Next Article in Journal
P2X7 Receptor in Dendritic Cells and Macrophages: Implications in Antigen Presentation and T Lymphocyte Activation
Next Article in Special Issue
Prostaglandin Transporter and Dipeptidyl Peptidase-4 as New Pharmacological Targets in the Prevention of Acute Kidney Injury in Diabetes: An In Vitro Study
Previous Article in Journal
Importance of Non-Covalent Interactions in Yeast Cell Wall Molecular Organization
Previous Article in Special Issue
Beta Blockade Prevents Cardiac Morphological and Molecular Remodelling in Experimental Uremia
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Type I IFN in Glomerular Disease: Scarring beyond the STING

by
Alexis Paulina Jimenez-Uribe
,
Steve Mangos
and
Eunsil Hahm
*
Department of Internal Medicine, Division of Nephrology, Rush University Medical Center, Chicago, IL 60612, USA
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2024, 25(5), 2497; https://doi.org/10.3390/ijms25052497
Submission received: 31 December 2023 / Revised: 13 February 2024 / Accepted: 19 February 2024 / Published: 21 February 2024
(This article belongs to the Special Issue Molecular Pathology, Diagnostics and Therapeutics of Nephropathy 3.0)

Abstract

:
The field of nephrology has recently directed a considerable amount of attention towards the stimulator of interferon genes (STING) molecule since it appears to be a potent driver of chronic kidney disease (CKD). STING and its activator, the cyclic GMP-AMP synthase (cGAS), along with intracellular RIG-like receptors (RLRs) and toll-like receptors (TLRs), are potent inducers of type I interferon (IFN-I) expression. These cytokines have been long recognized as part of the mechanism used by the innate immune system to battle viral infections; however, their involvement in sterile inflammation remains unclear. Mounting evidence pointing to the involvement of the IFN-I pathway in sterile kidney inflammation provides potential insights into the complex interplay between the innate immune system and damage to the most sensitive segment of the nephron, the glomerulus. The STING pathway is often cited as one cause of renal disease not attributed to viral infections. Instead, this pathway can recognize and signal in response to host-derived nucleic acids, which are also recognized by RLRs and TLRs. It is still unclear, however, whether the development of renal diseases depends on subsequent IFN-I induction or other processes involved. This review aims to explore the main endogenous inducers of IFN-I in glomerular cells, to discuss what effects autocrine and paracrine signaling have on IFN-I induction, and to identify the pathways that are implicated in the development of glomerular damage.

1. Introduction

Over one hundred years ago, Paul Ehrlich introduced the term “horror autotoxicus” to the field of immunopathology. He used this term to refer to the process whereby the immune system directs its attention to attack the host rather than protect it from foreign harmful agents. This term finds increased relevance today and can be used to describe many types of hypersensitivity reactions, including autoimmune diseases (part of the type II and III hypersensitivity reactions) and disorders stemming from a hyperinflammatory response, such as cytokine storm syndrome and other autoinflammatory diseases that damage the kidney.
Recently, molecules typically associated with an antiviral immune response, cyclic GMP-AMP synthase (cGAS) and stimulator of interferon genes (STING), have been linked to the development of chronic kidney disease (CKD) in the absence of infection. Instead, cGAS/STING activation is through host-derived endogenous nucleic acids. Most relevant, STING blockade results in reduced kidney damage [1], especially at the level of the glomerulus [2,3]. Interestingly, although the expression of type I interferons (IFN-I) is one of the most expected outcomes after the activation of cGAS/STING and other intracellular pattern-recognition receptors (PRRs), it is still not clear if the renoprotective effects of cGAS/STING blockade rely on the abrogation of IFN-I production more so than other mechanisms.
The IFN-I pathway has been extensively studied as part of the innate immune response against viral infections. Signaling begins with the recognition of viral or endogenous nucleic acids by cGAS-like receptors (cGLRs), RIG-like receptors (RLRs), or toll-like receptors (TLRs), all members of the intracellular pattern-recognition receptor family. Once this recognition occurs, IFN-I expression is induced and acts in an autocrine or paracrine fashion. Ultimately, several genes are expressed that reduce transcription and translation rates and alter nucleic acid and lipid metabolism to block viral replication [4]. However, the function of this pathway in sterile inflammation is not yet fully understood. With the description more than a decade ago of interferonopathies, a group of diseases characterized by overactivation of the IFN-I pathway, much attention turned to focus on the relationship between IFN-I and autoinflammatory effects [5,6]. New light was shed on how endogenous stress, through self-nucleic acid recognition, drives sterile inflammation.
Glomerular disease (GD) represents a group of immune-mediated diseases characterized by inflammation and fibrosis within the glomerulus causing well-described histopathological lesions that encompass: (1) membranoproliferative glomerulonephritis (MPGN), (2) minimal change disease (MCD), (3) focal segmental glomerulosclerosis (FSGS), (4) membranous nephropathy (MN), (5) IgA nephropathy (IgAN), (6) crescentic glomerulonephritis (CGN), and (7) lupus nephritis (LN) [7]. Although the current classification is helpful for diagnosis, several and often different etiologies are involved in the development of each. Indeed, among the different etiologies, infections are responsible for only a fraction of GD cases. Most are caused by auto-antibodies against a broad spectrum of glomerular cell-specific molecules, genetic defects leading to autoinflammatory disorders, or as a consequence of other non-communicable diseases such as diabetes [8], making many GDs part of the aforementioned “horror autotoxicus”.
Among the different molecular mechanisms involved in the pathogenesis of GDs, there is accumulating evidence that the IFN-I pathway participates in the progression of glomerular damage. Much evidence stems from patient data derived from kidney biopsies, identifying the upregulation of gene products involved in the IFN-I pathway. These patient-derived data are summarized in Table 1.
The most compelling evidence suggesting the direct involvement of the IFN-I pathway in the development and progression of GD comes from patients diagnosed with multiple sclerosis, hepatitis C, and some types of cancer, all developing glomerular lesions after receiving recombinant IFN therapy [16,17,18,19]. Moreover, patients with inborn errors causing enhanced IFN-I production such as Aicardi–Goutières syndrome [20], STING-associated vasculopathy with onset in infancy (SAVI) [21,22], and systemic lupus erythematosus (SLE) [23] also present with glomerular lesions.
Results from experiments using animal models have provided additional support for the direct effect of IFN-I on the glomerulus. A murine model of nephritis, generated by injecting anti-glomerular basement membrane (GBM) antibodies, showed an increase in IFNα in renal tissue and a correlation with renal dysfunction demonstrated by increased levels of blood urea nitrogen (BUN) and proteinuria. Moreover, the exogenous overexpression of IFNα using an adenoviral vector increased proteinuria and led to more glomerular damage in treated animals [24]. In a similar approach, Migliorini et al. combined a model of adriamycin nephropathy with the administration of IFNα and IFNβ and showed enhanced kidney dysfunction through the perturbation of parietal epithelial cells (PEC) and podocyte loss [25]. In testing a novel model of murine SLE, Nacionales et al. showed that by blocking IFN-I activity by knocking out an IFN-I receptor (IFNAR), kidney dysfunction and glomerular injury could be ameliorated [26]. Taken together, results from these experiments suggest that the different causes of GD drive a diverse set of complex immunopathologies, including the stimulation of the IFN-I pathway, which, in turn, participates in further aggravating the disease.
The goal of this review is to (1) present a general overview of the IFN-pathway; (2) explore how the IFN-I pathway is induced, focusing on endogenous stress activators such as host-derived self-nucleic acids; and (3) outline how kidney-specific cells are affected by these cytokines.

2. IFN-I Induction in Sterile Inflammation

An early immune response to foreign pathogens, such as viruses, is the activation of PRRs, leading to the induction of IFN-I expression. This response can also be triggered by endogenous danger signals, mainly self-nucleic acids detected in the cytosol [27], but also by proteotoxic stress [28] and elevated cellular oxidative stress [29]. In physiological homeostatic conditions, mechanisms are in place to avoid aberrant PRR activation by self-nucleic acids. These include quick degradation using DNA and RNA nucleases, protective epigenetic and posttranscriptional modifications, and physically compartmentalizing the nucleus and mitochondria. However, nucleic acids can be damaged under certain stress-inducing conditions such as exposure to xenobiotics, metabolic stress, elevated reactive oxygen species, and the unfolded protein response. Nucleic acid damage can lead to their release and accumulation in the cytoplasm in the form of free cytosolic nucleic acids, micronuclei, chromatin fragments, or transposable elements [30,31], as illustrated in Figure 1.
In many cases of CKD, including several types of GD, patient biopsies show evidence of nucleic acid damage [32]. In a retrospective study using human biopsy samples, Ott et al. observed DNA fragmentation across a spectrum of different types of GD [33]. More specifically, in mice, it was shown that double-stranded DNA breaks in podocytes caused by the endonuclease I-Ppol correlated with proteinuria, glomerulosclerosis, and tubulointerstitial fibrosis [34]. This study also identified portions of the immune system as being responsible for the development of disease. Similarly, Dhillon et al. show that the increased levels of transposable elements (TE) and retrotransposons associated with DNA damage were detected in diseased samples from human patients and mouse models of renal fibrosis, suggesting a link with IFN-I pathway activation [35]. In support of these findings, De Cecco et al. showed an association between retrotransposable elements and IFN-I pathway signaling through the detection of cytoplasmic cDNA [36].
As previously mentioned, the first response in avoiding intracellular PRR activation is to degrade nucleic acids. Mechanisms to guard against immune activation caused by self-nucleic acid detection exist in the kidney. For example, case study data show a link between mutations in TREX1 and RNaseH2, key nucleases involved in cytoplasmic nucleic acid degradation, and thrombotic microangiopathy (TMA) or Aicardi–Goutières syndrome with kidney alterations [20,37], respectively.
If self-derived nucleic acids accumulate in the cytoplasm due to their release from the nucleus or a lack of degradation, there is a vast variety of PRRs able to recognize them, including cGLRs, RLRs, and TLRs [27]. Table 2 provides a general summary of the distinct types of intracellular PRRs and their cognate ligands.
Although different nucleic acid receptors activate different signal transduction pathways, most converge at the nexus of activating TANK-binding kinase 1 (TBK1) and, in turn, the phosphorylation of interferon regulatory factors (IRF), transcription factors that allow IFN-I expression. Figure 1 provides a graphical representation of the main molecular interactions that occur to initiate and propagate signaling through the IFN-I pathway. The remainder of this review will highlight certain aspects of these interactions as they relate to the available data concerning glomerular-specific diseases and other prominent kidney diseases.

3. cGAS/STING Activation in the Kidney

cGAS and its analogs, cGAS-like receptors (cGLR), are evolutionary conserved receptors in metazoans that detect cytosolic double-stranded (ds) DNA [38]. Once these receptors recognize their ligands, they have the ability to enzymatically convert triphosphate nucleotides into signaling linear or cyclic dinucleotides. For example, cGAS can generate 2′3′-cGAMP, a second messenger that binds and activates STING in the endoplasmic reticulum [39,40,41]. STING then oligomerizes and translocates to the Golgi, leading to the activation of TBK1 and IRF3, which in turn translocates to the nucleus and induces IFN-I expression, as shown in Figure 1.
Primarily due to its cellular localization and inability to discriminate between host and foreign dsDNA, the cGAS/STING system could be the first mechanism to respond to nuclear and mitochondrial (mt)-derived DNA present in cytoplasm. Interest in the cGAS/STING pathway has recently increased within the field of nephrology with the discovery that key glomerular cells express components of the pathway at the early stage of kidney dysfunction. Zang et al. show that podocyte injury can be triggered in a mouse model of diabetic kidney disease (DKD) through lipotoxicity-induced mitochondrial damage and mtDNA leakage leading to activation of the cGAS/STING pathway [3]. Mitrofanova et al. observed similar results in cultured human and mouse podocytes, as well as in mouse models of DKD and Alport syndrome that were exposed to a STING-specific agonist [2]. In an attempt to identify molecular pathways responsible for apolipoprotein 1 (APOL1)-induced kidney dysfunction, Wu et al. identified the expression of STING as an early event leading to podocyte cytotoxicity and cell death [42]. In all cases, pharmacologic inhibition or genetic deletion of the STING pathway reduced glomerular damage in these mouse models of disease. A more detailed exploration of the involvement of the cGAS/STING pathway in the progression to LN end-stage renal disease (LN-ESRD) using cultured human podocytes was recently reported. Davis et al. showed that if podocytes are treated with nucleosome-associated dsDNA fragments, a common marker found in the blood of Lupus patients, IFNγ-inducible protein 16 (IFI16) triggers the expression of both IFNβ and APOL1 and the activation of the cGAS/STING pathway [43]. More directly, this group showed that exposing podocytes to IFNβ promotes the expression of IFI16, suggesting a positive feedback loop between dsDNA recognition and cGAS/STING pathway activation. Additional evidence implicating the cGAS/STING pathway in lupus-mediated podocyte damage was provided by Li et al., who treated mouse podocytes with LN patient serum. It was shown that in these podocytes, the cGAS/STING pathway is activated, IFN-I expression is stimulated, and endoplasmic reticulum stress is induced, leading to apoptosis [44]. Podocytes from diabetic mice also show increased levels of cGAS and STING as well as activation of TBK1. Palmitic acid, a fatty acid associated with DKD, can elicit mitochondrial damage and leakage of mtDNA into the cytoplasm to activate the STING pathway. As a result, several inflammatory markers are induced, and levels of apoptosis increase. Interestingly, IFN-I expression does not seem to be elevated in this mouse model [3].
The bulk of research linking the activation of the cGAS/STING pathway to glomerular disease has focused on the podocyte; however, some evidence exists showing that other cells within the glomerulus are affected by this pathway and could contribute to renal dysfunction. There are several reports that implicate the STING pathway, either directly or indirectly, in glomerular endothelial cell (GEC) damage. In diabetic mice, Qi et al. present evidence that mtDNA contributes to glomerular damage indirectly through GECs by inducing podocyte loss, leading to proteinuria [45], while Caselena et al. show that GECs display severe mitochondrial damage when treated with high-glucose media [46]. Mitochondrial dysfunction is also seen in GECs from a transgenic mouse model of FSGS. This study showed crosstalk between glomerular cells, where the activation of transforming growth factor β receptor 1 (TGFβR1) in podocytes is associated with an increase in mitochondrial stress and release of mtDNA in GECs, leading to a subsequent loss of podocytes [47]. More indirect evidence comes from studies using mice expressing an APOL1 renal risk variant (RSK). The endothelial cells of these mice display high levels of mitophagy with mtDNA leakage into the cytoplasm and IFN-I pathway activation [48]. Although mostly indirect, there is evidence that the cGAS/STING pathway is activated in GECs and that this activation leads to glomerular damage. It is interesting to note that in 2009, Hagele et al. reported that GECs could be activated by dsDNA in a TLR-independent manner, showing increased IFN-I pathway expression [49]; this report was published prior to the cGAS/STING pathway being discovered [50,51].
Activation of the cGAS/STING pathway in mesangial cells (MCs) is still somewhat unexplored. There is some evidence, however, that mitochondrial damage leading to leakage of mtDNA occurs in cultured human mesangial cells (HMCs). This group showed that culturing HMCs with galactose deficient IgA from IgAN patients leads to mitochondrial damage due to the reduction in peroxisome proliferator-activated receptor alpha (PPARα) expression [52]. Curiously, MCs grown in hyperglycemic conditions have also shown markers of mitochondrial damage, suggesting that leakage of mtDNA and the subsequent activation of the cGAS/STING pathway could also be operating in some instances of DN.
It should be included here that there are other cell types not typically associated with the glomerulus that could also contribute to glomerular injury through the STING pathway. As an example, plasmacytoid dendritic cells (pDCs) are a special type of immune cell characterized, in part, by their high expression of IFN-I. Indeed, Deng et al. showed that pDCs rapidly infiltrate the kidney after AKI and produce IFNα, leading to kidney damage [53]. These cells have also been implicated in LN [54,55], where it has been reported that STING activation promotes the maturation of pDCs with their subsequent participation in glomerular injury [56]. However, it must be emphasized that it is still not clear if pDCs’ contribution to renal disease is through their involvement systemically or at the level of the kidney.

4. RLR Activation in the Kidney

Melanoma differentiation-associated gene 5 (MDA5) and retinoic acid-inducible gene I (RIG-I) are two RLR family members that reside in the cytoplasm, act as receptors for dsRNA, and, in a manner similar to cGAS, activate the mitochondrial antiviral-signaling protein (MAVS), TBK1, and IRF3 to elicit IFN-I expression. Although cytoplasmic endogenous dsRNA is a less common indicator of cellular stress as compared to cytoplasmic dsDNA, sources include retrotransposons, mt-dsRNA, and other secondary RNA structures that arise from epigenetic alterations, mitochondrial damage, or defects in RNA processing [57,58,59,60,61,62]. Murine models of autoimmune disease have been instrumental in showing that mt-dsRNA plays a relevant role in driving sterile inflammation through IFN-I pathway activation [63,64].
The activation of RLRs has been documented in a variety of kidney diseases, both in human patient samples and animal models of AKI and several different types of GD. A number of studies have shown that tubule-specific damage is linked to RLR activation. For example, Zhu et al. showed that renal tubule injury in mice caused by ischemia-reperfusion injury (IRI) or unilateral ureteral obstruction was closely tied to an accumulation of mt-dsRNA [65]. Similar results were reported by Doke et al., also using the murine IRI method [66]. Exploring the mechanisms responsible for AKI caused by crush syndrome (CS), Wang et al. utilized rats and were able to show a significant increase in RIG-I-associated signaling [67]. A study using whole-exome sequencing in LN patients identified a gain-of-function mutation in RIG-I leading to its constitutive activation [68], and in a separate study using kidney samples from CKD patients, RLR activation was associated with increased levels of retrotransposons [35].
Cells within the glomerulus also express the major sensors for dsRNA and can activate the associated immune signaling pathways. Yamashita et al. used cultured human and mouse podocytes to show that regardless of the source of dsRNA, be it from mitochondria, retrotransposons, or secondary structures spontaneously formed, RLR activation in podocytes leads to the expression of IFN-I, IL-6, and cytoskeleton alterations [69], the latter being instrumental in causing effacement and eventual cell loss [70]. The remaining bulk of evidence linking RLRs to podocyte dysfunction comes from investigations into the mechanisms whereby APOL1 causes kidney damage. Fang et al. used long-term injections of recombinant APOL1 in mice and observed increased expression of RIG-I in podocytes. Similar findings were seen in cultured human podocytes engineered to express APOL1 RSK. Knock-down of RIG-I, either using siRNA against RIG-I in in vitro experiments or adeno-associated virus short harpins (AAV-sh) in in vivo mouse experiments, blunted the expression of several pro-inflammatory genes and attenuated podocyte and glomerular damage, respectively [71]. One caveat emerging from this study is that it remains unclear what the direct activator of RIG-I is, and if RIG-I drives the expression of IFN-I along with other proinflammatory genes. Further confounding the involvement of RLRs in APOL1-mediated podocyte damage is the discovery that APOL1 mRNA itself possesses structural characteristics that allow for the formation of dsRNA secondary structures [72], which indeed have been shown to be recognized by MDA5 in podocytes [73]. However, as just mentioned, it is yet to be determined if these APOL1 secondary structures can be also recognized by RIG-I.
There is a dearth of evidence for the expression of RLRs in other glomerular cells. Results from in vitro experiments support the notion that RLRs can contribute to glomerular injury. Hagele et al. used a synthetic agonist of RLR to treat cultured murine GECs. These experiments revealed that there is an increase in the expression of IFN-I, IL-6, and intracellular adhesion molecule 1 (ICAM-I). It can be extrapolated that these changes in GECs could lead to increased albumin permeability [74]. The same synthetic agonist was used to treat MCs in vitro. The result was an increase in the expression of RIG-I chemokine ligand 5 (CCL5) and the C-X-C motif chemokine ligand 10 (CXCL10) [75,76]. Taken together, these results suggest that if GECs and MCs are under stress and exposed to RLR ligands, they are able to promote inflammation, favoring chemotaxis and the adhesion of leukocytes. However, since the synthetic ligand for RLRs can also activate TLR3, signaling through TLR3-mediated pathways cannot be ruled out.

5. TLR Activation in the Kidney

TLRs comprise a large family of PRRs that are expressed on the plasma membrane and intracellularly in endosomes, the latter location being activated by nucleic acids and highly associated with sterile inflammation [77]. Endosomally restricted TLRs include: TLR3 (which recognizes dsRNA), TLR7/8 (which recognizes ssRNA), and TLR9 (which recognizes dsDNA). It must be highlighted that intracellular TLRs cannot participate in the first response to self-derived nucleic acids in the cytoplasm as the other RLRs mentioned above can since they are physically sequestered within endosomes. Instead, these TLRs are only activated in the event that extracellular DNA or RNA from damaged or dead bystander cells has been endocytosed and the nucleic-acid-containing endosome fuses with an acidified endosome containing the TLRs [78]. Indeed, there are several reports in the literature where extracellular mRNA or mitochondrial RNA derived from damaged cells has been engulfed by neighboring cells, resulting in TLR3 activation and sterile inflammation [63,79,80]. Highlighting a route for entry for nucleic acids, Bertheloot et al. showed that RNA released from damaged cells can be detected by the receptor for advanced glycation end products (RAGE) to elicit their internalization and activate TLR7 and TLR8 [81]. It is thought that a similar mechanism exists for the detection and internalization of mtDNA from damaged mitochondria, leading the activation of TLR9 [82]. Extracellular nucleic acids are shielded from degradation if they are bound to other molecules. These shielding molecules include antibodies, antimicrobial peptides, neutrophil extracellular traps (NETS), and microvesicles, all of which have been detected in patients with lupus [83,84,85,86]. Once formed, these nucleic acid–protein complexes can be taken up through endocytosis by specialized cells, including macrophages and dendritic cells, but also by podocytes, MCs, and GECs [87,88,89]. However, we should consider that the activation of this class of TLRs could be the consequence of previous IFN-I or other pathway activation and/or as a result of actions carried out by any number of immune cell types, rather than directly by cells within the glomerulus. Interestingly, some RNA-based prophylactics and therapeutics have been associated with altered kidney function [90,91,92], suggesting that these exogenous nucleic acids could be implicated in the induction of the IFN-I pathway. On the other hand, there is also experimental evidence pointing to the advantages of RNA-based drugs that target coding and non-coding RNAs associated with the development of kidney disease [93,94].
The function of TLRs in CKD and GD has been extensively reviewed in previous publications [95,96]; therefore, this review will narrowly focus on the involvement of TLR signaling in podocytes, GECs, and MCs.
Experiments using cultured human podocytes confirm that these cells highly express TLR3 and, upon its activation, were shown to induce the expression of IFNβ, several chemokines (IL-6, MCP-1, CCL5), the costimulatory molecule CD80, and APOL1, but not IFNα [97,98,99]. TLR7 agonists have been widely used to generate mouse models of lupus, which display varying degrees of podocyte injury; however, podocyte damage can be traced to the action of other cell types, including those that belong to the immune system [100,101]. A more direct involvement of TLR8 in podocyte damage has been seen in mice models of lupus and in rodents that have undergone unilateral ureteral obstruction. In these experimental models, TLR8 expression is increased in podocytes and is associated with the elevation of micro-RNA 21 (miR21) [102,103], an endogenous ligand for TLR8 [104]. Interestingly, it has been reported that miR21 is increased in the urine and renal tissue of CKD patients, including those diagnosed with DN and IgAN [105,106,107]. In a rat model of nephrotic syndrome and in a mouse model of autoimmune GN, TLR9 expression is increased and associated with podocyte injury [108,109]. Moreover, mtDNA and/or dsDNA seems to be the ligand for this receptor in podocytes, inducing their apoptosis in vitro. Seemingly contradictory results were reported by Bossaller et al., who showed that TLR9 deficiency aggravates LN induced by TLR7 agonists in mice [110], suggesting a role for TLR9 as a negative regulator to limit glomerular damage.
Only the involvement of TLR3 in GECs has been reported. A number of publications originating from the same group using cultured human cells showed that the in vitro activation of TLR3 elicits the expression of IFN-I, RLRs, chemokines, and plasminogen activator inhibitor-1 (PAI-1), a negative regulator of fibrinolysis [111,112,113,114,115,116]. Taken together, this body of work suggests that the activation of TLR3 in GECs may contribute to inflammatory and fibrotic responses, events that could also be a result of a secondary response to the IFN-I pathway signaling.
Similar to reports for GECs, much of the work exploring the action of TLRs in MCs was accomplished using cultured human mesangial cells. These studies show that the in vitro activation of TLR3 in MCs results in the expression of several different chemokines, adhesion molecules, and matrix metalloproteinase 9 [117,118,119,120]. It is worth mentioning that this metalloproteinase is an enzyme relevant to the inflammatory response and is detected in the biopsies of patients diagnosed with active and chronic GD [121], as well as in the urine of patients with MN [122]. A report by Shen et al. revealed that TLR9 was significantly elevated in MCs grown in vitro under hyperglycemic conditions and that knockdown of TLR9 under these same conditions reduces the expression of several inflammatory markers and the incidence of apoptosis [123]. In the same study using a mouse model of diabetes, silencing TLR9 reduced glomerular matrix cell expansion.
Taken together, these various reports suggest that under conditions that result in cellular stress, nucleic acids may be released into the extracellular space, where they can be taken up by any number of different cells, including cells within the glomerulus. This, in turn, results in the activation of the IFN-I pathway and may include other signal pathways, such those that induce nuclear factor κB (NF-κB) activation. The end result is to prolong and propagate sterile inflammation in the kidney.

6. Effect of IFN-I in Glomerular Cells

As described above, the activation of intracellular PRRs leads to the synthesis of IFN-I. The term ‘IFN’ encompasses a group of cytokines divided into three family types referred as type I, type II (also named γ), and type III (also named λ). Interferons were initially discovered for their essential role in the anti-viral and anti-cancer immune response, and more recently, for their participation in autoimmune and autoinflammatory disorders [6,124,125,126]. Among the IFN-I family members, several subtypes have been described in mammals, including α, β, δ, ε, κ, τ, and ω. In humans, IFN-I contains 13 isoforms of IFNα and one isoform of IFNβ, and they are primarily recognized by their biological function [124,127].
Type IFN-Is are recognized by a heterodimeric receptor composed by interferon α/β receptor subunits 1 and 2 (IFNAR1 and IFNAR2). Although both subunits are necessary for IFN recognition, the signal transduction is initiated by IFNAR1. Attached to the cytoplasmic tail of IFNAR1 is tyrosine kinase 2 (TYK2), which is initially activated, followed by the phosphorylation of Janus kinase 1 (JAK1) and the phosphorylation of signal transduction and activator of transcription (STAT)1 and STAT2, which are associated with the cytoplasmic tail of IFNAR2. Next is the formation of ISGF3, a heterotrimeric transcriptional factor made up of STAT1, STAT2, and IRF9, which functions to promote the expression of interferon-stimulated genes (ISGs), as shown Figure 1. Cellular stress, including ribosomal stress, drives crosstalk between the above-mentioned canonical IFN-I pathway and other signaling pathways to boost the expression of hundreds of ISGs. The end result is the alteration of several cellular processes, such as decreased transcription due to RNA degradation triggered by ISG20, a diminished translation rate due to protein kinase R activity, and changes in lipid metabolism by cholesterol 25-hydroxylase [4,128,129,130].
In mice, IFNAR1/2 have been reported to be ubiquitously expressed in all tissues, and after systemic activation with recombinant IFN-I injection, the kidney shows a strong response to these cytokines [131,132,133]. In human kidney cells, there is little information related to their expression levels. Chang et al. provide some peripheral evidence of IFNAR1 expression in para-tumoral healthy tissue from renal cell carcinoma biopsies [134]. Several glomerular diseases display enrichment of the IFN-I pathway (summarized in Table 1), strongly suggesting the activation of IFNAR1/2. However, there is a substantial lack of direct evidence related to their expression levels in different glomerular cells, either in normal or pathological conditions.
Recently, Manoharan et al. reported that IFNAR1 expression on podocytes, where it can physically interact with tissue factor (TF) under steady-state conditions due to its homology with IFNAR2, acts to restrict IFN-I pathway activation. Moreover, TF deletion in podocytes promotes IFN-I pathway activation and the development of glomerular damage. Interestingly, biopsies from patients diagnosed with different forms of GD show a reduction in TF levels within the glomerulus [135], suggesting that the loss of IFNAR1 and TF interaction could promote IFN-I pathway activity.
The effects of IFNα signaling in podocytes includes the induction of autophagy and the inhibition of the mammalian target of rapamycin complex 1 (mTORC1) pathway [136]. These observations provide an additional link between IFN-I signaling and the development of GD since the loss of mTORC1 in podocytes causes proteinuria associated with glomerular damage [137]. Furthermore, IFNβ induces the expression of APOL1 [99] and apoptosis in mature podocytes, along with the reduction of nephrin expression during podocyte differentiation in vitro [25]. Li et al. also showed that IFN-I activation leads to the induction of viperin (also known as RSAD2), a common ISG that has been reported to be a negative regulator of podocyte differentiation [138]. Similar effects have been described in other kidney-specific cells, such as parietal epithelial cells (PECs) and tubule epithelial cells (TECs), where IFN-I signaling disrupts the cell cycle and promotes cell death [25,139]. It is becoming clear that elevated IFN-I expression in podocytes can drive autophagy, impair cellular differentiation, and promote podocyte apoptosis, events that can exacerbate glomerular disease. On the other hand, Lee et al. showed that by targeting IFNAR1/2 signals using bariticinib, a JAK inhibitor, podocyte damage in a mouse LN model could be reduced, along with a general reduction in inflammation, pointing to a systemic effect of the inhibitor [140].
There is little direct evidence of what effect IFN-I has on glomerular endothelial cells. A series of publications from Hiroshi Tanaka’s group using cultured human GECs identified a set of ISGs with the potential to be used as biomarkers for LN [111,141,142]. Instead, we might draw on work accomplished using other types of endothelial cells to extrapolate IFN-I signaling effects on GECs. For example, work using brain, liver, and lung endothelial cells has indicated that IFN-I cytokines, mainly IFNβ, affect tight junctions through the down-regulation of VE-cadherin, reducing the synthesis of nitric oxide, the induction of PAI-1 secretion, and the reduction in caveolin 1 expression [143,144,145]. Combined, these effects could promote endothelial permeability and stiffness. Additionally, interferon-stimulated gene 15 (ISG15), one of the byproducts of IFNAR1/2 activation, has been linked to the development of vascular stiffness in cases of hypertension [146].
In MCs, it has been demonstrated that IFNβ promotes the expression of IL-6 and participates in regulating proliferation [147]. Using primary HMCs, Zhang et al. discovered a feed-forward regulatory pathway where IFN-I stimulates the expression of miR744, which boosts the activation of JAK1/STAT1/STAT2 to promote the expression of several chemokines, including CXCL10 [148]. More recently, Gao et al. showed that CXCL10 can stimulate mesangial cell proliferation and migration in vitro and participates in mesangial cell expansion in a mouse model of glomerulonephritis [149]. Figure 2 summarizes the key findings of this review with respect to the effect of the IFN-I pathway on glomerular cells. Many questions, however, remain unanswered. For example, is it possible that IFN-I pathway activation is merely an indirect reaction to an initial insult rather than the main cause of GD? Since GD is a group of diseases with each group displaying a complex pathophysiology, it may prove difficult to accurately uncover the responsible mechanisms for each since in vivo and in vitro models seldom fully resemble the pathology displayed by human patients. Nonetheless, and despite the complexity of the molecular mechanisms involved, it is clear that the IFN-I pathway is participating in the development and progression of GD.

7. Concluding Remarks and Future Perspectives

The evidence of IFN-I pathway function in glomerular cells presents an opportunity to explore the potential exploitation of this pathway for therapeutic purposes in GD. For example, manipulation of the IFN-I pathway and related cytokines has been reported to boost the production of antibodies in vaccination [150,151]. Therefore, in the context of autoimmune-associated GD, abrogating IFN-I pathway signaling could potentially have the opposite effect by decreasing the production of auto-antibodies. This idea is supported by a clinical trial that demonstrated the reduction of auto-antibodies in systemic lupus using a JAK1/2 inhibitor [152]. In a different study, blocking upstream of this pathway by targeting STING appears to be a promising strategy given recent progress in drug discovery [153].
Another avenue is to explore the effect of the IFN-I pathway on the metabolism of glomerular cells, including glycolysis, oxidative phosphorylation, and lipid synthesis, as has been reported for macrophages and T cells, affecting their function [154,155,156], or its relation with other cellular stress responses, such as the activation of inflammasomes and the unfolded protein response (UPR) [28,157]. Finally, new insights into renal fibrosis, glomerular cell senescence, and aging could be gained by exploring IFN-I pathway crosstalk with the transforming growth factor β (TGF-β) pathway [158], DNA damage and epigenetic changes [159], or the inflammaging process [160], respectively.

Author Contributions

Conceptualization, A.P.J.-U., S.M. and E.H.; original draft preparation, A.P.J.-U. and S.M.; visualization, E.H.; review and editing, A.P.J.-U., S.M. and E.H. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Institutes of Diabetes and Digestive and Kidney Diseases (NIDDK) grants R01DK125394 and R01DK132072.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Chung, K.W.; Dhillon, P.; Huang, S.; Sheng, X.; Shrestha, R.; Qiu, C.; Kaufman, B.A.; Park, J.; Pei, L.; Baur, J.; et al. Mitochondrial Damage and Activation of the STING Pathway Lead to Renal Inflammation and Fibrosis. Cell Metab. 2019, 30, 784–799.e5. [Google Scholar] [CrossRef]
  2. Mitrofanova, A.; Fontanella, A.; Tolerico, M.; Mallela, S.; David, J.M.; Zuo, Y.; Boulina, M.; Kim, J.-J.; Santos, J.; Ge, M.; et al. Activation of Stimulator of IFN Genes (STING) Causes Proteinuria and Contributes to Glomerular Diseases. J. Am. Soc. Nephrol. 2022, 33, 2153–2173. [Google Scholar] [CrossRef]
  3. Zang, N.; Cui, C.; Guo, X.; Song, J.; Hu, H.; Yang, M.; Xu, M.; Wang, L.; Hou, X.; He, Q.; et al. cGAS-STING activation contributes to podocyte injury in diabetic kidney disease. iScience 2022, 25, 105145. [Google Scholar] [CrossRef]
  4. Schoggins, J.W. Interferon-Stimulated Genes: What Do They All Do? Annu. Rev. Virol. 2019, 6, 567–584. [Google Scholar] [CrossRef]
  5. Crow, Y.J.; Lebon, P.; Casanova, J.-L.; Gresser, I. A Brief Historical Perspective on the Pathological Consequences of Excessive Type I Interferon Exposure In vivo. J. Clin. Immunol. 2018, 38, 694–698. [Google Scholar] [CrossRef] [PubMed]
  6. Crow, Y.J.; Stetson, D.B. The type I interferonopathies: 10 years on. Nat. Rev. Immunol. 2022, 22, 471–483. [Google Scholar] [CrossRef] [PubMed]
  7. Tinawi, M. Update on the etiology, classification, and management of glomerular diseases. Avicenna J. Med. 2020, 10, 61–67. [Google Scholar] [CrossRef] [PubMed]
  8. Anders, H.-J.; Kitching, A.R.; Leung, N.; Romagnani, P. Glomerulonephritis: Immunopathogenesis and immunotherapy. Nat. Rev. Immunol. 2023, 23, 453–471. [Google Scholar] [CrossRef] [PubMed]
  9. Koh, J.H.; Kang, M.; Park, S.; Cho, J.M.; Cho, S.; Kim, Y.; Lee, S.; Lee, H.; Joo, K.-W.; Kim, Y.S.; et al. Spatially resolved transcriptomic profiling for glomerular and tubulointerstitial gene expression in C3 glomerulopathy. medRxiv 2023, 23292064. [Google Scholar]
  10. Yu, B.C.; Moon, A.; Lee, K.H.; Oh, Y.S.; Park, M.Y.; Choi, S.J.; Kim, J.K. Minimal Change Disease Is Associated with Mitochondrial Injury and STING Pathway Activation. J. Clin. Med. 2022, 11, 557. [Google Scholar] [CrossRef]
  11. Hodgin, J.B.; Mariani, L.H.; Zee, J.; Liu, Q.; Smith, A.R.; Eddy, S.; Hartman, J.; Hamidi, H.; Gaut, J.P.; Palmer, M.B.; et al. Quanti-fication of Glomerular Structural Lesions: Associations with Clinical Outcomes and Transcriptomic Profiles in Nephrotic Syndrome. Am. J. Kidney Dis. 2022, 79, 807–819.e1. [Google Scholar] [CrossRef]
  12. Menon, R.; Otto, E.A.; Hoover, P.; Eddy, S.; Mariani, L.; Godfrey, B.; Berthier, C.C.; Eichinger, F.; Subramanian, L.; Harder, J.; et al. Single cell transcriptomics identifies focal segmental glomerulosclerosis remission endothelial biomarker. J. Clin. Investig. 2020, 5, 133267. [Google Scholar] [CrossRef] [PubMed]
  13. Xu, J.; Shen, C.; Lin, W.; Meng, T.; Ooi, J.D.; Eggenhuizen, P.J.; Tang, R.; Xiao, G.; Jin, P.; Ding, X.; et al. Single-Cell Profiling Reveals Transcriptional Signatures and Cell-Cell Crosstalk in An-ti-PLA2R Positive Idiopathic Membranous Nephropathy Patients. Front. Immunol. 2021, 12, 683330. [Google Scholar] [CrossRef] [PubMed]
  14. Deng, Y.; Zheng, Y.; Li, D.; Hong, Q.; Zhang, M.; Li, Q.; Fu, B.; Wu, L.; Wang, X.; Shen, W.; et al. Expression characteristics of interferon-stimulated genes and possible regulatory mechanisms in lupus patients using transcriptomics analyses. EBioMedicine 2021, 70, 103477. [Google Scholar] [CrossRef] [PubMed]
  15. Zhang, Y.; Liao, Y.; Hang, Q.; Sun, D.; Liu, Y. GBP2 acts as a member of the interferon signalling pathway in lupus nephritis. BMC Immunol. 2022, 23, 44. [Google Scholar] [CrossRef]
  16. Irifuku, T.; Okimoto, K.; Masuzawa, N.; Masaki, T. Nephrotic-range proteinuria and membranoproliferative glomerulone-phritis-like pattern caused by interferon-β1b in a patient with multiple sclerosis. CEN Case Rep. 2023, 12, 275–280. [Google Scholar] [CrossRef]
  17. Gianassi, I.; Allinovi, M.; Caroti, L.; Cirami, L.C. Broad spectrum of interferon-related nephropathies-glomerulonephritis, systemic lupus erythematosus-like syndrome and thrombotic microangiopathy: A case report and review of literature. World J. Nephrol. 2019, 8, 109–117. [Google Scholar]
  18. Markowitz, G.S.; Nasr, S.H.; Stokes, M.B.; D’Agati, V.D. Treatment with IFN-α, -β, or -γ is associated with collapsing focal segmental glomerulosclerosis. Clin. J. Am. Soc. Nephrol. 2010, 5, 607–615. [Google Scholar] [CrossRef]
  19. Kayar, Y.; Kayar, N.B.; Alpay, N.; Hamdard, J.; Ekinci, I.; Emegil, S.; Soydas, R.B.; Baysal, B. Interferon Induced Focal Segmental Glomerulosclerosis. Case Rep. Nephrol. 2016, 2016, 6967378. [Google Scholar] [CrossRef] [PubMed]
  20. He, T.; Xia, Y.; Yang, J. Systemic inflammation and chronic kidney disease in a patient due to the RNASEH2B defect. Pediatr. Rheumatol. 2021, 19, 9. [Google Scholar] [CrossRef]
  21. Abid, Q.; Rocha, A.B.; Larsen, C.P.; Schulert, G.; Marsh, R.; Yasin, S.; Patty-Resk, C.; Valentini, R.P.; Adams, M.; Baracco, R. APOL1-Associated Collapsing Focal Segmental Glomerulosclerosis in a Patient with Stimulator of Interferon Genes (STING)-Associated Vasculopathy with Onset in Infancy (SAVI). Am. J. Kidney Dis. 2020, 75, 287–290. [Google Scholar] [CrossRef] [PubMed]
  22. Staels, F.; Betrains, A.; Doubel, P.; Willemsen, M.; Cleemput, V.; Vanderschueren, S.; Corveleyn, A.; Meyts, I.; Sprangers, B.; Crow, Y.J.; et al. Adult-Onset ANCA-Associated Vasculitis in SAVI: Extension of the Phenotypic Spectrum, Case Report and Review of the Literature. Front. Immunol. 2020, 11, 575219. [Google Scholar] [CrossRef] [PubMed]
  23. Hou, G.; Zhou, T.; Xu, N.; Yin, Z.; Zhu, X.; Zhang, Y.; Cui, Y.; Ma, J.; Tang, Y.; Cheng, Z.; et al. Integrative Functional Genomics Identifies Systemic Lupus Erythematosus Causal Genetic Variant in the IRF5 Risk Locus. Arthritis Rheumatol. 2023, 75, 574–585. [Google Scholar] [CrossRef] [PubMed]
  24. Fairhurst, A.-M.; Xie, C.; Fu, Y.; Wang, A.; Boudreaux, C.; Zhou, X.J.; Cibotti, R.; Coyle, A.; Connolly, J.E.; Wakeland, E.K.; et al. Type I Interferons Produced by Resident Renal Cells May Promote End-Organ Disease in Autoantibody-Mediated Glomerulonephritis. J. Immunol. 2009, 183, 6831–6838. [Google Scholar] [CrossRef] [PubMed]
  25. Migliorini, A.; Angelotti, M.L.; Mulay, S.R.; Kulkarni, O.O.; Demleitner, J.; Dietrich, A.; Sagrinati, C.; Ballerini, L.; Peired, A.; Shankland, S.J.; et al. The antiviral cytokines IFN-α and IFN-β modulate parietal epithelial cells and promote podocyte loss: Implications for IFN toxicity, viral glomerulonephritis, and glomerular regeneration. Am. J. Pathol. 2013, 183, 431–440. [Google Scholar] [CrossRef]
  26. Nacionales, D.C.; Kelly-Scumpia, K.M.; Lee, P.Y.; Weinstein, J.S.; Lyons, R.; Sobel, E.; Satoh, M.; Reeves, W.H. Deficiency of the type I interferon receptor protects mice from experimental lupus. Arthritis Rheum. 2007, 56, 3770–3783. [Google Scholar] [CrossRef] [PubMed]
  27. Okude, H.; Ori, D.; Kawai, T. Signaling Through Nucleic Acid Sensors and Their Roles in Inflammatory Diseases. Front. Immunol. 2021, 11, 625833. [Google Scholar] [CrossRef] [PubMed]
  28. Davidson, S.; Yu, C.-H.; Steiner, A.; Ebstein, F.; Baker, P.J.; Jarur-Chamy, V.; Schaale, K.H.; Laohamonthonkul, P.; Kong, K.; Calleja, D.J.; et al. Protein kinase R is an innate immune sensor of proteotoxic stress via accumulation of cytoplasmic IL-24. Sci. Immunol. 2022, 7, eabi6763. [Google Scholar] [CrossRef]
  29. Hwang, I.; Uchida, H.; Dai, Z.; Li, F.; Sanchez, T.; Locasale, J.W.; Cantley, L.C.; Zheng, H.; Paik, J. Cellular stress signaling activates type-I IFN response through FOXO3-regulated lamin posttranslational modification. Nat. Commun. 2021, 12, 640. [Google Scholar] [CrossRef]
  30. Miller, K.N.; Victorelli, S.G.; Salmonowicz, H.; Dasgupta, N.; Liu, T.; Passos, J.F.; Adams, P.D. Cytoplasmic DNA: Sources, sensing, and role in aging and disease. Cell 2021, 184, 5506–5526. [Google Scholar] [CrossRef]
  31. Chen, Y.G.; Hur, S. Cellular origins of dsRNA, their recognition and consequences. Nat. Rev. Mol. Cell Biol. 2022, 23, 286–301. [Google Scholar] [CrossRef]
  32. Yoshimoto, N.; Hayashi, K.; Hishikawa, A.; Hashiguchi, A.; Nakamichi, R.; Sugita-Nishimura, E.; Yoshida-Hama, E.; Azegami, T.; Nakayama, T.; Itoh, H. Significance of podocyte DNA damage and glomerular DNA methylation in CKD patients with proteinuria. Hypertens. Res. 2023, 46, 1000–1008. [Google Scholar] [CrossRef] [PubMed]
  33. Ott, U.; Aschoff, A.; Pocock, J.; Funfstuck, R.; Jirikowski, G.; Stein, G.; Wolf, G. DNA fragmentation in chronic glomerulone-phritis: An immunohistological analysis. Nephron Clin. Pract. 2007, 105, c18–c28. [Google Scholar] [CrossRef]
  34. Nakamichi, R.; Hishikawa, A.; Chikuma, S.; Yoshimura, A.; Sasaki, T.; Hashiguchi, A.; Abe, T.; Tokuhara, T.; Yoshimoto, N.; Nishimura, E.S.; et al. DNA-damaged podocyte-CD8 T cell crosstalk exacerbates kidney injury by altering DNA methylation. Cell Rep. 2023, 42, 112302. [Google Scholar] [CrossRef] [PubMed]
  35. Dhillon, P.; Mulholland, K.A.; Hu, H.; Park, J.; Sheng, X.; Abedini, A.; Liu, H.; Vassalotti, A.; Wu, J.; Susztak, K. Increased levels of endogenous retroviruses trigger fibroinflammation and play a role in kidney disease development. Nat. Commun. 2023, 14, 559. [Google Scholar] [CrossRef] [PubMed]
  36. De Cecco, M.; Ito, T.; Petrashen, A.P.; Elias, A.E.; Skvir, N.J.; Criscione, S.W.; Caligiana, A.; Brocculi, G.; Adney, E.M.; Boeke, J.D.; et al. L1 drives IFN in senescent cells and promotes age-associated inflammation. Nature 2019, 566, 73–78. [Google Scholar] [CrossRef] [PubMed]
  37. Song, Z.R.; Jiang, L.; Li, Y.; Xiang, C.G.; Liu, Z.Y.; Li, M.S.; Zhang, H.; Zhou, X.J. Kidney-Predominant Thrombotic Microangiopathy Associated with TREX1 Frameshift Mutation. Kidney Int. Rep. 2023, 8, 2172–2176. [Google Scholar] [CrossRef]
  38. Li, Y.; Slavik, K.M.; Toyoda, H.C.; Morehouse, B.R.; Mann, C.C.d.O.; Elek, A.; Levy, S.; Wang, Z.; Mears, K.S.; Liu, J.; et al. cGLRs are a diverse family of pattern recognition receptors in innate immunity. Cell 2023, 186, 3261–3276.e20. [Google Scholar] [CrossRef] [PubMed]
  39. Hall, J.; Ralph, E.C.; Shanker, S.; Wang, H.; Byrnes, L.J.; Horst, R.; Wong, J.; Brault, A.; Dumlao, D.; Smith, J.F.; et al. The catalytic mechanism of cyclic GMP-AMP synthase (cGAS) and implications for innate immunity and inhibition. Protein Sci. 2017, 26, 2367–2380. [Google Scholar] [CrossRef]
  40. Shang, G.; Zhang, C.; Chen, Z.J.; Bai, X.C.; Zhang, X. Cryo-EM structures of STING reveal its mechanism of activation by cyclic GMP-AMP. Nature 2019, 567, 389–393. [Google Scholar] [CrossRef]
  41. Motani, K.; Saito-Tarashima, N.; Nishino, K.; Yamauchi, S.; Minakawa, N.; Kosako, H. The Golgi-resident protein ACBD3 concentrates STING at ER-Golgi contact sites to drive export from the ER. Cell Rep. 2022, 41, 111868. [Google Scholar] [CrossRef]
  42. Wu, J.; Raman, A.; Coffey, N.J.; Sheng, X.; Wahba, J.; Seasock, M.J.; Ma, Z.; Beckerman, P.; Laczkó, D.; Palmer, M.B.; et al. The key role of NLRP3 and STING in APOL1-associated podocytopathy. J. Clin. Investig. 2021, 131, jci136329. [Google Scholar] [CrossRef]
  43. Davis, S.E.; Khatua, A.K.; Popik, W. Nucleosomal dsDNA Stimulates APOL1 Expression in Human Cultured Podocytes by Activating the cGAS/IFI16-STING Signaling Pathway. Sci. Rep. 2019, 9, 15485. [Google Scholar] [CrossRef] [PubMed]
  44. Li, X.; Tao, S.; Xu, Z.; Ren, Y.; Xiang, W.; He, X. SMURF1 activates the cGAS/STING/IFN-1 signal axis by mediating YY1 ubiquitination to accelerate the progression of lupus nephritis. Autoimmunity 2023, 56, 2281235. [Google Scholar] [CrossRef] [PubMed]
  45. Qi, H.; Casalena, G.; Shi, S.; Yu, L.; Ebefors, K.; Sun, Y.; Zhang, W.; D’Agati, V.; Schlondorff, D.; Haraldsson, B.; et al. Glomerular Endothelial Mitochondrial Dysfunction Is Essential and Characteristic of Diabetic Kidney Disease Susceptibility. Diabetes 2017, 66, 763–778. [Google Scholar] [CrossRef] [PubMed]
  46. Casalena, G.A.; Yu, L.; Gil, R.; Rodriguez, S.; Sosa, S.; Janssen, W.; Azeloglu, E.U.; Leventhal, J.S.; Daehn, I.S. The diabetic microenvironment causes mitochondrial oxidative stress in glomerular endothelial cells and pathological crosstalk with podocytes. Cell Commun. Signal. 2020, 18, 105. [Google Scholar] [CrossRef] [PubMed]
  47. Daehn, I.; Casalena, G.; Zhang, T.; Shi, S.; Fenninger, F.; Barasch, N.; Yu, L.; D’Agati, V.; Schlondorff, D.; Kriz, W.; et al. Endothelial mitochondrial oxidative stress determines podocyte depletion in segmental glomerulosclerosis. J. Clin. Investig. 2014, 124, 1608–1621. [Google Scholar] [CrossRef] [PubMed]
  48. Wu, J.; Ma, Z.; Raman, A.; Beckerman, P.; Dhillon, P.; Mukhi, D.; Palmer, M.; Chen, H.C.; Cohen, C.R.; Dunn, T.; et al. APOL1 risk variants in individuals of African genetic ancestry drive endothelial cell defects that exacerbate sepsis. Immunity 2021, 54, 2632–2649.e6. [Google Scholar] [CrossRef] [PubMed]
  49. Hägele, H.; Allam, R.; Pawar, R.D.; Reichel, C.A.; Krombach, F.; Anders, H.-J. Double-Stranded DNA Activates Glomerular Endothelial Cells and Enhances Albumin Permeability via a Toll-Like Receptor-Independent Cytosolic DNA Recognition Pathway. Am. J. Pathol. 2009, 175, 1896–1904. [Google Scholar] [CrossRef] [PubMed]
  50. Ishikawa, H.; Ma, Z.; Barber, G.N. STING regulates intracellular DNA-mediated, type I interferon-dependent innate immunity. Nature 2009, 461, 788–792. [Google Scholar] [CrossRef]
  51. Wu, J.; Sun, L.; Chen, X.; Du, F.; Shi, H.; Chen, C.; Chen, Z.J. Cyclic GMP-AMP Is an Endogenous Second Messenger in Innate Immune Signaling by Cytosolic DNA. Science 2013, 339, 826–830. [Google Scholar] [CrossRef] [PubMed]
  52. Wu, J.; Shao, X.; Shen, J.; Lin, Q.; Zhu, X.; Li, S.; Li, J.; Zhou, W.; Qi, C.; Ni, Z. Downregulation of PPARα mediates FABP1 expression, contributing to IgA nephropathy by stimulating ferroptosis in human mesangial cells. Int. J. Biol. Sci. 2022, 18, 5438–5458. [Google Scholar] [CrossRef] [PubMed]
  53. Deng, B.; Lin, Y.; Chen, Y.; Ma, S.; Cai, Q.; Wang, W.; Li, B.; Liu, T.; Zhou, P.; He, R.; et al. Plasmacytoid dendritic cells promote acute kidney injury by producing interferon-α. Cell. Mol. Immunol. 2021, 18, 219–229. [Google Scholar] [CrossRef]
  54. Nehar-Belaid, D.; Hong, S.; Marches, R.; Chen, G.; Bolisetty, M.; Baisch, J.; Walters, L.; Punaro, M.; Rossi, R.J.; Chung, C.H.; et al. Mapping systemic lupus erythematosus heterogeneity at the single-cell level. Nat. Immunol. 2020, 21, 1094–1106. [Google Scholar] [CrossRef]
  55. Iwamoto, T.; Dorschner, J.M.; Selvaraj, S.; Mezzano, V.; Jensen, M.A.; Vsetecka, D.; Amin, S.; Makol, A.; Osborn, T.; Moder, K.; et al. High Systemic Type I Interferon Activity Is Associated with Active Class III/IV Lupus Nephritis. J. Rheumatol. 2022, 49, 388–397. [Google Scholar] [CrossRef] [PubMed]
  56. Thim-Uam, A.; Prabakaran, T.; Tansakul, M.; Makjaroen, J.; Wongkongkathep, P.; Chantaravisoot, N.; Saethang, T.; Lee-lahavanichkul, A.; Benjachat, T.; Paludan, S.; et al. STING Mediates Lupus via the Activation of Conventional Dendritic Cell Maturation and Plasmacytoid Dendritic Cell Differentiation. iScience 2020, 23, 101530. [Google Scholar] [CrossRef] [PubMed]
  57. Mehdipour, P.; Marhon, S.A.; Ettayebi, I.; Chakravarthy, A.; Hosseini, A.; Wang, Y.; de Castro, F.A.; Loo Yau, H.; Ishak, C.; Abelson, S.; et al. Epigenetic therapy induces transcription of inverted SINEs and ADAR1 dependency. Nature 2020, 588, 169–173. [Google Scholar] [CrossRef]
  58. Tunbak, H.; Enriquez-Gasca, R.; Tie, C.H.C.; Gould, P.A.; Mlcochova, P.; Gupta, R.K.; Fernandes, L.; Holt, J.; van der Veen, A.G.; Giampazolias, E.; et al. The HUSH complex is a gatekeeper of type I interferon through epigenetic regulation of LINE-1s. Nat. Commun. 2020, 11, 5387. [Google Scholar] [CrossRef]
  59. Silva, S.; Camino, L.P.; Aguilera, A. Human mitochondrial degradosome prevents harmful mitochondrial R loops and mito-chondrial genome instability. Proc. Natl. Acad. Sci. USA 2018, 115, 11024–11029. [Google Scholar] [CrossRef]
  60. Dhir, A.; Dhir, S.; Borowski, L.S.; Jimenez, L.; Teitell, M.; Rotig, A.; Crow, Y.J.; Rice, G.I.; Duffy, D.; Tamby, C.; et al. Mitochondrial double-stranded RNA triggers antiviral signalling in humans. Nature 2018, 560, 238–242. [Google Scholar] [CrossRef]
  61. Tigano, M.; Vargas, D.C.; Tremblay-Belzile, S.; Fu, Y.; Sfeir, A. Nuclear sensing of breaks in mitochondrial DNA enhances immune surveillance. Nature 2021, 591, 477–481. [Google Scholar] [CrossRef]
  62. Garcia-Gonzalez, C.; Dieterich, C.; Maroli, G.; Wiesnet, M.; Wietelmann, A.; Li, X.; Yuan, X.; Graumann, J.; Stellos, K.; Kubin, T.; et al. ADAR1 Prevents Autoinflammatory Processes in the Heart Mediated by IRF7. Circ. Res. 2022, 131, 580–597. [Google Scholar] [CrossRef]
  63. Kim, S.; Lee, K.; Choi, Y.S.; Ku, J.; Kim, H.; Kharbash, R.; Yoon, J.; Lee, Y.S.; Kim, J.-H.; Lee, Y.J.; et al. Mitochondrial double-stranded RNAs govern the stress response in chondrocytes to promote osteoarthritis development. Cell Rep. 2022, 40, 111178. [Google Scholar] [CrossRef] [PubMed]
  64. Yoon, J.; Lee, M.; Ali, A.A.; Oh, Y.R.; Choi, Y.S.; Kim, S.; Lee, N.; Jang, S.G.; Park, S.; Chung, J.-H.; et al. Mitochondrial double-stranded RNAs as a pivotal mediator in the pathogenesis of Sjӧgren’s syndrome. Mol. Ther. Nucleic Acids 2022, 30, 257–269. [Google Scholar] [CrossRef] [PubMed]
  65. Zhu, Y.; Zhang, M.; Wang, W.; Qu, S.; Liu, M.; Rong, W.; Yang, W.; Liang, H.; Zeng, C.; Zhu, X.; et al. Polynucleotide phosphorylase protects against renal tubular injury via blocking mt-dsRNA-PKR-eIF2α axis. Nat. Commun. 2023, 14, 1223. [Google Scholar] [CrossRef] [PubMed]
  66. Doke, T.; Mukherjee, S.; Mukhi, D.; Dhillon, P.; Abedini, A.; Davis, J.G.; Chellappa, K.; Chen, B.; Baur, J.A.; Susztak, K. NAD(+) precursor supplementation prevents mtRNA/RIG-I-dependent inflammation during kidney injury. Nat. Metab. 2023, 5, 414–430. [Google Scholar] [CrossRef]
  67. Wang, P.T.; Li, N.; Wang, X.Y.; Chen, J.L.; Geng, C.H.; Liu, Z.Q.; Fan, H.J.; Lv, Q.; Hou, S.K.; Gong, Y.H. RIG-I, a novel DAMPs sensor for myoglobin activates NF-kappaB/caspase-3 signaling in CS-AKI model. Mil. Med. Res. 2021, 8, 37. [Google Scholar]
  68. Peng, J.; Wang, Y.; Han, X.; Zhang, C.; Chen, X.; Jin, Y.; Yang, Z.; An, Y.; Zhang, J.; Liu, Z.; et al. Clinical Implications of a New DDX58 Pathogenic Variant That Causes Lupus Nephritis due to RIG-I Hyperactivation. J. Am. Soc. Nephrol. 2023, 34, 258–272. [Google Scholar] [CrossRef]
  69. Yamashita, M.; Millward, C.A.; Inoshita, H.; Saikia, P.; Chattopadhyay, S.; Sen, G.C.; Emancipator, S.N. Antiviral innate immunity disturbs podocyte cell function. J. Innate Immun. 2013, 5, 231–241. [Google Scholar] [CrossRef]
  70. Schell, C.; Huber, T.B. The Evolving Complexity of the Podocyte Cytoskeleton. J. Am. Soc. Nephrol. 2017, 28, 3166–3174. [Google Scholar] [CrossRef]
  71. Fang, J.; Yao, X.; Hou, M.; Duan, M.; Xing, L.; Huang, J.; Wang, Y.; Zhu, B.; Chen, Q.; Wang, H. ApoL1 induces kidney in-flammation through RIG-I/NF-κB activation. Biochem. Biophys. Res. Commun. 2020, 527, 466–473. [Google Scholar] [CrossRef]
  72. Okamoto, K.; Rausch, J.W.; Wakashin, H.; Fu, Y.; Chung, J.-Y.; Dummer, P.D.; Shin, M.K.; Chandra, P.; Suzuki, K.; Shrivastav, S.; et al. APOL1 risk allele RNA contributes to renal toxicity by activating protein kinase R. Commun. Biol. 2018, 1, 188. [Google Scholar] [CrossRef]
  73. Riella, C.V.; McNulty, M.; Ribas, G.T.; Tattersfield, C.F.; Perez-Gill, C.; Eichinger, F.; Kelly, J.; Chun, J.; Subramanian, B.; Guizelini, D.; et al. ADAR regulates APOL1 via A-to-I RNA editing by inhibition of MDA5 activation in a paradoxical biological circuit. Proc. Natl. Acad. Sci. USA 2022, 119, e2210150119. [Google Scholar] [CrossRef]
  74. Hägele, H.; Allam, R.; Pawar, R.D.; Anders, H.-J. Double-stranded RNA activates type I interferon secretion in glomerular endothelial cells via retinoic acid-inducible gene (RIG)-1. Nephrol. Dial. Transplant. 2009, 24, 3312–3318. [Google Scholar] [CrossRef]
  75. Imaizumi, T.; Tanaka, H.; Matsumiya, T.; Yoshida, H.; Tanji, K.; Tsuruga, K.; Oki, E.; Aizawa-Yashiro, T.; Ito, E.; Satoh, K. Retinoic acid-inducible gene-I is induced by double-stranded RNA and regulates the expression of CC chemokine ligand (CCL) 5 in human mesangial cells. Nephrol. Dial. Transplant. 2010, 25, 3534–3539. [Google Scholar] [CrossRef]
  76. Imaizumi, T.; Aizawa-Yashiro, T.; Tsuruga, K.; Tanaka, H.; Matsumiya, T.; Yoshida, H.; Tatsuta, T.; Xing, F.; Hayakari, R.; Satoh, K. Melanoma Differentiation-Associated Gene 5 Regulates the Expression of a Chemokine CXCL10 in Human Mesangial Cells: Implications for Chronic Inflammatory Renal Diseases. Tohoku J. Exp. Med. 2012, 228, 17–26. [Google Scholar] [CrossRef]
  77. Patra, M.C.; Achek, A.; Kim, G.-Y.; Panneerselvam, S.; Shin, H.-J.; Baek, W.-Y.; Lee, W.H.; Sung, J.; Jeong, U.; Cho, E.-Y.; et al. A Novel Small-Molecule Inhibitor of Endosomal TLRs Reduces Inflammation and Alleviates Autoimmune Disease Symptoms in Murine Models. Cells 2020, 9, 1648. [Google Scholar] [CrossRef]
  78. Lind, N.A.; Rael, V.E.; Pestal, K.; Liu, B.; Barton, G.M. Regulation of the nucleic acid-sensing Toll-like receptors. Nat. Rev. Immunol. 2022, 22, 224–235. [Google Scholar] [CrossRef]
  79. Gollmann-Tepeköylü, C.; Graber, M.; Pölzl, L.; Nägele, F.; Moling, R.; Esser, H.; Summerer, B.; Mellitzer, V.; Ebner, S.; Hirsch, J.; et al. Toll-like receptor 3 mediates ischaemia/reperfusion injury after cardiac transplantation. Eur. J. Cardio-Thoracic Surg. 2020, 57, 826–835. [Google Scholar] [CrossRef]
  80. Stolberg-Stolberg, J.; Boettcher, A.; Sambale, M.; Stuecker, S.; Sherwood, J.; Raschke, M.; Pap, T.; Bertrand, J. Toll-like receptor 3 activation promotes joint degeneration in osteoarthritis. Cell Death Dis. 2022, 13, 224. [Google Scholar] [CrossRef]
  81. Bertheloot, D.; Naumovski, A.L.; Langhoff, P.; Horvath, G.L.; Jin, T.; Xiao, T.S.; Garbi, N.; Agrawal, S.; Kolbeck, R.; Latz, E. RAGE Enhances TLR Responses through Binding and Internalization of RNA. J. Immunol. 2016, 197, 4118–4126. [Google Scholar] [CrossRef]
  82. Oka, T.; Hikoso, S.; Yamaguchi, O.; Taneike, M.; Takeda, T.; Tamai, T.; Oyabu, J.; Murakawa, T.; Nakayama, H.; Nishida, K.; et al. Mitochondrial DNA that escapes from autophagy causes inflammation and heart failure. Nature 2012, 485, 251–255. [Google Scholar] [CrossRef]
  83. Lande, R.; Ganguly, D.; Facchinetti, V.; Frasca, L.; Conrad, C.; Gregorio, J.; Meller, S.; Chamilos, G.; Sebasigari, R.; Riccieri, V.; et al. Neutrophils activate plasmacytoid dendritic cells by releasing self-DNA-peptide complexes in systemic lupus erythematosus. Sci. Transl. Med. 2011, 3, 73ra19. [Google Scholar] [CrossRef]
  84. Bruschi, M.; Bonanni, A.; Petretto, A.; Vaglio, A.; Pratesi, F.; Santucci, L.; Migliorini, P.; Bertelli, R.; Galetti, M.; Belletti, S.; et al. Neutrophil Extracellular Traps Profiles in Patients with Incident Systemic Lupus Erythematosus and Lupus Nephritis. J. Rheumatol. 2020, 47, 377–386. [Google Scholar] [CrossRef]
  85. Jorgensen, M.H.; Rekvig, O.P.; Jacobsen, R.S.; Jacobsen, S.; Fenton, K.A. Circulating levels of chromatin fragments are in-versely correlated with anti-dsDNA antibody levels in human and murine systemic lupus erythematosus. Immunol. Lett. 2011, 138, 179–186. [Google Scholar] [CrossRef]
  86. Rasmussen, N.S.; Nielsen, C.H.; Jacobsen, S. Microvesicles in active lupus nephritis show Toll-like receptor 9-dependent co-expression of galectin-3 binding protein and double-stranded DNA. Clin. Exp. Immunol. 2021, 204, 64–77. [Google Scholar] [CrossRef]
  87. Liu, Y.; Su, H.; Ma, C.; Ji, D.; Zheng, X.; Wang, P.; Zheng, S.; Wang, L.; Wang, Z.; Xu, D. IQGAP1 mediates podocyte injury in diabetic kidney disease by regulating nephrin endocytosis. Cell. Signal. 2019, 59, 13–23. [Google Scholar] [CrossRef]
  88. Bryniarski, M.A.; Yee, B.M.; Chaves, L.D.; Stahura, C.M.; Yacoub, R.; Morris, M.E. Megalin-mediated albumin endocytosis in cultured murine mesangial cells. Biochem. Biophys. Res. Commun. 2020, 529, 740–746. [Google Scholar] [CrossRef]
  89. Moriyama, T.; Karasawa, K.; Hasegawa, F.; Uchida, K.; Nitta, K. Sertraline Reduces Albuminuria by Interfering with Caveolae-Mediated Endocytosis through Glomerular Endothelial and Epithelial Cells. Am. J. Nephrol. 2019, 50, 444–453. [Google Scholar] [CrossRef]
  90. Lim, J.-H.; Kim, M.-S.; Kim, Y.-J.; Han, M.-H.; Jung, H.-Y.; Choi, J.-Y.; Cho, J.-H.; Kim, C.-D.; Kim, Y.-L.; Park, S.-H. New-Onset Kidney Diseases after COVID-19 Vaccination: A Case Series. Vaccines 2022, 10, 302. [Google Scholar] [CrossRef]
  91. Cho, I.; Kim, J.-K.; Kim, S.G. IgA vasculitis presenting as nephrotic syndrome following COVID-19 vaccination: A case report. BMC Nephrol. 2022, 23, 403. [Google Scholar] [CrossRef]
  92. Lazareth, H.; Poli, A.; Bignon, Y.; Mirmiran, A.; Rabant, M.; Cohen, R.; Schmitt, C.; Puy, H.; Karras, A.; Gouya, L.; et al. Renal Function Decline with Small Interfering RNA Silencing Aminolevulinic Acid Synthase 1 (ALAS1). Kidney Int. Rep. 2021, 6, 1904–1911. [Google Scholar] [CrossRef]
  93. Wang, Y.; Wu, Q.; Wang, J.; Li, L.; Sun, X.; Zhang, Z.; Zhang, L. Co-delivery of p38α MAPK and p65 siRNA by novel liposomal glomerulus-targeting nano carriers for effective immunoglobulin a nephropathy treatment. J. Control. Release 2020, 320, 457–468. [Google Scholar] [CrossRef]
  94. Kölling, M.; Kaucsar, T.; Schauerte, C.; Hübner, A.; Dettling, A.; Park, J.-K.; Busch, M.; Wulff, X.; Meier, M.; Scherf, K.; et al. Therapeutic miR-21 Silencing Ameliorates Diabetic Kidney Disease in Mice. Mol. Ther. 2017, 25, 165–180. [Google Scholar] [CrossRef]
  95. Liu, F.; Chen, H.; Cao, C.; Liang, Y.; Zhou, Y. The role of toll-like receptors (TLRs) and their therapeutic applications in glo-merulonephritis. Int. Urol. Nephrol. 2023, 55, 2845–2856. [Google Scholar] [CrossRef]
  96. Liu, M.; Zen, K. Toll-Like Receptors Regulate the Development and Progression of Renal Diseases. Kidney Dis. 2021, 7, 14–23. [Google Scholar] [CrossRef]
  97. Umetsu, H.; Watanabe, S.; Imaizumi, T.; Aizawa, T.; Tsugawa, K.; Kawaguchi, S.; Seya, K.; Matsumiya, T.; Tanaka, H. Inter-leukin-6 via Toll-Like Receptor 3 Signaling Attenuates the Expression of Proinflammatory Chemokines in Human Podocytes. Kidney Blood Press. Res. 2021, 46, 207–218. [Google Scholar] [CrossRef]
  98. Shimada, M.; Ishimoto, T.; Lee, P.Y.; Lanaspa, M.A.; Rivard, C.J.; Roncal-Jimenez, C.A.; Wymer, D.T.; Yamabe, H.; Mathieson, P.W.; Saleem, M.A.; et al. Toll-like receptor 3 ligands induce CD80 expression in human podocytes via an NF-κB-dependent pathway. Nephrol. Dial. Transplant. 2012, 27, 81–89. [Google Scholar] [CrossRef]
  99. Nichols, B.; Jog, P.; Lee, J.H.; Blackler, D.; Wilmot, M.; D’Agati, V.; Markowitz, G.; Kopp, J.B.; Alper, S.L.; Pollak, M.R.; et al. Innate immunity pathways regulate the nephropathy gene Apolipoprotein L1. Kidney Int. 2015, 87, 332–342. [Google Scholar] [CrossRef]
  100. Carlucci, F.; Ishaque, A.; Ling, G.S.; Szajna, M.; Sandison, A.; Donatien, P.; Cook, H.T.; Botto, M. C1q Modulates the Response to TLR7 Stimulation by Pristane-Primed Macrophages: Implications for Pristane-Induced Lupus. J. Immunol. 2016, 196, 1488–1494. [Google Scholar] [CrossRef]
  101. Zhang, D.; Xu, J.; Ren, J.; Ding, L.; Shi, G.; Li, D.; Dou, H.; Hou, Y. Myeloid-Derived Suppressor Cells Induce Podocyte Injury Through Increasing Reactive Oxygen Species in Lupus Nephritis. Front. Immunol. 2018, 9, 1443. [Google Scholar] [CrossRef]
  102. Masum, A.; Ichii, O.; Elewa, Y.H.A.; Kon, Y. Podocyte Injury Through Interaction Between Tlr8 and Its Endogenous Ligand miR-21 in Obstructed and Its Collateral Kidney. Front. Immunol. 2021, 11, 606488. [Google Scholar] [CrossRef]
  103. Kimura, J.; Ichii, O.; Miyazono, K.; Nakamura, T.; Horino, T.; Otsuka-Kanazawa, S.; Kon, Y. Overexpression of Toll-like receptor 8 correlates with the progression of podocyte injury in murine autoimmune glomerulonephritis. Sci. Rep. 2014, 4, 7290. [Google Scholar] [CrossRef]
  104. Zhang, Z.-J.; Guo, J.-S.; Li, S.-S.; Wu, X.-B.; Cao, D.-L.; Jiang, B.-C.; Jing, P.-B.; Bai, X.-Q.; Li, C.-H.; Wu, Z.-H.; et al. TLR8 and its endogenous ligand miR-21 contribute to neuropathic pain in murine DRG. J. Exp. Med. 2018, 215, 3019–3037. [Google Scholar] [CrossRef]
  105. Lange, T.; Artelt, N.; Kindt, F.; Stracke, S.; Rettig, R.; Lendeckel, U.; Chadjichristos, C.E.; Kavvadas, P.; Chatziantoniou, C.; Endlich, K.; et al. MiR-21 is up-regulated in urinary exosomes of chronic kidney disease patients and after glomerular injury. J. Cell. Mol. Med. 2019, 23, 4839–4843. [Google Scholar] [CrossRef]
  106. Zang, J.; Maxwell, A.P.; Simpson, D.A.; McKay, G.J. Differential Expression of Urinary Exosomal MicroRNAs miR-21-5p and miR-30b-5p in Individuals with Diabetic Kidney Disease. Sci. Rep. 2019, 9, 10900. [Google Scholar] [CrossRef]
  107. Szeto, C.-C.; Ng, J.K.-C.; Fung, W.W.-S.; Luk, C.C.-W.; Wang, G.; Chow, K.-M.; Lai, K.-B.; Li, P.K.-T.; Lai, F.M.-M. Kidney microRNA-21 Expression and Kidney Function in IgA Nephropathy. Radiology 2021, 3, 76–82. [Google Scholar] [CrossRef]
  108. Bao, W.; Xia, H.; Liang, Y.; Ye, Y.; Lu, Y.; Xu, X.; Duan, A.; He, J.; Chen, Z.; Wu, Y.; et al. Toll-like Receptor 9 Can be Activated by Endogenous Mitochondrial DNA to Induce Podocyte Apoptosis. Sci. Rep. 2016, 6, 22579. [Google Scholar] [CrossRef]
  109. Masum, A.; Ichii, O.; Elewa, Y.H.A.; Nakamura, T.; Otani, Y.; Hosotani, M.; Kon, Y. Overexpression of toll-like receptor 9 correlates with podocyte injury in a murine model of autoimmune membranoproliferative glomerulonephritis. Autoimmunity 2018, 51, 386–398. [Google Scholar] [CrossRef]
  110. Bossaller, L.; Christ, A.; Pelka, K.; Nündel, K.; Chiang, P.-I.; Pang, C.; Mishra, N.; Busto, P.; Bonegio, R.G.; Schmidt, R.E.; et al. TLR9 Deficiency Leads to Accelerated Renal Disease and Myeloid Lineage Abnormalities in Pristane-Induced Murine Lupus. J. Immunol. 2016, 197, 1044–1053. [Google Scholar] [CrossRef]
  111. Karasawa, T.; Sato, R.; Imaizumi, T.; Fujita, M.; Aizawa, T.; Tsugawa, K.; Mattinzoli, D.; Kawaguchi, S.; Seya, K.; Terui, K.; et al. Expression of interferon-stimulated gene 20 (ISG20), an antiviral effector protein, in glomerular endothelial cells: Possible involvement of ISG20 in lupus nephritis. Ren. Fail. 2023, 45, 2224890. [Google Scholar] [CrossRef]
  112. Liu, Q.; Imaizumi, T.; Aizawa, T.; Hirono, K.; Kawaguchi, S.; Watanabe, S.; Tsugawa, K.; Matsumiya, T.; Seya, K.; Yoshida, H.; et al. Cytosolic Sensors of Viral RNA Are Involved in the Production of Interleukin-6 via Toll-Like Receptor 3 Signaling in Human Glomerular Endothelial Cells. Kidney Blood Press. Res. 2019, 44, 62–71. [Google Scholar] [CrossRef]
  113. Hirono, K.; Imaizumi, T.; Aizawa, T.; Watanabe, S.; Tsugawa, K.; Shiratori, T.; Kawaguchi, S.; Seya, K.; Matsumiya, T.; Ito, E.; et al. Endothelial expression of fractalkine (CX3CL1) is induced by Toll-like receptor 3 signaling in cultured human glomerular endothelial cells. Mod. Rheumatol. 2020, 30, 1074–1081. [Google Scholar] [CrossRef]
  114. Sato, R.; Imaizumi, T.; Aizawa, T.; Watanabe, S.; Tsugawa, K.; Kawaguchi, S.; Seya, K.; Matsumiya, T.; Tanaka, H. Inhibitory effect of anti-malarial agents on the expression of proinflammatory chemokines via Toll-like receptor 3 signaling in human glomerular endothelial cells. Ren. Fail. 2021, 43, 643–650. [Google Scholar] [CrossRef]
  115. Aizawa, T.; Imaizumi, T.; Hirono, K.; Watanabe, S.; Tsugawa, K.; Tanaka, H. Chloroquine attenuates TLR3-mediated plasminogen activator inhibitor-1 expression in cultured human glomerular endothelial cells. Clin. Exp. Nephrol. 2019, 23, 448–454. [Google Scholar] [CrossRef]
  116. Imaizumi, T.; Hashimoto, S.; Sato, R.; Umetsu, H.; Aizawa, T.; Watanabe, S.; Kawaguchi, S.; Matsumiya, T.; Seya, K.; Ding, J.; et al. IFIT Proteins Are Involved in CXCL10 Expression in Human Glomerular Endothelial Cells Treated with a Toll-Like Receptor 3 Agonist. Kidney Blood Press. Res. 2021, 46, 74–83. [Google Scholar] [CrossRef]
  117. Merkle, M.; Ribeiro, A.; Köppel, S.; Pircher, J.; Mannell, H.; Roeder, M.; Wörnle, M. TLR3-dependent immune regulatory functions of human mesangial cells. Cell. Mol. Immunol. 2012, 9, 334–340. [Google Scholar] [CrossRef]
  118. Imaizumi, T.; Aizawa, T.; Segawa, C.; Shimada, M.; Tsuruga, K.; Kawaguchi, S.; Matsumiya, T.; Yoshida, H.; Joh, K.; Tanaka, H. Toll-like receptor 3 signaling contributes to the expression of a neutrophil chemoattractant, CXCL1 in human mesangial cells. Clin. Exp. Nephrol. 2015, 19, 761–770. [Google Scholar] [CrossRef]
  119. Imaizumi, T.; Hayakari, R.; Watanabe, S.; Aizawa, T.; Matsumiya, T.; Yoshida, H.; Tsuruga, K.; Kawaguchi, S.; Tanaka, H. Cylindromatosis (CYLD), a Deubiquitinase, Attenuates Inflammatory Signaling Pathways by Activating Toll-Like Receptor 3 in Human Mesangial Cells. Kidney Blood Press. Res. 2017, 42, 942–950. [Google Scholar] [CrossRef]
  120. Merkle, M.; Ribeiro, A.; Koppel, S.; Wornle, M. TNFα enhances TLR3-dependent effects on MMP-9 expression in human mesangial cells. Cell Biol. Int. 2012, 36, 1155–1160. [Google Scholar] [CrossRef]
  121. Phillips, T.M.; Fadia, M.; Lea-Henry, T.N.; Smiles, J.; Walters, G.D.; Jiang, S.H. MMP2 and MMP9 associate with crescentic glomerulonephritis. Clin. Kidney J. 2017, 10, 215–220. [Google Scholar] [CrossRef]
  122. Gilbert, A.; Changjuan, A.; Guixue, C.; Jianhua, L.; Xiaosong, Q. Urinary Matrix Metalloproteinase-9 and Nephrin in Idiopathic Membranous Nephropathy: A Cross-Sectional Study. Dis. Mark. 2021, 2021, 1620545. [Google Scholar] [CrossRef]
  123. Shen, J.; Dai, Z.; Li, Y.; Zhu, H.; Zhao, L. TLR9 regulates NLRP3 inflammasome activation via the NF-kB signaling pathway in diabetic nephropathy. Diabetol. Metab. Syndr. 2022, 14, 26. [Google Scholar] [CrossRef]
  124. Bourdon, M.; Manet, C.; Montagutelli, X. Host genetic susceptibility to viral infections: The role of type I interferon induction. Genes Immun. 2020, 21, 365–379. [Google Scholar] [CrossRef]
  125. Aricò, E.; Castiello, L.; Capone, I.; Gabriele, L.; Belardelli, F. Type I Interferons and Cancer: An Evolving Story Demanding Novel Clinical Applications. Cancers 2019, 11, 1943. [Google Scholar] [CrossRef]
  126. Zhang, Q.; Bastard, P.; Liu, Z.; Le Pen, J.; Moncada-Velez, M.; Chen, J.; Ogishi, M.; Sabli, I.K.D.; Hodeib, S.; Korol, C.; et al. Inborn errors of type I IFN immunity in patients with life-threatening COVID-19. Science 2020, 370, eabd4570. [Google Scholar] [CrossRef]
  127. Peters, S.O.; Hussain, T.; Adenaike, A.S.; Hazzard, J.; Morenikeji, O.B.; De Donato, M.; Paul, S.; Babar, M.; Yakubu, A.; Imumorin, I.G. Evolutionary Pattern of Interferon Alpha Genes in Bovidae and Genetic Diversity of IFNAA in the Bovine Genome. Front. Immunol. 2020, 11, 580412. [Google Scholar] [CrossRef]
  128. Shemesh, M.; Lochte, S.; Piehler, J.; Schreiber, G. IFNAR1 and IFNAR2 play distinct roles in initiating type I interferon-induced JAK-STAT signaling and activating STATs. Sci. Signal. 2021, 14, eabe4627. [Google Scholar] [CrossRef]
  129. Boccuni, L.; Podgorschek, E.; Schmiedeberg, M.; Platanitis, E.; Traxler, P.; Fischer, P.; Schirripa, A.; Novoszel, P.; Nebreda, A.R.; Arthur, J.S.C.; et al. Stress signaling boosts interferon-induced gene transcription in macrophages. Sci. Signal. 2022, 15, eabq5389. [Google Scholar] [CrossRef]
  130. Hubel, P.; Urban, C.; Bergant, V.; Schneider, W.M.; Knauer, B.; Stukalov, A.; Scaturro, P.; Mann, A.; Brunotte, L.; Hoffmann, H.H.; et al. A protein-interaction network of interfer-on-stimulated genes extends the innate immune system landscape. Nat. Immunol. 2019, 20, 493–502. [Google Scholar] [CrossRef]
  131. Hardy, M.P.; Sanij, E.P.; Hertzog, P.J.; Owczarek, C.M. Characterization and transcriptional analysis of the mouse Chromosome 16 cytokine receptor gene cluster. Mamm. Genome 2003, 14, 105–118. [Google Scholar] [CrossRef]
  132. Hardy, M.P.; Owczarek, C.M.; Trajanovska, S.; Liu, X.; Kola, I.; Hertzog, P.J. The soluble murine type I interferon receptor Ifnar-2 is present in serum, is independently regulated, and has both agonistic and antagonistic properties. Blood 2001, 97, 473–482. [Google Scholar] [CrossRef]
  133. Pulverer, J.E.; Rand, U.; Lienenklaus, S.; Kugel, D.; Zietara, N.; Kochs, G.; Naumann, R.; Weiss, S.; Staeheli, P.; Hauser, H.; et al. Temporal and spatial resolution of type I and III interferon responses in vivo. J. Virol. 2010, 84, 8626–8638. [Google Scholar] [CrossRef]
  134. Chang, K.; Xu, F.; Zhang, X.; Zeng, B.; Zhang, W.; Shi, G.; Ye, D. Construction of an Immune Escape-Related Signature in Clear Cell Renal Cell Carcinoma and Identification of the Relationship between IFNAR1 and Immune Infiltration by Multiple Immunohistochemistry. Cancers 2022, 15, 169. [Google Scholar] [CrossRef]
  135. Manoharan, J.; Rana, R.; Kuenze, G.; Gupta, D.; Elwakiel, A.; Ambreen, S.; Wang, H.; Banerjee, K.; Zimmermann, S.; Singh, K.; et al. Tissue factor binds to and inhibits interferon-α receptor 1 signaling. Immunity 2024, 57, 68–85.e11. [Google Scholar] [CrossRef]
  136. Qi, Y.-Y.; Zhou, X.-J.; Cheng, F.-J.; Hou, P.; Ren, Y.-L.; Wang, S.-X.; Zhao, M.-H.; Yang, L.; Martinez, J.; Zhang, H. Increased autophagy is cytoprotective against podocyte injury induced by antibody and interferon-α in lupus nephritis. Ann. Rheum. Dis. 2018, 77, 1799–1809. [Google Scholar] [CrossRef]
  137. Godel, M.; Hartleben, B.; Herbach, N.; Liu, S.; Zschiedrich, S.; Lu, S.; Debreczeni-Mor, A.; Lindenmeyer, M.T.; Rastaldi, M.P.; Hartleben, G.; et al. Role of mTOR in podocyte function and diabetic nephropathy in humans and mice. J. Clin. Investig. 2011, 121, 2197–2209. [Google Scholar] [CrossRef]
  138. Li, Z.; Yin, H.; Hao, S.; Wang, L.; Gao, J.; Tan, X.; Yang, Z. miR-200 family promotes podocyte differentiation through repression of RSAD2. Sci. Rep. 2016, 6, 27105. [Google Scholar] [CrossRef] [PubMed]
  139. Cordoba-David, G.; Garcia-Gimenez, J.; Cardoso Castelo-Branco, R.; Carrasco, S.; Cannata, P.; Ortiz, A.; Ramos, A.M. Crosstalk between TBK1/IKKε and the type I interferon pathway contributes to tubulointerstitial inflammation and kidney tubular injury. Front. Pharmacol. 2022, 13, 987979. [Google Scholar] [CrossRef] [PubMed]
  140. Lee, J.; Park, Y.; Jang, S.G.; Hong, S.M.; Song, Y.S.; Kim, M.J.; Baek, S.; Park, S.H.; Kwok, S.K. Baricitinib Attenuates Autoimmune Phenotype and Podocyte Injury in a Murine Model of Systemic Lupus Erythematosus. Front. Immunol. 2021, 12, 704526. [Google Scholar] [CrossRef] [PubMed]
  141. Karasawa, T.; Sato, R.; Imaizumi, T.; Hashimoto, S.; Fujita, M.; Aizawa, T.; Tsugawa, K.; Kawaguchi, S.; Seya, K.; Terui, K.; et al. Glomerular endothelial expression of type I IFN-stimulated gene, DExD/H-Box helicase 60 via toll-like receptor 3 signaling: Possible involvement in the pathogenesis of lupus nephritis. Ren. Fail. 2022, 44, 137–145. [Google Scholar] [CrossRef]
  142. Hashimoto, S.; Imaizumi, T.; Watanabe, S.; Aizawa, T.; Tsugawa, K.; Kawaguchi, S.; Seya, K.; Matsumiya, T.; Tanaka, H. Expression of IFN-induced transmembrane protein 1 in glomerular endothelial cells. Pediatr. Int. 2021, 63, 1075–1081. [Google Scholar] [CrossRef]
  143. Jana, A.; Wang, X.; Leasure, J.W.; Magana, L.; Wang, L.; Kim, Y.-M.; Dodiya, H.; Toth, P.T.; Sisodia, S.S.; Rehman, J. Increased Type I interferon signaling and brain endothelial barrier dysfunction in an experimental model of Alzheimer’s disease. Sci. Rep. 2022, 12, 16488. [Google Scholar] [CrossRef]
  144. Jia, H.; Thelwell, C.; Dilger, P.; Bird, C.; Daniels, S.; Wadhwa, M. Endothelial cell functions impaired by interferon in vitro: Insights into the molecular mechanism of thrombotic microangiopathy associated with interferon therapy. Thromb. Res. 2018, 163, 105–116. [Google Scholar] [CrossRef]
  145. Gairhe, S.; Awad, K.S.; Dougherty, E.J.; Ferreyra, G.A.; Wang, S.; Yu, Z.X.; Takeda, K.; Demirkale, C.Y.; Torabi-Parizi, P.; Austin, E.D.; et al. Type I interferon activation and endothelial dysfunction in caveolin-1 insufficiency-associated pulmonary arterial hypertension. Proc. Natl. Acad. Sci. USA 2021, 118, e2010206118. [Google Scholar] [CrossRef]
  146. Gonzalez-Amor, M.; Garcia-Redondo, A.B.; Jorge, I.; Zalba, G.; Becares, M.; Ruiz-Rodriguez, M.J.; Rodriguez, C.; Bermeo, H.; Rodrigues-Diez, R.; Rios, F.J.; et al. Interferon-stimulated gene 15 pathway is a novel mediator of endothelial dysfunction and aneurysms development in angiotensin II infused mice through increased oxidative stress. Cardiovasc. Res. 2022, 118, 3250–3268. [Google Scholar] [CrossRef]
  147. Flür, K.; Allam, R.; Zecher, D.; Kulkarni, O.P.; Lichtnekert, J.; Schwarz, M.; Beutler, B.; Vielhauer, V.; Anders, H.-J. Viral RNA Induces Type I Interferon-Dependent Cytokine Release and Cell Death in Mesangial Cells via Melanoma-Differentiation-Associated Gene-5: Implications for Viral Infection-Associated Glomerulonephritis. Am. J. Pathol. 2009, 175, 2014–2022. [Google Scholar] [CrossRef] [PubMed]
  148. Zhang, X.; Han, X.; Tang, Y.; Wu, Y.; Qu, B.; Shen, N. miR-744 enhances type I interferon signaling pathway by targeting PTP1B in primary human renal mesangial cells. Sci. Rep. 2015, 5, 12987. [Google Scholar] [CrossRef] [PubMed]
  149. Gao, J.; Wu, L.; Wang, Y.; Cui, S.; Duan, S.; Dong, Z.; Feng, Z.; Chen, X. Knockdown of Cxcl10 Inhibits Mesangial Cell Pro-liferation in Murine Habu Nephritis Via ERK Signaling. Cell. Physiol. Biochem. 2017, 42, 2118–2129. [Google Scholar] [CrossRef]
  150. Ye, L.; Ohnemus, A.; Ong, L.C.; Gad, H.H.; Hartmann, R.; Lycke, N.; Staeheli, P. Type I and Type III Interferons Differ in Their Adjuvant Activities for Influenza Vaccines. J. Virol. 2019, 93, 23. [Google Scholar] [CrossRef]
  151. Le Bon, A.; Schiavoni, G.; D’Agostino, G.; Gresser, I.; Belardelli, F.; Tough, D.F. Type I Interferons Potently Enhance Humoral Immunity and Can Promote Isotype Switching by Stimulating Dendritic Cells In Vivo. Immunity 2001, 14, 461–470. [Google Scholar] [CrossRef]
  152. Dorner, T.; van Vollenhoven, R.F.; Doria, A.; Jia, B.; Ross Terres, J.A.; Silk, M.E.; de Bono, S.; Fischer, P.; Wallace, D.J. Baricitinib decreases anti-dsDNA in patients with systemic lupus erythematosus: Results from a phase II double-blind, randomized, placebo-controlled trial. Arthritis Res. Ther. 2022, 24, 112. [Google Scholar] [CrossRef]
  153. Kong, X.; Zuo, H.; Huang, H.D.; Zhang, Q.; Chen, J.; He, C.; Hu, Y. STING as an emerging therapeutic target for drug discovery: Perspectives from the global patent landscape. J. Adv. Res. 2023, 44, 119–133. [Google Scholar] [CrossRef]
  154. Olson, G.S.; Murray, T.A.; Jahn, A.N.; Mai, D.; Diercks, A.H.; Gold, E.S.; Aderem, A. Type I interferon decreases macrophage energy metabolism during mycobacterial infection. Cell Rep. 2021, 35, 109195. [Google Scholar] [CrossRef]
  155. Buang, N.; Tapeng, L.; Gray, V.; Sardini, A.; Whilding, C.; Lightstone, L.; Cairns, T.D.; Pickering, M.C.; Behmoaras, J.; Ling, G.S.; et al. Type I interferons affect the metabolic fitness of CD8+T cells from patients with systemic lupus erythematosus. Nat. Commun. 2021, 12, 1980. [Google Scholar] [CrossRef] [PubMed]
  156. York, A.G.; Williams, K.J.; Argus, J.P.; Zhou, Q.D.; Brar, G.; Vergnes, L.; Gray, E.E.; Zhen, A.; Wu, N.C.; Yamada, D.H.; et al. Limiting Cholesterol Biosynthetic Flux Spontaneously Engages Type I IFN Signaling. Cell 2015, 163, 1716–1729. [Google Scholar] [CrossRef] [PubMed]
  157. Chipurupalli, S.; Samavedam, U.; Robinson, N. Crosstalk Between ER Stress, Autophagy and Inflammation. Front. Med. 2021, 8, 758311. [Google Scholar] [CrossRef]
  158. Ahodantin, J.; Nio, K.; Funaki, M.; Zhai, X.; Wilson, E.; Kottilil, S.; Cheng, L.; Li, G.; Su, L. Type I interferons and TGF-β cooperate to induce liver fibrosis during HIV-1 infection under antiretroviral therapy. JCI Insight 2022, 7, e152738. [Google Scholar] [CrossRef]
  159. Yu, Q.; Katlinskaya, Y.V.; Carbone, C.J.; Zhao, B.; Katlinski, K.V.; Zheng, H.; Guha, M.; Li, N.; Chen, Q.; Yang, T.; et al. DNA-Damage-Induced Type I Interferon Promotes Senescence and Inhibits Stem Cell Function. Cell Rep. 2015, 11, 785–797. [Google Scholar] [CrossRef]
  160. Rasa, S.M.M.; Annunziata, F.; Krepelova, A.; Nunna, S.; Omrani, O.; Gebert, N.; Adam, L.; Käppel, S.; Höhn, S.; Donati, G.; et al. Inflammaging is driven by upregulation of innate immune receptors and systemic interferon signaling and is ameliorated by dietary restriction. Cell Rep. 2022, 39, 111017. [Google Scholar] [CrossRef]
Figure 1. Origins and intracellular sensors of self-nucleic acids promoting the IFN-I signaling pathway. During cellular stress, self-derived nucleic acids (DNA or RNA) are released from damaged mitochondria and the nucleus, consequently accumulating in the cytosol. In addition, extracellular nucleic acids liberated from neighboring dying or damaged cells are internalized by endocytosis. These intracellular (cytosolic or endosomal) nucleic acids are recognized by diverse intracellular nucleic acid sensors, triggering the activation of the signaling pathways that produce IFN-I and proinflammatory cytokines. Specifically, sensors for endosomal nucleic acids include TLR3 (for double-stranded DNA), TLR7 (for single-stranded RNA), and TLR9 (for single-stranded DNA). Cytosolic double-stranded RNA is detected by RIG-I or MDA5, while cytosolic double-stranded DNA is recognized by members of the cGAS-STING pathway. Activation of these intracellular nucleic acid sensors stimulates TBK1 activation, prompting the translocation of IRFs and NF-kB into the nucleus. There, they orchestrate the expression of IFN-I and proinflammatory cytokine genes. Subsequently, binding of IFN-I to IFNAR1/IFNAR2 triggers the activation of the JAK-STAT pathway, culminating in the induction of ISGs. This cascade of events illustrates the intricate molecular mechanisms involved in the recognition of self-nucleic acids, the subsequent activation of signaling pathways leading to the expression and secretion of IFN-I and proinflammatory cytokines, and the subsequent induction of ISGs via the JAK-STAT pathway. This figure was created with BioRender.com. IFN-I, type I interferons; TLR, toll-like receptor; RIG-I, retinoic acid-inducible gene I; MDA5, melanoma differentiation-associated protein 5; cGAS, cyclic GMP-AMP (cGAMP) synthase; STING, stimulator of interferon genes; TBK1, TANK-binding kinase 1; IFNAR, type I interferon receptor; ISGs, interferon-stimulated genes.
Figure 1. Origins and intracellular sensors of self-nucleic acids promoting the IFN-I signaling pathway. During cellular stress, self-derived nucleic acids (DNA or RNA) are released from damaged mitochondria and the nucleus, consequently accumulating in the cytosol. In addition, extracellular nucleic acids liberated from neighboring dying or damaged cells are internalized by endocytosis. These intracellular (cytosolic or endosomal) nucleic acids are recognized by diverse intracellular nucleic acid sensors, triggering the activation of the signaling pathways that produce IFN-I and proinflammatory cytokines. Specifically, sensors for endosomal nucleic acids include TLR3 (for double-stranded DNA), TLR7 (for single-stranded RNA), and TLR9 (for single-stranded DNA). Cytosolic double-stranded RNA is detected by RIG-I or MDA5, while cytosolic double-stranded DNA is recognized by members of the cGAS-STING pathway. Activation of these intracellular nucleic acid sensors stimulates TBK1 activation, prompting the translocation of IRFs and NF-kB into the nucleus. There, they orchestrate the expression of IFN-I and proinflammatory cytokine genes. Subsequently, binding of IFN-I to IFNAR1/IFNAR2 triggers the activation of the JAK-STAT pathway, culminating in the induction of ISGs. This cascade of events illustrates the intricate molecular mechanisms involved in the recognition of self-nucleic acids, the subsequent activation of signaling pathways leading to the expression and secretion of IFN-I and proinflammatory cytokines, and the subsequent induction of ISGs via the JAK-STAT pathway. This figure was created with BioRender.com. IFN-I, type I interferons; TLR, toll-like receptor; RIG-I, retinoic acid-inducible gene I; MDA5, melanoma differentiation-associated protein 5; cGAS, cyclic GMP-AMP (cGAMP) synthase; STING, stimulator of interferon genes; TBK1, TANK-binding kinase 1; IFNAR, type I interferon receptor; ISGs, interferon-stimulated genes.
Ijms 25 02497 g001
Figure 2. The potential deleterious effects of IFN-I on different glomerular cell types. IFN-I can exhibit a direct influence on diverse cell types within the glomerular compartment, altering their cellular functions. In addition, increased IFN-I levels induce the expression of interferon-stimulated genes (ISGs). This could intensify inflammatory processes by recruiting inflammatory immune cells to the kidney and increasing the production of autoantibodies. This figure was created with BioRender.com. APOL-1, apolipoprotein L1; VE-cadherin, vascular endothelial-cadherin; NO, nitric oxide. Red arrows indicate increased responses.
Figure 2. The potential deleterious effects of IFN-I on different glomerular cell types. IFN-I can exhibit a direct influence on diverse cell types within the glomerular compartment, altering their cellular functions. In addition, increased IFN-I levels induce the expression of interferon-stimulated genes (ISGs). This could intensify inflammatory processes by recruiting inflammatory immune cells to the kidney and increasing the production of autoantibodies. This figure was created with BioRender.com. APOL-1, apolipoprotein L1; VE-cadherin, vascular endothelial-cadherin; NO, nitric oxide. Red arrows indicate increased responses.
Ijms 25 02497 g002
Table 1. IFN-I pathway components enriched in glomerular disease.
Table 1. IFN-I pathway components enriched in glomerular disease.
Glomerular DiseaseObservationRef.
MPGN
C3 glomerulopathy
Interferon-stimulated genes (ISG) IFI44L, IFIT1, MX1, and OAS2 enriched[9]
MCDImmunohistochemical detection of STING in the glomerulus
ISG and PRR genes enriched (IFIH1, IRF7, OAS1, TLR7, TLR8)
[10]
[11]
FSGSISG and PRR genes enriched (IFIH1, IRF7, OAS1, TLR7, TLR8)
IFN-I signaling pathway enrichment in glomerular endothelial cells
[11]
[12]
MN
anti-PLA2R
IFN-I signaling pathway in glomerular mesangial cells (IFI16 gene enrichment)[13]
IgANImmunohistochemical detection of STING in glomerulus, tubules, and interstitium, associated with urinary mitochondrial DNA[10]
LNIFN-I signaling pathway enrichment
ISG genes enriched (IRF5, OAS1, MX2, DHX58, IFITM1, ADAR, GBP2)
[14]
[15]
Table 2. Intracellular pattern recognition receptors (PRRs) involved in IFN-I induction.
Table 2. Intracellular pattern recognition receptors (PRRs) involved in IFN-I induction.
FamilyNameLigandsCell DistributionSource of Endogenous Activators
cGLRscGASdsDNACytoplasm
  • Nuclear DNA
  • Mitochondrial DNA
RLRsRIG-IdsRNACytoplasm
  • Mitochondrial dsRNA
  • Transposable elements (TE) in the form of Alu RNA, RNA:DNA duplex
  • Misedited RNA with secondary structures
MDA5dsRNACytoplasm
TLRsTLR3dsRNAEndosomes
  • Extracellular nucleic acids from damaged bystander cells
TLR7ssRNA
TLR8ssRNA
TLR9dsDNA
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

Jimenez-Uribe, A.P.; Mangos, S.; Hahm, E. Type I IFN in Glomerular Disease: Scarring beyond the STING. Int. J. Mol. Sci. 2024, 25, 2497. https://doi.org/10.3390/ijms25052497

AMA Style

Jimenez-Uribe AP, Mangos S, Hahm E. Type I IFN in Glomerular Disease: Scarring beyond the STING. International Journal of Molecular Sciences. 2024; 25(5):2497. https://doi.org/10.3390/ijms25052497

Chicago/Turabian Style

Jimenez-Uribe, Alexis Paulina, Steve Mangos, and Eunsil Hahm. 2024. "Type I IFN in Glomerular Disease: Scarring beyond the STING" International Journal of Molecular Sciences 25, no. 5: 2497. https://doi.org/10.3390/ijms25052497

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