Back to Journals » International Journal of General Medicine » Volume 18
Inflammation-Induced Klotho Deficiency: A Possible Key Driver of Chronic Kidney Disease Progression
Authors Liang Y, Zhang Q, Qian JR, Li SS, Liu QF
Received 20 December 2024
Accepted for publication 5 May 2025
Published 11 May 2025 Volume 2025:18 Pages 2507—2520
DOI https://doi.org/10.2147/IJGM.S513497
Checked for plagiarism Yes
Review by Single anonymous peer review
Peer reviewer comments 4
Editor who approved publication: Dr Franco Musio
Yan Liang,1 Qi Zhang,1 Jing-Rong Qian,2 Sha-Sha Li,3 Qi-Feng Liu1,2
1Gusu School, Nanjing Medical University, The First People’s Hospital of Kunshan, Kunshan, Jiangsu, 215300, People’s Republic of China; 2Department of Nephrology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, 215300, People’s Republic of China; 3Clinical Research and Laboratory Centre, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, 215300, People’s Republic of China
Correspondence: Sha-Sha Li, Email [email protected] Qi-Feng Liu, Email [email protected]
Abstract: Chronic kidney disease (CKD) is influenced by inflammation, a critical factor in its progression. However, the underlying mechanism through which inflammation contributes to CKD is still obscure. The Klotho protein, which is predominantly found in the kidneys, is known for its protective functions, including anti-inflammatory, anti-aging, antioxidant, and anti-fibrotic effects. A myriad of studies have suggested that inflammation in CKD leads to a decrease in Klotho expression, diminishing Klotho protection capabilities and exacerbating kidney damage, thereby promoting CKD progression. These findings suggest that Klotho deficiency could be a crucial link between inflammation and CKD progression. However, the mechanism regarding their relationship is still unclear. The reduction in Klotho due to inflammation may be attributed to epigenetic mechanisms, such as DNA methylation, histone deacetylation, transcription factor, microRNA (miRNA) regulation and long non-coding RNA (lncRNA) regulation or non-epigenetic factors, such as endoplasmic reticulum (ER) stress and ER-associated degradation (ERAD), which affect Klotho protein metabolism. Through these pathways, inflammation triggers a decrease in Klotho expression, further driving CKD progression. Notably, Klotho also exerts a strong anti-inflammatory effect by inhibiting key inflammatory factors and pathways, suggesting that there is intricate crosstalk between inflammatory factors and Klotho in CKD. This review highlights how inflammation suppresses the expression of Klotho and further contributes to the development and exacerbation of CKD. By focusing on the interplay between inflammation and Klotho, the present review provides novel potential therapeutic strategies such as correcting epigenetic and non-epigenetic abnormalities for treating CKD by targeting this specific axis.
Keywords: inflammation, Klotho, chronic kidney disease, epigenetics, non-epigenetics, kidney fibrosis
Introduction
Recent epidemiological surveys have suggested that chronic kidney disease (CKD) affects 13.4% of the global population and 8.2% of China as of early 2019.1 CKD-associated prevalence rate and mortality remain high, underscoring the critical need to improve patient outcomes.2 A consistent, low-grade inflammatory response is a central characteristic of CKD and plays a key role in its progression as well as in the transition from acute kidney injury (AKI) to CKD.3,4 Systemic or localized inflammatory responses contribute to increased oxidative stress, which damages the intrinsic cells of the kidneys.5 Inflammation also causes endothelial dysfunction, exacerbating renal ischemia.6 Furthermore, it induces the trans-differentiation of tubular cells into myofibroblasts, leading to enhanced collagen deposition.7 As a result, the persistent inflammatory response ultimately results in renal fibrosis and a decline in kidney function. Thus, anti-inflammatory therapies are essential for delaying CKD progression and improving patient prognosis.8,9 However, the specific mechanisms by which inflammation accelerates CKD progression are complex and not yet completely understood. Consequently, gaining deeper insights into these inflammatory pathways could provide potential strategies for the prevention and treatment of CKD.
Klotho protein was originally identified as an anti-aging protein10 and exists in three isoforms: α-Klotho, β-Klotho, and γ-Klotho, with α-Klotho being predominantly synthesized in the kidneys.11 Klotho occurs in two forms: membrane-bound Klotho (mKlotho) and soluble Klotho (sKlotho). mKlotho is a transmembrane protein featuring a long extracellular region that contains two internal repeats, KL1 and KL2. These repeats can be cleaved by proteases such as A Disintegrin and Metalloproteinase (ADAM) 10 and ADAM 17, producing the functionally active circulating form, sKlotho.12 mKlotho is highly expressed in the kidneys and forms a complex with fibroblast growth factor (FGF) receptors, acting as a coreceptor for FGF-23 and mediating phosphate excretion.13 In contrast, sKlotho functions as an endocrine substance, participating in both FGF-23-dependent and -independent processes.14 Several studies have shown that levels of sKlotho decline in CKD8,15–22 (Table 1), indicating a negative correlation between sKlotho levels and kidney function. Moreover, the downregulation of sKlotho is associated with adverse outcomes, such as creatinine doubling, renal replacement therapy or death during the CKD progression21,23–30(Table 2).
![]() |
Table 1 Characteristics of the Clinical Studies Regarding the Relationship of sKlotho with Kidney Function |
![]() |
Table 2 Characteristics of the Clinical Studies Regarding the Relationship of sKlotho with Adverse Clinical Outcomes in CKD |
The Klotho protein, which is predominantly detected in the kidneys, is critical for several protective functions, such as anti-inflammatory, anti-aging, antioxidant, anti-fibrotic,31 anti-apoptotic32 and regulating calcium-phosphorus metabolism.15,33 Previous studies have demonstrated a significant reduction in sKlotho expression in individuals with CKD, which is linked to disease progression and adverse outcomes.23,34 Thus, sKlotho deficiency plays an important role in CKD pathogenesis and development and is emerging as a potential target for CKD management nowadays.35 The downregulation of Klotho is driven by a variety of factors, including both epigenetic mechanisms and non-epigenetic mechanisms.36 A key contributor to this decrease in Klotho levels is abnormal epigenetic regulation, including DNA methylation, histone deacetylation, transcription factor activity, microRNA (miRNA) and long non-coding RNA (lncRNA) activity, which inhibits Klotho synthesis.
Clinical studies have shown that reduced sKlotho levels in the general population are associated with an increase in inflammatory biomarkers such as uric acid, C-reactive protein (CRP), and white blood cell count.37 In CKD patients, an inverse relationship between inflammation and sKlotho was observed as well, suggesting a significant interplay where inflammation may lead to reduced sKlotho levels.33,38 Animal studies on kidney diseases have shown that inflammatory biomarkers can decrease mKlotho expression through various mechanisms.39 Critically, the downregulation of Klotho aggravates kidney damage and accelerates CKD progression in the presence of chronic inflammation.40,41
Indeed, enhanced inflammatory responses and Klotho deficiency commonly occur together in CKD, suggesting their potential role in CKD pathogenesis and progression. These findings indicate that the inflammation-mediated suppression of Klotho may be a novel mechanism through which inflammation contributes to CKD progression. However, the underlying mechanisms are not yet fully understood. Understanding how inflammation leads to Klotho downregulation is crucial for developing novel therapeutic approaches for CKD. Increasing evidence suggests that inflammation can significantly reduce Klotho transcription through epigenetic mechanisms and influence Klotho protein degradation via non-epigenetic pathways, ultimately playing a role in CKD development and progression, which may not have been covered comprehensively in existing reviews. Thus, in this review, we summarized the potential mechanisms involved in the downregulation of Klotho associated with inflammation and we aimed to provide a theoretical basis for better CKD management by targeting the inflammation-Klotho axis in the future.
The Epigenetic Mechanisms Through Which Inflammation Suppresses Klotho Expression
Epigenetic regulation involves hereditary changes in gene expression without alterations to the DNA sequence. These alterations include DNA methylation, histone modifications, transcription factor interactions, non-coding RNA interactions, RNA modifications, and chromatin remodeling. A myriad of studies have shown that abnormal epigenetic mechanisms play a significant role in reducing Klotho transcription activity, which is a key factor contributing to decreased Klotho expression.42 Inflammation can inhibit Klotho expression by targeting various epigenetic processes, ultimately impacting gene regulation and contributing to disease progression.43
DNA Methylation of Klotho
DNA methylation involves the addition of a methyl group to the 5′-carbon of the cytosine residue in the CpG dinucleotide, a process facilitated by DNA methyltransferase (DNMT), leading to gene silencing. Previous research has shown that the Klotho gene promoter and first exon region are rich in CpG islands. Unlike regions with TATA or CAAT boxes, the Klotho promoter contains five GC boxes, making it susceptible to high levels of methylations.44 Related studies have indicated that the inflammation can lead to telomere shortening, which is often associated with increased DNA methylation.45 Additionally, inflammatory factors such as CRP have been shown to directly promote DNA methylation,46 an effect that can be reversed through anti-inflammatory treatment.47 Therefore, inflammation is not only positively correlated with DNA methylation but also serves as an inducer of this epigenetic modification.
A cross-sectional study revealed that the level of sKlotho protein in the peripheral blood of cardiovascular disease patients was inversely correlated with that of tumor necrosis factor-α (TNF-α), and pro-inflammatory markers were directly connected to DNMT1 gene expression. Furthermore, the observed downregulation of sKlotho expression was negatively associated with DNMT1-mediated hypermethylation in the Klotho promoter region.48 These findings suggest that vascular inflammation associated with atherosclerosis may cause Klotho promoter methylation, leading to reduced sKlotho expression. Further animal studies have demonstrated that in fibrotic kidney tissues, TGF-β promotes the methylation of Klotho in renal tubular epithelial cells, thereby inhibiting mKlotho expression.49 Thus, elevated levels of inflammatory cytokines may lead to increased methylation of Klotho in renal tissues, resulting in reduced Klotho expression.
DNMTs are critical enzymes responsible for DNA methylation, with DNMT1 playing a significant role in regulating the methylation of the Klotho gene. Several studies have demonstrated that inflammation can influence DNMT1 expression through multiple mechanisms, subsequently affecting the methylation of the Klotho gene. First, inflammation can activate NF-κB, which further binds to the DNMT1 promoter region, increasing DNMT1 expression.50–52 This increase in DNMT1 activity results in increased methylation levels of Klotho. Moreover, inflammatory cytokines such as IL-1β and TNF-α can upregulate miRNAs such as miR-29C, miR-26B, and miR-20A, which in turn inhibit the methylcytosine dioxygenase gene, further increasing DNMT activity.53 Furthermore, the inflammatory marker C-C chemokine ligand 5 (CCL5) can activate signal transducer and activator of transcription 3 (STAT3), promoting downstream DNMT1 transcription and accelerating Klotho promoter hypermethylation.54
Thus, inflammation may target DNMT1-mediated DNA methylation to suppress Klotho transcription, leading to reduced Klotho expression and the progression of CKD (Figure 1). Additionally, the role of DNA demethylases may be significant in Klotho deficiency, particularly in the context of inflammation. However, further investigation is needed, as there are currently very few studies exploring this relationship.
![]() |
Figure 1 Roles of DNA methylation and miRNA in inflammation-induced Klotho deficiency. By Figdraw. |
Histone Deacetylation
Histone deacetylation is a process mediated by histone deacetylase (HDAC), which removes acetyl groups from lysine residues on core histones, restoring the positive charge of the histone. This positively charged histone binds more tightly to the negatively charged DNA, causing chromatin condensation. This condensation blocks the transcription factor access and subsequently inhibits gene transcription. The kidneys are particularly rich in acetylated lysine protein,55 and histone deacetylation has been shown to promote the development of renal interstitial fibrosis in CKD.56,57 Inflammatory processes can activate histone deacetylation.58,59 For example, TNF-α and IL-1β can recruit HDAC2 to gene promoter sites, thereby activating histone deacetylation.60 Research has shown that HDAC8 inhibitors can reverse mKlotho reduction induced by unilateral ureteral obstruction (UUO)61 and that HDAC3 can promote histone deacetylation at the Klotho promoter, inhibiting Klotho transcription and aggravating kidney damage.62 These findings suggest that inflammation can activate histone deacetylation to regulate the mKlotho expression. Animal experiments further confirmed that TNF-like weak inducer of apoptosis can promote the binding of the NF-κB family protein RelA to the mouse Klotho promoter, enhancing histone H3 and H4 deacetylation and suppressing Klotho transcription.39 Additionally, inflammation can activate HDAC3 via the TGF-β/Smad signaling pathway. HDAC3 forms a repressive complex with the nuclear receptor inhibitor protein/NF-κB that binds to the Klotho promoter, leading to histone deacetylation and gene transcription suppression.62 Therefore, inflammation may downregulate Klotho transcription by activating histone deacetylation, thereby inhibiting the renoprotective effect of Klotho, which is linked to CKD progression (Figure 2). Similarly, histone acetyl transferases (HATs) are also involved in Klotho regulation. P300-mediated histone acetylation has been shown to suppress Klotho expression through regulating Akt signaling pathway.63 Yet it remains unclear whether HATs also contribute to the regulation of Klotho deficiency induced by inflammation and further research is needed to clarify this relationship.
![]() |
Figure 2 Roles of histone deacetylation and transcription factors in Klotho regulation. By Figdraw. |
Transcription Factors
Transcription factors influence the initiation and extension of gene expression, regulating it by binding to enhancer elements and recruiting co-activators and RNA polymerase II to target genes. The Klotho promoter region contains binding sites for multiple transcription factors, which means that Klotho expression is regulated by these factors.64 These transcription factors can directly bind to their specific sites on the Klotho promoter to regulate its expression. Additionally, they can indirectly modulate Klotho transcription through other mechanisms, thereby affecting its expression levels.
Direct Regulation of Klotho Expression by Transcription Factors That Bind to the Klotho Promoter
Certain transcription factors, such as specificity protein 1 (Sp1) and peroxisome proliferator-activated receptor-γ (PPAR-γ), can directly bind to the Klotho promoter to regulate its expression. The Sp family of transcription factors typically functions as transcription activator. For example, Sp1 can bind to GC-rich sequences in the regulatory regions of genes, preventing them from undergoing methylation and activating transcription.65 The Klotho promoter region contains five GC boxes that serve as binding sites for Sp1. This interaction can activate gene transcription and increase Klotho expression. Studies have demonstrated that the overexpression of Sp1 not only induces mKlotho expression but also mitigates renal fibrosis, suggesting the involvement of Sp1 in mKlotho regulation.66 Under inflammatory conditions, such as lipopolysaccharide (LPS)-induced damage, LPS can activate NF-κB, which in turn increases the activity of Sp1-degrading enzymes.67 This may lead to a further degradation of Sp1 and a subsequent reduction in Klotho expression. These findings suggest that inflammation-induced Klotho suppression may be partly mediated through Sp1 modulation (Figure 1).
PPAR-γ, a member of the ligand-activated nuclear hormone receptor and transcription factor family, plays a significant role in regulating Klotho expression. Studies have shown that a functional non-classical PPAR response element sequence located in the 5′-flank region of the Klotho gene can be recognized by PPAR-γ, thereby promoting Klotho transcription.68 However, inflammation can reduce PPAR-γ expression, subsequently affecting mKlotho levels. Some studies have shown that inflammation can lead to PPAR-γ deacetylation, which weakens its binding to the Klotho promoter, inhibiting Klotho transcription and expression69 (Figure 2).
Other Mechanisms by Which Transcription Factors Affect Klotho Expression
Transcription factors, such as STATs, can influence Klotho expression through various pathways, in addition to directly binding to promoters. STATs, which are activated by phosphorylation, regulate gene transcription and are implicated in CKD progression.70,71 When activated in mesangial cells, the JAK/STAT3 pathway promotes renal fibrosis by increasing TGF-β1, IV collagen and fibronectin production.72 Furthermore, phosphorylated STAT3 (p-STAT3) binds to the DNMT1 promoter, inducing DNMT1 expression and affecting Klotho transcription. Inflammation triggers STAT activation,73 and our previous studies revealed that, in CKD, the inflammatory factor CCL5 activates p-STAT3/DNMT1, leading to Klotho methylation and reduced expression54 (Figure 1).
Kelch-like ECH-associated protein 1 (Keap1) and nuclear factor erythroid 2- related factor 2 (Nrf2) play critical roles in anti-inflammatory and antioxidant responses. Normally, Keap1 binds Nrf2, leading to its ubiquitination and degradation. However, during inflammation, Keap1 undergoes sulfhydryl modification, releasing Nrf2, which then translocates to the nucleus to exert anti-inflammatory effects. Nrf2 inhibits NF-κB by upregulating heme oxygenase-1 and blocking NF-κB inhibitory protein α phosphorylation. Additionally, Nrf2 competes with NF-κB for CREB binding proteins, preventing Klotho methylation and histone deacetylation.74,75 Studies have shown that in CKD, chronic inflammation can upregulate Keap1, enhancing Keap1-Nrf2 binding and Nrf2 degradation,76 ultimately reducing Klotho transcription through NF-κB-dependent epigenetic mechanisms and promoting CKD progression (Figure 2).
miRNAs
miRNA is an endogenous, non-coding small RNA that can directly bind to the 3’UTR of the target mRNA, negatively regulating gene expression.77 In CKD, inflammation leads to the upregulation of certain miRNAs, such as miR-29-5p, miR-21-5p and miR-196a-5p,78 indicating that inflammatory responses can increase the expression of miRNAs. Notably, the inflammatory factor TGF-β1 activates p53/Smad3 signaling, resulting in increased miR-34a expression, which in turn reduces Klotho expression by directly binding to its 3′UTR.79,80 Furthermore, TGF-β suppresses the expression of the miR-152 and miR-30 families. The 3′UTRs of DNMT1 and DNMT3a mRNAs contain binding sequences for these miRNA families. Inflammation can prevent miRNA from binding to the 3′UTR of DNMT mRNA, increasing the post-transcriptional translation of DNMT, promoting hypermethylation of the Klotho gene and a subsequent decrease in Klotho expression.80 In summary, the targeted regulation of miRNAs may be another epigenetic mechanism by which inflammation modulates Klotho expression, contributing to CKD progression (Figure 1).
lncRNAs
LncRNA represent a large and diverse class of transcripts longer than 200 nucleotides, involved in gene transcription and post-transcriptional regulation.81,82 Recent studies have identified lncRNAs as potential biomarkers for CKD.83 For instance, lncRNA TCONS_00088786 has been shown to promote renal interstitial fibrosis through the regulation of miR-132.84 Conversely, the knockdown of lncRNA-ATB can inhibit the progression of renal fibrosis in vitro,85 indicating that lncRNAs play a significant role in CKD progression. Furthermore, available studies have revealed that inflammation can upregulate lncRNA expression.86 In high glucose-induced human renal glomerular endothelial cells (HRGECs), both lncRNA MALAT1 and inflammatory markers, such as IL-6 and TNF-α, were found to be elevated. This phenomenon has also been observed in the vitreous humor of diabetic patients.87 Inflammation serves as a positive inducer of lncRNAs, where MALAT1 can recruit the methyltransferase G9a to increase H3K9me1 levels, subsequently binding to the Klotho promoter and epigenetically inhibiting mKlotho expression, leading to HRGEC injury.88 Thus, inflammation promotes lncRNA interactions with the Klotho promoter, resulting in reduced mKlotho expression. Additionally, some lncRNAs can interact with miRNAs to decrease the levels of fibrotic or inflammatory miRNAs, thereby functioning in an anti-inflammatory and anti-fibrotic capacity.89 However, the precise relationship between these lncRNAs and Klotho remains unclear. In summary, the regulation of lncRNAs that target the Klotho promoter may represent one of the mechanisms by which inflammation suppresses Klotho expression, thereby exacerbating the progression of CKD.
Non-Epigenetic Mechanisms by Which Inflammation Inhibits Klotho Expression
Inflammation can suppress Klotho expression via several epigenetic mechanisms, such as DNA methylation, histone deacetylation, transcription factor expression, miRNA and lncRNA expression. However, in addition to these epigenetic mechanisms, inflammation can also inhibit Klotho expression via non-epigenetic mechanisms, which include endoplasmic reticulum (ER) stress and ER-associated degradation (ERAD).
ER Stress and ERAD
The ER is a primary organelle responsible for protein synthesis, folding, and post-translational modification. CKD is often associated with increased ER stress, which plays a critical role in its progression.90 While ER stress can help restore ER homeostasis, persistent ER stress, which is commonly observed in CKD, disrupts protein folding and post-translational modifications, leading to the accumulation of unfolded or misfolded proteins in the ER and preventing proper protein maturation.91 Studies have shown that inhibiting ER stress increases mKlotho protein levels without altering Klotho mRNA levels.92 Additionally, the inhibition of ER stress has been demonstrated to restore mKlotho expression and ameliorate AKI.93 These findings suggest that ER stress may promote the degradation of the mKlotho protein. Given that inflammation can trigger ER stress, the mechanisms by which inflammation suppresses mKlotho expression may involve ER stress94 (Figure 3).
![]() |
Figure 3 Roles of ER stress and ERAD in inflammation-induced Klotho deficiency. By Figdraw. |
ERAD and the unfolded protein response (UPR) are two critical pathways activated by ER stress. When unfolded or misfolded proteins accumulate in the ER, both ERAD and UPR are triggered to restore ER homeostasis.95 If the UPR fails to restore ER homeostasis, ERAD is activated to ubiquitinate and degrade these unfolded or misfolded proteins.96 Our previous research involved a mouse model of renal interstitial fibrosis induced by UUO and revealed that blocking ERAD with Eeyarestatin I significantly restored mKlotho expression and alleviated kidney fibrosis, suggesting that mKlotho may be a potential substrate of ERAD.97 Therefore, inflammation may degrade mKlotho by inducing ER stress and activating ERAD, contributing to further kidney damage (Figure 3).
Other Non-Epigenetic Mechanisms
Other non-epigenetic mechanisms, such as the renin-angiotensin-aldosterone system (RAAS) and mineral imbalances, influence Klotho regulation. Inflammation can activate the RAAS, which in turn increases the expression of pro-inflammatory cytokines.98 The activated RAAS is known to suppress sKlotho expression, whereas blocking the RAAS can help restore sKlotho levels in CKD.21,99 Additionally, RAAS activation is associated with triggering ER stress, which can be alleviated via the use of RAAS inhibitors.100 Inflammation can also impact vitamin D regulation, leading to decreased Klotho expression; however, the underlying mechanism is not yet fully understood and requires further research.101–103
Mechanisms by Which Klotho Inhibits the Inflammatory Response
Klotho protein also exhibits strong anti-inflammatory properties by inhibiting key inflammatory factors and pathways. In eukaryotic cells, members of the NF-κB family exist as dimers in the cytoplasm. IκB acts as an NF-κB dimer inhibitor by binding to NF-κB, thereby preventing its translocation to the nucleus and keeping it inactive in the cytoplasm. Klotho inhibits inflammation by modulating the NF-κB signaling pathway.
First, Klotho prevents NF-κB pathway activation via the inhibition of IκB phosphorylation. Both in vitro and in vivo, Klotho reduces phosphorylated IκB levels and consequently lowers inflammatory factor levels.104 Second, Klotho also inhibits the non-classical NF-κB pathway via RelA-Ser536 phosphorylation, preventing NF-κB from binding to target gene promoters and thereby inhibiting the inflammatory response.105 Additionally, Klotho upregulates the expression of PDZ and LIM domain-containing proteins, which function as ubiquitin E3 ligases. These proteins target the NF-κB p65 subunit, promoting its ubiquitination and degradation, which in turn inhibits NF-κB activation.106
Furthermore, sKlotho exhibits glycoside activity that inhibits downstream inflammatory signals by deglycosylation of toll-like receptor 4.107 mKlotho also reduces CKD-associated systemic inflammation by suppressing the RIG-I signaling pathway and monocyte activation.41 Studies have reported that Klotho promotes the polarization of anti-inflammatory M2 macrophages, mitigating inflammation induced by indoxyl sulfate and resulting in reduced levels of urine, serum urea nitrogen, and creatinine.108,109 Finally, the anti-inflammatory effects of Klotho may also involve modulation of the Wnt/β-catenin signaling pathway.110 Overall, Klotho can effectively suppress inflammation and potentially improve CKD outcomes by targeting various inflammatory pathways (Figure 4).
![]() |
Figure 4 Anti-inflammatory actions of Klotho. By Figdraw. |
Taken together, a complex network exists between inflammation and Klotho in CKD. Inflammation may suppress Klotho expression via both epigenetic mechanisms, such as DNA methylation, histone deacetylation, transcription factors, miRNAs and lncRNAs, and non-epigenetic mechanisms, including ER stress and ERAD. This suppression of Klotho compromises its protective effects, thereby aggravating inflammation and kidney function in CKD and promoting its progression. This establishes a vicious cycle between inflammation and Klotho, underscoring the critical role of the inflammation-Klotho axis in the initiation and progression of CKD.
Therapeutic Applications of the Inflammation-Klotho Axis
Recent research exploring the inflammation-Klotho axis has been on the rise. Numerous pharmacological agents have been shown to enhance Klotho expression through anti-inflammatory mechanisms. For instance, rapamycin has been demonstrated to reverse the decline in mKlotho and sKlotho levels by inhibiting inflammatory pathways such as mTOR signaling.111 The TNF-α inhibitor infliximab inactivates Wnt/β-catenin signaling by upregulating mKlotho, which helps suppress the progression of CKD.112 Animal studies indicate that baicalin can exert anti-inflammatory effects in diabetic kidneys and mitigate renal fibrosis by reversing Klotho suppression induced by DNA methylation.113 However, it remains to be determined whether this compound upregulates Klotho expression through the inhibition of DNA methylation in an anti-inflammatory context. Additionally, the selective class IIa HDAC inhibitor MC1568 has been shown to upregulate Klotho expression and may impede the initiation and progression of inflammation-induced renal fibrosis by suppressing NF-κB phosphorylation.114 As a key transcription factor, NF-κB regulates Klotho through multiple epigenetic mechanisms, suggesting that MC1568 may target NF-κB to counteract the inflammation-induced downregulation of Klotho. Current research on the epigenetic regulation of Klotho expression through anti-inflammatory mechanisms is still limited, and this study provides novel insights into potential therapeutic strategies in this underexplored area.
Furthermore, several interventions have been found to upregulate Klotho expression, thereby mediating anti-inflammatory effects. For example, pentoxifylline has been shown to ameliorate inflammatory responses during the atherosclerotic process by increasing sKlotho levels.8 Recombinant human α-Klotho used as an adjunctive therapy to telmisartan has demonstrated anti-inflammatory effects, thereby attenuating diabetic kidney disease.115 Additionally, novel therapeutic targets related to the Klotho gene have been identified. Mutations in the Klotho gene may be linked to severe systemic inflammatory responses.116 Clinical trials have shown that the KL-VS mutation of the Klotho gene is associated with lower serum levels of inflammatory markers, such as CRP and TNF-α.117 Moreover, delivering Klotho-expressing plasmid DNA via stem cell-homing hydrogels has been found to alleviate osteoarthritis.118 Therefore, targeting Klotho gene interventions may offer innovative strategies for combating inflammation.
Conclusion
Overall, Klotho deficiency, particularly under inflammatory conditions, is a significant factor in CKD. The suppression of Klotho by inflammation involves various epigenetic and non-epigenetic mechanisms, which may represent a novel mechanism by which inflammation promotes CKD progression. Conversely, Klotho exerts a protective effect by inhibiting key inflammatory factors and pathways, thereby reducing inflammation. This interplay forms a closed-loop relationship, suggesting that targeting the inflammation-Klotho axis could serve as a potential therapeutic strategy for CKD. Existing pharmacological therapies and genetic interventions targeting this axis have been reported, while the approaches remain relatively limited in scope. Understanding the regulatory dynamics between inflammation and Klotho could provide new insights for managing inflammation, regulating Klotho expression, and ultimately preventing and treating CKD. Although anti-inflammatory therapy has been shown to effectively protect kidney function and improve prognosis, the therapeutic effects of enhancing Klotho by targeting Klotho regulators in clinical settings have not yet been investigated under the inflammatory conditions during CKD. Therefore, translational research is needed to evaluate and confirm the clinical significance of the inflammation-Klotho axis in the CKD population in the future.
Acknowledgments
This work was funded by (i) “National Tutorial System” Training Program for Key Young Health Professionals in Suzhou (Grant No.Qngg2023048); (ii) Medical Research Project, Jiangsu Provincial Health Commission (Grant No.Z2023039); (iii) Science and Technology Development Plan Project: Investigating Applications of Medical Innovations, Suzhou (Grant No.SKY2023095); (iV) Medical Education Collaborative Innovation Fund, Jiangsu University (Grant No.JDY2023016). All figures were created by Figdraw (https://www.figdraw.com) (ID: WPIOU242d2; OWUUO7aba6; USSSUa1baf; PAAPU25552).
Disclosure
The authors report no conflicts of interest in this work.
References
1. Wang L, Xu X, Zhang M, et al. Prevalence of chronic kidney disease in china: results from the sixth china chronic disease and risk factor surveillance. JAMA Intern Med. 2023;183(4):298–310. doi:10.1001/jamainternmed.2022.6817
2. Dong B, Zhao Y, Wang J, et al. Epidemiological analysis of chronic kidney disease from 1990 to 2019 and predictions to 2030 by Bayesian age-period-cohort analysis. Ren Fail. 2024;46(2):2403645. doi:10.1080/0886022X.2024.2403645
3. Yan Z, Shao T. Chronic inflammation in chronic kidney disease. Nephron Clin Pract. 2024;148(3):143–151.
4. Yeh TH, Tu KC, Wang HY, et al. From acute to chronic: unraveling the pathophysiological mechanisms of the progression from acute kidney injury to acute kidney disease to chronic kidney disease. Int J Mol Sci. 2024;25(3):1755. doi:10.3390/ijms25031755
5. Ma T, Li X, Zhu Y, et al. Excessive activation of notch signaling in macrophages promote kidney inflammation, fibrosis, and necroptosis. Front Immunol. 2022;13:835879. doi:10.3389/fimmu.2022.835879
6. Jourde-Chiche N, Fakhouri F, Dou L, et al. Endothelium structure and function in kidney health and disease. Nat Rev Nephrol. 2019;15(2):87–108. doi:10.1038/s41581-018-0098-z
7. Li M, Yan Y, He J, et al. Jolkinolide B alleviates renal fibrosis via anti-inflammation and inhibition of epithelial-mesenchymal transition in unilateral ureteral obstruction mice. J Asian Nat Prod Res. 2022;24(1):76–87. doi:10.1080/10286020.2021.2016715
8. Donate-Correa J, Ferri CM, Mora-Fernandez C, et al. Pentoxifylline ameliorates subclinical atherosclerosis progression in patients with type 2 diabetes and chronic kidney disease: a randomized pilot trial. Cardiovasc Diabetol. 2024;23(1):314. doi:10.1186/s12933-024-02393-x
9. Perez-Villalobos MC, Barba-Gonzalez A, Garcia-Carrillo N, et al. Nephroprotective effect of pioglitazone in a Wistar rat model of adenine-induced chronic kidney disease. Exp Ther Med. 2024;28(4):392. doi:10.3892/etm.2024.12681
10. Kuro-o M, Matsumura Y, Aizawa H, et al. Mutation of the mouse klotho gene leads to a syndrome resembling ageing. Nature. 1997;390(6655):45–51. doi:10.1038/36285
11. Kuro-O M. The Klotho proteins in health and disease. Nat Rev Nephrol. 2019;15(1):27–44. doi:10.1038/s41581-018-0078-3
12. Chen CD, Podvin S, Gillespie E, et al. Insulin stimulates the cleavage and release of the extracellular domain of Klotho by ADAM10 and ADAM17. Proc Natl Acad Sci U S A. 2007;104(50):19796–19801. doi:10.1073/pnas.0709805104
13. Chen G, Liu Y, Goetz R, et al. alpha-Klotho is a non-enzymatic molecular scaffold for FGF23 hormone signalling. Nature. 2018;553(7689):461–466. doi:10.1038/nature25451
14. Hu MC, Kuro-o M, Moe OW. Klotho and kidney disease. J Nephrol. 2010;23 Suppl 16(Suppl 16):S136–S144.
15. Hu MC, Shi M, Zhang J, et al. Klotho deficiency causes vascular calcification in chronic kidney disease. J Am Soc Nephrol. 2011;22(1):124–136. doi:10.1681/ASN.2009121311
16. Akimoto T, Yoshizawa H, Watanabe Y, et al. Characteristics of urinary and serum soluble Klotho protein in patients with different degrees of chronic kidney disease. BMC Nephrol. 2012;13:155. doi:10.1186/1471-2369-13-155
17. Martin-Virgala J, Fernandez-Villabrille S, Martin-Carro B, et al. Serum and urinary soluble alpha-klotho as markers of kidney and vascular impairment. Nutrients. 2023;15(6):1470. doi:10.3390/nu15061470
18. de Araujo TB, de Luca CH, de Deus LA, et al. The effects of home-based progressive resistance training in chronic kidney disease patients. Exp Gerontol. 2023;171:112030. doi:10.1016/j.exger.2022.112030
19. Iyengar A, Kamath N, V RH, et al. Determining the optimal cholecalciferol dosing regimen in children with CKD: a randomized controlled trial. Nephrol Dial Transplant. 2022;37(2):326–334. doi:10.1093/ndt/gfaa369
20. Liu J, Wang H, Liu Q, et al. Klotho exerts protection in chronic kidney disease associated with regulating inflammatory response and lipid metabolism. Cell Biosci. 2024;14(1):46. doi:10.1186/s13578-024-01226-4
21. Qian J, Zhong J, Yan M, et al. Circulating alpha-Klotho is related to plasma aldosterone and its follow-up change predicts CKD progression. Kidney Blood Press Res. 2018;43(3):836–846. doi:10.1159/000490138
22. Rotondi S, Pasquali M, Tartaglione L, et al. Soluble alpha-Klotho serum levels in chronic kidney disease. Int J Endocrinol. 2015;2015:872193. doi:10.1155/2015/872193
23. Han S, Zhang X, Wang X, et al. Association between serum Klotho and all-cause mortality in chronic kidney disease: evidence from a prospective cohort study. Am J Nephrol. 2024;55(3):273–283. doi:10.1159/000535808
24. Liu L, Jia J, Cheng X, et al. The optimal cut-off values of Klotho for predicting all-cause and cardiovascular mortality among chronic kidney disease: results from NHANES. Sci Rep. 2024;14(1):4647. doi:10.1038/s41598-024-52701-4
25. Milovanova LY, Nezhdanov KS, Milovanova SY, et al. alpha-Klotho is associated with cardiovascular and all-cause mortality in patients with stage 3b and 4 chronic kidney disease (CKD): a long-term prospective cohort study. J Nephrol. 2024;38:171–179. doi:10.1007/s40620-024-02069-5
26. Edmonston D, Fuchs M, J BE, et al. Klotho and clinical outcomes in CKD: findings from the chronic renal insufficiency cohort (CRIC) study. Am J Kidney Dis. 2024;84(3):349–360. doi:10.1053/j.ajkd.2024.02.008
27. Gao Y, J ZC, Liu Q, et al. Relationship between serum indoxyl sulfate and Klotho protein and vascular calcification in patients with chronic kidney disease stages 3-5. Int J Endocrinol. 2024;2024:8229604. doi:10.1155/2024/8229604
28. Li F, Ye X, Yang G, et al. Relationships between blood bone metabolic biomarkers and anemia in patients with chronic kidney disease. Ren Fail. 2023;45(1):2210227. doi:10.1080/0886022X.2023.2210227
29. Martins AR, Azeredo-Lopes S, Pereira SA, et al. Klotho and lean mass as novel cardiovascular risk factors in hemodialysis patients. Clin Kidney J. 2023;16(12):2587–2596. doi:10.1093/ckj/sfad166
30. Manou E, Thodis E, Arsos G, et al. Fibroblast growth factor 23 and alpha-Klotho protein are associated with adverse clinical outcomes in non-dialysis CKD patients. Kidney Blood Press Res. 2020;45(6):900–915. doi:10.1159/000510351
31. Doi S, Zou Y, Togao O, et al. Klotho inhibits transforming growth factor-beta1 (TGF-beta1) signaling and suppresses renal fibrosis and cancer metastasis in mice. J Biol Chem. 2011;286(10):8655–8665. doi:10.1074/jbc.M110.174037
32. Neyra JA, Hu MC, Moe OW. Klotho in clinical nephrology: diagnostic and therapeutic implications. Clin J Am Soc Nephrol. 2020;16(1):162–176. doi:10.2215/CJN.02840320
33. Sakan H, Nakatani K, Asai O, et al. Reduced renal alpha-Klotho expression in CKD patients and its effect on renal phosphate handling and vitamin D metabolism. PLoS One. 2014;9(1):e86301. doi:10.1371/journal.pone.0086301
34. Zhang Z, Zhou X, Deng L, et al. The association between serum soluble Klotho and chronic kidney disease among us adults ages 40 to 79 years: cross-sectional study. Front Public Health. 2022;10:995314. doi:10.3389/fpubh.2022.995314
35. Kanbay M, Brinza C, Ozbek L, et al. The association between klotho and kidney and cardiovascular outcomes: a comprehensive systematic review and meta-analysis. Clin Kidney J. 2024;17(9):sfae255. doi:10.1093/ckj/sfae255
36. S LS, J SM, Y SZ, et al. Upstream and downstream regulators of Klotho expression in chronic kidney disease. Metabolism. 2023;142:155530. doi:10.1016/j.metabol.2023.155530
37. Wu SE, Chen WL. Soluble klotho as an effective biomarker to characterize inflammatory states. Ann Med. 2022;54(1):1520–1529. doi:10.1080/07853890.2022.2077428
38. Jena S, Sarangi P, Das UK, et al. Serum alpha-Klotho protein can be an independent predictive marker of oxidative stress (OS) and declining glomerular function rate in chronic kidney disease (CKD) patients. Cureus. 2022;14(6):e25759. doi:10.7759/cureus.25759
39. A MJ, C IM, D S-NM, et al. The inflammatory cytokines TWEAK and TNFalpha reduce renal klotho expression through NFkappaB. J Am Soc Nephrol. 2011;22(7):1315–1325. doi:10.1681/ASN.2010101073
40. Lai L, Li Y, Liu J, et al. Bovine serum albumin aggravates macrophage M1 activation and kidney injury in heterozygous Klotho-deficient mice via the gut microbiota-immune axis. Int J Biol Sci. 2021;17(3):742–755. doi:10.7150/ijbs.56424
41. Zeng Y, H WP, Zhang M, et al. Aging-related renal injury and inflammation are associated with downregulation of Klotho and induction of RIG-I/NF-kappaB signaling pathway in senescence-accelerated mice. Aging Clin Exp Res. 2016;28(1):69–76. doi:10.1007/s40520-015-0371-y
42. Xia J, Cao W. Epigenetic modifications of Klotho expression in kidney diseases. J Mol Med. 2021;99(5):581–592. doi:10.1007/s00109-021-02044-8
43. Ogura Y, Mimura I. Epigenetic roles in clonal hematopoiesis and aging kidney-related chronic kidney disease. Front Cell Dev Biol. 2023;11:1281850. doi:10.3389/fcell.2023.1281850
44. Azuma M, Koyama D, Kikuchi J, et al. Promoter methylation confers kidney-specific expression of the Klotho gene. FASEB J. 2012;26(10):4264–4274. doi:10.1096/fj.12-211631
45. L SE, Ton R, Boner W, et al. Associations between DNA methylation and telomere length during early life: insight from wild zebra finches (Taeniopygia guttata). Mol Ecol. 2022;31(23):6261–6272. doi:10.1111/mec.16187
46. Wielscher M, R MP, Kuehnel B, et al. DNA methylation signature of chronic low-grade inflammation and its role in cardio-respiratory diseases. Nat Commun. 2022;13(1):2408. doi:10.1038/s41467-022-29792-6
47. K SH, Venkateswaran S, Kilaru V, et al. Blood-derived DNA methylation signatures of crohn’s disease and severity of intestinal inflammation. Gastroenterology. 2019;156(8):2254–2265. doi:10.1053/j.gastro.2019.01.270
48. Martin-Nunez E, Perez-Castro A, G TV, et al. Klotho expression in peripheral blood circulating cells is associated with vascular and systemic inflammation in atherosclerotic vascular disease. Sci Rep. 2022;12(1):8422. doi:10.1038/s41598-022-12548-z
49. Yin S, Zhang Q, Yang J, et al. TGFbeta-incurred epigenetic aberrations of miRNA and DNA methyltransferase suppress Klotho and potentiate renal fibrosis. Biochim Biophys Acta Mol Cell Res. 2017;1864(7):1207–1216. doi:10.1016/j.bbamcr.2017.03.002
50. Hong J, Li D, Wands J, et al. Role of NADPH oxidase NOX5-S, NF-κB, and DNMT1 in acid-induced p16 hypermethylation in Barrett’s cells. Am J Physiol Cell Physiol. 2013;305(10):C1069–C1079. doi:10.1152/ajpcell.00080.2013
51. G OF, Soria-Valles C, Santiago-Fernandez O, et al. NF-kappaB signaling as a driver of ageing. Int Rev Cell Mol Biol. 2016;326:133–174.
52. Taniguchi K, Karin M. NF-kappaB, inflammation, immunity and cancer: coming of age. Nat Rev Immunol. 2018;18(5):309–324. doi:10.1038/nri.2017.142
53. Takeshima H, Niwa T, Yamashita S, et al. TET repression and increased DNMT activity synergistically induce aberrant DNA methylation. J Clin Invest. 2020;130(10):5370–5379. doi:10.1172/JCI124070
54. Liu Q, Li S, Yu L, et al. CCL5 suppresses Klotho expression via p-STAT3/DNA methyltransferase1-mediated promoter hypermethylation. Front Physiol. 2022;13:856088. doi:10.3389/fphys.2022.856088
55. Hyndman KA. Histone deacetylases in kidney physiology and acute kidney injury. Semin Nephrol. 2020;40(2):138–147. doi:10.1016/j.semnephrol.2020.01.005
56. Liu H. The roles of histone deacetylases in kidney development and disease. Clin Exp Nephrol. 2021;25(3):215–223. doi:10.1007/s10157-020-01995-5
57. Pan S, Yuan T, Xia Y, et al. Role of histone modifications in kidney fibrosis. Medicina. 2024;60(6):888. doi:10.3390/medicina60060888
58. Lin Y, Qiu T, Wei G, et al. Role of histone post-translational modifications in inflammatory diseases. Front Immunol. 2022;13:852272. doi:10.3389/fimmu.2022.852272
59. Wang Y, Jiao B, Hu Z, et al. Critical Role of histone deacetylase 3 in the regulation of kidney inflammation and fibrosis. Kidney Int. 2024;105(4):775–790. doi:10.1016/j.kint.2024.01.010
60. Bedenbender K, Scheller N, Fischer S, et al. Inflammation-mediated deacetylation of the ribonuclease 1 promoter via histone deacetylase 2 in endothelial cells. FASEB J. 2019;33(8):9017–9029. doi:10.1096/fj.201900451R
61. Zhang Y, Zou J, Tolbert E, et al. Identification of histone deacetylase 8 as a novel therapeutic target for renal fibrosis. FASEB J. 2020;34(6):7295–7310. doi:10.1096/fj.201903254R
62. Chen F, Gao Q, Wei A, et al. Histone deacetylase 3 aberration inhibits Klotho transcription and promotes renal fibrosis. Cell Death Differ. 2021;28(3):1001–1012. doi:10.1038/s41418-020-00631-9
63. Li S, Kao Y, Chung C, et al. Activated p300 acetyltransferase activity modulates aortic valvular calcification with osteogenic transdifferentiation and downregulation of Klotho. Int J Cardiol. 2017;232:271–279. doi:10.1016/j.ijcard.2017.01.005
64. Jung D, Xu Y, Sun Z. Induction of anti-aging gene klotho with a small chemical compound that demethylates CpG islands. Oncotarget. 2017;8(29):46745–46755. doi:10.18632/oncotarget.18608
65. O’Connor L, Gilmour J, Bonifer C. The role of the ubiquitously expressed transcription factor Sp1 in tissue-specific transcriptional regulation and in disease. Yale J Biol Med. 2016;89(4):513–525.
66. Li Y, Liu Y, Wang K, et al. Klotho is regulated by transcription factor Sp1 in renal tubular epithelial cells. BMC Mol Cell Biol. 2020;21(1):45. doi:10.1186/s12860-020-00292-z
67. Ye X, Liu H, S GY, et al. LPS down-regulates specificity protein 1 activity by activating NF-kappaB pathway in endotoxemic mice. PLoS One. 2015;10(6):e130317.
68. Zhang H, Li Y, Fan Y, et al. Klotho is a target gene of PPAR-gamma. Kidney Int. 2008;74(6):732–739. doi:10.1038/ki.2008.244
69. Maquigussa E, Paterno JC, De Oliveira PG, et al. Klotho and PPAR Gamma Activation Mediate the Renoprotective Effect of Losartan in the 5/6 Nephrectomy Model. Front Physiol. 2018;9:1033. doi:10.3389/fphys.2018.01033
70. Bienaime F, Muorah M, Yammine L, et al. Stat3 controls tubulointerstitial communication during CKD. J Am Soc Nephrol. 2016;27(12):3690–3705. doi:10.1681/ASN.2015091014
71. Park JY, Yoo KD, Bae E, et al. Blockade of STAT3 signaling alleviates the progression of acute kidney injury to chronic kidney disease through antiapoptosis. Am J Physiol Renal Physiol. 2022;322(5):F553–F572. doi:10.1152/ajprenal.00595.2020
72. Zheng C, Huang L, Luo W, et al. Inhibition of STAT3 in tubular epithelial cells prevents kidney fibrosis and nephropathy in STZ-induced diabetic mice. Cell Death Dis. 2019;10(11):848. doi:10.1038/s41419-019-2085-0
73. Platanitis E, Decker T. Regulatory networks involving STATs, IRFs, and NFkappaB in inflammation. Front Immunol. 2018;9:2542. doi:10.3389/fimmu.2018.02542
74. M AS, Luo L, Namani A, et al. Nrf2 signaling pathway: pivotal roles in inflammation. Biochim Biophys Acta Mol Basis Dis. 2017;1863(2):585–597. doi:10.1016/j.bbadis.2016.11.005
75. Aranda-Rivera AK, Cruz-Gregorio A, Pedraza-Chaverri J, et al. Nrf2 activation in chronic kidney disease: promises and pitfalls. Antioxidants. 2022;11(6):1112.
76. Kim HJ, Vaziri ND. Contribution of impaired Nrf2-Keap1 pathway to oxidative stress and inflammation in chronic renal failure. Am J Physiol Renal Physiol. 2010;298(3):F662–F671. doi:10.1152/ajprenal.00421.2009
77. Correia DSM, Gjorgjieva M, Dolicka D, et al. Deciphering miRNAs’ action through miRNA editing. Int J Mol Sci. 2019;20(24):6249.
78. Donderski R, Szczepanek J, Naruszewicz N, et al. Analysis of profibrogenic microRNAs (miRNAs) expression in urine and serum of chronic kidney disease (CKD) stage 1-4 patients and their relationship with proteinuria and kidney function. Int Urol Nephrol. 2022;54(4):937–947. doi:10.1007/s11255-021-02928-1
79. Overstreet JM, Samarakoon R, Meldrum KK, et al. Redox control of p53 in the transcriptional regulation of TGF-beta1 target genes through SMAD cooperativity. Cell Signal. 2014;26(7):1427–1436. doi:10.1016/j.cellsig.2014.02.017
80. Liu Y, Bi X, Xiong J, et al. MicroRNA-34a promotes renal fibrosis by downregulation of Klotho in tubular epithelial cells. Mol. 2019;27(5):1051–1065.
81. Quinn JJ, Chang HY. Unique features of long non-coding RNA biogenesis and function. Nat Rev Genet. 2016;17(1):47–62. doi:10.1038/nrg.2015.10
82. Giannuzzi F, Maiullari S, Gesualdo L, et al. The mission of long non-coding RNAs in human adult renal stem/progenitor cells and renal diseases. Cells. 2023;12(8):1115. doi:10.3390/cells12081115
83. Zhao C, Hu J, Wang Z, et al. Serum LncRNA PANDAR may act as a novel serum biomarker of diabetic nephropathy in patients with type 2 diabetes. Clin Lab. 2020;66(6). doi:10.7754/Clin.Lab.2019.191032.
84. G ZS, Zhang W, J MH, et al. Silencing of LncRNA TCONS_00088786 reduces renal fibrosis through miR-132. Eur Rev Med Pharmacol Sci. 2018;22(1):166–173. doi:10.26355/eurrev_201801_14114
85. Zhou J, Jiang H. Livin is involved in TGF-beta1-induced renal tubular epithelial-mesenchymal transition through lncRNA-ATB. Ann Transl Med. 2019;7(18):463. doi:10.21037/atm.2019.08.29
86. Vierbuchen T, Agarwal S, Johnson JL, et al. The lncRNA LUCAT1 is elevated in inflammatory disease and restrains inflammation by regulating the splicing and stability of NR4A2. Proc Natl Acad Sci U S A. 2023;120(1):e2081252176. doi:10.1073/pnas.2213715120
87. Biswas S, Thomas AA, Chen S, et al. MALAT1: An epigenetic regulator of inflammation in diabetic retinopathy. Sci Rep. 2018;8(1):6526. doi:10.1038/s41598-018-24907-w
88. Li Y, Ren D, Xu G. Long noncoding RNA MALAT1 mediates high glucose-induced glomerular endothelial cell injury by epigenetically inhibiting klotho via methyltransferase G9a. IUBMB Life. 2019;71(7):873–881. doi:10.1002/iub.2009
89. Gu YY, Dou JY, Huang XR, et al. Transforming growth factor-beta and long non-coding RNA in renal inflammation and fibrosis. Front Physiol. 2021;12:684236. doi:10.3389/fphys.2021.684236
90. Mohammed-Ali Z, Lu C, K MM, et al. Endoplasmic reticulum stress inhibition attenuates hypertensive chronic kidney disease through reduction in proteinuria. Sci Rep. 2017;7:41572. doi:10.1038/srep41572
91. Zhang Y, Guo S, Fu X, et al. Emerging insights into the role of NLRP3 inflammasome and endoplasmic reticulum stress in renal diseases. Int Immunopharmacol. 2024;136:112342. doi:10.1016/j.intimp.2024.112342
92. Delitsikou V, Jarad G, Rajaram RD, et al. Klotho regulation by albuminuria is dependent on ATF3 and endoplasmic reticulum stress. FASEB J. 2020;34(2):2087–2104. doi:10.1096/fj.201900893R
93. Kale A, Shelke V, Habshi T, et al. ER stress modulated Klotho restoration: a prophylactic therapeutic strategy against acute kidney injury-diabetes comorbidity. Biochim Biophys Acta Mol Basis Dis. 2024;1870(1):166905. doi:10.1016/j.bbadis.2023.166905
94. Ao Q, Hu H, Huang Y. Ferroptosis and endoplasmic reticulum stress in rheumatoid arthritis. Front Immunol. 2024;15:1438803. doi:10.3389/fimmu.2024.1438803
95. Bester D, Blignaut M, Huisamen B. ATM facilitates autophagy and protects against oxidative stress and apoptosis in response to ER stress in vitro. Biochem Biophys Res Commun. 2024;732:150422. doi:10.1016/j.bbrc.2024.150422
96. Zhou S, Cheng K, Peng Y, et al. Regulation mechanism of endoplasmic reticulum stress on metabolic enzymes in liver diseases. Pharmacol Res. 2024;207:107332. doi:10.1016/j.phrs.2024.107332
97. Li S, Kong J, Yu L, et al. Abnormally decreased renal Klotho is linked to endoplasmic reticulum-associated degradation in mice. Int J Med Sci. 2022;19(2):321–330. doi:10.7150/ijms.68137
98. Tibi S, Zeynalvand G, Mohsin H. Role of the renin angiotensin aldosterone system in the pathogenesis of sepsis-induced acute kidney injury: a systematic review. J Clin Med. 2023;12(14):4566. doi:10.3390/jcm12144566
99. Karalliedde J, Maltese G, Hill B, et al. Effect of renin-angiotensin system blockade on soluble Klotho in patients with type 2 diabetes, systolic hypertension, and albuminuria. Clin J Am Soc Nephrol. 2013;8(11):1899–1905. doi:10.2215/CJN.02700313
100. Kale A, Sankrityayan H, Anders HJ, et al. Klotho in kidney diseases: a crosstalk between the renin-angiotensin system and endoplasmic reticulum stress. Nephrol Dial Transplant. 2023;38(4):819–825. doi:10.1093/ndt/gfab340
101. Czaya B, Faul C. The role of fibroblast growth factor 23 in inflammation and anemia. Int J Mol Sci. 2019;20(17). doi:10.3390/ijms20174195
102. Liu S, Tang W, Zhou J, et al. Fibroblast growth factor 23 is a counter-regulatory phosphaturic hormone for vitamin D. J Am Soc Nephrol. 2006;17(5):1305–1315. doi:10.1681/ASN.2005111185
103. Forster RE, Jurutka PW, Hsieh JC, et al. Vitamin D receptor controls expression of the anti-aging klotho gene in mouse and human renal cells. Biochem Biophys Res Commun. 2011;414(3):557–562. doi:10.1016/j.bbrc.2011.09.117
104. Zhou Y, Kuang Y, Zhou J. Klotho protects against LPS-induced inflammation injury by inhibiting Wnt and NF-kappaB pathways in HK-2 cells. Pharmazie. 2017;72(4):227–231. doi:10.1691/ph.2017.6867
105. Zhao Y, Banerjee S, Dey N, et al. Klotho depletion contributes to increased inflammation in kidney of the db/db mouse model of diabetes via RelA (serine)536 phosphorylation. Diabetes. 2011;60(7):1907–1916. doi:10.2337/db10-1262
106. Jin M, Lv P, Chen G, et al. Klotho ameliorates cyclosporine A-induced nephropathy via PDLIM2/NF-kB p65 signaling pathway. Biochem Biophys Res Commun. 2017;486(2):451–457. doi:10.1016/j.bbrc.2017.03.061
107. Bi F, Chen F, Li Y, et al. Klotho preservation by Rhein promotes toll-like receptor 4 proteolysis and attenuates lipopolysaccharide-induced acute kidney injury. J Mol Med. 2018;96(9):915–927. doi:10.1007/s00109-018-1644-7
108. Wakamatsu T, Yamamoto S, Yoshida S, et al. Indoxyl sulfate-induced macrophage toxicity and therapeutic strategies in uremic atherosclerosis. Toxins. 2024;16(6):254. doi:10.3390/toxins16060254
109. Lv J, Chen J, Wang M, et al. Klotho alleviates indoxyl sulfate-induced heart failure and kidney damage by promoting M2 macrophage polarization. Aging. 2020;12(10):9139–9150. doi:10.18632/aging.103183
110. Chen X, Tan H, Xu J, et al. Klotho-derived peptide 6 ameliorates diabetic kidney disease by targeting Wnt/beta-catenin signaling. Kidney Int. 2022;102(3):506–520. doi:10.1016/j.kint.2022.04.028
111. Zhao Y, Zhao M, Cai Y, et al. Mammalian target of rapamycin signaling inhibition ameliorates vascular calcification via Klotho upregulation. Kidney Int. 2015;88(4):711–721. doi:10.1038/ki.2015.160
112. Younis NN, Mohamed HE, Shaheen MA, et al. Inactivation of Wnt/beta-catenin/renin angiotensin axis by tumor necrosis factor-alpha inhibitor, infliximab, ameliorates CKD induced in rats. Biochem Pharmacol. 2021;185:114426. doi:10.1016/j.bcp.2021.114426
113. Zhang X, Wang G, Ye L, et al. Baicalin reversal of DNA hypermethylation-associated Klotho suppression ameliorates renal injury in type 1 diabetic mouse model. Cell Cycle. 2020;19(23):3329–3347. doi:10.1080/15384101.2020.1843815
114. Xiong C, Guan Y, Zhou X, et al. Selective inhibition of class IIa histone deacetylases alleviates renal fibrosis. FASEB J. 2019;33(7):8249–8262. doi:10.1096/fj.201801067RR
115. Gaikwad AB, Hajare AD, Kulkarni H, et al. Combination of alpha-Klotho and telmisartan attenuates diabetic kidney disease via mitigating EMT, inflammation and apoptosis. Biochem Biophys Res Commun. 2025;760:151711. doi:10.1016/j.bbrc.2025.151711
116. Ramnitz MS, Gourh P, Goldbach-Mansky R, et al. Phenotypic and genotypic characterization and treatment of a cohort with familial tumoral calcinosis/hyperostosis-hyperphosphatemia syndrome. J Bone Miner Res. 2016;31(10):1845–1854. doi:10.1002/jbmr.2870
117. Slominski B, Ryba-Stanislawowska M, Skrzypkowska M, et al. The KL-VS polymorphism of KLOTHO gene is protective against retinopathy incidence in patients with type 1 diabetes. Biochim Biophys Acta Mol Basis Dis. 2018;1864(3):758–763. doi:10.1016/j.bbadis.2017.12.015
118. Wang P, Zhao Z, Li Z, et al. Attenuation of osteoarthritis progression via locoregional delivery of Klotho-expressing plasmid DNA and Tanshinon IIA through a stem cell-homing hydrogel. J Nanobiotechnology. 2024;22(1):325. doi:10.1186/s12951-024-02608-z
© 2025 The Author(s). This work is published and licensed by Dove Medical Press Limited. The
full terms of this license are available at https://www.dovepress.com/terms.php
and incorporate the Creative Commons Attribution
- Non Commercial (unported, 4.0) License.
By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted
without any further permission from Dove Medical Press Limited, provided the work is properly
attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.
Recommended articles

The Influence of Arginine Methylation in Immunity and Inflammation
Srour N, Khan S, Richard S
Journal of Inflammation Research 2022, 15:2939-2958
Published Date: 13 May 2022

Association of the Systemic Immune-Inflammation Index with Outcomes in Acute Coronary Syndrome Patients with Chronic Kidney Disease
Shi S, Kong S, Ni W, Lu Y, Li J, Huang Y, Chen J, Lin K, Li Y, Ke J, Zhou H
Journal of Inflammation Research 2023, 16:1343-1356
Published Date: 27 March 2023
Role and Mechanism of Growth Differentiation Factor 15 in Chronic Kidney Disease
Tang Y, Liu T, Sun S, Peng Y, Huang X, Wang S, Zhou Z
Journal of Inflammation Research 2024, 17:2861-2871
Published Date: 9 May 2024
Klotho Protein: A Multifaceted Guardian of Healthy Aging and Its Therapeutic Potential
Shen J, Bin W, Lin X, Lai Y, Lin X, Guan T, Liu H
International Journal of Nanomedicine 2025, 20:7251-7270
Published Date: 9 June 2025