1.Guidong People’s Hospital of Guangxi Zhuang Autonomous Region;2.Guilin Medical University
This work was supported by a grant from Horizontal Collaborative Research Project of Guidong People's Hospital of Guangxi Zhuang Autonomous Region (2025GDHX02) ,the Open Project Program of Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University (GKLCDSM-20230101-03) ,the Open Project Program of Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University (GKLCDSM-KF2025-08) ,Guangxi Medical and Health Key Discipline Construction Project ( Gui Wei Ke Jiao Fa [2021] No. 8), Guangxi Medical and Health Key Cultivation Discipline Construction Project ( Gui Wei Ke Jiao Fa [2023] No. 1), ,Basic Scientific Research Ability Enhancement Program for Young and Middle-aged Teachers in Guangxi Universities (2025KY0509) ,Guangxi Traditional Chinese Medicine Appropriate Technology Development and Promotion Program (GZSY2025082) ,Guangxi Medical and Health Appropriate Technology Development, Popularization and Application Program (S2023108)
Diabetic Nephropathy (DN) is the leading cause of End-Stage Renal Disease (ESRD) globally, representing a major global health burden with limited disease-modifying therapies. Podocyte injury serves as the core pathological hallmark of DN, and conventional treatments targeting metabolic disorders or hemodynamic abnormalities fail to reverse the progressive decline of renal function. Accumulating evidence over the past decade has established that high glucose-induced podocyte pyroptosis—a pro-inflammatory form of programmed cell death—is a key driving force in DN progression, whose core molecular mechanism hinges on the activation of the TXNIP-NLRP3 inflammasome axis. Under sustained hyperglycemic conditions, excessive Reactive Oxygen Species (ROS) are generated via pathways including the polyol pathway, advanced glycation end products (AGEs) accumulation, and mitochondrial dysfunction; concurrently, methylglyoxal (a glucose metabolite) mediates post-translational modification of Thioredoxin-Interacting Protein (TXNIP). These events collectively trigger the dissociation of TXNIP from Thioredoxin (TRX), a redox-regulating protein. The free TXNIP then translocates to the mitochondria, where it binds to NOD-Like Receptor Family Pyrin Domain-Containing Protein 3 (NLRP3) and promotes inflammasome assembly. This assembly activates Cysteine-Aspartic Acid Protease 1 (caspase-1), which cleaves Gasdermin D (GSDMD) to generate its N-terminal fragment (GSDMD-NT). GSDMD-NT oligomerizes to form membrane pores, leading to podocyte swelling, rupture, and the release of pro-inflammatory cytokines Interleukin-1β(IL-1β) and Interleukin-18 (IL-18). These cytokines amplify local inflammatory responses, induce mesangial cell proliferation, and accelerate extracellular matrix deposition, ultimately exacerbating glomerulosclerosis.MCC950, a highly selective NLRP3 inhibitor, exerts its therapeutic effects through a multi-layered mechanism: it binds to the NACHT domain (NAIP, CIITA, HET-E and TP1 Domain) of NLRP3 with nanomolar affinity, forming hydrogen bonds with key residues (Lys-42 and Asp-166) within the ATP-hydrolysis pocket to block ATP hydrolysis, thereby locking NLRP3 in an inactive conformational state. Additionally, MCC950 interferes with the protein-protein interaction between TXNIP and NLRP3, and regulates mitochondrial homeostasis to reduce ROS production. Preclinical studies have demonstrated that MCC950 dose-dependently reduces proteinuria, restores the expression of podocyte-specific markers (Nephrin Protein and Wilms Tumor 1 Protein, WT1), and alleviates podocyte foot process fusion and glomerulosclerosis in both Streptozotocin (STZ)-induced type 1 diabetic models (characterized by absolute insulin deficiency) and db/db type 2 diabetic models (driven by insulin resistance). However, discrepancies in therapeutic outcomes exist across different models—some studies report exacerbated renal inflammation and fibrosis in STZ-induced models—which may stem from differences in disease pathogenesis, intervention timing (early vs. mid-stage disease), and dosing duration.Despite its promising preclinical efficacy, MCC950 faces significant translational challenges, including low oral bioavailability, insufficient podocyte targeting, potential hepatotoxicity, and drug-drug interactions with statins (commonly prescribed to diabetic patients for cardiovascular risk management). Furthermore, off-target effects such as the inhibition of carbonic anhydrase 2 have been identified, raising concerns about its safety profile. Nevertheless, its unique mechanism of action—directly blocking podocyte pyroptosis by targeting the TXNIP-NLRP3 axis—endows it with substantial translational value.In the future, strategies to overcome these barriers are expected to advance its clinical application: targeted delivery via nanocarriers (e.g., PLGA-PEG nanoparticles or nephrin antibody-conjugated systems) to enhance renal accumulation and podocyte specificity; precise patient stratification based on biomarkers such as serum IL-18 and renal TXNIP/NLRP3 expression to identify "inflammation-phenotype" DN patients most likely to benefit; and combination therapy with Sodium-Glucose Cotransporter 2 (SGLT2) inhibitors—whose metabolic benefits synergize with MCC950’s anti-inflammatory effects. These approaches hold great potential to break through clinical translation bottlenecks, offering a novel precise anti-inflammatory treatment option for DN and addressing an unmet clinical need for therapies targeting the inflammatory underpinnings of the disease.
Zheng Hong, Mo Zhong Cheng, Liu Hang, Pan Xi Zhang, Wei Bing. MCC950 Targeted Inhibition of TXNIP-NLRP3 Axis-Mediated Podocyte Pyroptosis in Diabetic Nephropathy[J]. Progress in Biochemistry and Biophysics,,():
Copy

Scan code to follow ® 2025 Website Copyright ICP:京ICP备05023138号-1 京公网安备 11010502031771号
