Category Ranking

98%

Total Visits

921

Avg Visit Duration

2 minutes

Citations

20

Article Abstract

Target identification in natural products plays a critical role in the development of innovative drugs. Bufalin, a compound derived from traditional medicines, has shown promising anti-cancer activity; however, its precise molecular mechanism of action remains unclear. Here, we employ artificial intelligence, molecular docking, and molecular dynamics simulations to elucidate the molecular mechanism of Bufalin. Using an integrated multi-predictive strategy, we identify CYP17A1, ESR1, mTOR, AR, and PRKCD as the potential targets of Bufalin. Subsequent validation via surface plasmon resonance, biotin pulldown, and thermal shift assays confirms Bufalin's direct binding to ESR1, which encodes estrogen receptor alpha (ERα). Molecular docking analyses pinpoint Bufalin's selective interaction with Arg394 on ERα. Molecular dynamic simulations further show that Bufalin acts as a molecular glue, enhancing the interaction between ERα and the E3 ligase STUB1, thereby promoting proteasomal degradation of ERα. Given the therapeutic potential of ERα degradation in overcoming endocrine resistance, we investigate the inhibitory effect of Bufalin on endocrine-resistant models and prove Bufalin reverses Tamoxifen resistance in vitro, in vivo, and in patient-derived breast cancer organoids from tamoxifen-relapsed cases. Collectively, our findings indicate that Bufalin functions as a molecular glue to degrade ERα, offering a potential therapeutic strategy for reversing Tamoxifen resistance.

Download full-text PDF

Source
http://www.ncbi.nlm.nih.gov/pmc/articles/PMC12373995PMC
http://dx.doi.org/10.1038/s41467-025-62288-7DOI Listing

Publication Analysis

Top Keywords

molecular glue
12
molecular
9
artificial intelligence
8
bufalin
8
estrogen receptor
8
receptor alpha
8
molecular mechanism
8
molecular docking
8
erα molecular
8
tamoxifen resistance
8

Similar Publications

Neurodegenerative diseases (NDs), including Alzheimer's, Huntington's, and Parkinson's disease, are associated with significant declines in cognitive function and mobility. The accumulation of misfolded proteins such as β-amyloid, tau, α-synuclein, and polyglutamates is a key factor in the progression of these conditions. Unfortunately, traditional small-molecule drugs face major obstacles in effectively targeting these proteins.

View Article and Find Full Text PDF

Rational optimization of molecular glue degraders (MGD) remains a challenging and lengthy process even after identification of a promising scaffold. Unlike proteolysis targeting chimeras (PROTAC), MGDs rely on induced protein-protein interactions as opposed to direct binding in order to target a protein of interest for degradation. Here, we report the synthesis of MGDs targeting the transcription factor ZBTB11 guided by protein complex modeling.

View Article and Find Full Text PDF

Stabilization of Native Protein-Protein Interactions with Molecular Glues: A 14-3-3 Case Study.

Acc Chem Res

September 2025

Department of Pharmaceutical Chemistry and Small Molecule Discovery Center, University of California, San Francisco 94158, United States.

ConspectusProtein-protein interactions (PPIs) play a key role in homeostasis and are often dysregulated in disease. PPIs were traditionally considered "undruggable" due to their flat surfaces and disordered domains. Recently, the identification of PPI stabilizers, or molecular glues (MGs), compounds that bind cooperatively to PPI interfaces, has provided a new direction for the field.

View Article and Find Full Text PDF

Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by persistent inflammation and joint damage, accompanied by the accumulation of plasma cells, which contributes to its pathogenesis. Understanding the genetic alterations occurring during plasma cell differentiation in RA can deepen our comprehension of its pathogenesis and guide the development of targeted therapeutic interventions. Here, our study elucidates the intricate molecular mechanisms underlying plasma cell differentiation by demonstrating that PRDX1 interacts with DOK3 and modulates its degradation by the autophagy-lysosome pathway.

View Article and Find Full Text PDF

Cancer cells are acutely dependent on nuclear transport due to elevated transcriptional activity, suggesting an unrealized opportunity for selective therapeutic inhibition of the nuclear pore complex. Through large-scale phenotypic profiling of cancer cell lines, genome-scale functional genomic modifier screens, and mass spectrometry-based proteomics, we discovered that the clinical drug PRLX-93936 is a molecular glue that binds and reprograms the TRIM21 ubiquitin ligase to degrade the nuclear pore complex. Upon compound-induced TRIM21 recruitment, the nuclear pore is ubiquitylated and degraded, resulting in the loss of short-lived cytoplasmic mRNA transcripts and induction of cancer cell apoptosis.

View Article and Find Full Text PDF