diseases Leading to End-Stage Renal Failure in the United States, Europe, and Australia/New Zealand: Results from an International Comparative Study. Am. J. Kidney Dis. 2000, 35, 157–165. [Google Scholar] [CrossRef]
  55. Beck, L.H.; Bonegio, R.G.B.; Lambeau, G.; Beck, D.M.; Powell, D.W.; Cummins, T.D.; Klein, J.B.; Salant, D.J. M-Type Phospholipase A 2 Receptor as Target Antigen in Idiopathic Membranous Nephropathy. N. Engl. J. Med. 2009, 361, 11–21. [Google Scholar] [CrossRef] [PubMed]
  56. Tomas, N.M.; Beck, L.H.; Meyer-Schwesinger, C.; Seitz-Polski, B.; Ma, H.; Zahner, G.; Dolla, G.; Hoxha, E.; Helmchen, U.; Dabert-Gay, A.-S.; et al. Thrombospondin Type-1 Domain-Containing 7A in Idiopathic Membranous Nephropathy. N. Engl. J. Med. 2014, 371, 2277–2287. [Google Scholar] [CrossRef] [PubMed]
  57. Vidarsson, G.; Dekkers, G.; Rispens, T. IgG Subclasses and Allotypes: From Structure to Effector Functions. Front. Immunol. 2014, 5, 520. [Google Scholar] [CrossRef] [PubMed]
  58. Wiech, T.; Stahl, R.A.K.; Hoxha, E. Diagnostic Role of Renal Biopsy in PLA2R1-Antibody-Positive Patients with Nephrotic Syndrome. Mod. Pathol. 2019, 32, 1320–1328. [Google Scholar] [CrossRef] [PubMed]
  59. Hayashi, N.; Okada, K.; Matsui, Y.; Fujimoto, K.; Adachi, H.; Yamaya, H.; Matsushita, M.; Yokoyama, H. Glomerular Mannose-Binding Lectin Deposition in Intrinsic Antigen-Related Membranous Nephropathy. Nephrol. Dial. Transplant. 2018, 33, 832–840. [Google Scholar] [CrossRef] [PubMed]
  60. Hoxha, E.; Harendza, S.; Pinnschmidt, H.; Panzer, U.; Stahl, R.A.K. PLA2R Antibody Levels and Clinical Outcome in Patients with Membranous Nephropathy and Non-Nephrotic Range Proteinuria under Treatment with Inhibitors of the Renin-Angiotensin System. PLoS ONE 2014, 9, e110681. [Google Scholar] [CrossRef] [PubMed]
  61. Bally, S.; Debiec, H.; Ponard, D.; Dijoud, F.; Rendu, J.; Fauré, J.; Ronco, P.; Dumestre-Perard, C. Phospholipase A2 Receptor–Related Membranous Nephropathy and Mannan-Binding Lectin Deficiency. J. Am. Soc. Nephrol. JASN 2016, 27, 3539–3544. [Google Scholar] [CrossRef]
  62. Seifert, L.; Zahner, G.; Meyer-Schwesinger, C.; Hickstein, N.; Dehde, S.; Wulf, S.; Köllner, S.M.S.; Lucas, R.; Kylies, D.; Froembling, S.; et al. The Classical Pathway Triggers Pathogenic Complement Activation in Membranous Nephropathy. Nat. Commun. 2023, 14, 473. [Google Scholar] [CrossRef]
  63. Manral, P.; Caza, T.N.; Storey, A.J.; Beck, L.H.; Borza, D.-B. The Alternative Pathway Is Necessary and Sufficient for Complement Activation by Anti-THSD7A Autoantibodies, Which Are Predominantly IgG4 in Membranous Nephropathy. Front. Immunol. 2022, 13, 952235. [Google Scholar] [CrossRef]
  64. Sethi, S.; Fervenza, F.C. Membranoproliferative Glomerulonephritis—A New Look at an Old Entity. N. Engl. J. Med. 2012, 366, 1119–1131. [Google Scholar] [CrossRef]
  65. Sethi, S.; Fervenza, F.C. Membranoproliferative Glomerulonephritis: Pathogenetic Heterogeneity and Proposal for a New Classification. Semin. Nephrol. 2011, 31, 341–348. [Google Scholar] [CrossRef]
  66. Smith, R.J.H.; Appel, G.B.; Blom, A.M.; Cook, H.T.; D’Agati, V.D.; Fakhouri, F.; Fremeaux-Bacchi, V.; Józsi, M.; Kavanagh, D.; Lambris, J.D.; et al. C3 Glomerulopathy—Understanding a Rare Complement-Driven Renal Disease. Nat. Rev. Nephrol. 2019, 15, 129–143. [Google Scholar] [CrossRef]
  67. Pickering, M.C.; D’Agati, V.D.; Nester, C.M.; Smith, R.J.; Haas, M.; Appel, G.B.; Alpers, C.E.; Bajema, I.M.; Bedrosian, C.; Braun, M.; et al. C3 Glomerulopathy: Consensus Report. Kidney Int. 2013, 84, 1079–1089. [Google Scholar] [CrossRef]
  68. Zipfel, P.F.; Wiech, T.; Gröne, H.-J.; Skerka, C. Complement Catalyzing Glomerular Diseases. Cell Tissue Res. 2021, 385, 355–370. [Google Scholar] [CrossRef] [PubMed]
  69. Zipfel, P.F.; Wiech, T.; Stea, E.D.; Skerka, C. CFHR Gene Variations Provide Insights in the Pathogenesis of the Kidney Diseases Atypical Hemolytic Uremic Syndrome and C3 Glomerulopathy. J. Am. Soc. Nephrol. JASN 2020, 31, 241–256. [Google Scholar] [CrossRef] [PubMed]
  70. Iatropoulos, P.; Noris, M.; Mele, C.; Piras, R.; Valoti, E.; Bresin, E.; Curreri, M.; Mondo, E.; Zito, A.; Gamba, S.; et al. Complement Gene Variants Determine the Risk of Immunoglobulin-Associated MPGN and C3 Glomerulopathy and Predict Long-Term Renal Outcome. Mol. Immunol. 2016, 71, 131–142. [Google Scholar] [CrossRef] [PubMed]
  71. Donadelli, R.; Pulieri, P.; Piras, R.; Iatropoulos, P.; Valoti, E.; Benigni, A.; Remuzzi, G.; Noris, M. Unraveling the Molecular Mechanisms Underlying Complement Dysregulation by Nephritic Factors in C3G and IC-MPGN. Front. Immunol. 2018, 9, 2329. [Google Scholar] [CrossRef]
  72. Ortega, L.M.; Schultz, D.R.; Lenz, O.; Pardo, V.; Contreras, G.N. Review: Lupus Nephritis: Pathologic Features, Epidemiology and a Guide to Therapeutic Decisions. Lupus 2010, 19, 557–574. [Google Scholar] [CrossRef]
  73. Markowitz, G.S.; D’Agati, V.D. Classification of Lupus Nephritis. Curr. Opin. Nephrol. Hypertens. 2009, 18, 220–225. [Google Scholar] [CrossRef]
  74. Birmingham, D.; Irshaid, F.; Nagaraja, H.; Zou, X.; Tsao, B.; Wu, H.; Yu, C.; Hebert, L.; Rovin, B. The Complex Nature of Serum C3 and C4 as Biomarkers of Lupus Renal Flare. Lupus 2010, 19, 1272–1280. [Google Scholar] [CrossRef]
  75. Kim, A.H.J.; Strand, V.; Sen, D.P.; Fu, Q.; Mathis, N.L.; Schmidt, M.J.; Bruchas, R.R.; Staten, N.R.; Olson, P.K.; Stiening, C.M.; et al. Association of Blood Concentrations of Complement Split Product iC 3b and Serum C3 With Systemic Lupus Erythematosus Disease Activity. Arthritis Rheumatol. 2019, 71, 420–430. [Google Scholar] [CrossRef]
  76. Martin, M.; Trattner, R.; Nilsson, S.C.; Björk, A.; Zickert, A.; Blom, A.M.; Gunnarsson, I. Plasma C4d Correlates with C4d Deposition in Kidneys and With Treatment Response in Lupus Nephritis Patients. Front. Immunol. 2020, 11, 582737. [Google Scholar] [CrossRef]
  77. Sethi, S.; Palma, L.M.P.; Theis, J.D.; Fervenza, F.C. Proteomic Analysis of Complement Proteins in Glomerular Diseases. Kidney Int. Rep. 2023, 8, 827–836. [Google Scholar] [CrossRef]
  78. Drachenberg, C.B.; Papadimitriou, J.C.; Chandra, P.; Haririan, A.; Mendley, S.; Weir, M.R.; Rubin, M.F. Epidemiology and Pathophysiology of Glomerular C4d Staining in Native Kidney Biopsies. Kidney Int. Rep. 2019, 4, 1555–1567. [Google Scholar] [CrossRef]
  79. Yu, F.; Haas, M.; Glassock, R.; Zhao, M.-H. Redefining Lupus Nephritis: Clinical Implications of Pathophysiologic Subtypes. Nat. Rev. Nephrol. 2017, 13, 483–495. [Google Scholar] [CrossRef]
  80. Gaya Da Costa, M.; Poppelaars, F.; Berger, S.P.; Daha, M.R.; Seelen, M.A. The Lectin Pathway in Renal Disease: Old Concept and New Insights. Nephrol. Dial. Transplant. 2018, 33, 2073–2079. [Google Scholar] [CrossRef]
  81. Asanuma, Y.; Nozawa, K.; Matsushita, M.; Kusaoi, M.; Abe, Y.; Yamaji, K.; Tamura, N. Critical Role of Lectin Pathway Mediated by MBL-Associated Serine Proteases in Complement Activation for the Pathogenesis in Systemic Lupus Erythematosus. Heliyon 2023, 9, e19072. [Google Scholar] [CrossRef]
  82. D’Agati, V.D.; Kaskel, F.J.; Falk, R.J. Focal Segmental Glomerulosclerosis. N. Engl. J. Med. 2011, 365, 2398–2411. [Google Scholar] [CrossRef]
  83. Van De Lest, N.A.; Zandbergen, M.; Wolterbeek, R.; Kreutz, R.; Trouw, L.A.; Dorresteijn, E.M.; Bruijn, J.A.; Bajema, I.M.; Scharpfenecker, M.; Chua, J.S. Glomerular C4d Deposition Can Precede the Development of Focal Segmental Glomerulosclerosis. Kidney Int. 2019, 96, 738–749. [Google Scholar] [CrossRef]
  84. Huang, J.; Cui, Z.; Gu, Q.; Zhang, Y.; Qu, Z.; Wang, X.; Wang, F.; Cheng, X.; Meng, L.; Liu, G.; et al. Complement Activation Profile of Patients with Primary Focal Segmental Glomerulosclerosis. PLoS ONE 2020, 15, e0234934. [Google Scholar] [CrossRef]
  85. Thurman, J.M.; Wong, M.; Renner, B.; Frazer-Abel, A.; Giclas, P.C.; Joy, M.S.; Jalal, D.; Radeva, M.K.; Gassman, J.; Gipson, D.S.; et al. Complement Activation in Patients with Focal Segmental Glomerulosclerosis. PLoS ONE 2015, 10, e0136558. [Google Scholar] [CrossRef]
  86. Liu, J.; Xie, J.; Zhang, X.; Tong, J.; Hao, X.; Ren, H.; Wang, W.; Chen, N. Serum C3 and Renal Outcome in Patients with Primary Focal Segmental Glomerulosclerosis. Sci. Rep. 2017, 7, 4095. [Google Scholar] [CrossRef]
  87. Tato, A.M.; Carrera, N.; García-Murias, M.; Shabaka, A.; Ávila, A.; Mora Mora, M.T.; Rabasco, C.; Soto, K.; de la Prada Alvarez, F.J.; Fernández-Lorente, L.; et al. Genetic Testing in Focal Segmental Glomerulosclerosis: In Whom and When? Clin. Kidney J. 2023, 16, 2011–2022. [Google Scholar] [CrossRef]
  88. Trachtman, H.; Laskowski, J.; Lee, C.; Renner, B.; Feemster, A.; Parikh, S.; Panzer, S.E.; Zhong, W.; Cravedi, P.; Cantarelli, C.; et al. Natural Antibody and Complement Activation Characterize Patients with Idiopathic Nephrotic Syndrome. Am. J. Physiol. Ren. Physiol. 2021, 321, F505–F516. [Google Scholar] [CrossRef]
  89. Koopman, J.J.E.; Van Essen, M.F.; Rennke, H.G.; De Vries, A.P.J.; Van Kooten, C. Deposition of the Membrane Attack Complex in Healthy and Diseased Human Kidneys. Front. Immunol. 2021, 11, 599974. [Google Scholar] [CrossRef]
  90. Chugh, S.S.; Clement, L.C. “Idiopathic” Minimal Change Nephrotic Syndrome: A Podocyte Mystery Nears the End. Am. J. Physiol. Ren. Physiol. 2023, 325, F685–F694. [Google Scholar] [CrossRef]
  91. Muruve, D.A.; Debiec, H.; Dillon, S.T.; Gu, X.; Plaisier, E.; Can, H.; Otu, H.H.; Libermann, T.A.; Ronco, P. Serum Protein Signatures Using Aptamer-Based Proteomics for Minimal Change Disease and Membranous Nephropathy. Kidney Int. Rep. 2022, 7, 1539–1556. [Google Scholar] [CrossRef]
  92. Wendt, R.; Siwy, J.; He, T.; Latosinska, A.; Wiech, T.; Zipfel, P.F.; Tserga, A.; Vlahou, A.; Rupprecht, H.; Catanese, L.; et al. Molecular Mapping of Urinary Complement Peptides in Kidney Diseases. Proteomes 2021, 9, 49. [Google Scholar] [CrossRef]
  93. Legendre, C.M.; Licht, C.; Muus, P.; Greenbaum, L.A.; Babu, S.; Bedrosian, C.; Bingham, C.; Cohen, D.J.; Delmas, Y.; Douglas, K.; et al. Terminal Complement Inhibitor Eculizumab in Atypical Hemolytic–Uremic Syndrome. N. Engl. J. Med. 2013, 368, 2169–2181. [Google Scholar] [CrossRef]
  94. Ruggenenti, P.; Daina, E.; Gennarini, A.; Carrara, C.; Gamba, S.; Noris, M.; Rubis, N.; Peraro, F.; Gaspari, F.; Pasini, A.; et al. C5 Convertase Blockade in Membranoproliferative Glomerulonephritis: A Single-Arm Clinical Trial. Am. J. Kidney Dis. 2019, 74, 224–238. [Google Scholar] [CrossRef]
  95. Sethi, S.; Fervenza, F.C.; Zhang, Y.; Zand, L.; Meyer, N.C.; Borsa, N.; Nasr, S.H.; Smith, R.J.H. Atypical Postinfectious Glomerulonephritis Is Associated with Abnormalities in the Alternative Pathway of Complement. Kidney Int. 2013, 83, 293–299. [Google Scholar] [CrossRef]
  96. Wong, E.; Nester, C.; Cavero, T.; Karras, A.; Le Quintrec, M.; Lightstone, L.; Eisenberger, U.; Soler, M.J.; Kavanagh, D.; Daina, E.; et al. Efficacy and Safety of Iptacopan in Patients with C3 Glomerulopathy. Kidney Int. Rep. 2023, 8, 2754–2764. [Google Scholar] [CrossRef]
  97. Tarragon Estebanez, B.; Bomback, A.S. C3 Glomerulopathy: Novel Treatment Paradigms. Kidney Int. Rep. 2023, 93, 977–985. [Google Scholar] [CrossRef]
  98. Fervenza, F.C.; Appel, G.B.; Barbour, S.J.; Rovin, B.H.; Lafayette, R.A.; Aslam, N.; Jefferson, J.A.; Gipson, P.E.; Rizk, D.V.; Sedor, J.R.; et al. Rituximab or Cyclosporine in the Treatment of Membranous Nephropathy. N. Engl. J. Med. 2019, 381, 36–46. [Google Scholar] [CrossRef]
  99. Kistler, A.D.; Salant, D.J. Complement Activation and Effector Pathways in Membranous Nephropathy. Kidney Int. 2023, 5, 572–574. [Google Scholar] [CrossRef]
  100. Pickering, M.C.; Botto, M.; Taylor, P.R.; Lachmann, P.J.; Walport, M.J. Systemic Lupus Erythematosus, Complement Deficiency, and Apoptosis. In Advances in Immunology; Elsevier: Amsterdam, The Netherlands, 2001; Volume 76, pp. 227–324. ISBN 978-0-12-022476-0. [Google Scholar]
  101. McGrogan, A.; Franssen, C.F.M.; de Vries, C.S. The Incidence of Primary Glomerulonephritis Worldwide: A Systematic Review of the Literature. Nephrol. Dial. Transplant. 2011, 26, 414–430. [Google Scholar] [CrossRef]
  102. Suzuki, H.; Kiryluk, K.; Novak, J.; Moldoveanu, Z.; Herr, A.B.; Renfrow, M.B.; Wyatt, R.J.; Scolari, F.; Mestecky, J.; Gharavi, A.G.; et al. The Pathophysiology of IgA Nephropathy. J. Am. Soc. Nephrol. 2011, 22, 1795–1803. [Google Scholar] [CrossRef]
  103. Caravaca-Fontán, F.; Gutiérrez, E.; Sevillano, Á.M.; Praga, M. Targeting Complement in IgA Nephropathy. Clin. Kidney J. 2023, 16, ii28–ii39. [Google Scholar] [CrossRef]
  104. Zhang, H.; Rizk, D.V.; Perkovic, V.; Maes, B.; Kashihara, N.; Rovin, B.; Trimarchi, H.; Sprangers, B.; Meier, M.; Kollins, D.; et al. Results of a Randomized Double-Blind Placebo-Controlled Phase 2 Study Propose Iptacopan as an Alternative Complement Pathway Inhibitor for IgA Nephropathy. Kidney Int. 2024, 105, 189–199. [Google Scholar] [CrossRef] [PubMed]
  105. Wang, S.; Broder, A.; Shao, D.; Kesarwani, V.; Boderman, B.; Aguilan, J.; Sidoli, S.; Suzuki, M.; Greally, J.M.; Saenger, Y.M.; et al. Urine Proteomics Link Complement Activation with Interstitial Fibrosis/Tubular Atrophy in Lupus Nephritis Patients. Semin. Arthritis Rheum. 2023, 63, 152263. [Google Scholar] [CrossRef] [PubMed]
  106. Schmidt, T.; Afonso, S.; Perie, L.; Heidenreich, K.; Wulf, S.; Krebs, C.F.; Zipfel, P.F.; Wiech, T. An Interdisciplinary Diagnostic Approach to Guide Therapy in C3 Glomerulopathy. Front. Immunol. 2022, 13, 826513. [Google Scholar] [CrossRef]
  107. Person, F.; Petschull, T.; Wulf, S.; Buescheck, F.; Biniaminov, S.; Fehrle, W.; Oh, J.; Skerka, C.; Zipfel, P.F.; Wiech, T. In Situ Visualization of C3/C5 Convertases to Differentiate Complement Activation. Kidney Int. Rep. 2020, 5, 927–930. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Activation (gray) and effects (pink) of the complement system through the three pathways (alternative, lectin, classical) with regulatory factors (yellow) and therapeutic complement inhibitors (light green).
Figure 1. Activation (gray) and effects (pink) of the complement system through the three pathways (alternative, lectin, classical) with regulatory factors (yellow) and therapeutic complement inhibitors (light green).
Biomedicines 12 00455 g001
Table 1. Overview of current studies on complement system inhibition in various renal diseases.
Table 1. Overview of current studies on complement system inhibition in various renal diseases.
DiseaseInhibitorPathwayPhaseNCT-Number
Primary membranous nephropathyIptacopanAlternative (factor B)IINCT04154787
PelecopanAlternative (factor D)IINCT05162066
NarsoplimabLectin (MASP2)IINCT02682407
PegcetacoplanCentral (C3)IINCT03453619
GefurulimabTerminal (C5)INCT05314231
C3GIptacopanAlternative (factor B)II, III, OLENCT03832114, NCT04817618, NCT03955445
NM8074Alternative (factor Bb)IbNCT05647811
PelecopanAlternative (factor D)IINCT05162066
DanicopanAlternative (factor D)IIa, IIbNCT03124368, NCT03369236, NCT03459443, NCT03723512
NarsoplimabLectin (MASP2)IINCT02682407
TP10Inhibition (scR1)IIaNCT02302755
PegcetacoplanCentral (C3)II, III, OLENCT04572854, NCT03453619, NCT05067127, NCT05809531
ARO-C3Central (C3)I/IINCT05083364
AMY-101Central (C3)INCT03316521
KP104Inhibition (CFH/C5)IINCT05517980
AvacopanInflammation (C5aR)IINCT03301467
IC-MPGNIptacopanAlternative (factor B)IIINCT05755386
DanicopanAlternative (factor D)IIa, IIbNCT03124368, NCT03459443, NCT03723512, NCT03369236
PegcetacoplanCentral (C3)II, III, OLENCT04572854, NCT05067127, NCT05809531
Lupus NephritisIptacopanAlternative (factor B)IINCT05268289
Vemircopan Alternative (factor D)IINCT05097989
NarsoplimabLectin (MASP2)IINCT02682407
ANX009 Classical (C1q)I NCT05780515
Pegcetacoplan Central (C3)IINCT03453619
Gefurulimab Terminal (C5)INCT05314231
RavulizumabTerminal (C5)IINCT04564339
IgA NephropathyIONIS-FB-LRxAlternative (factor B)II, IIINCT04014335, NCT05797610
IptacopanAlternative (factor B)II, IIINCT03373461, NCT04557462, NCT04578834
VemircopanAlternative (factor D)IINCT05097989
PelecopanAlternative (factor D)IINCT05162066
NarsoplimabLectin (MASP2)II, IIINCT02682407, NCT03608033
ARO-C3 Central (C3)INCT05083364
KP104Inhibition (CFH/C5)IINCT05517980
RavulizumabTerminal (C5)IINCT04564339
Avacopan Inflammation (C5aR)INCT06004947
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Nell, D.; Wolf, R.; Podgorny, P.M.; Kuschnereit, T.; Kuschnereit, R.; Dabers, T.; Stracke, S.; Schmidt, T. Complement Activation in Nephrotic Glomerular Diseases. Biomedicines 2024, 12, 455. https://doi.org/10.3390/biomedicines12020455

AMA Style

Nell D, Wolf R, Podgorny PM, Kuschnereit T, Kuschnereit R, Dabers T, Stracke S, Schmidt T. Complement Activation in Nephrotic Glomerular Diseases. Biomedicines. 2024; 12(2):455. https://doi.org/10.3390/biomedicines12020455

Chicago/Turabian Style

Nell, Dominik, Robert Wolf, Przemyslaw Marek Podgorny, Tobias Kuschnereit, Rieke Kuschnereit, Thomas Dabers, Sylvia Stracke, and Tilman Schmidt. 2024. "Complement Activation in Nephrotic Glomerular Diseases" Biomedicines 12, no. 2: 455. https://doi.org/10.3390/biomedicines12020455

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop