Open Access

TGF‑β/Smad signaling in chronic kidney disease: Exploring post‑translational regulatory perspectives (Review)

  • Authors:
    • Jianchun Li
    • Yuanxia Zou
    • Jiraporn Kantapan
    • Hongwei Su
    • Li Wang
    • Nathupakorn Dechsupa
  • View Affiliations

  • Published online on: June 18, 2024     https://doi.org/10.3892/mmr.2024.13267
  • Article Number: 143
  • Copyright: © Li et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

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


Abstract

The TGF‑β/Smad signaling pathway plays a pivotal role in the onset of glomerular and tubulointerstitial fibrosis in chronic kidney disease (CKD). The present review delves into the intricate post‑translational modulation of this pathway and its implications in CKD. Specifically, the impact of the TGF‑β/Smad pathway on various biological processes was investigated, encompassing not only renal tubular epithelial cell apoptosis, inflammation, myofibroblast activation and cellular aging, but also its role in autophagy. Various post‑translational modifications (PTMs), including phosphorylation and ubiquitination, play a crucial role in modulating the intensity and persistence of the TGF‑β/Smad signaling pathway. They also dictate the functionality, stability and interactions of the TGF‑β/Smad components. The present review sheds light on recent findings regarding the impact of PTMs on TGF‑β receptors and Smads within the CKD landscape. In summary, a deeper insight into the post‑translational intricacies of TGF‑β/Smad signaling offers avenues for innovative therapeutic interventions to mitigate CKD progression. Ongoing research in this domain holds the potential to unveil powerful antifibrotic treatments, aiming to preserve renal integrity and function in patients with CKD.

Introduction

Chronic kidney disease (CKD) poses a significant challenge to healthcare systems, affecting an estimated 8–15% of the global population (1,2). This condition is signified by the gradual deterioration of kidney function over time, culminating in end-stage renal disease, which requires treatment through dialysis or kidney transplantation (3). Fibrosis originates from kidney damage stemming from a range of factors such as diabetes, hypertension, glomerular diseases, toxins, infections and autoimmune conditions, resulting in excessive accumulation of the extracellular matrix (ECM), which in turn disrupts the normal tissue structure (4,5). At present, no treatments have received approval to specifically address the fibrotic process in an effort to prevent, arrest or reverse CKD (6). The limited efficacy observed in addressing kidney fibrosis underscores the need for a comprehensive review of its foundational mechanisms and the exploration of emerging therapeutic targets.

Transforming growth factor-β1 (TGF-β1) signaling is intrinsically associated with the advancement of renal fibrosis (RF) in CKD (5). Extensive animal research underscores TGF-β1 as the key pathogenic driver, propelling both glomerular and tubulointerstitial fibrosis (5). TGF-β1 signaling is regulated at several stages to ensure homeostasis (7). This encompasses TGF-β activation, the creation, activation and breakdown of functional TGF-β receptor complexes, the modulation of Smad both in activation and degradation, and the assembly of Smad transcription complexes (7). These complexes co-operate with various DNA-binding transcription factors and coregulators at gene regulatory sequences (8). During these procedures, the hyperactivation of TGF-β1/Smad3 signaling may occur, which subsequently causes tubular dysfunction, interstitial fibroblast proliferation, inflammation and augmented ECM deposition (8). Reestablishing the balance of TGF-β1 signaling offers a compelling antifibrotic strategy to halt CKD progression (7). In this context, the present study aimed to delve deeper into the complex functions of the TGF-β/Smad pathway in detail, examining its influence on biological activities such as renal tubular epithelial cell apoptosis, inflammation, myofibroblast activation, cellular aging and its involvement in autophagy (Fig. 1). Of particular emphasis in the present review, recent findings regarding the roles of post-translational modifications (PTMs) including but not limited to phosphorylation, ubiquitination, SUMOylation, and acetylation (Fig. 2) in determining the strength and duration of TGF-β/Smad signaling were comprehensively summarized.

Pathogenic role of TGF-β signaling in CKD

Activation of TGF-β signaling

TGF-β ligands comprise three distinct isoforms, namely TGF-β1, TGF-β2 and TGF-β3 (9). These isoforms are ubiquitously distributed across various cellular and tissue contexts, with TGF-β1 being the most prevalent (10). TGF-β ligands initially emerge as precursor proteins, which undergo a cleavage process at the N-terminal region (11). The cleavage process results in the formation of the latency-associated peptide, which stays bound to the mature TGF-β homodimer at the C-terminal (Fig. 1). This complex association with the latent TGF-β-binding proteins ensures that TGF-β remains inactive, commonly termed latent TGF-βs (12,13). To become biologically active, these latent forms require the intervention of certain environmental triggers such as specific enzymes or an acidic milieu (5). In the extracellular environment, associations with the ECM and subsequent cleavage by various proteases, including plasmin and specific matrix metalloproteinases, facilitate the liberation and activation of the TGF-β ligands (11).

Upon engagement with the TGF-β1 ligand, the TGF-β receptor (TGF-βR) II triggers the activation of TGF-βRI through phosphorylation (5). This series of activations subsequently culminates in the activation of Smad2/3 transcription factors through phosphorylation, commencing the standard signaling process (5). Integral to this cascade is Smad4, which associates with Smad2/3 after phosphorylation, directing the Smad2/3/4 complex towards the nucleus (7) (Fig. 1). This migration to the nucleus is a pivotal step for transcribing genes, which includes key genes such as NADPH oxidase 4 (NOX4), connective tissue growth factor (CTGF) and others [receptor interacting protein kinase 3 and proto-oncogene tyrosine-protein kinase Src (Src)] involved in tissue repair and cellular regulation (1421). Furthermore, Smad7, an inhibitory molecule, becomes active in response to Smad3, and engages in competitive binding with TGF-βRI, thereby hindering the phosphorylation process of Smad2 and Smad3 (22). The transcriptional modulation orchestrated by Smad3-containing complexes is influenced by an array of non-Smad co-activators, including the transcriptional coactivator p300, activator protein 1 and yes-associated protein, as well as co-suppressors such as SKI-like proto-oncogene (SKI) and Ski-like oncoprotein N (SnoN) (2326). The transcriptional effects of TGF-β1 necessitate the involvement of these accessory factors, driving structural alterations in Smad2/3 to interact effectively with target motifs (8). In addition, TGF-β1 activates an array of pathways independent of Smad signaling, which contribute to its biological activities. These routes include TGF-β-activated kinase 1, phosphatidylinositol 3-kinase/Akt and Rho-like GTPase signaling pathways (27,28).

TGF-β and myofibroblast activation

Activation of myofibroblasts and the ensuing accumulation of ECM are pivotal events in RF (29). The activated myofibroblast serves as the key driver of RF, given its significant capacity to produce the majority of the matrix (29). While myofibroblasts are scarce under normal conditions, their numbers surge in fibrotic kidneys (7,29). Proposed precursors for myofibroblasts include pericytes, cells of epithelial and endothelial origin, circulating cells derived from bone marrow and local fibroblasts (3035). For epithelial cells, research has clarified the fibrosis-promoting influence of TGF-β1, emphasizing the critical roles of key molecules in the Smad signaling pathway, such as TGF-βRI, TGF-βRII and Smad3, in epithelial-to-mesenchymal transition (EMT) (3639). Furthermore, a number of miRNAs and long non-coding RNAs (lncRNAs) have been identified that are reliant on Smad3 function in different capacities to regulate EMT (7,4042). For circulating bone marrow-derived cells, mounting evidence indicates that macrophages originating from bone marrow can directly transition into myofibroblasts (MMT) (43,44). In fibrotic kidneys, the recruited Smad3-deficient macrophages do not differentiate into myofibroblasts (44). Additionally, a series of factors transcriptionally regulated by Smad3, such as Pou4f1, P2Y12 and Src, have been proven to be involved in the MMT process (20,4548). This suggests that the progression of MMT is closely governed by the TGF-β/Smad3 signaling pathway. For endothelium, in models of RF triggered by either folic acid or unilateral ureteral obstruction, the specific reduction of TGF-βRII levels in endothelial cells led to a mitigation of fibrotic remodeling and an inhibition of endothelial-to-mesenchymal transition (49). For fibroblasts, researchers discovered that half of the myofibroblasts arise from the proliferation of local resident fibroblasts, while an additional 35% originate from bone marrow fibroblasts (50). The TGF-β pathway appears instrumental in this process, as evidenced by the fact that specific deletion of TGF-βRII in α-smooth muscle actin (+) cells leads to a marked decrease in fibroblast numbers (50). Furthermore, initiating the conditional deletion of Smad2 in fibroblasts under the influence of the fibroblast-specific protein 1 promoter, diminishes RF in streptozotocin (STZ)-triggered diabetic nephropathy (DN) (51). In summary, the TGF-β/Smad pathway plays a crucial role in guiding cellular dynamics and transitions vital for RF.

Renal inflammation

Sterile inflammation, characterized by the inflammatory response devoid of any infectious agents or specific immunogens, serves as a key trigger for the development of RF (52). TGF-β is instrumental in the formation, balance, diversification and tolerance of immune cells (53,54). Diminishment of TGF-β1 can result in the hyperactivation of immunocytes and trigger the occurrence of autoimmune diseases, a phenomenon noted in mice lacking either TGF-β1 or its receptors. In such cases, excessive inflammatory responses with massive lymphocyte and macrophage infiltration were observed in many organs, primarily in the heart and lungs (55,56). Consequently, broadly blocking upstream TGF-β signaling might exacerbate renal inflammation.

Smad3 acts as a central regulator in renal inflammation, uniquely modulating its dynamics by either inhibiting or promoting the functions of macrophages and T cells (8). As a crucial effector molecule, Smad3 is involved in the TGF-β1-driven suppression of macrophage activation, as demonstrated by its capability to inhibit the regulatory actions of the inducible nitric oxide synthase and matrix metalloproteinase-12 promoters within these cells (57). Furthermore, Smad3 has a critical function in preserving the equilibrium between Treg and Th17 immune responses. This is verified by the observation that a lack of Smad3 results in diminished forkhead box P3 induction, while simultaneously enhancing Th17 cell generation by inhibition of retinoid acid receptor-related orphan receptor γt transcriptional activity within both controlled (in vitro) and living (in vivo) contexts (58). Conversely, Smad3 potentially facilitates macrophage recruitment during renal inflammation through its chemotactic effects, as it has been found to engage with macrophage chemotactic protein-1 (MCP-1), in turn amplifying the renal inflammation driven by macrophages (59). Furthermore, LRNA9884, a lncRNA that is transcriptionally regulated by Smad3 has been shown to influence DN in db/db mice by stimulating the synthesis of MCP-1, further amplifying renal inflammation (60). Additionally, in obstructed nephropathy, AT-rich interaction domain 2 intronic transcript, facilitated by TGF-β/Smad3, enhances the inflammation triggered by IL-1β via the NF-κB pathway, without influencing the fibrosis modulated by the TGF-β/Smad3 process (61). Notably, Smad7 serves as a negative regulator of TGF-β/Smad signaling (62). Previous investigations have further demonstrated that Smad7 functions as a key adjuster by stimulating IκBα, a suppressor of NF-κB, consequently mitigating renal inflammation (63,64). The observed phenomenon suggests that a deficiency in Smad3 inhibits renal inflammation, which is driven by NF-κB in the unilateral ureteral obstruction (UUO) model (65). Presumably, the removal of Smad3 inhibits the breakdown of Smad7 by E3 ubiquitin-protein ligases, such as Smad ubiquitin regulatory factor (Smurf)1/Smurf2 (66). In summary, Smad3 has been identified as a key controller in renal inflammation, orchestrating various molecular interactions and pathways to either amplify or mitigate inflammatory responses.

Cellular senescence and autophagy

Cellular senescence describes the process wherein cells lose their ability to replicate and permanently exit the cell cycle after repeated duplications (67). These senescent cells resist apoptosis and consistently release a diverse secretome, termed the senescence-associated secretory phenotype, which includes pro-inflammatory and pro-fibrotic mediators (67). In recent studies, cellular senescence in renal tubular epithelial cells has been identified as a primary contributor to the onset of RF, and consequently, delaying this senescence presents an effective intervention to curb RF and offers a crucial strategy for decelerating the progression of CKD (6769). A previous study indicates that the TGF-β/Smad pathway promotes cellular senescence by reducing histone 4 lysine 20 tri-methylation through miR-29, impacting DNA repair and genome stability (70). Furthermore, a recent study has demonstrated that ubiquitin-specific protease 11 (USP11) promotes cellular senescence and fibrosis regulated by the Smad/P53 complex, by inhibiting the ubiquitination of TGF-βRII (71). In the context of the aging kidney, heightened stimulation of the TGF-β/Smad3 pathway in podocyte-specific TGF-β overexpressing mice leads to cell senescence through processes that involve transference of p16 and initiation of p21 (72).

Autophagy, a preserved lysosomal pathway, facilitates the degradation of cytoplasmic constituents (73). Yet, its role in kidney fibrosis, whether protective or pathological, remains ambiguous (73). Moreover, the exact mechanisms and signaling pathways that govern autophagy responses across various kidney cell types and disease spectra require further elucidation (73). Recent studies highlighted the influence of Smad3 on autophagy and its prospective role as a treatment focus for fibrotic diseases. A study has demonstrated that Smad3 contributes to lysosomal depletion by inhibiting transcription factor EB-mediated lysosome biogenesis, resulting in impaired autophagy during the advancement of DN (74). Moreover, TGFβ, through an epigenetic mechanism that involves the Smad3-mediated decrease of histone acetyltransferase KAT8 (also termed as MYST1), activates autophagy which promotes fibrotic diseases, including dermal and pulmonary fibrosis, suggesting a potential therapeutic target (75).

Cell death

Preventing the death of renal tubular epithelial cells is crucial in halting the progression of CKD (76). It is widely acknowledged that TGF-β is known to facilitate cell death by the interruption of the cell cycle at its G1 phase, orchestrated through the Smad pathway (7779). A previous study revealed that Smad3 contributes to acute kidney injury (AKI) by directly interacting with cyclin-dependent kinase inhibitor proteins p21 and p27 (78). This interaction results in the death of renal tubular epithelial cells (TECs) due to G1 phase cell cycle arrest (79). Moreover, a recent study has indicated that Smad3 can also transcriptionally activate the receptor-interacting protein kinase 3/mixed lineage kinase domain-like protein necroptosis pathway, subsequently resulting in the death of TECs (21).

Ferroptosis, a regulated form of cell death induced by oxidative stress and dependent on iron-mediated lipid peroxidation, is intricately linked with numerous renal and fibrotic diseases, including AKI, CKD and diabetic kidney diseases (80,81). However, the precise mechanisms driving RF through ferroptosis are yet to be fully understood. Recently published studies demonstrated that Smad3 induces ferroptosis in TECs, primarily through the modulation of NOX4 gene transcription (18,82,83). It also works in conjunction with activating transcription factor 3 (ATF3) to suppress the gene expression of solute carrier family 7 member 11 (SLC7A11), thereby modulating the ferroptosis process (84). In light of these findings, the present research group has made some interesting discoveries. Preliminary investigations have revealed that active natural compounds exhibit specificity towards key components involved in the ferroptosis process. For example, tectorigenin has been found to specifically target Smad3, regulating NOX4 and thereby inhibiting the ferroptosis pathway, suggesting a potential therapeutic role against RF (18). Formononetin, on the other hand, suppresses the binding of the SMAD3/ATF3 complex, promoting the expression of SLC7A11 and highlighting its potential as a novel modulator for ferroptosis in RF (84). Lastly, kaempferitrin has demonstrated the ability to bind with NOX4, leading to an improvement in tubular ferroptosis within the kidneys, proposing a new approach to the treatment of RF through its impact on ferroptosis (83). These promising steps forward provide valuable insights into how natural compounds can potentially be utilized in the modulation of ferroptosis and treatment of RF.

PTMs of TGF-β1 signaling

PTMs are chemical alterations essential for regulating protein functions (85). They modulate the activity, localization, stability, and interactions of proteins with other cellular components, including other proteins, nucleic acids, lipids and cofactors (85). PTMs can occur on amino acid side chains or at the C- or N-termini of proteins (85). These modifications enhance the chemical diversity of the 20 standard amino acids, either by altering an existing functional group or introducing novel ones such as phosphate (85). Common PTMs include (de-)phosphorylation, (de-)ubiquitination, (de-)SUMOylation, reversible acetylation and O-GlcNAcylation (86). In the present study, related reports of PTMs in the TGF-β/Smad signaling pathway and their roles in TGF-β/Smad signal transduction are discussed.

Phosphorylation and de-phosphorylation

Phosphorylation is a process occurring after protein translation, characterized by the addition of a phosphate group to certain amino acids in a protein, typically serine, threonine or tyrosine residues (87). Normally, Smad2 and Smad3, which are among the receptor-regulated Smads (R-Smads), undergo activation via ligand-induced phosphorylation at two serine residues within their carboxy-terminal SSXS motif, mediated by TGF-βRI (88,89). Beyond the established TGF-β signaling pathway involving Smad2 and Smad3, the TGF-β/Smad signaling pathway is further influenced by multiple kinases, providing further refinement, expansion or modulation of the signaling output (90). The mitogen-activated protein kinase (MAPK) family, comprising three primary kinases including p38 MAPK, c-Jun N-terminal kinase (JNK) and extracellular signal-regulated kinase (ERK), has been shown to be activated in humans with both acute and CKD, as indicated by a number of studies (9193). In addition, through employing pharmacological and genetic interventions, the blocking of p38 and/or JNK effectively mitigates RF across diverse animal studies (9496). Notably, each of the three MAPKs are capable of phosphorylating specific sites within the linker regions of both Smad2 and Smad3 (97). These regions are situated in the interdomain space that separates the mad homology domain 1 (MH1) and MH2 domains within a Smad protein, subsequently altering the transcription of Smad3-dependent genes (97). Hence, besides being activated by TGF-β1, Smad3 activation can be triggered by various stress-inducing agents such as angiotensin II (Ang II), advanced end products (AGE) and C-reactive protein (CRP) through MAPK-dependent pathways. Specifically, research reveals that Ang II has the capability to directly stimulate Smad3, thereby promoting the upregulation of both CTGF and collagen I via the AT1-ERK/p38 MAPK interaction route (98). In addition, there is a strong link between AGEs and the expression of CTGF, and EMT. Through the Smad3 pathway that functions independently of TGF-β1, introducing AGEs induces CTGF expression in TECs devoid of TGF-β1, evidenced by the swift activation of Smad2/3, ERK1/2 and p38 (99). Furthermore, CRP, reported as an inflammatory marker and mediator, is known to regulate the fibrotic process, primarily by modulating the CD32b-ERK/p38 MAPK pathway, subsequently activating Smad3 (100).

Protein dephosphorylation, mediated by protein phosphatases, serves as a key control process that influences the operation of a number of proteins within signal transduction routes (87). Chen et al (101) discovered that loss of integrin α1β1, a regulator for collagen synthesis, exacerbated RF in a UUO model via a TGF-β/Smad-dependent manner. In terms of the mechanism, integrin α1β1 promotes the recruitment of the phosphatase, T cell protein tyrosine phosphatase (TCPTP) to TGF-βRII which results in the dephosphorylation of tyrosine residues in the TGF-βRII cytoplasmic tail, subsequently impairing TGF-βR-dependent fibrotic signaling transmission (101). In addition, protein phosphatase magnesium-dependent 1A (PPM1A) facilitates the dephosphorylation of TGF-β-activated Smad2/3 within their carboxy-terminal SSXS motif, subsequently promoting their nuclear export (102). In obstructive and aristolochic acid-induced nephropathy, a decrease in PPM1A levels within the tubulointerstitium has been noted and this diminution plays a role in enhancing Smad3 phosphorylation, leading to subsequent RF (103). In obstructive nephropathy, maxacalcitol, an analog of vitamin D, enhances the function of the PPM1A/vitamin D receptor complex, resulting in the dephosphorylation of Smad3, thereby reducing tubulointerstitial fibrosis (104). Furthermore, PPM1A and PTEN collaboratively work to diminish the phosphorylation of Smad3 and the activation of genes associated with fibrosis (105).

Ubiquitination and deubiquitination

In all organ tissues, intracellular proteins undergo continuous turnover through degradation and synthesis (106). The ubiquitin-proteasome system (UPS) primarily degrades intracellular proteins (106). This multi-enzyme process covalently attaches ubiquitin to a substrate protein, which the proteasome, a core proteolytic complex of the UPS, then recognizes and degrades (106).

Ubiquitination alters different elements within the TGF-β signaling pathway (87). The Smurfs, identified as the first ubiquitin E3 ligases for Smads, exemplify such ubiquitin E3 ligases (107). Extensive research has focused on these ligases, which specifically act upon R-Smads, Smad7 and certain Smad-associated proteins for proteasomal degradation (108). Smurf1 and Smurf2 are both E3 ubiquitin ligases for Smad7 (87). Additionally, Smurf2 is capable of initiating the polyubiquitylation and degradation of Smad2, as well as the Smad gene silencing cofactors SKI and SnoN within kidneys undergoing fibrosis (107,109). This is evidenced by the fact that the overexpression of Smurf2 has been shown to encourage the activation of the TGF-β-responsive promoter and induce EMT within human kidney cortical epithelial cells (107). Recent research has demonstrated that flavin-containing monooxygenase 2 can enhance the expression of Smurf2 and facilitate its nuclear translocation, resulting in tubular cell fibrogenesis and paracrine secretion (110). In addition, the E3 ligase, Arkadia/RNF111, regulates TGF-β signaling by degrading Smad7 (111,112). This degradation consequently fostered the advancement of fibrosis in a rat model of tubulointerstitial fibrosis (111,112). Overexpression of latent TGF-β1 has been shown to ameliorate DN through the inhibition of Arkadia-induced Smad7 imbalance, which subsequently provoked renal inflammation and tissue fibrosis in type 1 diabetes mice induced by STZ (113). In addition, neural precursor cell expressed developmentally down-regulated 4-2 (NEDD4-2) has been identified as the ubiquitin ligase that facilitates proteasome-mediated degradation of TGF-β-induced phosphorylated Smad2/3 (114,115). Deficiency of NEDD4-2 in mice results in progressive kidney injury, marked by fibrosis, tubular epithelial cell apoptosis and various characteristics of CKD, including dilated/cystic tubules, and elevated expression of kidney injury markers (116). In conclusion, the UPS may either promote or inhibit fibrotic outcomes, depending on the TGF-β signaling elements that undergo degradation.

Deubiquitination, the counterpart to ubiquitination, is a key cellular procedure encompassing the removal of ubiquitin molecules that have been added to proteins (117,118). This dynamic interplay between ubiquitination and deubiquitination ensures the precise regulation of protein function, stability and interactions within the cell (117). Deubiquitinating enzymes, also known as deubiquitinases (DUBs), constitute a vast group of proteases responsible for removing ubiquitin from proteins (117). Moreover, they aid in the creation of independent entities from freshly translated polyubiquitins and repurpose ubiquitins after the breakdown of polyubiquitinated protein substrates (119). While the control of TGF-β signaling through ubiquitination has been widely studied in the past decades, the function of deubiquitination steered by DUBs in the TGF-β signaling pathway, especially in the context of CKD, has only begun to gain attention recently.

PR-619, a comprehensive DUB inhibitor, mitigated fibrosis in mice undergoing UUO and in rat kidney fibroblast cells, namely NRK-49F cells, triggered by TGF-β1 (120). Furthermore, PR-619 demonstrates an inhibitory effect on Smad4 levels, while it does not affect the production of TGF-βR, Smad2 or Smad3 (120). This indicates that DUBs may regulate fibrosis by modulating Smad4 (120). At present, members of the ubiquitin specific proteases family (USPs), which have been reported to regulate Smad4 deubiquitination, include USP9X (121), USP10 (122), USP13 (123), USP17 (124) and USP25 (125). USPs are currently known as the most extensive and predominant family of enzymes associated with deubiquitination (117). In fact, USP9X is documented to suppress fibrosis triggered by the stimulation of AGEs in mesangial cells, as well as EMT in renal tubular cells (126,127). USP10 has been reported to counteract renal impairment caused by sepsis, primarily by reducing apoptosis in TECs and mitigating oxidative stress (128). Furthermore, recent research has confirmed that USP25 is instrumental in advancing hypertensive renal disease (125). Knockout of USP25 in mice has been shown to reduce kidney malfunctions and fibrotic conditions (125). From a mechanistic viewpoint, USP25 is associated with the regulation of TGF-β signaling activation (125). Specifically, USP25 functions by reducing Smad4 K63-linked polyubiquitination (125). For R-Smads and Smad7, although some DUBs have been reported to regulate their ubiquitination processes (129133), their functions in CKD remain to be further elucidated.

Additionally, several DUBs, such as 26S proteasome-associated PAD1 homolog 1 (134) and ubiquitin C-terminal hydrolase 37 (131), have been reported to regulate the deubiquitination process of TGF-βRI, thereby promoting TGF signaling. However, only USP11 has been documented to modulate the fibrotic process in CKD (135,136). Ni et al (71) conducted an intervention using the USP11 inhibitor mitoxantrone on mice experiencing both UUO and folic acid-triggered RF. They discovered that this intervention could inhibit TGF-βRII expression and associated fibrotic and aging phenotypes. The use of USP11 conditional knockout mice further confirmed this phenomenon (135). It is noteworthy that these DUBs often have multiple substrates. For instance, USP11 can regulate the deubiquitination of both TGF-βRII and epidermal growth factor receptor (EGFR) (135,136).

SUMOylation and de-SUMOylation

SUMOylation, a process where SUMO covalently attaches to specific protein targets, plays a pivotal role in modulating signal transduction through changes in subcellular localization, influencing enzymatic activity and directing the ubiquitin-mediated breakdown of its target substrates (137). This modification process is driven by a series of enzymes requiring ATP, encompassing the E1 activator, the E2 conjugator known as Ubc9 and various E3 ligases (138). Notably, the function of SUMOylation in the TGF-β signaling pathway is attracting more and more interest (139).

To date, researchers have identified five SUMO proteins, labeled SUMO1-5 (140). Given the considerable sequence resemblance between SUMO2 and SUMO3, they are commonly categorized as SUMO2/3. Among these, SUMO1 and SUMO2/3 are expressed ubiquitously, while the distribution of SUMO4 is confined to specific organs, such as the spleen and kidney (137). The ability for SUMOylation to be reversed is maintained by SUMO-specific proteases (SENPs), which can detach SUMO proteins from their targets (108). Currently, seven distinct SENPs, ranging from SENP1-3 to SENP5-8, have been identified (141,142). Apart from reversing SUMOylation, these enzymes also mature pro-SUMO into a conjugatable form (142).

Within the scope of TGF-β signaling, the SUMOylation process of TGF-βR1 amplifies its capability to interact with Smad3, which promotes the phosphorylation of Smad3 (139). SENP2 counteracts this alteration, and an upsurge in SENP2 expression curtails the EMT instigated by TGF-β (143). The role of Smad4 SUMOylation in TGF-β transcription regulation remains contentious. Some researchers posit a detrimental effect of Smad4 SUMOylation on TGF-β signaling, highlighting that the K113R/K159R mutation curtails the polyubiquitination of Smad4 (144). In the context of renal mesangial cells under high glucose conditions, the SUMO2/3-driven SUMOylation of Smad4 triggers the TGF-β/Smad pathway, subsequently elevating fibronectin levels (145). These contrasting perspectives might arise from distinct cellular contexts (146). Conclusively, adjusting the (de-)SUMOylation of the TGF-β/Smad pathway presents a hopeful approach for CKD treatment.

Reversible acetylation

Protein acetylation is recognized as a significant and reversible post-translational modification, underscoring its various cellular and physiological activities (147). Reversible acetylation is orchestrated by two primary enzyme classes: Acetyltransferases (KATs) and deacetylases (KDACs) (147). KATs enable the addition of acetyl groups onto lysine residues and encompass the general control non-derepressible 5, p300 and MYST families, along with other unclassified KATs (147). Although KATs primarily acetylate histones, enzymes such as p300 also influence the TGF-β/Smad pathway, and are also recognized for enhancing TGF-β activity through the acetylation of Smad2 or Smad3 (148,149). Furthermore, the inhibition of p300 with a novel FATp300 inhibitor, L002, mitigates RF caused by hypertension and opposes fibrogenic responses in fibroblasts (150).

Unlike KATs, KDACs are divided into two main categories: Classical histone deacetylases, which are Zn2+-dependent, and sirtuin deacetylases (SIRTs), which rely on NAD+ (147). In recent studies, seven mammalian counterparts of the yeast Sir2, specifically labeled as SIRT1 to SIRT7, have been discerned (151). Within the kidney, SIRT1 stands out as the predominant SIRTs under investigation. Primarily localized in the nucleus, it orchestrates the acetylation patterns of nucleosome histones and influences the dynamics of multiple transcriptional regulators (151). As a result, SIRT1 plays a pivotal role in cellular defense by mitigating processes such as apoptosis, inflammation and fibrosis (151). SIRT1 deficiency promotes Smad3 acetylation, subsequently activating TGF-β signaling and exacerbating the progression of CKD (152). Resveratrol intervention facilitates the interaction between SIRT1 and Smad3, thereby attenuating Smad3 acetylation (153). Moreover, elevating SIRT1 levels in tubular cells impedes the transition from AKI to tubulointerstitial fibrosis (151). This also curtails the subsequent accumulation of matrix metalloproteinase-7 in the kidney through the deacetylation of Smad4 (154). Hence, SIRT1 emerges as a promising candidate for therapy in treating CKD (152). Unlike SIRT1, SIRT2 predominantly resides in the cytoplasm and plays a role in hindering fibrosis within renal tubules (155). The specific removal of SIRT2 from TECs aggravates RF, while its deliberate overexpression in these cells reduces RF (155). In terms of mechanism, SIRT2 forms a direct association with Smad2 and Smad3, leading to their deacetylation; this interaction subsequently mitigates the fibrotic effects triggered by TGF-β (155). This highlights the therapeutic potential of SIRT2 in addressing fibrosis. In the context of CKD, there are limited reports on the regulatory roles of other SIRT family members, specifically SIRT3-7, concerning the TGF-β/Smad pathway. To sum up, the complex equilibrium and interaction between these enzymes underscore their potential as therapeutic targets in CKD and fibrotic conditions.

O-GlcNAcylation

O-GlcNAcylation, a fluctuating and reversible type of PTM, entails the addition of a β-D-N-acetylglucosamine (GlcNAc) molecule onto serine or threonine residues within proteins (156). This modification, predominantly occurring within the cytoplasm and nucleus, is distinct from the conventional N- and O-glycosylations that take place in the endoplasmic reticulum and Golgi apparatus (156). The enzymes O-GlcNAc transferase and O-GlcNAcase are key in controlling O-GlcNAcylation, which is vital for numerous cellular functions such as signal transduction and transcription, as well as maintaining protein stability (156,157). Due to its significance, changes in O-GlcNAcylation are linked to various diseases including diabetes, neurodegenerative conditions and cancer (158160).

A previous study has indicated that an increase in O-GlcNAcylation levels in kidney tissues following UUO, and that glucosamine-driven O-GlcNAcylation can promote fibrosis in the renal parenchymal cell, HK2 TECs (161). Notably, a previous study observed that Smad4 has been identified as a newly discovered protein undergoing O-GlcNAc modification in human lung cancer cells. In this context, O-GlcNAc hinders the linkage between Smad4 and GSK-3β, interactions that are crucial for the proteasomal breakdown of Smad4 (162). Nevertheless, the precise function of O-GlcNAcylation in fibrosis, along with its interplay with the TGF-β1/Smad3 signaling pathway remains to be further elucidated.

Myristoylation

Myristoylation, a post-translational modification process, involves covalently linking myristate (a 14-carbon fatty acid) to the N-terminal glycine residue of the protein via an amide bond (163). This modification is critical in numerous protein signaling systems as it imparts various effects such as modulating protein stability, facilitating protein-protein interactions and enhancing subcellular localization to organelles or the plasma membrane (163).

There is relatively scant research focusing on the role of myristoylation in the progression of CKD. Notably, a previous study noted that myristoylated TGF-βRI and TGF-βRII can induce transcriptional activation of Smad2, suggesting a potential role for myristoylation in the activation of the TGF-β pathway (164). In addition, myristoylation may exert an indirect influence on the regulation of the TGF-β signaling pathway. For instance, the myristoylation of PPM1A could enhance the phosphatase activity of PPM1A, as promoted by the cellular senescence-inhibited gene, thereby inhibiting TGF-β signaling further (165). This suggests that the modulation of myristoylation states could represent a novel strategy for managing TGF-β-driven processes in CKD.

Perspectives

The TGF-β/Smad signaling pathway stands as a primary contributor to RF in CKD (5). Its intricate post-translational regulation dictates both the intensity and temporal specificity of the signaling (90). A myriad of PTMs, encompassing phosphorylation, ubiquitination, acetylation and SUMOylation, govern the stability, activity and interplay of TGF-β receptors, Smad proteins, and associated co-regulators (87,90). This fine-tunes the fibrotic signaling cascade. Delving into these multifaceted post-translational mechanisms offers profound insights, paving the way for innovative antifibrotic strategies to combat CKD progression (87,90).

However, despite these insights, there remain several considerable limitations that hinder a comprehensive understanding. A complete grasp of individual PTMs and how each modification precisely regulates this process is still lacking, necessitating further exploration into each PTM to clarify their specific roles and impacts on overall signaling. In addition, the intricate and complex interactions among various PTMs and their collective influence on TGF-β/Smad signaling presents a challenging aspect that is not yet fully understood due to several reasons. First, the crosstalk between different PTMs can be complex and context-dependent, making it difficult to predict the net effect on Smad signaling (87). Second, many PTMs can target multiple proteins within the pathway, further complicating the overall picture (136,155,166). Finally, technological limitations can hinder our ability to comprehensively analyze these interactions and their dynamic regulation within cells (167). Despite these challenges, unraveling these complexities holds immense promise for developing more targeted therapeutic strategies for fibrotic diseases. More research is needed to elucidate these dynamic relationships and create a more holistic view of the signaling network. Furthermore, despite advances in proteomics techniques, current approaches are limited in their ability to uncover disease-specific regulations in patient-derived samples (168). The development and application of more sophisticated tools could enable a finer resolution of these regulatory intricacies, providing invaluable insight into disease progression and response to treatment (168). Moreover, the current landscape of therapeutic strategies lacks personalization, with most approaches not considering the unique progression patterns of CKD in different individuals (167). This represents a significant gap in effectively managing and treating CKD. Lastly, inefficiencies in existing drug delivery systems, particularly those targeting the kidneys, pose another challenge by reducing therapeutic efficacy and leading to potential side effects (169). Existing kidney-targeting drug delivery systems face challenges related to nanoparticle size. Small nanoparticles (<6-8 nm) can pass through the glomerular filtration barrier but are quickly cleared by urine, limiting their use for sustained drug delivery (170). Larger nanoparticles (350–400 nm) may accumulate in the kidneys but struggle with bioavailability and filtration (170). Additionally, the protein corona on nanoparticles can reduce targeting efficiency, complicating effective treatment (170).

Emerging research horizons include finding ways to translate cellular insights into physiologically relevant disease models, elucidating the interconnections between diverse PTMs, broadening the spectrum of known post-translational regulators and leveraging advanced proteomics techniques to decode previously concealed, disease-specific regulations in patient-derived samples. A more detailed and stage-specific understanding of TGF-β/Smad signaling regulation could pave the way for personalized therapeutic strategies tailored to individual CKD progressions, thus enhancing CKD treatment outcomes and preserving renal architecture and functionality in patients.

Acknowledgements

Not applicable.

Funding

This study received financial support from the following sources: National Natural Science Foundation of China (grant no. 82104665); The Science and Technology Department of Sichuan Province (grant nos. 2023NSFSC1763 and 2022YFS0621); and The Innovation Team Project of the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University (grant no. 2022-CXTD-03).

Availability of data and materials

Not applicable.

Authors' contributions

The manuscript was initially drafted by JL and YZ. It was then edited and revised by JK, HS, LW and ND. All authors read and approved the final version of the manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Webster AC, Nagler EV, Morton RL and Masson P: Chronic kidney disease. Lancet. 389:1238–1252. 2017. View Article : Google Scholar : PubMed/NCBI

2 

GBD Chronic Kidney Disease Collaboration, . Global, regional, and national burden of chronic kidney disease, 1990-2017: A systematic analysis for the Global Burden of Disease Study 2017. Lancet. 395:709–733. 2020. View Article : Google Scholar : PubMed/NCBI

3 

Chen TK, Knicely DH and Grams ME: Chronic kidney disease diagnosis and management: A review. JAMA. 322:1294–1304. 2019. View Article : Google Scholar : PubMed/NCBI

4 

Liu Y: Cellular and molecular mechanisms of renal fibrosis. Nat Rev Nephrol. 7:684–696. 2011. View Article : Google Scholar : PubMed/NCBI

5 

Meng X, Nikolic-Paterson DJ and Lan HY: TGF-β: The master regulator of fibrosis. Nat Rev Nephrol. 12:325–338. 2016. View Article : Google Scholar : PubMed/NCBI

6 

Ruiz-Ortega M, Rayego-Mateos S, Lamas S, Ortiz A and Rodrigues-Diez RR: Targeting the progression of chronic kidney disease. Nat Rev Nephrol. 16:269–288. 2020. View Article : Google Scholar : PubMed/NCBI

7 

Gu YY, Liu XS, Huang XR, Yu XQ and Lan HY: TGF-β in renal fibrosis: Triumphs and challenges. Future Med Chem. 12:853–866. 2020. View Article : Google Scholar : PubMed/NCBI

8 

Wu W, Wang X, Yu X and Lan HY: Smad3 Signatures in Renal Inflammation and Fibrosis. Int J Biol Sci. 18:2795–2806. 2022. View Article : Google Scholar : PubMed/NCBI

9 

Meng XM, Tang PM, Li J and Lan HY: TGF-β/Smad signaling in renal fibrosis. Front Physiol. 6:822015. View Article : Google Scholar : PubMed/NCBI

10 

Yu L, Border WA, Huang Y and Noble NA: TGF-beta isoforms in renal fibrogenesis. Kidney Int. 64:844–856. 2003. View Article : Google Scholar : PubMed/NCBI

11 

Weiss A and Attisano L: The TGFbeta superfamily signaling pathway. Wiley Interdiscip Rev Dev Biol. 2:47–63. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Annes JP, Munger JS and Rifkin DB: Making sense of latent TGFbeta activation. J Cell Sci. 116:217–224. 2003. View Article : Google Scholar : PubMed/NCBI

13 

Robertson IB, Horiguchi M, Zilberberg L, Dabovic B, Hadjiolova K and Rifkin DB: Latent TGF-β-binding proteins. Matrix Biol. 47:44–53. 2015. View Article : Google Scholar : PubMed/NCBI

14 

Macconi D, Remuzzi G and Benigni A: Key fibrogenic mediators: Old players. Renin-angiotensin system. Kidney Int. Suppl (2011):4:58–64. 2014. View Article : Google Scholar : PubMed/NCBI

15 

Loeffler I and Wolf G: Transforming growth factor-β and the progression of renal disease. Nephrol Dial Transplant. 29 (Suppl 1):i37–i45. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Samarakoon R, Overstreet JM and Higgins PJ: TGF-β signaling in tissue fibrosis: Redox controls, target genes and therapeutic opportunities. Cell Signal. 25:264–268. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Samarakoon R, Overstreet JM, Higgins SP and Higgins PJ: TGF-β1 → SMAD/p53/USF2 → PAI-1 transcriptional axis in ureteral obstruction-induced renal fibrosis. Cell Tissue Res. 347:117–128. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Li J, Yang J, Zhu B, Fan J, Hu Q and Wang L: Tectorigenin protects against unilateral ureteral obstruction by inhibiting Smad3-mediated ferroptosis and fibrosis. Phytother Res. 36:475–487. 2022. View Article : Google Scholar : PubMed/NCBI

19 

Yang Q, Gao L, Hu XW, Wang JN, Zhang Y, Dong YH, Lan HY and Meng XM: Smad3-Targeted Therapy Protects against Cisplatin-Induced AKI by attenuating programmed cell death and inflammation via a NOX4-dependent mechanism. Kidney Dis (Basel). 7:372–390. 2021. View Article : Google Scholar : PubMed/NCBI

20 

Tang PM, Zhou S, Li CJ, Liao J, Xiao J, Wang QM, Lian GY, Li J, Huang XR, To KF, et al: The proto-oncogene tyrosine protein kinase Src is essential for macrophage-myofibroblast transition during renal scarring. Kidney Int. 93:173–187. 2018. View Article : Google Scholar : PubMed/NCBI

21 

Liang L, Wang W, Chen J, Wu W, Huang XR, Wei B, Zhong Y, Ma RCW, Yu X and Lan HY: SARS-CoV-2 N protein induces acute kidney injury in diabetic mice via the Smad3-Ripk3/MLKL necroptosis pathway. Signal Transduct Target Ther. 8:1472023. View Article : Google Scholar : PubMed/NCBI

22 

Hayashi H, Abdollah S, Qiu Y, Cai J, Xu YY, Grinnell BW, Richardson MA, Topper JN, Gimbrone MA Jr, Wrana JL and Falb D: The MAD-related protein Smad7 associates with the TGFbeta receptor and functions as an antagonist of TGFbeta signaling. Cell. 89:1165–1173. 1997. View Article : Google Scholar : PubMed/NCBI

23 

Zhang Y, Feng XH and Derynck R: Smad3 and Smad4 cooperate with c-Jun/c-Fos to mediate TGF-beta-induced transcription. Nature. 394:909–913. 1998. View Article : Google Scholar : PubMed/NCBI

24 

Samarakoon R, Dobberfuhl AD, Cooley C, Overstreet JM, Patel S, Goldschmeding R, Meldrum KK and Higgins PJ: Induction of renal fibrotic genes by TGF-β1 requires EGFR activation, p53 and reactive oxygen species. Cell Signal. 25:2198–2209. 2013. View Article : Google Scholar : PubMed/NCBI

25 

Samarakoon R, Higgins SP, Higgins CE and Higgins PJ: TGF-beta1-induced plasminogen activator inhibitor-1 expression in vascular smooth muscle cells requires pp60(c-src)/EGFR(Y845) and Rho/ROCK signaling. J Mol Cell Cardiol. 44:527–538. 2008. View Article : Google Scholar : PubMed/NCBI

26 

Meng XM, Chung ACK and Lan HY: Role of the TGF-β/BMP-7/Smad pathways in renal diseases. Clin Sci (Lond). 124:243–254. 2013. View Article : Google Scholar : PubMed/NCBI

27 

Zhang YE: Non-Smad signaling pathways of the TGF-β Family. Cold Spring Harb Perspect Biol. 9:a0221292017. View Article : Google Scholar : PubMed/NCBI

28 

Kim SI and Choi ME: TGF-β-activated kinase-1: New insights into the mechanism of TGF-β signaling and kidney disease. Kidney Res Clin Pract. 31:94–105. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Humphreys BD: Mechanisms of Renal Fibrosis. Annu Rev Physiol. 80:309–326. 2018. View Article : Google Scholar : PubMed/NCBI

30 

Lin SL, Kisseleva T, Brenner DA and Duffield JS: Pericytes and perivascular fibroblasts are the primary source of collagen-producing cells in obstructive fibrosis of the kidney. Am J Pathol. 173:1617–1627. 2008. View Article : Google Scholar : PubMed/NCBI

31 

Nakamura J, Sato Y, Kitai Y, Wajima S, Yamamoto S, Oguchi A, Yamada R, Kaneko K, Kondo M, Uchino E, et al: Myofibroblasts acquire retinoic acid-producing ability during fibroblast-to-myofibroblast transition following kidney injury. Kidney Int. 95:526–539. 2019. View Article : Google Scholar : PubMed/NCBI

32 

Chen YT, Chang FC, Wu CF, Chou YH, Hsu HL, Chiang WC, Shen J, Chen YM, Wu KD, Tsai TJ, et al: Platelet-derived growth factor receptor signaling activates pericyte-myofibroblast transition in obstructive and post-ischemic kidney fibrosis. Kidney Int. 80:1170–1181. 2011. View Article : Google Scholar : PubMed/NCBI

33 

Kramann R, Schneider RK, DiRocco DP, Machado F, Fleig S, Bondzie PA, Henderson JM, Ebert BL and Humphreys BD: Perivascular Gli1+ progenitors are key contributors to injury-induced organ fibrosis. Cell Stem Cell. 16:51–66. 2015. View Article : Google Scholar : PubMed/NCBI

34 

Li J, Qu X and Bertram JF: Endothelial-myofibroblast transition contributes to the early development of diabetic renal interstitial fibrosis in streptozotocin-induced diabetic mice. Am J Pathol. 175:1380–1388. 2009. View Article : Google Scholar : PubMed/NCBI

35 

Meng X, Jin J and Lan HY: Driving role of macrophages in transition from acute kidney injury to chronic kidney disease. Chin Med J (Engl). 135:757–766. 2022. View Article : Google Scholar : PubMed/NCBI

36 

Zeisberg M and Kalluri R: The role of epithelial-to-mesenchymal transition in renal fibrosis. J Mol Med (Berl). 82:175–181. 2004. View Article : Google Scholar : PubMed/NCBI

37 

Kim KK, Sheppard D and Chapman HA: TGF-β1 signaling and tissue fibrosis. Cold Spring Harb Perspect Biol. 10:a0222932018. View Article : Google Scholar : PubMed/NCBI

38 

Meng XM, Huang XR, Chung AC, Qin W, Shao X, Igarashi P, Ju W, Bottinger EP and Lan HY: Smad2 protects against TGF-beta/Smad3-mediated renal fibrosis. J Am Soc Nephrol. 21:1477–1487. 2010. View Article : Google Scholar : PubMed/NCBI

39 

Sato M, Muragaki Y, Saika S, Roberts AB and Ooshima A: Targeted disruption of TGF-beta1/Smad3 signaling protects against renal tubulointerstitial fibrosis induced by unilateral ureteral obstruction. J Clin Invest. 112:1486–1494. 2003. View Article : Google Scholar : PubMed/NCBI

40 

Zarjou A, Yang S, Abraham E, Agarwal A and Liu G: Identification of a microRNA signature in renal fibrosis: Role of miR-21. Am J Physiol Renal Physiol. 301:F793–F801. 2011. View Article : Google Scholar : PubMed/NCBI

41 

Zhong X, Chung AC, Chen HY, Meng XM and Lan HY: Smad3-mediated upregulation of miR-21 promotes renal fibrosis. J Am Soc Nephrol. 22:1668–1681. 2011. View Article : Google Scholar : PubMed/NCBI

42 

Wang B, Komers R, Carew R, Winbanks CE, Xu B, Herman-Edelstein M, Koh P, Thomas M, Jandeleit-Dahm K, Gregorevic P, et al: Suppression of microRNA-29 expression by TGF-β1 promotes collagen expression and renal fibrosis. J Am Soc Nephrol. 23:252–265. 2012. View Article : Google Scholar : PubMed/NCBI

43 

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

44 

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

45 

Wei J, Xu Z and Yan X: The role of the macrophage-to-myofibroblast transition in renal fibrosis. Front Immunol. 13:9343772022. View Article : Google Scholar : PubMed/NCBI

46 

Tang PM, Zhang YY, Xiao J, Tang PC, Chung JY, Li J, Xue VW, Huang XR, Chong CC, Ng CF, et al: Neural transcription factor Pou4f1 promotes renal fibrosis via macrophage-myofibroblast transition. Proc Natl Acad Sci USA. 117:20741–20752. 2020. View Article : Google Scholar : PubMed/NCBI

47 

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

48 

Chen J, Tang Y, Zhong Y, Wei B, Huang XR, Tang PM, Xu A and Lan HY: P2Y12 inhibitor clopidogrel inhibits renal fibrosis by blocking macrophage-to-myofibroblast transition. Mol Ther. 30:3017–3033. 2022. View Article : Google Scholar : PubMed/NCBI

49 

Xavier S, Vasko R, Matsumoto K, Zullo JA, Chen R, Maizel J, Chander PN and Goligorsky MS: Curtailing endothelial TGF-β signaling is sufficient to reduce endothelial-mesenchymal transition and fibrosis in CKD. J Am Soc Nephrol. 26:817–829. 2015. View Article : Google Scholar : PubMed/NCBI

50 

LeBleu VS, Taduri G, O'Connell J, Teng Y, Cooke VG, Woda C, Sugimoto H and Kalluri R: Origin and function of myofibroblasts in kidney fibrosis. Nat Med. 19:1047–1053. 2013. View Article : Google Scholar : PubMed/NCBI

51 

Loeffler I, Liebisch M, Allert S, Kunisch E, Kinne RW and Wolf G: FSP1-specific SMAD2 knockout in renal tubular, endothelial, and interstitial cells reduces fibrosis and epithelial-to-mesenchymal transition in murine STZ-induced diabetic nephropathy. Cell Tissue Res. 372:115–133. 2018. View Article : Google Scholar : PubMed/NCBI

52 

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

53 

Li MO and Flavell RA: TGF-beta: A master of all T cell trades. Cell. 134:392–404. 2008. View Article : Google Scholar : PubMed/NCBI

54 

Li MO, Wan YY, Sanjabi S, Robertson AKL and Flavell RA: Transforming growth factor-beta regulation of immune responses. Annu Rev Immunol. 24:99–146. 2006. View Article : Google Scholar : PubMed/NCBI

55 

Kulkarni AB, Huh CG, Becker D, Geiser A, Lyght M, Flanders KC, Roberts AB, Sporn MB, Ward JM and Karlsson S: Transforming growth factor beta 1 null mutation in mice causes excessive inflammatory response and early death. Proc Natl Acad Sci USA. 90:770–774. 1993. View Article : Google Scholar : PubMed/NCBI

56 

Yaswen L, Kulkarni AB, Fredrickson T, Mittleman B, Schiffman R, Payne S, Longenecker G, Mozes E and Karlsson S: Autoimmune manifestations in the transforming growth factor-beta 1 knockout mouse. Blood. 87:1439–1445. 1996. View Article : Google Scholar : PubMed/NCBI

57 

Werner F, Jain MK, Feinberg MW, Sibinga NE, Pellacani A, Wiesel P, Chin MT, Topper JN, Perrella MA and Lee ME: Transforming growth factor-beta 1 inhibition of macrophage activation is mediated via Smad3. J Biol Chem. 275:36653–36658. 2000. View Article : Google Scholar : PubMed/NCBI

58 

Martinez GJ, Zhang Z, Chung Y, Reynolds JM, Lin X, Jetten AM, Feng XH and Dong C: Smad3 differentially regulates the induction of regulatory and inflammatory T cell differentiation. J Biol Chem. 284:35283–35286. 2009. View Article : Google Scholar : PubMed/NCBI

59 

Zhang F, Tsai S, Kato K, Yamanouchi D, Wang C, Rafii S, Liu B and Kent KC: Transforming growth factor-beta promotes recruitment of bone marrow cells and bone marrow-derived mesenchymal stem cells through stimulation of MCP-1 production in vascular smooth muscle cells. J Biol Chem. 284:17564–17574. 2009. View Article : Google Scholar : PubMed/NCBI

60 

Zhang YY, Tang PM, Tang PC, Xiao J, Huang XR, Yu C, Ma RCW and Lan HY: LRNA9884, a Novel Smad3-Dependent long noncoding rna, promotes diabetic kidney injury in db/db Mice via Enhancing MCP-1-Dependent renal inflammation. Diabetes. 68:1485–1498. 2019. View Article : Google Scholar : PubMed/NCBI

61 

Zhou Q, Huang XR, Yu J, Yu X and Lan HY: Long Noncoding RNA Arid2-IR Is a Novel Therapeutic Target for Renal Inflammation. Mol Ther. 23:1034–1043. 2015. View Article : Google Scholar : PubMed/NCBI

62 

Wang W, Huang XR, Li AG, Liu F, Li JH, Truong LD, Wang XJ and Lan HY: Signaling mechanism of TGF-beta1 in prevention of renal inflammation: Role of Smad7. J Am Soc Nephrol. 16:1371–1383. 2005. View Article : Google Scholar : PubMed/NCBI

63 

Lan HY: Smad7 as a therapeutic agent for chronic kidney diseases. Front Biosci. 13:4984–4992. 2008. View Article : Google Scholar : PubMed/NCBI

64 

Chung AC, Huang XR, Zhou L, Heuchel R, Lai KN and Lan HY: Disruption of the Smad7 gene promotes renal fibrosis and inflammation in unilateral ureteral obstruction (UUO) in mice. Nephrol Dial Transplant. 24:1443–1454. 2009. View Article : Google Scholar : PubMed/NCBI

65 

You YK, Wu WF, Huang XR, Li HD, Ren YP, Zeng JC, Chen H and Lan HY: Deletion of Smad3 protects against C-reactive protein-induced renal fibrosis and inflammation in obstructive nephropathy. Int J Biol Sci. 17:3911–3922. 2021. View Article : Google Scholar : PubMed/NCBI

66 

Liu Z, Huang XR, Chen HY, Fung E, Liu J and Lan HY: Deletion of angiotensin-converting enzyme-2 promotes hypertensive nephropathy by targeting Smad7 for ubiquitin degradation. Hypertension. 70:822–830. 2017. View Article : Google Scholar : PubMed/NCBI

67 

Wang Y, Wang Y, Yang M and Ma X: Implication of cellular senescence in the progression of chronic kidney disease and the treatment potencies. Biomed Pharmacother. 135:1111912021. View Article : Google Scholar : PubMed/NCBI

68 

Zhang JQ, Li YY, Zhang XY, Tian ZH, Liu C, Wang ST and Zhang FR: Cellular senescence of renal tubular epithelial cells in renal fibrosis. Front Endocrinol (Lausanne). 14:10856052023. View Article : Google Scholar : PubMed/NCBI

69 

Li C, Shen Y, Huang L, Liu C and Wang J: Senolytic therapy ameliorates renal fibrosis postacute kidney injury by alleviating renal senescence. FASEB J. 35:e212292021.PubMed/NCBI

70 

Lyu G, Guan Y, Zhang C, Zong L, Sun L, Huang X, Huang L, Zhang L, Tian XL, Zhou Z and Tao W: TGF-β signaling alters H4K20me3 status via miR-29 and contributes to cellular senescence and cardiac aging. Nat Commun. 9:25602018. View Article : Google Scholar : PubMed/NCBI

71 

Ni JY, Wang X, Xie HY, Yang NH, Li JY, Sun XA, Guo HJ, Zhou L, Zhang W, Liu J and Lu LM: Deubiquitinating enzyme USP11 promotes renal tubular cell senescence and fibrosis via inhibiting the ubiquitin degradation of TGF-β receptor II. Acta Pharmacol Sin. 44:584–595. 2023. View Article : Google Scholar : PubMed/NCBI

72 

Ueda S, Tominaga T, Ochi A, Sakurai A, Nishimura K, Shibata E, Wakino S, Tamaki M and Nagai K: TGF-β1 is involved in senescence-related pathways in glomerular endothelial cells via p16 translocation and p21 induction. Sci Rep. 11:216432021. View Article : Google Scholar : PubMed/NCBI

73 

Tang C, Livingston MJ, Liu Z and Dong Z: Autophagy in kidney homeostasis and disease. Nat Rev Nephrol. 16:489–508. 2020. View Article : Google Scholar : PubMed/NCBI

74 

Yang C, Chen XC, Li ZH, Wu HL, Jing KP, Huang XR, Ye L, Wei B, Lan HY and Liu HF: SMAD3 promotes autophagy dysregulation by triggering lysosome depletion in tubular epithelial cells in diabetic nephropathy. Autophagy. 17:2325–2344. 2021. View Article : Google Scholar : PubMed/NCBI

75 

Zehender A, Li YN, Lin NY, Stefanica A, Nüchel J, Chen CW, Hsu HH, Zhu H, Ding X, Huang J, et al: TGFβ promotes fibrosis by MYST1-dependent epigenetic regulation of autophagy. Nat Commun. 12:44042021. View Article : Google Scholar : PubMed/NCBI

76 

Sanz AB, Sanchez-Niño MD, Ramos AM and Ortiz A: Regulated cell death pathways in kidney disease. Nat Rev Nephrol. 19:281–299. 2023. View Article : Google Scholar : PubMed/NCBI

77 

Massagué J, Blain SW and Lo RS: TGFbeta signaling in growth control, cancer, and heritable disorders. Cell. 103:295–309. 2000. View Article : Google Scholar : PubMed/NCBI

78 

Wang W, Chen J, Hu D, Pan P, Liang L, Wu W, Tang Y, Huang XR, Yu X, Wu J and Lan HY: SARS-CoV-2 N protein induces acute kidney injury via Smad3-Dependent G1 cell cycle arrest mechanism. Adv Sci (Weinh). 9:e21032482022. View Article : Google Scholar : PubMed/NCBI

79 

Fu S, Tang Y, Huang XR, Feng M, Xu AP and Lan HY: Smad7 protects against acute kidney injury by rescuing tubular epithelial cells from the G1 cell cycle arrest. Clin Sci (Lond). 131:1955–1969. 2017. View Article : Google Scholar : PubMed/NCBI

80 

Zhang HY, Cheng M, Zhang L and Wang YP: Ferroptosis and renal fibrosis: A new target for the future (Review). Exp Ther Med. 25:132022. View Article : Google Scholar : PubMed/NCBI

81 

Wang J, Wang Y, Liu Y, Cai X, Huang X, Fu W, Wang L, Qiu L, Li J and Sun L: Ferroptosis, a new target for treatment of renal injury and fibrosis in a 5/6 nephrectomy-induced CKD rat model. Cell Death Discov. 8:1272022. View Article : Google Scholar : PubMed/NCBI

82 

Wang JN, Yang Q, Yang C, Cai YT, Xing T, Gao L, Wang F, Chen X, Liu XQ, He XY, et al: Smad3 promotes AKI sensitivity in diabetic mice via interaction with p53 and induction of NOX4-dependent ROS production. Redox Biol. 32:1014792020. View Article : Google Scholar : PubMed/NCBI

83 

Li J, Yang J, Xian Q, Su H, Ni Y and Wang L: Kaempferitrin attenuates unilateral ureteral obstruction-induced renal inflammation and fibrosis in mice by inhibiting NOX4-mediated tubular ferroptosis. Phytother Res. Mar 15–2024.(Epub ahead of print). View Article : Google Scholar

84 

Zhu B, Ni Y, Gong Y, Kang X, Guo H, Liu X, Li J and Wang L: Formononetin ameliorates ferroptosis-associated fibrosis in renal tubular epithelial cells and in mice with chronic kidney disease by suppressing the Smad3/ATF3/SLC7A11 signaling. Life Sci. 315:1213312023. View Article : Google Scholar : PubMed/NCBI

85 

Streets A and Ong A: Post-translational modifications of the polycystin proteins. Cell Signal. 72:1096442020. View Article : Google Scholar : PubMed/NCBI

86 

Duan G and Walther D: The roles of post-translational modifications in the context of protein interaction networks. PLoS Comput Biol. 11:e10040492015. View Article : Google Scholar : PubMed/NCBI

87 

Xu P, Liu J and Derynck R: Post-translational regulation of TGF-β receptor and Smad signaling. FEBS Lett. 586:1871–1884. 2012. View Article : Google Scholar : PubMed/NCBI

88 

Huse M, Muir TW, Xu L, Chen YG, Kuriyan J and Massagué J: The TGF beta receptor activation process: An inhibitor- to substrate-binding switch. Mol Cell. 8:671–682. 2001. View Article : Google Scholar : PubMed/NCBI

89 

Shi Y and Massagué J: Mechanisms of TGF-beta signaling from cell membrane to the nucleus. Cell. 113:685–700. 2003. View Article : Google Scholar : PubMed/NCBI

90 

Xu P, Lin X and Feng XH: Posttranslational Regulation of Smads. Cold Spring Harb Perspect Biol. 8:a0220872016. View Article : Google Scholar : PubMed/NCBI

91 

Adhikary L, Chow F, Nikolic-Paterson DJ, Stambe C, Dowling J, Atkins RC and Tesch GH: Abnormal p38 mitogen-activated protein kinase signalling in human and experimental diabetic nephropathy. Diabetologia. 47:1210–1222. 2004. View Article : Google Scholar : PubMed/NCBI

92 

De Borst MH, Prakash J, Melenhorst WB, van den Heuvel MC, Kok RJ, Navis G and van Goor H: Glomerular and tubular induction of the transcription factor c-Jun in human renal disease. J Pathol. 213:219–228. 2007. View Article : Google Scholar : PubMed/NCBI

93 

Ma FY, Sachchithananthan M, Flanc RS and Nikolic-Paterson DJ: Mitogen activated protein kinases in renal fibrosis. Front Biosci (Schol Ed). 1:171–187. 2009. View Article : Google Scholar : PubMed/NCBI

94 

Stambe C, Atkins RC, Tesch GH, Masaki T, Schreiner GF and Nikolic-Paterson DJ: The role of p38alpha mitogen-activated protein kinase activation in renal fibrosis. J Am Soc Nephrol. 15:370–379. 2004. View Article : Google Scholar : PubMed/NCBI

95 

Ma FY, Flanc RS, Tesch GH, Bennett BL, Friedman GC and Nikolic-Paterson DJ: Blockade of the c-Jun amino terminal kinase prevents crescent formation and halts established anti-GBM glomerulonephritis in the rat. Lab Invest. 89:470–484. 2009. View Article : Google Scholar : PubMed/NCBI

96 

Müller R, Daniel C, Hugo C, Amann K, Mielenz D, Endlich K, Braun T, van der Veen B, Heeringa P, Schett G and Zwerina J: The mitogen-activated protein kinase p38α regulates tubular damage in murine anti-glomerular basement membrane nephritis. PLoS One. 8:e563162013. View Article : Google Scholar : PubMed/NCBI

97 

Kamato D, Burch ML, Piva TJ, Rezaei HB, Rostam MA, Xu S, Zheng W, Little PJ and Osman N: Transforming growth factor-β signalling: Role and consequences of Smad linker region phosphorylation. Cell Signal. 25:2017–2024. 2013. View Article : Google Scholar : PubMed/NCBI

98 

Yang F, Chung ACK, Huang XR and Lan HY: Angiotensin II induces connective tissue growth factor and collagen I expression via transforming growth factor-beta-dependent and -independent Smad pathways: The role of Smad3. Hypertension. 54:877–884. 2009. View Article : Google Scholar : PubMed/NCBI

99 

Chung AC, Zhang H, Kong YZ, Tan JJ, Huang XR, Kopp JB and Lan HY: Advanced glycation end-products induce tubular CTGF via TGF-beta-independent Smad3 signaling. J Am Soc Nephrol. 21:249–260. 2010. View Article : Google Scholar : PubMed/NCBI

100 

You YK, Huang XR, Chen HY, Lyu XF, Liu HF and Lan HY: C-Reactive protein promotes diabetic kidney disease in db/db Mice via the CD32b-Smad3-mTOR signaling pathway. Sci Rep. 6:267402016. View Article : Google Scholar : PubMed/NCBI

101 

Chen X, Wang H, Liao HJ, Hu W, Gewin L, Mernaugh G, Zhang S, Zhang ZY, Vega-Montoto L, Vanacore RM, et al: Integrin-mediated type II TGF-β receptor tyrosine dephosphorylation controls SMAD-dependent profibrotic signaling. J Clin Invest. 124:3295–3310. 2014. View Article : Google Scholar : PubMed/NCBI

102 

Lin X, Duan X, Liang YY, Su Y, Wrighton KH, Long J, Hu M, Davis CM, Wang J, Brunicardi F, et al: PPM1A functions as a Smad phosphatase to terminate TGFbeta signaling. Cell. 125:915–928. 2006. View Article : Google Scholar : PubMed/NCBI

103 

Samarakoon R, Rehfuss A, Khakoo NS, Falke LL, Dobberfuhl AD, Helo S, Overstreet JM, Goldschmeding R and Higgins PJ: Loss of expression of protein phosphatase magnesium-dependent 1A during kidney injury promotes fibrotic maladaptive repair. FASEB J. 30:3308–3320. 2016. View Article : Google Scholar : PubMed/NCBI

104 

Inoue K, Matsui I, Hamano T, Fujii N, Shimomura A, Nakano C, Kusunoki Y, Takabatake Y, Hirata M, Nishiyama A, et al: Maxacalcitol ameliorates tubulointerstitial fibrosis in obstructed kidneys by recruiting PPM1A/VDR complex to pSmad3. Lab Invest. 92:1686–1697. 2012. View Article : Google Scholar : PubMed/NCBI

105 

Tang J, Goldschmeding R, Samarakoon R and Higgins PJ: Protein phosphatase Mg2+/Mn2+ dependent-1A and PTEN deregulation in renal fibrosis: Novel mechanisms and co-dependency of expression. FASEB J. 34:2641–2656. 2020. View Article : Google Scholar : PubMed/NCBI

106 

Meyer-Schwesinger C: The ubiquitin-proteasome system in kidney physiology and disease. Nat Rev Nephrol. 15:393–411. 2019. View Article : Google Scholar : PubMed/NCBI

107 

Tan R, He W, Lin X, Kiss LP and Liu Y: Smad ubiquitination regulatory factor-2 in the fibrotic kidney: Regulation, target specificity, and functional implication. Am J Physiol Renal Physiol. 294:F1076–F1083. 2008. View Article : Google Scholar : PubMed/NCBI

108 

Iyengar PV: Regulation of ubiquitin enzymes in the TGF-β Pathway. Int J Mol Sci. 18:8772017. View Article : Google Scholar : PubMed/NCBI

109 

Bonni S, Wang HR, Causing CG, Kavsak P, Stroschein SL, Luo K and Wrana JL: TGF-beta induces assembly of a Smad2-Smurf2 ubiquitin ligase complex that targets SnoN for degradation. Nat Cell Biol. 3:587–595. 2001. View Article : Google Scholar : PubMed/NCBI

110 

Wang L, Zha H, Huang J and Shi L: Flavin containing monooxygenase 2 regulates renal tubular cell fibrosis and paracrine secretion via SMURF2 in AKI-CKD transformation. Int J Mol Med. 52:1102023. View Article : Google Scholar : PubMed/NCBI

111 

Liu FY, Li XZ, Peng YM, Liu H and Liu YH: Arkadia-Smad7-mediated positive regulation of TGF-beta signaling in a rat model of tubulointerstitial fibrosis. Am J Nephrol. 27:176–183. 2007. View Article : Google Scholar : PubMed/NCBI

112 

Liu FY and Li XZ: The roles of Arkadia in renal tubular epithelial to mesenchymal transition. Med Hypotheses. 67:1205–1207. 2006. View Article : Google Scholar : PubMed/NCBI

113 

Wu W, Huang XR, You Y, Xue L, Wang XJ, Meng X, Lin X, Shen J, Yu X, Lan HY and Chen H: Latent TGF-β1 protects against diabetic kidney disease via Arkadia/Smad7 signaling. Int J Biol Sci. 17:3583–3594. 2021. View Article : Google Scholar : PubMed/NCBI

114 

Gao S, Alarcón C, Sapkota G, Rahman S, Chen PY, Goerner N, Macias MJ, Erdjument-Bromage H, Tempst P and Massagué J: Ubiquitin ligase Nedd4L targets activated Smad2/3 to limit TGF-beta signaling. Mol Cell. 36:457–468. 2009. View Article : Google Scholar : PubMed/NCBI

115 

Manning JA, Shah SS, Nikolic A, Henshall TL, Khew-Goodall Y and Kumar S: The ubiquitin ligase NEDD4-2/NEDD4L regulates both sodium homeostasis and fibrotic signaling to prevent end-stage renal disease. Cell Death Dis. 12:3982021. View Article : Google Scholar : PubMed/NCBI

116 

Henshall TL, Manning JA, Alfassy OS, Goel P, Boase NA, Kawabe H and Kumar S: Deletion of Nedd4-2 results in progressive kidney disease in mice. Cell Death Differ. 24:2150–2160. 2017. View Article : Google Scholar : PubMed/NCBI

117 

Nijman SM, Luna-Vargas MP, Velds A, Brummelkamp TR, Dirac AM, Sixma TK and Bernards R: A genomic and functional inventory of deubiquitinating enzymes. Cell. 123:773–786. 2005. View Article : Google Scholar : PubMed/NCBI

118 

Zhang J, Zhang X, Xie F, Zhang Z, van Dam H, Zhang L and Zhou F: The regulation of TGF-β/SMAD signaling by protein deubiquitination. Protein Cell. 5:503–517. 2014. View Article : Google Scholar : PubMed/NCBI

119 

Komander D, Clague MJ and Urbé S: Breaking the chains: structure and function of the deubiquitinases. Nat Rev Mol Cell Biol. 10:550–563. 2009. View Article : Google Scholar : PubMed/NCBI

120 

Soji K, Doi S, Nakashima A, Sasaki K, Doi T and Masaki T: Deubiquitinase inhibitor PR-619 reduces Smad4 expression and suppresses renal fibrosis in mice with unilateral ureteral obstruction. PLoS One. 13:e02024092018. View Article : Google Scholar : PubMed/NCBI

121 

Dupont S, Mamidi A, Cordenonsi M, Montagner M, Zacchigna L, Adorno M, Martello G, Stinchfield MJ, Soligo S, Morsut L, et al: FAM/USP9×, a deubiquitinating enzyme essential for TGFbeta signaling, controls Smad4 monoubiquitination. Cell. 136:123–135. 2009. View Article : Google Scholar : PubMed/NCBI

122 

Xie S, Xing Y, Shi W, Zhang M, Chen M, Fang W, Liu S, Zhang T, Zeng X, Chen S, et al: Cardiac fibroblast heat shock protein 47 aggravates cardiac fibrosis post myocardial ischemia-reperfusion injury by encouraging ubiquitin specific peptidase 10 dependent Smad4 deubiquitination. Acta Pharm Sin B. 12:4138–4153. 2022. View Article : Google Scholar : PubMed/NCBI

123 

Liao X, Li Y, Liu J, Zhang Y, Tan J, Kass DJ, Rojas M, Mallampalli RK, Zhao J and Zhao Y: Deubiquitinase USP13 promotes extracellular matrix expression by stabilizing Smad4 in lung fibroblast cells. Transl Res. 223:15–24. 2020. View Article : Google Scholar : PubMed/NCBI

124 

Song C, Liu W and Li J: USP17 is upregulated in osteosarcoma and promotes cell proliferation, metastasis, and epithelial-mesenchymal transition through stabilizing SMAD4. Tumour Biol. 39:10104283177171382017. View Article : Google Scholar : PubMed/NCBI

125 

Zhao Y, Chen X, Lin Y, Li Z, Su X, Fan S, Chen Y, Wang X and Liang G: USP25 inhibits renal fibrosis by regulating TGFβ-SMAD signaling pathway in Ang II-induced hypertensive mice. Biochim Biophys Acta Mol Basis Dis. 1869:1667132023. View Article : Google Scholar : PubMed/NCBI

126 

Sun XH, Xiao HM, Zhang M, Lin ZY, Yang Y, Chen R, Liu PQ, Huang KP and Huang HQ: USP9X deubiquitinates connexin43 to prevent high glucose-induced epithelial-to-mesenchymal transition in NRK-52E cells. Biochem Pharmacol. 188:1145622021. View Article : Google Scholar : PubMed/NCBI

127 

Huang K and Zhao X: USP9X prevents AGEs-induced upregulation of FN and TGF-β1 through activating Nrf2-ARE pathway in rat glomerular mesangial cells. Exp Cell Res. 393:1121002020. View Article : Google Scholar : PubMed/NCBI

128 

Gao F, Qian M, Liu G, Ao W, Dai D and Yin C: USP10 alleviates sepsis-induced acute kidney injury by regulating Sirt6-mediated Nrf2/ARE signaling pathway. J Inflamm (Lond). 18:252021. View Article : Google Scholar : PubMed/NCBI

129 

Huang Z, Shen S, Wang M, Li W, Wu G, Huang W, Luo W and Liang G: Mouse endothelial OTUD1 promotes angiotensin II-induced vascular remodeling by deubiquitinating SMAD3. EMBO Rep. 24:e561352023. View Article : Google Scholar : PubMed/NCBI

130 

Huang YT, Cheng AC, Tang HC, Huang GC, Cai L, Lin TH, Wu KJ, Tseng PH, Wang GG and Chen WY: USP7 facilitates SMAD3 autoregulation to repress cancer progression in p53-deficient lung cancer. Cell Death Dis. 12:8802021. View Article : Google Scholar : PubMed/NCBI

131 

Wicks SJ, Haros K, Maillard M, Song L, Cohen RE, Dijke PT and Chantry A: The deubiquitinating enzyme UCH37 interacts with Smads and regulates TGF-beta signalling. Oncogene. 24:8080–8084. 2005. View Article : Google Scholar : PubMed/NCBI

132 

Tian Y, Liao F and Wu G, Chang D, Yang Y, Dong X, Zhang Z, Zhang Y and Wu G: Ubiquitination and regulation of Smad7 in the TGF-β1/Smad signaling of aristolochic acid nephropathy. Toxicol Mech Methods. 25:645–652. 2015. View Article : Google Scholar : PubMed/NCBI

133 

Zhao Y, Thornton AM, Kinney MC, Ma CA, Spinner JJ, Fuss IJ, Shevach EM and Jain A: The Deubiquitinase CYLD targets Smad7 protein to regulate transforming growth factor β (TGF-β) signaling and the development of regulatory T cells. J Biol Chem. 286:40520–40530. 2011. View Article : Google Scholar : PubMed/NCBI

134 

Wang B, Xu X, Yang Z, Zhang L and Liu Y, Ma A, Xu G, Tang M, Jing T, Wu L and Liu Y: POH1 contributes to hyperactivation of TGF-β signaling and facilitates hepatocellular carcinoma metastasis through deubiquitinating TGF-β receptors and caveolin-1. EBioMedicine. 41:320–332. 2019. View Article : Google Scholar : PubMed/NCBI

135 

Shi Y, Tao M, Chen H, Ma X, Wang Y, Hu Y, Zhou X, Li J, Cui B, Qiu A, et al: Ubiquitin-specific protease 11 promotes partial epithelial-to-mesenchymal transition by deubiquitinating the epidermal growth factor receptor during kidney fibrosis. Kidney Int. 103:544–564. 2023. View Article : Google Scholar : PubMed/NCBI

136 

Jacko AM, Nan L, Li S, Tan J, Zhao J, Kass DJ and Zhao Y: De-ubiquitinating enzyme, USP11, promotes transforming growth factor β-1 signaling through stabilization of transforming growth factor β receptor II. Cell Death Dis. 7:e24742016. View Article : Google Scholar : PubMed/NCBI

137 

Du C, Chen X, Su Q, Lu W, Wang Q, Yuan H, Zhang Z, Wang X, Wu H and Qi Y: The function of SUMOylation and Its critical roles in cardiovascular diseases and potential clinical implications. Int J Mol Sci. 22:106182021. View Article : Google Scholar : PubMed/NCBI

138 

Wang X, Liu T, Huang Y, Dai Y and Lin H: Regulation of transforming growth factor-beta signalling by SUMOylation and its role in fibrosis. Open Biol. 11:2100432021. View Article : Google Scholar : PubMed/NCBI

139 

Kang JS, Saunier EF, Akhurst RJ and Derynck R: The type I TGF-beta receptor is covalently modified and regulated by sumoylation. Nat Cell Biol. 10:654–664. 2008. View Article : Google Scholar : PubMed/NCBI

140 

Enserink JM: Sumo and the cellular stress response. Cell Div. 10:42015. View Article : Google Scholar : PubMed/NCBI

141 

Reverter D and Lima CD: A basis for SUMO protease specificity provided by analysis of human Senp2 and a Senp2-SUMO complex. Structure. 12:1519–1531. 2004. View Article : Google Scholar : PubMed/NCBI

142 

Gong L and Yeh ETH: Characterization of a family of nucleolar SUMO-specific proteases with preference for SUMO-2 or SUMO-3. J Biol Chem. 281:15869–15877. 2006. View Article : Google Scholar : PubMed/NCBI

143 

Tan M, Zhang D, Zhang E, Xu D, Liu Z, Qiu J, Fan Y and Shen B: SENP2 suppresses epithelial-mesenchymal transition of bladder cancer cells through deSUMOylation of TGF-βRI. Mol Carcinog. 56:2332–2341. 2017. View Article : Google Scholar : PubMed/NCBI

144 

Long J, Wang G, He D and Liu F: Repression of Smad4 transcriptional activity by SUMO modification. Biochem J. 379((Pt 1)): 23–29. 2004. View Article : Google Scholar : PubMed/NCBI

145 

Zhou X, Gao C, Huang W, Yang M, Chen G, Jiang L, Gou F, Feng H, Ai N and Xu Y: High glucose induces sumoylation of Smad4 via SUMO2/3 in mesangial cells. Biomed Res Int. 2014:7826252014. View Article : Google Scholar : PubMed/NCBI

146 

Liu P, Zhang J, Wang Y, Wang C, Qiu X and Chen DQ: Natural products against renal fibrosis via modulation of SUMOylation. Front Pharmacol. 13:8008102022. View Article : Google Scholar : PubMed/NCBI

147 

Liu W, Yuan Q, Cao S, Wang G, Liu X, Xia Y, Bian Y, Xu F and Chen Y: Review: Acetylation mechanisms and targeted therapies in cardiac fibrosis. Pharmacol Res. 193:1068152023. View Article : Google Scholar : PubMed/NCBI

148 

Bugyei-Twum A, Advani A, Advani SL, Zhang Y, Thai K, Kelly DJ and Connelly KA: High glucose induces Smad activation via the transcriptional coregulator p300 and contributes to cardiac fibrosis and hypertrophy. Cardiovasc Diabetol. 13:892014. View Article : Google Scholar : PubMed/NCBI

149 

Inoue Y, Itoh Y, Abe K, Okamoto T, Daitoku H, Fukamizu A, Onozaki K and Hayashi H: Smad3 is acetylated by p300/CBP to regulate its transactivation activity. Oncogene. 26:500–508. 2007. View Article : Google Scholar : PubMed/NCBI

150 

Rai R, Verma SK, Kim D, Ramirez V, Lux E, Li C, Sahoo S, Wilsbacher LD, Vaughan DE, Quaggin SE and Ghosh AK: A novel acetyltransferase p300 inhibitor ameliorates hypertension-associated cardio-renal fibrosis. Epigenetics. 12:1004–1013. 2017. View Article : Google Scholar : PubMed/NCBI

151 

Morigi M, Perico L and Benigni A: Sirtuins in renal health and disease. J Am Soc Nephrol. 29:1799–1809. 2018. View Article : Google Scholar : PubMed/NCBI

152 

Huang XZ, Wen D, Zhang M, Xie Q, Ma L, Guan Y, Ren Y, Chen J and Hao CM: Sirt1 activation ameliorates renal fibrosis by inhibiting the TGF-β/Smad3 pathway. J Cell Biochem. 115:996–1005. 2014. View Article : Google Scholar : PubMed/NCBI

153 

Chen Q, Zeng Y, Yang X, Wu Y, Zhang S, Huang S, Zhong Y and Chen M: Resveratrol ameliorates myocardial fibrosis by regulating Sirt1/Smad3 deacetylation pathway in rat model with dilated cardiomyopathy. BMC Cardiovasc Disord. 22:172022. View Article : Google Scholar : PubMed/NCBI

154 

Simic P, Williams EO, Bell EL, Gong JJ, Bonkowski M and Guarente L: SIRT1 suppresses the epithelial-to-mesenchymal transition in cancer metastasis and organ fibrosis. Cell Rep. 3:1175–1186. 2013. View Article : Google Scholar : PubMed/NCBI

155 

Yang S, Yang G, Wang X, Xiang J, Kang L and Liang Z: SIRT2 alleviated renal fibrosis by deacetylating SMAD2 and SMAD3 in renal tubular epithelial cells. Cell Death Dis. 14:6462023. View Article : Google Scholar : PubMed/NCBI

156 

Ma J and Hart GW: O-GlcNAc profiling: From proteins to proteomes. Clin Proteomics. 11:82014. View Article : Google Scholar : PubMed/NCBI

157 

Harosh-Davidovich SB and Khalaila I: O-GlcNAcylation affects β-catenin and E-cadherin expression, cell motility and tumorigenicity of colorectal cancer. Exp Cell Res. 364:42–49. 2018. View Article : Google Scholar : PubMed/NCBI

158 

He XF, Hu X, Wen GJ, Wang Z and Lin WJ: O-GlcNAcylation in cancer development and immunotherapy. Cancer Lett. 566:2162582023. View Article : Google Scholar : PubMed/NCBI

159 

Ma J and Hart GW: Protein O-GlcNAcylation in diabetes and diabetic complications. Expert Rev Proteomics. 10:365–380. 2013. View Article : Google Scholar : PubMed/NCBI

160 

Park J, Lai MKP, Arumugam TV and Jo DG: O-GlcNAcylation as a therapeutic target for Alzheimer's disease. Neuromolecular Med. 22:171–193. 2020. View Article : Google Scholar : PubMed/NCBI

161 

Feng D, Sheng-Dong L, Tong W and Zhen-Xian D: O-GlcNAcylation of RAF1 increases its stabilization and induces the renal fibrosis. Biochim Biophys Acta Mol Basis Dis. 1866:1655562020. View Article : Google Scholar : PubMed/NCBI

162 

Kim YJ, Kang MJ, Kim E, Kweon TH, Park YS, Ji S, Yang WH, Yi EC and Cho JW: O-GlcNAc stabilizes SMAD4 by inhibiting GSK-3β-mediated proteasomal degradation. Sci Rep. 10:199082020. View Article : Google Scholar : PubMed/NCBI

163 

Yuan M, Song ZH, Ying MD, Zhu H, He QJ, Yang B and Cao J: N-myristoylation: From cell biology to translational medicine. Acta Pharmacol Sin. 41:1005–1015. 2020. View Article : Google Scholar : PubMed/NCBI

164 

Stockwell BR and Schreiber SL: TGF-beta-signaling with small molecule FKBP12 antagonists that bind myristoylated FKBP12-TGF-beta type I receptor fusion proteins. Chem Biol. 5:385–395. 1998. View Article : Google Scholar : PubMed/NCBI

165 

Zhu F, Xie N, Jiang Z, Li G, Ma L and Tong T: The cellular senescence-inhibited gene is essential for PPM1A myristoylation to modulate transforming growth factor β signaling. Mol Cell Biol. 38:e00414–18. 2018. View Article : Google Scholar : PubMed/NCBI

166 

Al-Salihi MA, Herhaus L, Macartney T and Sapkota GP: USP11 augments TGFβ signalling by deubiquitylating ALK5. Open Biol. 2:1200632012. View Article : Google Scholar : PubMed/NCBI

167 

Siwy J, Mischak H and Zürbig P: Proteomics and personalized medicine: A focus on kidney disease. Expert Rev Proteomics. 16:773–782. 2019. View Article : Google Scholar : PubMed/NCBI

168 

Giudice G and Petsalaki E: Proteomics and phosphoproteomics in precision medicine: Applications and challenges. Brief Bioinform. 20:767–777. 2019. View Article : Google Scholar : PubMed/NCBI

169 

Xu H, Wu T and Huang L: Therapeutic and delivery strategies of phytoconstituents for renal fibrosis. Adv Drug Deliv Rev. 177:1139112021. View Article : Google Scholar : PubMed/NCBI

170 

Trac N, Ashraf A, Giblin J, Prakash S, Mitragotri S and Chung EJ: Spotlight on genetic kidney diseases: A call for drug delivery and nanomedicine solutions. ACS Nano. 17:6165–6177. 2023. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2024
Volume 30 Issue 2

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li J, Zou Y, Kantapan J, Su H, Wang L and Dechsupa N: TGF‑&beta;/Smad signaling in chronic kidney disease: Exploring post‑translational regulatory perspectives (Review). Mol Med Rep 30: 143, 2024
APA
Li, J., Zou, Y., Kantapan, J., Su, H., Wang, L., & Dechsupa, N. (2024). TGF‑&beta;/Smad signaling in chronic kidney disease: Exploring post‑translational regulatory perspectives (Review). Molecular Medicine Reports, 30, 143. https://doi.org/10.3892/mmr.2024.13267
MLA
Li, J., Zou, Y., Kantapan, J., Su, H., Wang, L., Dechsupa, N."TGF‑&beta;/Smad signaling in chronic kidney disease: Exploring post‑translational regulatory perspectives (Review)". Molecular Medicine Reports 30.2 (2024): 143.
Chicago
Li, J., Zou, Y., Kantapan, J., Su, H., Wang, L., Dechsupa, N."TGF‑&beta;/Smad signaling in chronic kidney disease: Exploring post‑translational regulatory perspectives (Review)". Molecular Medicine Reports 30, no. 2 (2024): 143. https://doi.org/10.3892/mmr.2024.13267