1
|
Jin B, Lu Z, Cheng C, Pei Y, Chen L, Yue Z, Lin A, Yang S, Mo Y, Jiang X. Factors associated with chronic calcineurin inhibitor nephrotoxicity in children with minimal-change disease. Ren Fail 2025; 47:2474743. [PMID: 40091628 PMCID: PMC11915743 DOI: 10.1080/0886022x.2025.2474743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 02/22/2025] [Accepted: 02/26/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND Calcineurin inhibitors (CNIs), such as cyclosporine (CsA) and tacrolimus (TAC), are commonly used to treat children with complicated minimal change nephrotic syndrome. However, chronic nephrotoxicity associated with CNIs poses a significant safety concern. This study aimed to identify the risk factors that contribute to chronic nephrotoxicity in these patients. MATERIAL AND METHODS Clinical and pathological data of MCD children treated with CsA or TAC in our center between 1 January 2003 and 31 December 2022, were retrospectively reviewed. Kidney biopsies were performed on 80 patients who received CNI treatment for more than 6 months. RESULTS Chronic CNI nephrotoxicity (striped interstitial fibrosis with tubular atrophy) was observed in 15% (12/80) of patients. Higher CNI culminating amounts were shown in patients who developed nephrotoxicity regardless of CsA or TAC treatment. Risk factors for chronic CNI nephrotoxicity included persistent nephrotic-range proteinuria for more than 30 days during CNI treatment, increased urinary NAG level, and CNI resistance. Multivariate analysis revealed that increased urinary NAG level and CNI resistance were the independent risk factors for chronic CNI nephrotoxicity in children with MCD. CONCLUSION MCD children who developed CNI resistance were susceptible to chronic CNI nephrotoxicity. Urinary NAG might be a valuable biomarker for CNI nephrotoxicity prediction in MCD children.
Collapse
Affiliation(s)
- Bei Jin
- Department of Pediatric Nephrology and Rheumatology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Ziji Lu
- Department of Pathology, Zhongshan people’s hospital, Guangdong, China
| | - Cheng Cheng
- Department of Pediatric Nephrology and Rheumatology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yuxin Pei
- Department of Pediatric Nephrology and Rheumatology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Lizhi Chen
- Department of Pediatric Nephrology and Rheumatology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zhihui Yue
- Department of Pediatric Nephrology and Rheumatology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Aihua Lin
- Office of the School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | - Shicong Yang
- Department of Pathology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Ying Mo
- Department of Pediatric Nephrology and Rheumatology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xiaoyun Jiang
- Department of Pediatric Nephrology and Rheumatology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
2
|
Pan Y, Wang Y, Gou S. Proteolysis targeting chimera, molecular glue degrader and hydrophobic tag tethering degrader for targeted protein degradation: Mechanisms, strategies and application. Bioorg Chem 2025; 161:108491. [PMID: 40306190 DOI: 10.1016/j.bioorg.2025.108491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/13/2025] [Accepted: 04/16/2025] [Indexed: 05/02/2025]
Abstract
Targeted protein degradation (TPD) represents a revolutionary approach to drug discovery, offering a novel mechanism that outperforms traditional inhibitors. This approach employs small molecule drugs to induce the ubiquitination and subsequent degradation of target protein via the proteasome or lysosomal pathways. Key strategies within TPD include proteolysis targeting chimeras (PROTACs), hydrophobic tag tethering degraders (HyTTDs), and molecular glue degraders (MGDs). PROTACs have been undergone clinical evaluations, MGDs have been used in the clinic, and HyTTDs have shown significant progress in cancer treatment. Each of these strategies presents unique advantages and approaches to target protein degradation. This review summarizes five years of research on PROTACs, HyTTDs, and MGDs, highlighting their design principles, advantages, limitations, and future challenges to provide clear guidance and in-depth insights for advancing drug development.
Collapse
Affiliation(s)
- Yanchang Pan
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Yuanjiang Wang
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China; Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing 211189, PR China
| | - Shaohua Gou
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China; Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing 211189, PR China.
| |
Collapse
|
3
|
Omar I, Alakhras A, Mutwali S, Bakhiet M. Molecular insights into T cell development, activation and signal transduction (Review). Biomed Rep 2025; 22:94. [PMID: 40247929 PMCID: PMC12001230 DOI: 10.3892/br.2025.1972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 03/28/2025] [Indexed: 04/19/2025] Open
Abstract
T cell modulation plays a fundamental role to adaptive and innate immunity, which aids the recognition and defense against pathogens while also maintaining self-tolerance. Numerous molecular pathways participate in this process including thymic selection, T cell receptor and antigen-presenting cells cross linkage, along with co-stimulatory signaling cascades. The present review demonstrates a holistic analysis of various classic and novel mechanisms that govern T cell regulation and emerging therapeutic applications. Recent advancements have introduced novel roles in the journey of T cell modulation that can have a pivotal impact on the understanding of this process; for example, phase separation of the linker for activation of T cells, and the newer application of chimeric antigen receptor (CAR) T cell therapy in autoimmune diseases. While discoveries of proximal and distal signal transduction pathways have contributed to the comprehension of T cell anergy, cytokine-mediated differentiation and the delicate balance between immune activation and tolerance, there are still unresolved debates about further molecular mechanisms. There are also still questions about the long-term side effects of CAR-T cell therapy. Deeper research and analysis are required to further aid the understanding and use of this novel therapeutic approach.
Collapse
Affiliation(s)
- Isra Omar
- Department of Clinical Medicine, College of Medicine, Almaarefa University, 11597 Riyadh, Kingdom of Saudi Arabia
- Department of Clinical Medicine, College of Medicine, University of Medical Sciences and Technology, 3523 Kigali, Rwanda
- Royal College of Physicians of Ireland, Dublin D02 E434, Ireland
| | - Ahmed Alakhras
- Department of Clinical Medicine, College of Medicine, Almaarefa University, 11597 Riyadh, Kingdom of Saudi Arabia
| | - Samahir Mutwali
- TeleGeriatric Research Fellowship Program, Michigan State University, MI 48824, USA
| | - Moiz Bakhiet
- Department of Molecular Medicine and Medical Science, Arabian Gulf University, Manama 328, Kingdom of Bahrain
| |
Collapse
|
4
|
Umar I, Gulzar SEJ, Sundaramurthy V. M. tuberculosis surface sulfoglycolipid SL-1 activates the mechanosensitive channel TRPV4 to enhance lysosomal biogenesis and exocytosis in macrophages. Mol Biol Cell 2025; 36:ar76. [PMID: 40305098 DOI: 10.1091/mbc.e24-12-0560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2025] Open
Abstract
Intracellular pathogens manipulate host cellular pathways to ensure their survival. Mycobacterium tuberculosis (Mtb) disrupts phagosomal trafficking to prevent fusion with lysosomes. Beyond this localized effect, Mtb globally remodels the host lysosomal system, predominantly through its virulence-associated surface lipid, sulfolipid-1 (SL-1). SL-1 enhances lysosomal biogenesis via the mTORC1-TFEB axis; however, the upstream mediators remain unknown. Here, we show that SL-1 induces calcium influx into macrophages and identify the mechanosensitive calcium channel transient receptor potential vanilloid subtype 4 (TRPV4) as a crucial upstream mediator of SL-1-induced lysosomal remodeling. TRPV4 influences multiple aspects of lysosomal function, including biogenesis, acidification, enzymatic activity, phagosome maturation, and lysosomal exocytosis. These effects are recapitulated during Mtb infection, underscoring the relevance of SL-1- and TRPV4-dependent lysosomal remodeling in an infection context. TRPV4 expression is upregulated during Mtb infection and partially localizes to both lysosomes and the Mtb-containing vacuole. Remarkably, TRPV4 activation, independent of SL-1, is sufficient to enhance lysosomal biogenesis, identifying TRPV4 as a key regulator of lysosomal homeostasis. Together, these findings uncover a novel mechanism of lysosomal remodeling driven by a pathogen lipid virulence factor and reveal a previously unrecognized role for TRPV4 in modulating lysosomal homeostasis in macrophages.
Collapse
Affiliation(s)
- Ibrahim Umar
- National Center for Biological Sciences, Bangalore 560065, India
- SASTRA University Thanjavur 613401, India
| | - Shah-E-Jahan Gulzar
- National Center for Biological Sciences, Bangalore 560065, India
- SASTRA University Thanjavur 613401, India
| | | |
Collapse
|
5
|
Li F, Cai C, Wang F, Zhang N, Zhao Q, Chen Y, Cui X, Wang S, Zhang W, Liu D, Cai Y, Jin J. 20(S)-ginsenoside Rg3 suppresses gastric cancer cell proliferation by inhibiting E2F-DP dimerization. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156740. [PMID: 40252583 DOI: 10.1016/j.phymed.2025.156740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 03/24/2025] [Accepted: 04/07/2025] [Indexed: 04/21/2025]
Abstract
BACKGROUND Gastric cancer (GC) is a common and aggressive malignancy, with treatment options often limited by drug resistance and the adverse effects of targeted therapies and immunotherapy. Ginsenoside Rg3, a bioactive compound derived from ginseng, has shown promise in inhibiting the growth of various tumor types, including GC. However, the molecular mechanisms underlying its therapeutic effects against GC remain insufficiently understood. OBJECTIVE This study aimed to elucidate the molecular mechanisms underlying the anti-cancer effects of ginsenoside Rg3 against GC. METHODS To explore the molecular mechanisms underlying Rg3's anti-GC effects, RNA sequencing (RNA-Seq) was conducted to identify potential Rg3-regulated targets. The interaction between Rg3 and E2F was analyzed using several approaches, including the cellular thermal shift assay (CETSA), Rg3-PEGA pull-down, Rg3 pull-down protein mass spectrometry, and 3D molecular docking. Additionally, quantitative reverse transcription PCR (qRT-PCR), co-transfection followed by immunoprecipitation, Western blotting, flow cytometry, Annexin V-FITC staining, Hoechst staining, and luciferase reporter assays were employed to elucidate the molecular effects of Rg3. The inhibitory effect of Rg3 on GC proliferation was assessed through colony formation assays in vitro and tumor xenograft experiments in C57BL/6 mice in vivo. RESULTS Rg3-mediated gene expression profiling in GC cells revealed several transcription factors, including E2F, and biological processes potentially influenced by Rg3. Consistent with these findings, Rg3 suppressed E2F expression and impeded GC cell proliferation by inducing G1/S cell cycle arrest, reducing cell growth both in vitro and in vivo, enhancing apoptosis, and inhibiting CDC6 transactivation. CETSA and Rg3 pull-down assays confirmed an interaction between Rg3 and E2F. Additionally, 3D molecular docking analysis demonstrated that Rg3 binds with high affinity to E2F at the heterodimeric domain via hydrogen bonding, potentially disrupting E2F-DP heterodimer formation and subsequently inhibiting cell cycle gene expression. In agreement with this, Rg3-treated GC cells exhibited reduced expression of cyclin D1, CDK4, cyclin A, CDK1, and CDK2. Moreover, Rg3 activated the tumor suppressors p53 and p21, further inhibiting RB phosphorylation by suppressing cyclin/CDK activity, thereby blocking transcription of G1/S transition-related genes. CONCLUSION This study provides the first evidence that Rg3 directly binds to E2F proteins, disrupting E2F-DP heterodimer formation and inhibiting the transcription of E2F-DP-regulated target genes. Furthermore, Rg3 activates the p53-p21 pathway while suppressing the cyclin/CDK-RB signaling pathway, effectively inhibiting cancer cell proliferation. These findings highlight a potential therapeutic strategy for developing small molecules structurally similar to Rg3 to target tumors with high E2F expression.
Collapse
Affiliation(s)
- Fuqiang Li
- School of Life Sciences, Jilin University, 2699 Qianjin Street, Chaoyang District, Changchun, Jilin 130012, China; School of Pharmacy, Changchun University of Chinese Medicine, Boshuo Road, Jingyue Development Zone, Changchun, Jilin 130117, China
| | - Chengyu Cai
- School of Life Sciences, Jilin University, 2699 Qianjin Street, Chaoyang District, Changchun, Jilin 130012, China
| | - Fei Wang
- School of Life Sciences, Jilin University, 2699 Qianjin Street, Chaoyang District, Changchun, Jilin 130012, China
| | - Na Zhang
- School of Life Sciences, Jilin University, 2699 Qianjin Street, Chaoyang District, Changchun, Jilin 130012, China
| | - Qingzhi Zhao
- School of Life Sciences, Jilin University, 2699 Qianjin Street, Chaoyang District, Changchun, Jilin 130012, China
| | - Yuyang Chen
- School of Life Sciences, Jilin University, 2699 Qianjin Street, Chaoyang District, Changchun, Jilin 130012, China
| | - Xueli Cui
- School of Life Sciences, Jilin University, 2699 Qianjin Street, Chaoyang District, Changchun, Jilin 130012, China
| | - Siyang Wang
- School of Life Sciences, Jilin University, 2699 Qianjin Street, Chaoyang District, Changchun, Jilin 130012, China
| | - Wenjie Zhang
- School of Life Sciences, Jilin University, 2699 Qianjin Street, Chaoyang District, Changchun, Jilin 130012, China
| | - Da Liu
- School of Pharmacy, Changchun University of Chinese Medicine, Boshuo Road, Jingyue Development Zone, Changchun, Jilin 130117, China.
| | - Yong Cai
- School of Life Sciences, Jilin University, 2699 Qianjin Street, Chaoyang District, Changchun, Jilin 130012, China.
| | - Jingji Jin
- School of Life Sciences, Jilin University, 2699 Qianjin Street, Chaoyang District, Changchun, Jilin 130012, China.
| |
Collapse
|
6
|
Repity ML, Deutscher RCE, Hausch F. Nondegradative Synthetic Molecular Glues Enter the Clinic. ChemMedChem 2025; 20:e202500048. [PMID: 40226972 PMCID: PMC12091845 DOI: 10.1002/cmdc.202500048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 03/07/2025] [Indexed: 04/15/2025]
Abstract
Molecular glues are small molecules that can induce or stabilize protein-protein interactions between proteins inside cells. Unlike classical small molecule drugs, molecular glues can target challenging disease-causing proteins lacking well-defined binding pockets. Nature has repeatedly used this mode of action, but identifying molecular glues for new target proteins has been a major challenge. Recently, manmade molecular glues, inspired by natural products, for KRas, entered clinical trials although KRas is a major cancer target long thought to be undruggable. Here, how these molecules are initially discovered and optimized to provide several advanced drug candidates for various KRas-dependent cancer types are outlined. The major insights obtained for this new class of drug modalities are further summarized. These results showcase how molecular glues that do not rely on protein degradation can provide clinical benefits for challenging drug targets.
Collapse
Affiliation(s)
- Maximilian L. Repity
- Department Chemistry and BiochemistryClemens‐Schöpf‐InstituteTechnical University DarmstadtPeter‐Grünberg Strasse 464287DarmstadtGermany
| | - Robin C. E. Deutscher
- Department Chemistry and BiochemistryClemens‐Schöpf‐InstituteTechnical University DarmstadtPeter‐Grünberg Strasse 464287DarmstadtGermany
| | - Felix Hausch
- Department Chemistry and BiochemistryClemens‐Schöpf‐InstituteTechnical University DarmstadtPeter‐Grünberg Strasse 464287DarmstadtGermany
- Centre for Synthetic BiologyTechnical University64287DarmstadtGermany
| |
Collapse
|
7
|
Kotlyar M, Guo Z, Rao AVS, Peng H, Wang J, Ma Z, Schiene-Fischer C, Fischer G, Liu JO. Identification of Rapaglutin E as an Isoform-Specific Inhibitor of Glucose Transporter 1. ACS Chem Biol 2025; 20:1004-1009. [PMID: 40226990 DOI: 10.1021/acschembio.5c00152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
Natural products rapamycin and FK506 are macrocyclic compounds with therapeutic benefits whose unique scaffold inspired the generation and exploration of hybrid macrocycle rapafucins. From this library, a potent inhibitor of the facilitative glucose transporter (GLUT), rapaglutin A (RgA), was previously identified. RgA is a pan-GLUT inhibitor of Class I isoforms GLUT1, GLUT3, and GLUT4. Herein, we report the discovery of rapaglutin E (RgE). Unlike RgA, RgE is highly specific for GLUT1. Further characterization revealed that RgE and RgA likely bound to distinct sites on GLUT1 despite their shared FKBP-binding domain, suggesting that the distinct effector domains of RgE and RgA play key roles in the recognition of GLUTs.
Collapse
Affiliation(s)
- Marnie Kotlyar
- Chemistry Biology Interface Graduate Program, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Department of Pharmacology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
- The SJ Yan and HJ Mao Laboratory of Chemical Biology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Zufeng Guo
- Department of Pharmacology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
- The SJ Yan and HJ Mao Laboratory of Chemical Biology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - A V Subba Rao
- Department of Pharmacology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
- The SJ Yan and HJ Mao Laboratory of Chemical Biology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Hanjing Peng
- Department of Pharmacology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
- The SJ Yan and HJ Mao Laboratory of Chemical Biology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Jingxin Wang
- Department of Pharmacology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
- The SJ Yan and HJ Mao Laboratory of Chemical Biology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Zhongnan Ma
- Department of Pharmacology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
- The SJ Yan and HJ Mao Laboratory of Chemical Biology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Cordelia Schiene-Fischer
- Department of Enzymology, Institute for Biochemistry and Biotechnology, Martin Luther University Halle-Wittenberg, 06120 Halle, Germany
| | - Gunter Fischer
- Max Planck Institute for Multidisciplinary Sciences, 37077 Göttingen, Germany
| | - Jun O Liu
- Chemistry Biology Interface Graduate Program, Johns Hopkins University, Baltimore, Maryland 21218, United States
- The SJ Yan and HJ Mao Laboratory of Chemical Biology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
- Departments of Pharmacology and Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| |
Collapse
|
8
|
Dreizler JK, Meyners C, Sugiarto WO, Repity ML, Maciel EVS, Purder PL, Lermyte F, Knapp S, Hausch F. Broad Target Screening Reveals Abundance of FKBP12-Based Molecular Glues in Focused Libraries. J Med Chem 2025; 68:9525-9536. [PMID: 40336336 DOI: 10.1021/acs.jmedchem.5c00220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
Competitive (nondegradative) molecular glues represent a promising drug modality that remains underexplored primarily due to the lack of adequate hit identification approaches. In this study, we screened our historically grown FKBP-focused library containing >1000 drug-like molecules to identify FKBP-assisted molecular glues targeting a diverse panel of 57 proteins. In addition to establishing a robust and generalizable screening approach, we discovered three novel FKBP-dependent molecular glues targeting PTPRN, BRD4BD2, and STAT4. Our results demonstrate that molecular glues are more common than previously thought and that they can be identified by repurposing existing focused libraries. An optimized, highly cooperative FKBP12-BRD4BD2 glue demonstrated the involvement of the BD2 pocket and exhibited selectivity over the closely related BD1 domain. Our results underscore the value of FKBP12-assisted molecular glues to target challenging proteins with the potential for high selectivity.
Collapse
Affiliation(s)
- Johannes K Dreizler
- Department of Chemistry and Biochemistry Clemens-Schöpf-Institute, Technical University Darmstadt, Peter-Grünberg Straße 4, 64287 Darmstadt, Germany
| | - Christian Meyners
- Department of Chemistry and Biochemistry Clemens-Schöpf-Institute, Technical University Darmstadt, Peter-Grünberg Straße 4, 64287 Darmstadt, Germany
| | - Wisely Oki Sugiarto
- Department of Chemistry and Biochemistry Clemens-Schöpf-Institute, Technical University Darmstadt, Peter-Grünberg Straße 4, 64287 Darmstadt, Germany
| | - Maximilian L Repity
- Department of Chemistry and Biochemistry Clemens-Schöpf-Institute, Technical University Darmstadt, Peter-Grünberg Straße 4, 64287 Darmstadt, Germany
| | - Edvaldo V S Maciel
- Department of Chemistry and Biochemistry Clemens-Schöpf-Institute, Technical University Darmstadt, Peter-Grünberg Straße 4, 64287 Darmstadt, Germany
| | - Patrick L Purder
- Department of Chemistry and Biochemistry Clemens-Schöpf-Institute, Technical University Darmstadt, Peter-Grünberg Straße 4, 64287 Darmstadt, Germany
| | - Frederik Lermyte
- Department of Chemistry and Biochemistry Clemens-Schöpf-Institute, Technical University Darmstadt, Peter-Grünberg Straße 4, 64287 Darmstadt, Germany
| | - Stefan Knapp
- Institute of Pharmaceutical Chemistry and Structural Genomics Consortium (SGC), Goethe University Frankfurt, 60438 Frankfurt am Main, Germany
| | - Felix Hausch
- Department of Chemistry and Biochemistry Clemens-Schöpf-Institute, Technical University Darmstadt, Peter-Grünberg Straße 4, 64287 Darmstadt, Germany
- Center for Synthetic Biology, Technical University Darmstadt, 64287 Darmstadt, Germany
| |
Collapse
|
9
|
Chen X, Shi C, Ye Y, Wang C, Li R, Wang H, Hou C, Song W, Mu C. FK506 activates the BMP signaling pathway to regulate ovarian development in Portunus trituberculatus. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2025; 166:105365. [PMID: 40169079 DOI: 10.1016/j.dci.2025.105365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 03/10/2025] [Accepted: 03/29/2025] [Indexed: 04/03/2025]
Abstract
Bone morphogenic proteins (BMPs) play important regulatory roles in the development of follicles in mammals. However, studies on the roles of BMPs in ovarian development in low-level aquatic animals, especially the swimming crab Portunus trituberculatus, are limited. In this study, a BMP Ⅰ-type receptor-specific activator (tacrolimus, FK506) was administered at different concentrations via in vivo injection, and the effects of FK506 on the regulation of the BMP signaling pathway during ovarian development in P. trituberculatus were examined. The tissue and cell morphology was observed, and a combined transcriptomics, proteomics and metabolomics analysis was carried out. Crabs administered FK506 exhibited elevated GSI alongside reduced HSI compared to control and blank groups. The main biological processes enriched by joint analysis included lipid metabolism, sugar metabolism, and amino acid metabolism. Fatty acid composition analysis revealed that the activator may activate the BMP signaling pathway to promote ovarian development and accelerate the transport of unsaturated fatty acids from the hepatopancreas to the ovaries. Amino acid metabolism and carbohydrate metabolism provide transporter proteins and energy for lipid metabolism. This study is highly important because it reveals the molecular mechanism by which the BMP signaling pathway regulates gonadal development in a crustacean.
Collapse
Affiliation(s)
- Xiaocong Chen
- Key Laboratory of Applied Aquacultral Biotechnology, Ministry of Education, Ningbo University, Ningbo, 315211, China; Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang, 222005, China; Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Ningbo, 315211, China
| | - Ce Shi
- Key Laboratory of Applied Aquacultral Biotechnology, Ministry of Education, Ningbo University, Ningbo, 315211, China; Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Ningbo, 315211, China
| | - Yangfang Ye
- Key Laboratory of Applied Aquacultral Biotechnology, Ministry of Education, Ningbo University, Ningbo, 315211, China; Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Ningbo, 315211, China
| | - Chunlin Wang
- Key Laboratory of Applied Aquacultral Biotechnology, Ministry of Education, Ningbo University, Ningbo, 315211, China; Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Ningbo, 315211, China
| | - Ronghua Li
- Key Laboratory of Applied Aquacultral Biotechnology, Ministry of Education, Ningbo University, Ningbo, 315211, China; Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Ningbo, 315211, China
| | - Huan Wang
- Key Laboratory of Applied Aquacultral Biotechnology, Ministry of Education, Ningbo University, Ningbo, 315211, China; Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Ningbo, 315211, China
| | - Congcong Hou
- Key Laboratory of Applied Aquacultral Biotechnology, Ministry of Education, Ningbo University, Ningbo, 315211, China; Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Ningbo, 315211, China
| | - Weiwei Song
- Key Laboratory of Applied Aquacultral Biotechnology, Ministry of Education, Ningbo University, Ningbo, 315211, China; Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Ningbo, 315211, China
| | - Changkao Mu
- Key Laboratory of Applied Aquacultral Biotechnology, Ministry of Education, Ningbo University, Ningbo, 315211, China; Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Ningbo, 315211, China
| |
Collapse
|
10
|
Rudgalvyte M, Hu Z, Kressler D, Dengjel J, Glauser DA. Antagonist actions of CMK-1/CaMKI and TAX-6/calcineurin along the C. elegans thermal avoidance circuit orchestrate adaptation of nociceptive response to repeated stimuli. eLife 2025; 14:RP103497. [PMID: 40305390 PMCID: PMC12043318 DOI: 10.7554/elife.103497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2025] Open
Abstract
Thermal nociception in Caenorhabditis elegans is regulated by the Ca²+/calmodulin-dependent protein kinase CMK-1, but its downstream effectors have remained unclear. Here, we combined in vitro kinase assays with mass-spectrometry-based phosphoproteomics to identify hundreds of CMK-1 substrates, including the calcineurin A subunit TAX-6, phosphorylated within its conserved regulatory domain. Genetic and pharmacological analyses reveal multiple antagonistic interactions between CMK-1 and calcineurin signaling in modulating both naive thermal responsiveness and adaptation to repeated noxious stimuli. Cell-specific manipulations indicate that CMK-1 acts in AFD and ASER thermo-sensory neurons, while TAX-6 functions in FLP thermo-sensory neurons and downstream interneurons. Since CMK-1 and TAX-6 act in distinct cell types, the phosphorylation observed in vitro might not directly underlie the behavioral phenotype. Instead, the opposing effects seem to arise from their distributed roles within the sensory circuit. Overall, our study provides (1) a resource of candidate CMK-1 targets for further dissecting CaM kinase signaling and (2) evidence of a previously unrecognized, circuit-level antagonism between CMK-1 and calcineurin pathways. These findings highlight a complex interplay of signaling modules that modulate thermal nociception and adaptation, offering new insights into potentially conserved mechanisms that shape nociceptive plasticity and pain (de)sensitization in more complex nervous systems.
Collapse
Affiliation(s)
| | - Zehan Hu
- Department of Biology, University of FribourgFribourgSwitzerland
| | - Dieter Kressler
- Department of Biology, University of FribourgFribourgSwitzerland
- Metabolomics and Proteomics Platform (MAPP), Department of Biology, University of FribourgFribourgSwitzerland
| | - Jörn Dengjel
- Department of Biology, University of FribourgFribourgSwitzerland
| | | |
Collapse
|
11
|
Hinterndorfer M, Spiteri VA, Ciulli A, Winter GE. Targeted protein degradation for cancer therapy. Nat Rev Cancer 2025:10.1038/s41568-025-00817-8. [PMID: 40281114 DOI: 10.1038/s41568-025-00817-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/21/2025] [Indexed: 04/29/2025]
Abstract
Targeted protein degradation (TPD) aims at reprogramming the target specificity of the ubiquitin-proteasome system, the major cellular protein disposal machinery, to induce selective ubiquitination and degradation of therapeutically relevant proteins. Since its conception over 20 years ago, TPD has gained a lot of attention mainly due to improvements in the design of bifunctional proteolysis targeting chimeras (PROTACs) and understanding the mechanisms underlying molecular glue degraders. Today, PROTACs are on the verge of a first clinical approval and recent structural and mechanistic insights combined with technological leaps promise to unlock the rational design of protein degraders, following the lead of lenalidomide and related clinically approved analogues. At the same time, the TPD universe is expanding at a record speed with the discovery of novel modalities beyond molecular glue degraders and PROTACs. Here we review the recent progress in the field, focusing on newly discovered degrader modalities, the current state of clinical degrader candidates for cancer therapy and upcoming design approaches.
Collapse
Affiliation(s)
- Matthias Hinterndorfer
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Valentina A Spiteri
- Centre for Targeted Protein Degradation, School of Life Sciences, University of Dundee, Dundee, UK
| | - Alessio Ciulli
- Centre for Targeted Protein Degradation, School of Life Sciences, University of Dundee, Dundee, UK.
| | - Georg E Winter
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.
| |
Collapse
|
12
|
Engler S, Buchner J. The evolution and diversification of the Hsp90 co-chaperone system. Biol Chem 2025:hsz-2025-0112. [PMID: 40261701 DOI: 10.1515/hsz-2025-0112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 04/11/2025] [Indexed: 04/24/2025]
Abstract
The molecular chaperone Hsp90 is the central element of a chaperone machinery in the cytosol of eukaryotic cells that is characterized by a large number of structurally and functionally different co-chaperones that influence the core chaperone component in different ways and increase its influence on the proteome. From yeast to humans, the number of Hsp90 co-chaperones has increased from 14 to over 40, and new co-chaperones are still being discovered. While Hsp90 itself has only undergone limited changes in structure and mechanism from yeast to humans, its increased importance and contribution to different processes in humans is based on the evolution and expansion of the cohort of co-chaperones. In this review, we provide an overview of Hsp90 co-chaperones, focusing on their roles in regulating Hsp90 function and their evolution from yeast to humans.
Collapse
Affiliation(s)
- Sonja Engler
- Center for Protein Assemblies (CPA), Department Bioscience, TUM School of Natural Sciences, Technical University of Munich, Ernst-Otto-Fischer-Strasse 8, D-85748 Garching, Germany
| | - Johannes Buchner
- Center for Protein Assemblies (CPA), Department Bioscience, TUM School of Natural Sciences, Technical University of Munich, Ernst-Otto-Fischer-Strasse 8, D-85748 Garching, Germany
| |
Collapse
|
13
|
Hann MM. A Career Doing STUFF. ChemMedChem 2025; 20:e202401017. [PMID: 40009618 DOI: 10.1002/cmdc.202401017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/10/2025] [Indexed: 02/28/2025]
Abstract
In this Guest Editorial for the MedChem Musings series, Mike, Former Senior Research Director at GSK, reflects on a career doing "STUFF": Science, Technology, Useful Functions and Fun. Mike summarises some of the many things he has been involved in throughout his 45 year-career in drug discovery, from which future generations of researchers can hopefully find practical advice and inspiration.
Collapse
Affiliation(s)
- Michael M Hann
- Former Senior Research Director GSK, Independent Consultant, Letchworth Garden City, UK
| |
Collapse
|
14
|
Tomlinson ACA, Knox JE, Brunsveld L, Ottmann C, Yano JK. The "three body solution": Structural insights into molecular glues. Curr Opin Struct Biol 2025; 91:103007. [PMID: 40014904 DOI: 10.1016/j.sbi.2025.103007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/22/2025] [Accepted: 01/28/2025] [Indexed: 03/01/2025]
Abstract
Molecular glues are small molecules that nucleate novel or stabilize natural, protein-protein interactions resulting in a ternary complex. Their success in targeting difficult to drug proteins of interest has led to ever-increasing interest in their use as therapeutics and research tools. While molecular glues and their targets vary in structure, inspection of diverse ternary complexes reveals commonalities. Whether of high or low molecular weight, molecular glues are often rigid and form direct hydrophobic interactions with their target protein. There is growing evidence that these hotspots can accommodate multiple ternary complex binding modes and enable targeting of traditionally undruggable targets. Advances in screening from the molecular glue degrader literature and insights in structure-based drug design, especially from the non-degrading tri-complex work, are likely intersectional.
Collapse
Affiliation(s)
| | | | - Luc Brunsveld
- Eindhoven University of Technology, Eindhoven, Netherlands
| | | | | |
Collapse
|
15
|
Yin L, Niu T, Li L, Yu W, Han B, Rehman A, Zeng K. Research advancements in molecular glues derived from natural product scaffolds: Chemistry, targets, and molecular mechanisms. CHINESE HERBAL MEDICINES 2025; 17:235-245. [PMID: 40256709 PMCID: PMC12009069 DOI: 10.1016/j.chmed.2025.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/30/2024] [Accepted: 01/03/2025] [Indexed: 04/22/2025] Open
Abstract
The mechanism of action of traditional Chinese medicine (TCM) remains unclear. Historically, research on TCM has mainly focused on exploring the mechanisms of active components acting on single targets. However, it is insufficient to explain the complex mechanisms by which these active components in TCM treat diseases. In recent years, the emergence of molecular glues (MGs) theory has provided new strategies to address this issue. MGs are small molecules that can promote interactions between proteins at their interface. The characteristic of MGs is to establish connections between diverse protein structures, thereby enabling a chemically-mediated proximity effect that triggers a wide spectrum of biological functions. Natural products are the result of billions of years of evolutionary processes in the natural environment. Thus, the extensive structural diversity of natural products renders them a rich source of MGs, including polyketides, terpenoids, steroids, lignans, organic acids, alkaloids and other classes. Currently, several well-known natural MGs, including the immunosuppressants cyclosporin A (CsA) and tacrolimus (FK506), as well as the anticancer agent taxol, have been incorporated into clinical practice. Meanwhile, the advancement of new technologies is propelling the discovery of novel MGs from natural products. Thus, we primarily summarize a growing variety of MGs from natural origins reported in recent years and categorize them based on the chemical structural types. Moreover, the main sources of TCM are natural products. The discovery of natural MGs promises to provide a new perspective for the elucidation of the molecular mechanism behind the efficiency of TCM. In summary, this review aims to provide insights from the perspective of natural products that could potentially influence TCM and modern drug development.
Collapse
Affiliation(s)
- Lina Yin
- School of Pharmacy/Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Shihezi University, Shihezi 832003, China
| | - Tingting Niu
- School of Pharmacy/Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Shihezi University, Shihezi 832003, China
| | - Ling Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Wei Yu
- School of Pharmacy/Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Shihezi University, Shihezi 832003, China
| | - Bo Han
- School of Pharmacy/Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Shihezi University, Shihezi 832003, China
| | - Asma Rehman
- National Institute for Biotechnology & Genetic Engineering College Pakistan Institute of Engineering & Applied Sciences (NIBGE-C, PIEAS), Faisalabad 38000, Pakistan
| | - Kewu Zeng
- School of Pharmacy/Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Shihezi University, Shihezi 832003, China
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| |
Collapse
|
16
|
Limbach MN, Lindberg ET, Shrestha C, Lou J, Steren CA, Best MD, Do TD. Biased Equilibrium Drives Cyclosporine Membrane Permeability: The Goldilocks Energy Barriers. J Med Chem 2025; 68:6588-6600. [PMID: 40077929 DOI: 10.1021/acs.jmedchem.4c03178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2025]
Abstract
Conformational flexibility allows macrocyclic peptides like cyclosporine A (CycA) to cross membranes, yet drug design leveraging this property has largely failed. A key challenge is linking specific conformers to function, as different conformers govern permeability versus target binding. We reveal a mechanism that enhances CycA and alisporivir (ALI) permeability: trans-to-cis isomerization at MeVal11-MeBmt1 creates conformers that remain "soluble" in both membrane-like and aqueous environments. A biased equilibrium favors this conformer in protic environments, while a lipophilic conformer with cis MeLeu9-MeLeu10 dominates in aprotic conditions. This mechanism explains why CycH, Valspodar (VALSPO), and O-acetyl CycA (OAc-CycA) fail to cross membranes─they adopt similar states but lack this biased equilibrium. Our findings provide a new strategy for designing membrane-permeable N-methylated macrocycles and underscore the role of high-energy conformers as transition states between membrane permeability and target engagement─offering critical insights for drug development.
Collapse
Affiliation(s)
- Miranda N Limbach
- Department of Chemistry, University of Tennessee, Knoxville, Tennessee 37996, United States
| | - Edward T Lindberg
- Department of Chemistry, University of Tennessee, Knoxville, Tennessee 37996, United States
| | - Cynthiya Shrestha
- Department of Chemistry, University of Tennessee, Knoxville, Tennessee 37996, United States
| | - Jinchao Lou
- Department of Chemistry, University of Tennessee, Knoxville, Tennessee 37996, United States
| | - Carlos A Steren
- Department of Chemistry, University of Tennessee, Knoxville, Tennessee 37996, United States
| | - Michael D Best
- Department of Chemistry, University of Tennessee, Knoxville, Tennessee 37996, United States
| | - Thanh D Do
- Department of Chemistry, University of Tennessee, Knoxville, Tennessee 37996, United States
| |
Collapse
|
17
|
Tan ZY, Adade JKA, Gu X, Hecht CJS, Salcius M, Tong B, Liu S, Hwang S, Zécri FJ, Graham DB, Schreiber SL, Xavier RJ. Development of an FKBP12-recruiting chemical-induced proximity DNA-encoded library and its application to discover an autophagy potentiator. Cell Chem Biol 2025; 32:498-510.e35. [PMID: 39753134 PMCID: PMC11928285 DOI: 10.1016/j.chembiol.2024.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 10/01/2024] [Accepted: 12/04/2024] [Indexed: 03/23/2025]
Abstract
Chemical inducers of proximity (CIPs) are molecules that recruit one protein to another and introduce new functionalities toward modulating protein states and activities. While CIP-mediated recruitment of E3 ligases is widely exploited for the development of degraders, other therapeutic modalities remain underexplored. We describe a non-degrader CIP-DNA-encoded library (CIP-DEL) that recruits FKBP12 to target proteins using non-traditional acyclic structures, with an emphasis on introducing stereochemically diverse and rigid connectors to attach the combinatorial library. We deployed this strategy to modulate ATG16L1 T300A, which confers genetic susceptibility to Crohn's disease (CD), and identified a compound that stabilizes the variant protein against caspase-3 (Casp3) cleavage in a FKBP12-independent manner. We demonstrate in cellular models that this compound potentiates autophagy, and reverses the xenophagy defects as well as increased cytokine secretion characteristic of ATG16L1 T300A. This study provides a platform to access unexplored chemical space for CIP design to develop therapeutic modalities guided by human genetics.
Collapse
Affiliation(s)
- Zher Yin Tan
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Joel K A Adade
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Xiebin Gu
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Cody J S Hecht
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Michael Salcius
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Bingqi Tong
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Shuang Liu
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Seungmin Hwang
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Frédéric J Zécri
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Daniel B Graham
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Stuart L Schreiber
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA; Arena BioWorks, Cambridge, MA 02139, USA.
| | - Ramnik J Xavier
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA.
| |
Collapse
|
18
|
Nix MN, Gourisankar S, Sarott RC, Dwyer BG, Nettles SA, Martinez MM, Abuzaid H, Yang H, Wang Y, Simanauskaite JM, Romero BA, Jones HM, Krokhotin A, Lowensohn TN, Chen L, Low C, Davis MM, Fernandez D, Zhang T, Green MR, Hinshaw SM, Gray NS, Crabtree GR. A Bivalent Molecular Glue Linking Lysine Acetyltransferases to Oncogene-induced Cell Death. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.14.643404. [PMID: 40166243 PMCID: PMC11956963 DOI: 10.1101/2025.03.14.643404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Developing cancer therapies that induce robust death of the malignant cell is critical to prevent relapse. Highly effective strategies, such as immunotherapy, exemplify this observation. Here we provide the structural and molecular underpinnings for an approach that leverages chemical induced proximity to produce specific cell killing of diffuse large B cell lymphoma, the most common non-Hodgkin's lymphoma. We develop KAT-TCIPs (lysine acetyltransferase transcriptional/epigenetic chemical inducers of proximity) that redirect p300 and CBP to activate programmed cell death genes normally repressed by the oncogenic driver, BCL6. Acute treatment rapidly reprograms the epigenome to initiate apoptosis and repress c-MYC. The crystal structure of the chemically induced p300-BCL6 complex reveals how chance interactions between the two proteins can be systematically exploited to produce the exquisite potency and selectivity of KAT-TCIPs. Thus, the malignant function of an oncogenic driver can be co-opted to activate robust cell death, with implications for precision epigenetic therapies.
Collapse
Affiliation(s)
- Meredith N. Nix
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA
- Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Sai Gourisankar
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA
| | - Roman C. Sarott
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA
| | - Brendan G. Dwyer
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA
| | | | - Michael M. Martinez
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA
| | - Hind Abuzaid
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Haopeng Yang
- Department of Lymphoma- & Myeloma, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yanlan Wang
- Department of Pathology, Stanford University, Stanford, CA, USA
| | | | - Bryan A. Romero
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA
| | - Hannah M. Jones
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA
| | | | | | - Lei Chen
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
| | - Cara Low
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA
| | - Mark M. Davis
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
| | - Daniel Fernandez
- Macromolecular Structure, Nucleus at Sarafan ChEM-H, Stanford University, Stanford, CA, USA
| | - Tinghu Zhang
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA
| | - Michael R. Green
- Department of Lymphoma- & Myeloma, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Stephen M. Hinshaw
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA
| | - Nathanael S. Gray
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA
| | - Gerald R. Crabtree
- Department of Pathology, Stanford University, Stanford, CA, USA
- Department of Developmental Biology, Stanford University, Stanford, CA, USA
| |
Collapse
|
19
|
Chandler F, Reddy PAN, Bhutda S, Ross RL, Datta A, Walden M, Walker K, Di Donato S, Cassel JA, Prakesch MA, Aman A, Datti A, Campbell LJ, Foglizzo M, Bell L, Stein DN, Ault JR, Al-Awar RS, Calabrese AN, Sicheri F, Del Galdo F, Salvino JM, Greenberg RA, Zeqiraj E. Molecular glues that inhibit deubiquitylase activity and inflammatory signaling. Nat Struct Mol Biol 2025:10.1038/s41594-025-01517-5. [PMID: 40097626 DOI: 10.1038/s41594-025-01517-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 02/13/2025] [Indexed: 03/19/2025]
Abstract
Deubiquitylases (DUBs) are crucial in cell signaling and are often regulated by interactions within protein complexes. The BRCC36 isopeptidase complex (BRISC) regulates inflammatory signaling by cleaving K63-linked polyubiquitin chains on type I interferon receptors (IFNAR1). As a Zn2+-dependent JAMM/MPN (JAB1, MOV34, MPR1, Pad1 N-terminal) DUB, BRCC36 is challenging to target with selective inhibitors. Here, we discover first-in-class inhibitors, termed BRISC molecular glues (BLUEs), which stabilize a 16-subunit human BRISC dimer in an autoinhibited conformation, blocking active sites and interactions with the targeting subunit, serine hydroxymethyltransferase 2. This unique mode of action results in selective inhibition of BRISC over related complexes with the same catalytic subunit, splice variants and other JAMM/MPN DUBs. BLUE treatment reduced interferon-stimulated gene expression in cells containing wild-type BRISC and this effect was abolished when using structure-guided, inhibitor-resistant BRISC mutants. Additionally, BLUEs increase IFNAR1 ubiquitylation and decrease IFNAR1 surface levels, offering a potential strategy to mitigate type I interferon-mediated diseases. Our approach also provides a template for designing selective inhibitors of large protein complexes by promoting rather than blocking protein-protein interactions.
Collapse
Affiliation(s)
- Francesca Chandler
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Poli Adi Narayana Reddy
- Medicinal Chemistry, Molecular and Cellular Oncogenesis (MCO) Program and The Wistar Cancer Center Molecular Screening, The Wistar Institute, Philadelphia, PA, USA
| | - Smita Bhutda
- Department of Cancer Biology, Penn Center for Genome Integrity, Basser Center for BRCA, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rebecca L Ross
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals, NHS Trust, Chapel Allerton Hospital, Leeds, UK
| | - Arindam Datta
- Department of Cancer Biology, Penn Center for Genome Integrity, Basser Center for BRCA, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Miriam Walden
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Kieran Walker
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Stefano Di Donato
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals, NHS Trust, Chapel Allerton Hospital, Leeds, UK
| | - Joel A Cassel
- Medicinal Chemistry, Molecular and Cellular Oncogenesis (MCO) Program and The Wistar Cancer Center Molecular Screening, The Wistar Institute, Philadelphia, PA, USA
| | - Michael A Prakesch
- Drug Discovery Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Ahmed Aman
- Drug Discovery Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Alessandro Datti
- Department of Agricultural, Food, and Environmental Sciences, University of Perugia, Perugia, Italy
| | - Lisa J Campbell
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Martina Foglizzo
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Lillie Bell
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Daniel N Stein
- Department of Cancer Biology, Penn Center for Genome Integrity, Basser Center for BRCA, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - James R Ault
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Rima S Al-Awar
- Drug Discovery Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Antonio N Calabrese
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Frank Sicheri
- Centre for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Francesco Del Galdo
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK.
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals, NHS Trust, Chapel Allerton Hospital, Leeds, UK.
| | - Joseph M Salvino
- Medicinal Chemistry, Molecular and Cellular Oncogenesis (MCO) Program and The Wistar Cancer Center Molecular Screening, The Wistar Institute, Philadelphia, PA, USA.
| | - Roger A Greenberg
- Department of Cancer Biology, Penn Center for Genome Integrity, Basser Center for BRCA, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Elton Zeqiraj
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK.
| |
Collapse
|
20
|
Fu H, Mo X, Ivanov AA. Decoding the functional impact of the cancer genome through protein-protein interactions. Nat Rev Cancer 2025; 25:189-208. [PMID: 39810024 DOI: 10.1038/s41568-024-00784-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/02/2024] [Indexed: 01/16/2025]
Abstract
Acquisition of genomic mutations enables cancer cells to gain fitness advantages under selective pressure and, ultimately, leads to oncogenic transformation. Interestingly, driver mutations, even within the same gene, can yield distinct phenotypes and clinical outcomes, necessitating a mutation-focused approach. Conversely, cellular functions are governed by molecular machines and signalling networks that are mostly controlled by protein-protein interactions (PPIs). The functional impact of individual genomic alterations could be transmitted through regulated nodes and hubs of PPIs. Oncogenic mutations may lead to modified residues of proteins, enabling interactions with other proteins that the wild-type protein does not typically interact with, or preventing interactions with proteins that the wild-type protein usually interacts with. This can result in the rewiring of molecular signalling cascades and the acquisition of an oncogenic phenotype. Here, we review the altered PPIs driven by oncogenic mutations, discuss technologies for monitoring PPIs and provide a functional analysis of mutation-directed PPIs. These driver mutation-enabled PPIs and mutation-perturbed PPIs present a new paradigm for the development of tumour-specific therapeutics. The intersection of cancer variants and altered PPI interfaces represents a new frontier for understanding oncogenic rewiring and developing tumour-selective therapeutic strategies.
Collapse
Affiliation(s)
- Haian Fu
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Emory University, Atlanta, GA, USA.
- Winship Cancer Institute of Emory University, Atlanta, GA, USA.
| | - Xiulei Mo
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Emory University, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Andrey A Ivanov
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Emory University, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| |
Collapse
|
21
|
Mazidi Z, Wieser M, Spinu N, Weidinger A, Kozlov AV, Vukovic K, Wellens S, Murphy C, Singh P, Lagares LM, Bobbili MR, Liendl L, Schosserer M, Diendorfer A, Bettelheim B, Eilenberg W, Exner T, Culot M, Jennings P, Wilmes A, Novic M, Benfenati E, Grillari-Voglauer R, Grillari J. Cyclosporin A toxicity on endothelial cells differentiated from induced pluripotent stem cells: Assembling an adverse outcome pathway. Toxicol In Vitro 2025; 103:105954. [PMID: 39550010 DOI: 10.1016/j.tiv.2024.105954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 06/15/2024] [Accepted: 10/15/2024] [Indexed: 11/18/2024]
Abstract
Cyclosporin A (CSA) is a potent immunosuppressive agent in pharmacologic studies. However, there is evidence for side effects, specifically regarding vascular dysfunction. Its mode of action inducing endothelial cell toxicity is partially unclear, and a connection with an adverse outcome pathway (AOP) is not established yet. Therefore, we designed this study to get deeper insights into the mechanistic toxicology of CSA on angiogenesis. Stem cells, especially induced pluripotent stem cells (iPSCs) with the ability of differentiation to all organs of the body, are considered a promising in vitro model to reduce animal experimentation. In this study, we differentiated iPSCs to endothelial cells (ECs) as one cell type that in other studies would allow to generate multi-cell type organoids from single donors. Flow cytometry and immunostaining confirmed our scalable differentiation protocol. Then dose and time course experiments assessing CSA cytotoxicity on iPS derived endothelial cells were performed. Transcriptomic data suggested CSA dependent induction of reactive oxygen species (ROS), mitochondrial dysfunction, and impaired angiogenesis via ROS induction which was confirmed by in vitro experiments. In order to put these data into a potential adverse outcome pathway (AOP) context, we performed a literature review for CSA-mediated endothelial cell toxicity and combined our experimental data with the publicly available knowledge. Such an AOP will help to design in vitro test batteries and to model events observed in human toxicity studies, as well in predictive toxicology.
Collapse
Affiliation(s)
- Zahra Mazidi
- Evercyte GmbH, Leberstrasse 20, 1110 Vienna, Austria; Institute of Molecular Biotechnology, Department of Biotechnology, BOKU University, Muthgasse 18, 1190 Vienna, Austria
| | | | - Nicoleta Spinu
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Byrom Street, Liverpool L3 3AF, UK
| | - Adelheid Weidinger
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Donaueschingenstrasse 13, 1200 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Andrey V Kozlov
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Donaueschingenstrasse 13, 1200 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Kristijan Vukovic
- Laboratory of Environmental Chemistry and Toxicology, Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche "Mario Negri"-IRCCS, Via Mario Negri 2, 20156 Milano, Italy
| | - Sara Wellens
- University of Artois, UR2465, Laboratoire de la Barrière Hémato-Encéphalique (LBHE), Faculté des sciences Jean Perrin, Rue Jean Souvraz SP18, F-62300 Lens, France
| | - Cormac Murphy
- Division of Molecular and Computational Toxicology, Department of Chemistry and Pharmaceutical Sciences, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081HZ Amsterdam, the Netherlands
| | - Pranika Singh
- Edelweiss Connect GmbH, Technology Park Basel, Hochbergerstrasse 60C, 4057 Basel, Switzerland; Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Liadys Mora Lagares
- Theory Department, Laboratory for Cheminformatics, National Institute of Chemistry, 1000 Ljubljana, Slovenia
| | - Madhusudhan Reddy Bobbili
- Institute of Molecular Biotechnology, Department of Biotechnology, BOKU University, Muthgasse 18, 1190 Vienna, Austria; Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Donaueschingenstrasse 13, 1200 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Lisa Liendl
- Institute of Molecular Biotechnology, Department of Biotechnology, BOKU University, Muthgasse 18, 1190 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Markus Schosserer
- Institute of Molecular Biotechnology, Department of Biotechnology, BOKU University, Muthgasse 18, 1190 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | | | - Bruno Bettelheim
- Department of Obstetrics and Gynecology, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Wolf Eilenberg
- Department of General Surgery, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Thomas Exner
- Seven Past Nine d.o.o., Hribljane 10, 1380 Cerknica, Slovenia
| | - Maxime Culot
- University of Artois, UR2465, Laboratoire de la Barrière Hémato-Encéphalique (LBHE), Faculté des sciences Jean Perrin, Rue Jean Souvraz SP18, F-62300 Lens, France
| | - Paul Jennings
- Division of Molecular and Computational Toxicology, Department of Chemistry and Pharmaceutical Sciences, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081HZ Amsterdam, the Netherlands
| | - Anja Wilmes
- Division of Molecular and Computational Toxicology, Department of Chemistry and Pharmaceutical Sciences, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081HZ Amsterdam, the Netherlands
| | - Marjana Novic
- Theory Department, Laboratory for Cheminformatics, National Institute of Chemistry, 1000 Ljubljana, Slovenia
| | - Emilio Benfenati
- Laboratory of Environmental Chemistry and Toxicology, Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche "Mario Negri"-IRCCS, Via Mario Negri 2, 20156 Milano, Italy
| | - Regina Grillari-Voglauer
- Evercyte GmbH, Leberstrasse 20, 1110 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Johannes Grillari
- Institute of Molecular Biotechnology, Department of Biotechnology, BOKU University, Muthgasse 18, 1190 Vienna, Austria; Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Donaueschingenstrasse 13, 1200 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria.
| |
Collapse
|
22
|
Jablonka-Shariff A, Broberg C, Snyder-Warwick AK. FK506 Enhancement of Neuromuscular Junction Recovery After Nerve Injury Is Macrophage-Dependent. Muscle Nerve 2025; 71:463-473. [PMID: 39780562 DOI: 10.1002/mus.28336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 12/20/2024] [Accepted: 12/26/2024] [Indexed: 01/11/2025]
Abstract
INTRODUCTION Motor recovery following nerve injury is dependent on time required for muscle reinnervation. This process is imperfect, however, and recovery is often incomplete. At the neuromuscular junction (NMJ), macrophage signaling aids muscle reinnervation. Tacrolimus (FK506) treatment speeds functional recovery through unknown mechanisms. This study investigated whether macrophages were required for FK506 neuroenhancing effects. METHODS Wildtype (WT) mice and mice with impaired macrophage recruitment to injury sites (Ccr2 -/- ) were injected subcutaneously with either saline or FK506 for 3 days prior to sciatic nerve transection and immediate repair and then daily for 4 weeks. Functional recovery was assessed by grid walk and muscle force. Morphometric NMJ and macrophage analyses were conducted in extensor digitorum longus muscles. RESULTS FK506-injected WT mice showed increased proportions of fully reinnervated NMJs and terminal Schwann cells/NMJ (p < 0.05), improved recovery of tetanic muscle force (p < 0.05), and improved grid walking (p < 0.05) relative to controls. Ccr2 -/- mice showed no enhancements in recovery; Ccr2 -/- mice treated with FK506 did not differ from controls on any tested metric. We also observed at the NMJ of WT mice increased macrophage numbers with FK506 treatment and increased macrophages expressing FK506 binding protein, FKBP52, after nerve injury. DISCUSSION These results show that macrophages are required for FK506-mediated improvements in NMJ reinnervation and muscle function. These data implicate macrophages in the mechanism underlying FK506-mediated enhancement of motor recovery after nerve injury. Enhanced knowledge of the neuroenhancing mechanism of FK506 may identify new clinically relevant therapeutic targets.
Collapse
Affiliation(s)
- Albina Jablonka-Shariff
- Division of Plastic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Curtis Broberg
- Washington University School of Medicine, St. Louis, Missouri, USA
| | - Alison K Snyder-Warwick
- Division of Plastic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
23
|
Newton LS, Gathmann C, Ridewood S, Smith RJ, Wijaya AJ, Hornsby TW, Morling KL, Annett D, Chiozzi RZ, Reuschl AK, Govasli ML, Tan YY, Thorne LG, Jolly C, Thalassinos K, Ciulli A, Towers GJ, Selwood DL. Macrocycle-based PROTACs selectively degrade cyclophilin A and inhibit HIV-1 and HCV. Nat Commun 2025; 16:1484. [PMID: 39929804 PMCID: PMC11811207 DOI: 10.1038/s41467-025-56317-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 01/14/2025] [Indexed: 02/13/2025] Open
Abstract
Targeting host proteins that are crucial for viral replication offers a promising antiviral strategy. We have designed and characterised antiviral PROteolysis TArgeting Chimeras (PROTACs) targeting the human protein cyclophilin A (CypA), a host cofactor for unrelated viruses including human immunodeficiency virus (HIV) and hepatitis C virus (HCV). The PROTAC warheads are based on fully synthetic macrocycles derived from sanglifehrin A, which are structurally different from the classical Cyp inhibitor, cyclosporine A. Our Cyp-PROTACs decrease CypA levels in cell lines and primary human cells and have high specificity for CypA confirmed by proteomics experiments. Critically, CypA degradation facilitates improved antiviral activity against HIV-1 in primary human CD4+ T cells compared to the non-PROTAC parental inhibitor, at limiting inhibitor concentrations. Similarly, we observe antiviral activity against HCV replicon in a hepatoma cell line. We propose that CypA-targeting PROTACs inhibit viral replication potently and anticipate reduced evolution of viral resistance and broad efficacy against unrelated viruses. Furthermore, they provide powerful tools for probing cyclophilin biology.
Collapse
Affiliation(s)
- Lydia S Newton
- Division of Infection and Immunity, University College London, London, UK
| | - Clara Gathmann
- Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Sophie Ridewood
- Division of Infection and Immunity, University College London, London, UK
| | - Robert J Smith
- Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Andre J Wijaya
- Centre for Targeted Protein Degradation, School of Life Sciences, University of Dundee, Dundee, UK
| | - Thomas W Hornsby
- Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Kate L Morling
- Division of Infection and Immunity, University College London, London, UK
- Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Dara Annett
- Division of Infection and Immunity, University College London, London, UK
| | - Riccardo Zenezini Chiozzi
- University College London Mass Spectrometry Science Technology Platform, Division of Biosciences, University College London, London, UK
| | | | - Morten L Govasli
- Division of Infection and Immunity, University College London, London, UK
- Department of Biomedicine, Centre for Cancer Biomarkers, University of Bergen, Bergen, Norway
| | - Ying Ying Tan
- Division of Infection and Immunity, University College London, London, UK
| | - Lucy G Thorne
- Division of Infection and Immunity, University College London, London, UK
- Department of Infectious Diseases, Imperial College London, London, UK
| | - Clare Jolly
- Division of Infection and Immunity, University College London, London, UK
| | - Konstantinos Thalassinos
- University College London Mass Spectrometry Science Technology Platform, Division of Biosciences, University College London, London, UK
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London, UK
| | - Alessio Ciulli
- Centre for Targeted Protein Degradation, School of Life Sciences, University of Dundee, Dundee, UK
| | - Greg J Towers
- Division of Infection and Immunity, University College London, London, UK.
| | - David L Selwood
- Wolfson Institute for Biomedical Research, University College London, London, UK.
| |
Collapse
|
24
|
Xie W, Kong Y, Ren C, Wen Y, Ying M, Xing H. Chemistries on the inner leaflet of the cell membrane. Chem Commun (Camb) 2025; 61:2387-2402. [PMID: 39810742 DOI: 10.1039/d4cc05186f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
The cell membrane, characterized by its inherent asymmetry, functions as a dynamic barrier that regulates numerous cellular activities. This Highlight aims to provide the chemistry community with a comprehensive overview of the intriguing and underexplored inner leaflet, encompassing both fundamental biology and emerging synthetic modification strategies. We begin by describing the asymmetric nature of the plasma membrane, with a focus on the distinct roles of lipids, proteins, and glycan chains, highlighting the composition and biofunctions of the inner leaflet and the biological mechanisms that sustain membrane asymmetry. Next, we explore chemical biological strategies for engineering the inner leaflet, including genetic engineering, transmembrane peptides, and liposome fusion-based transport. In the perspective section, we discuss the challenges in developing chemistries for the inner leaflet of the cell membrane, aiming to inspire researchers and collaborators to explore this field and address its unanswered biological questions.
Collapse
Affiliation(s)
- Wenxue Xie
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China.
| | - Yuhan Kong
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China.
| | - Cong Ren
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China.
| | - Yujian Wen
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China.
| | | | - Hang Xing
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China.
- Research Institute of Hunan University in Chongqing, Chongqing, 401100, China
| |
Collapse
|
25
|
Saca VR, Huber T, Sakmar TP. G protein-coupled receptor-targeted proteolysis-targeting chimeras in cancer therapeutics. Mol Pharmacol 2025; 107:100013. [PMID: 40023512 DOI: 10.1016/j.molpha.2024.100013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 12/05/2024] [Indexed: 03/04/2025] Open
Abstract
G protein-coupled receptors (GPCRs) comprise a family of heptahelical membrane proteins that mediate intracellular and intercellular transmembrane signaling. Defects in GPCR signaling pathways are implicated in the pathophysiology of many diseases, including cardiovascular disease, endocrinopathies, immune disorders, and cancer. Although GPCRs are attractive drug targets, only a small number of Food and Drug Administration-approved anticancer therapeutics target GPCRs. Targeted protein degradation (TPD) technology allows for the direct modulation of the cellular expression level of a protein of interest. TPD methods such as proteolysis-targeting chimeras (PROTACs) use the ubiquitin-proteasome system to degrade a protein of interest selectively. Although the PROTAC system has not been widely applied to GPCRs and other membrane proteins, there is evidence that PROTACs or other TPD methods could be applied to the GPCRome. Current GPCR PROTACs show the feasibility of using PROTACs to degrade GPCRs; however, the degradation mechanism for some of these GPCR PROTACs is uncertain. Additional studies aimed at elucidating the degradation mechanism of GPCRs with PROTACs are necessary. Discovery of new allosteric intracellular small molecule binders of GPCRs will be required for the development of intracellularly oriented PROTACs. Promising early results in targeted degradation of GPCRs suggest that TPD drug discovery platforms will be useful in developing PROTACs targeting pathological GPCRs. SIGNIFICANCE STATEMENT: Aberrant signaling of G protein-coupled receptors (GPCRs) can contribute to the pathophysiology of cancer. Although GPCRs are generally highly attractive drug targets, many individual GPCRs are currently undrugged using traditional drug discovery approaches. Targeted protein degradation technologies, such as proteolysis-targeting chimeras, provide a new approach to drug discovery for targeting previously undruggable GPCRs relevant to the molecular pathophysiology of cancer.
Collapse
Affiliation(s)
- Victoria R Saca
- Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University, New York, New York; Tri-Institutional PhD Program in Chemical Biology, New York, New York
| | - Thomas Huber
- Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University, New York, New York
| | - Thomas P Sakmar
- Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University, New York, New York.
| |
Collapse
|
26
|
Sordo L, Ubele MF, Boaz KA, Mefford JL, Jones ED, McCarty KL, van Rooyen HY, Smiley J, Bembenek Bailey SA, Perpich JA, Meacham B, Powell DK, Bresch F, Crump JW, Phelan MJ, Noche JA, Stark CE, Puskás LG, Norris CM, Head E. Calcineurin/NFAT inhibitors maintain cognition in a preclinical prevention study in an aging canine model of Alzheimer disease. Neurobiol Aging 2025; 146:1-14. [PMID: 39547056 DOI: 10.1016/j.neurobiolaging.2024.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 10/10/2024] [Accepted: 11/05/2024] [Indexed: 11/17/2024]
Abstract
Brain signaling of calcineurin (CN) and nuclear factor of activated T-cells (NFAT) transcription factor increases in Alzheimer disease (AD) and is associated with synaptic loss, neurodegeneration, neuroinflammation, amyloid-β (Aβ) production, and cognitive decline. CN/NFAT inhibitors ameliorate these neuropathologies in mouse models of AD. Further, chronic use of tacrolimus in transplant patients reduces risk of AD. Beagles naturally develop Aβ plaques and cognitive dysfunction. We evaluated the impact of FDA-approved CN inhibitor, tacrolimus, and experimental NFAT inhibitor, Q134R, on cognitive outcomes during a three-year prevention study in 37 middle-aged beagles. While beagles treated with CN/NFAT inhibitors showed differences in the pattern of cognitive maintenance and duration of their effect, there was improvement in spatial learning, as well as maintenance of memory, attention, and working memory relative to placebo dogs. CN/NFAT inhibition is a promising target for prevention of cognitive decline that may be rapidly implemented in human clinical trials.
Collapse
Affiliation(s)
- Lorena Sordo
- Department of Pathology and Laboratory Medicine, University of California, 837 Health Sciences Rd., Irvine, CA 92697, USA
| | - Margo F Ubele
- Sanders Brown Center on Aging, Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, 800 S. Limestone Street, Lexington, KY 19104, USA
| | - Kathy A Boaz
- Sanders Brown Center on Aging, Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, 800 S. Limestone Street, Lexington, KY 19104, USA
| | - Jennifer L Mefford
- Division of Laboratory Animal Resources, University of Kentucky, 204 Health Sciences Research Building, Lexington, KY 19104, USA
| | - Erin Dehnart Jones
- Division of Laboratory Animal Resources, University of Kentucky, 204 Health Sciences Research Building, Lexington, KY 19104, USA
| | - Katie L McCarty
- Division of Laboratory Animal Resources, University of Kentucky, 204 Health Sciences Research Building, Lexington, KY 19104, USA
| | - Hollie Y van Rooyen
- Division of Laboratory Animal Resources, University of Kentucky, 204 Health Sciences Research Building, Lexington, KY 19104, USA
| | - Jeffrey Smiley
- Division of Laboratory Animal Resources, University of Kentucky, 204 Health Sciences Research Building, Lexington, KY 19104, USA
| | - Stasia A Bembenek Bailey
- Division of Laboratory Animal Resources, University of Kentucky, 204 Health Sciences Research Building, Lexington, KY 19104, USA
| | - Jessica A Perpich
- Division of Laboratory Animal Resources, University of Kentucky, 204 Health Sciences Research Building, Lexington, KY 19104, USA
| | - Beverly Meacham
- Magnetic Resonance Imaging and Spectroscopy Center, University of Kentucky, 740 Rose Street, Lexington, KY 19104, USA
| | - David K Powell
- Magnetic Resonance Imaging and Spectroscopy Center, University of Kentucky, 740 Rose Street, Lexington, KY 19104, USA
| | - Frederick Bresch
- Sanders Brown Center on Aging, Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, 800 S. Limestone Street, Lexington, KY 19104, USA
| | - Jacob W Crump
- Sanders Brown Center on Aging, Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, 800 S. Limestone Street, Lexington, KY 19104, USA
| | - Michael J Phelan
- Department of Pathology and Laboratory Medicine, University of California, 837 Health Sciences Rd., Irvine, CA 92697, USA
| | - Jessica A Noche
- Department of Neurobiology and Behavior, Department of Cognitive Sciences, University of California, 2205 McGaugh Hall, Irvine, CA 92697, USA
| | - Craig E Stark
- Department of Neurobiology and Behavior, Department of Cognitive Sciences, University of California, 2205 McGaugh Hall, Irvine, CA 92697, USA
| | | | - Christopher M Norris
- Sanders Brown Center on Aging, Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, 800 S. Limestone Street, Lexington, KY 19104, USA
| | - Elizabeth Head
- Department of Pathology and Laboratory Medicine, University of California, 837 Health Sciences Rd., Irvine, CA 92697, USA.
| |
Collapse
|
27
|
Patalano SD, Fuxman Bass P, Fuxman Bass JI. Transcription factors in the development and treatment of immune disorders. Transcription 2025; 16:118-140. [PMID: 38100543 PMCID: PMC11970766 DOI: 10.1080/21541264.2023.2294623] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 12/05/2023] [Accepted: 12/08/2023] [Indexed: 12/17/2023] Open
Abstract
Immune function is highly controlled at the transcriptional level by the binding of transcription factors (TFs) to promoter and enhancer elements. Several TF families play major roles in immune gene expression, including NF-κB, STAT, IRF, AP-1, NRs, and NFAT, which trigger anti-pathogen responses, promote cell differentiation, and maintain immune system homeostasis. Aberrant expression, activation, or sequence of isoforms and variants of these TFs can result in autoimmune and inflammatory diseases as well as hematological and solid tumor cancers. For this reason, TFs have become attractive drug targets, even though most were previously deemed "undruggable" due to their lack of small molecule binding pockets and the presence of intrinsically disordered regions. However, several aspects of TF structure and function can be targeted for therapeutic intervention, such as ligand-binding domains, protein-protein interactions between TFs and with cofactors, TF-DNA binding, TF stability, upstream signaling pathways, and TF expression. In this review, we provide an overview of each of the important TF families, how they function in immunity, and some related diseases they are involved in. Additionally, we discuss the ways of targeting TFs with drugs along with recent research developments in these areas and their clinical applications, followed by the advantages and disadvantages of targeting TFs for the treatment of immune disorders.
Collapse
Affiliation(s)
- Samantha D. Patalano
- Biology Department, Boston University, Boston, MA, USA
- Molecular Biology, Cellular Biology and Biochemistry Program, Boston University, Boston, MA, USA
| | - Paula Fuxman Bass
- Facultad de Medicina, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Juan I. Fuxman Bass
- Biology Department, Boston University, Boston, MA, USA
- Molecular Biology, Cellular Biology and Biochemistry Program, Boston University, Boston, MA, USA
- Bioinformatics Program, Boston University, Boston, MA, USA
| |
Collapse
|
28
|
Müller S, Cañeque T, Solier S, Rodriguez R. Copper and iron orchestrate cell-state transitions in cancer and immunity. Trends Cell Biol 2025; 35:105-114. [PMID: 39079798 DOI: 10.1016/j.tcb.2024.07.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 07/03/2024] [Accepted: 07/08/2024] [Indexed: 02/09/2025]
Abstract
Whereas genetic mutations can alter cell properties, nongenetic mechanisms can drive rapid and reversible adaptations to changes in their physical environment, a phenomenon termed 'cell-state transition'. Metals, in particular copper and iron, have been shown to be rate-limiting catalysts of cell-state transitions controlling key chemical reactions in mitochondria and the cell nucleus, which govern metabolic and epigenetic changes underlying the acquisition of distinct cell phenotypes. Acquisition of a distinct cell identity, independently of genetic alterations, is an underlying phenomenon of various biological processes, including development, inflammation, erythropoiesis, aging, and cancer. Here, mechanisms that have been uncovered related to the role of these metals in the regulation of cell plasticity are described, illustrating how copper and iron can be exploited for therapeutic intervention.
Collapse
Affiliation(s)
- Sebastian Müller
- Institut Curie, CNRS, INSERM, PSL Research University, Equipe labellisée Ligue Contre Le Cancer, Paris, France
| | - Tatiana Cañeque
- Institut Curie, CNRS, INSERM, PSL Research University, Equipe labellisée Ligue Contre Le Cancer, Paris, France
| | - Stéphanie Solier
- Institut Curie, CNRS, INSERM, PSL Research University, Equipe labellisée Ligue Contre Le Cancer, Paris, France; Department of Genetics, Institut Curie, Paris, France; Paris Saclay University, UVSQ, Montigny-le-Bretonneux, France
| | - Raphaël Rodriguez
- Institut Curie, CNRS, INSERM, PSL Research University, Equipe labellisée Ligue Contre Le Cancer, Paris, France.
| |
Collapse
|
29
|
Rodriguez R, Müller S, Colombeau L, Solier S, Sindikubwabo F, Cañeque T. Metal Ion Signaling in Biomedicine. Chem Rev 2025; 125:660-744. [PMID: 39746035 PMCID: PMC11758815 DOI: 10.1021/acs.chemrev.4c00577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 11/10/2024] [Accepted: 12/10/2024] [Indexed: 01/04/2025]
Abstract
Complex multicellular organisms are composed of distinct tissues involving specialized cells that can perform specific functions, making such life forms possible. Species are defined by their genomes, and differences between individuals within a given species directly result from variations in their genetic codes. While genetic alterations can give rise to disease-causing acquisitions of distinct cell identities, it is now well-established that biochemical imbalances within a cell can also lead to cellular dysfunction and diseases. Specifically, nongenetic chemical events orchestrate cell metabolism and transcriptional programs that govern functional cell identity. Thus, imbalances in cell signaling, which broadly defines the conversion of extracellular signals into intracellular biochemical changes, can also contribute to the acquisition of diseased cell states. Metal ions exhibit unique chemical properties that can be exploited by the cell. For instance, metal ions maintain the ionic balance within the cell, coordinate amino acid residues or nucleobases altering folding and function of biomolecules, or directly catalyze specific chemical reactions. Thus, metals are essential cell signaling effectors in normal physiology and disease. Deciphering metal ion signaling is a challenging endeavor that can illuminate pathways to be targeted for therapeutic intervention. Here, we review key cellular processes where metal ions play essential roles and describe how targeting metal ion signaling pathways has been instrumental to dissecting the biochemistry of the cell and how this has led to the development of effective therapeutic strategies.
Collapse
Affiliation(s)
- Raphaël Rodriguez
- Institut
Curie, CNRS, INSERM, PSL Research University, 75005 Paris, France
| | - Sebastian Müller
- Institut
Curie, CNRS, INSERM, PSL Research University, 75005 Paris, France
| | - Ludovic Colombeau
- Institut
Curie, CNRS, INSERM, PSL Research University, 75005 Paris, France
| | - Stéphanie Solier
- Institut
Curie, CNRS, INSERM, PSL Research University, 75005 Paris, France
- Université
Paris-Saclay, UVSQ, 78180 Montigny-le-Bretonneux, France
| | | | - Tatiana Cañeque
- Institut
Curie, CNRS, INSERM, PSL Research University, 75005 Paris, France
| |
Collapse
|
30
|
Zhou KXT, Bujold KE. The Emergence of Oligonucleotide Building Blocks in the Multispecific Proximity-Inducing Drug Toolbox of Destruction. ACS Chem Biol 2025; 20:3-18. [PMID: 39704048 DOI: 10.1021/acschembio.4c00311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
Oligonucleotides are a rapidly emerging class of therapeutics. Their most well-known examples are informational drugs that modify gene expression by binding mRNA. Despite inducing proximity between biological machinery and mRNA when applied to modulating gene expression, oligonucleotides are not typically labeled as "proximity-inducing" in literature. Yet, they have recently been explored as building blocks for multispecific proximity-inducing drugs (MPIDs). MPIDs are unique because they can direct endogenous biological machinery to destroy targeted molecules and cells, in contrast to traditional drugs that inhibit only their functions. The unique mechanism of action of MPIDs has enabled the targeting of previously "undruggable" molecular entities that cannot be effectively inhibited. However, the development of MPIDs must ensure that these molecules will selectively direct a potent, destruction-based mechanism of action toward intended targets over healthy tissues to avoid causing life-threatening toxicities. Oligonucleotides have emerged as promising building blocks for the design of MPIDs because they are sequence-controlled molecules that can be rationally designed to program multispecific binding interactions. In this Review, we examine the emergence of oligonucleotide-containing MPIDs in the proximity induction space, which has been dominated by antibody and small molecule MPID modalities. Moreover, examples of oligonucleotides developed as MPID candidates in immunotherapy and protein degradation are discussed to demonstrate the utility of oligonucleotides in expanding the scope and selectivity of the MPID toolbox. Finally, we discuss the utility of programming "AND" gates into oligonucleotide scaffolds to encode conditional responses that have the potential to be incorporated into MPIDs, which can further enhance their selectivity, thus increasing the scope of this drug category.
Collapse
Affiliation(s)
- Kevin Xiao Tong Zhou
- Department of Chemistry & Chemical Biology, McMaster University, 1280 Main Street West, Hamilton, ONL8S 4M1, Canada
| | - Katherine E Bujold
- Department of Chemistry & Chemical Biology, McMaster University, 1280 Main Street West, Hamilton, ONL8S 4M1, Canada
| |
Collapse
|
31
|
Jing X, Mackay JP, Passioura T. Macrocyclic peptides as a new class of targeted protein degraders. RSC Chem Biol 2025:d4cb00199k. [PMID: 39822773 PMCID: PMC11733494 DOI: 10.1039/d4cb00199k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 01/01/2025] [Indexed: 01/19/2025] Open
Abstract
Targeted protein degraders, in the form of proteolysis targeting chimaeras (PROTACs) and molecular glues, leverage the ubiquitin-proteasome system to catalytically degrade specific target proteins of interest. Because such molecules can be extremely potent, they have attracted considerable attention as a therapeutic modality in recent years. However, while targeted degraders have great potential, they are likely to face many of the same challenges as more traditional small molecules when it comes to their development as therapeutics. In particular, existing targeted degrader design is largely only applicable to the same set of protein targets as traditional small molecules (i.e., ∼15% of the human proteome). Here, we consider the potential of macrocyclic peptides to overcome this limitation. Such molecules possess several features that make them well-suited for the role, including the ability to induce the formation of ternary protein complexes that can involve relatively flat surfaces and their structural commonality with E3 ligase-recruiting peptide degrons. For these reasons, macrocyclic peptides provide the opportunity both to broaden the number of targets accessible to degrader activity and to broaden the number of E3 ligases that can be harnessed to mediate that activity.
Collapse
Affiliation(s)
- Xuefei Jing
- School of Life and Environmental Sciences, The University of Sydney Sydney NSW 2006 Australia
| | - Joel P Mackay
- School of Life and Environmental Sciences, The University of Sydney Sydney NSW 2006 Australia
| | - Toby Passioura
- School of Chemistry, The University of Sydney Sydney NSW 2006 Australia
- Insamo South Pty Ltd Chippendale NSW 2008 Australia
| |
Collapse
|
32
|
Deutscher RCE, Meyners C, Repity ML, Sugiarto WO, Kolos JM, Maciel EVS, Heymann T, Geiger TM, Knapp S, Lermyte F, Hausch F. Discovery of fully synthetic FKBP12-mTOR molecular glues. Chem Sci 2025:d4sc06917j. [PMID: 39916884 PMCID: PMC11796051 DOI: 10.1039/d4sc06917j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 01/10/2025] [Indexed: 02/09/2025] Open
Abstract
Molecular glues are a new drug modality with the potential to engage otherwise undruggable targets. However, the rational discovery of molecular glues for desired targets is a major challenge and most known molecular glues have been discovered by serendipity. Here we present the first fully synthetic FKBP12-mTOR molecular glues, which were discovered from a FKBP-focused, target-unbiased ligand library. Our biochemical screening of >1000 in-house FKBP ligands yielded one hit that induced dimerization of FKBP12 and the FRB domain of mTOR. The crystal structure of the ternary complex revealed that the hit targeted a similar surface on the FRB domain compared to natural product rapamycin but with a radically different interaction pattern. Structure-guided optimization improved potency 500-fold, and led to compounds which initiate FKBP12-FRB complex formation in cells. Our results show that molecular glues targeting flat surfaces can be discovered by focused screening and support the use of FKBP12 as a versatile presenter protein for molecular glues.
Collapse
Affiliation(s)
- Robin C E Deutscher
- Institute for Organic Chemistry and Biochemistry, Technical University Darmstadt Peter-Grünberg-Straße 4 64287 Darmstadt Germany
| | - Christian Meyners
- Institute for Organic Chemistry and Biochemistry, Technical University Darmstadt Peter-Grünberg-Straße 4 64287 Darmstadt Germany
| | - Maximilian L Repity
- Institute for Organic Chemistry and Biochemistry, Technical University Darmstadt Peter-Grünberg-Straße 4 64287 Darmstadt Germany
| | - Wisely Oki Sugiarto
- Institute for Organic Chemistry and Biochemistry, Technical University Darmstadt Peter-Grünberg-Straße 4 64287 Darmstadt Germany
| | - Jürgen M Kolos
- Institute for Organic Chemistry and Biochemistry, Technical University Darmstadt Peter-Grünberg-Straße 4 64287 Darmstadt Germany
| | - Edvaldo V S Maciel
- Institute for Organic Chemistry and Biochemistry, Technical University Darmstadt Peter-Grünberg-Straße 4 64287 Darmstadt Germany
| | - Tim Heymann
- Institute for Organic Chemistry and Biochemistry, Technical University Darmstadt Peter-Grünberg-Straße 4 64287 Darmstadt Germany
| | - Thomas M Geiger
- Institute for Organic Chemistry and Biochemistry, Technical University Darmstadt Peter-Grünberg-Straße 4 64287 Darmstadt Germany
| | - Stefan Knapp
- Institut für Pharmazeutische Chemie, Goethe-University Frankfurt, Biozentrum Max-von-Laue-Str. 9 60438 Frankfurt am Main Germany
- Structural Genomics Consortium, Goethe-University Frankfurt, Buchmann Institute for Life Sciences Max-von-Laue-Str. 15 60438 Frankfurt am Main Germany
- German Cancer Consortium (DKTK)/German Cancer Research Center (DKFZ), DKTK Site Frankfurt-Mainz 69120 Heidelberg Germany
| | - Frederik Lermyte
- Institute for Organic Chemistry and Biochemistry, Technical University Darmstadt Peter-Grünberg-Straße 4 64287 Darmstadt Germany
| | - Felix Hausch
- Institute for Organic Chemistry and Biochemistry, Technical University Darmstadt Peter-Grünberg-Straße 4 64287 Darmstadt Germany
- Centre for Synthetic Biology, Technical University of Darmstadt 64287 Darmstadt Germany
| |
Collapse
|
33
|
Dome PA, Jeong P, Nam G, Jang H, Rivera A, Floyd Averette A, Park E, Liao TC, Ciofani M, Wu J, Chi JTA, Venters RA, Park HJ, Steinbach WJ, Juvvadi PR, Heitman J, Hong J. Structure-guided design and synthesis of C22- and C32-modified FK520 analogs with enhanced activity against human pathogenic fungi. Proc Natl Acad Sci U S A 2025; 122:e2419883121. [PMID: 39739817 PMCID: PMC11725869 DOI: 10.1073/pnas.2419883121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 11/22/2024] [Indexed: 01/02/2025] Open
Abstract
Invasive fungal infections are a leading cause of death worldwide. Translating molecular insights into clinical benefits is challenging because fungal pathogens and their hosts share similar eukaryotic physiology. Consequently, current antifungal treatments have limited efficacy, may be poorly fungicidal in the host, can exhibit toxicity, and are increasingly compromised by emerging resistance. We have established that the phosphatase calcineurin (CaN) is required for invasive fungal disease and an attractive target for antifungal drug development. CaN is a druggable target, and there is vast clinical experience with the CaN inhibitors FK506 and cyclosporin A (CsA). However, while FK506 and its natural analog FK520 exhibit antifungal activity, they are also immunosuppressive in the host and thus not fungal-selective. We leverage our pathogenic fungal CaN-FK506-FKBP12 complex X-ray structures and biophysical data to support structure-based ligand design as well as structure-activity relationship analyses of broad-spectrum FK506/FK520 derivatives with potent antifungal activity and reduced immunosuppressive activity. Here, we apply molecular docking studies to develop antifungal C22- or C32-modified FK520 derivatives with improved therapeutic index scores. Among them, the C32-modified FK520 derivative JH-FK-44 (7) demonstrates a significantly improved therapeutic index compared to JH-FK-08, our lead compound to date. NMR binding studies with C32-derivatives are consistent with our hypothesis that C32 modifications disrupt the hydrogen bonding network in the human complex while introducing favorable electrostatic and cation-π interactions with the fungal FKBP12 R86 residue. These findings further reinforce calcineurin inhibition as a promising strategy for antifungal therapy.
Collapse
Affiliation(s)
| | | | - Gibeom Nam
- School of Pharmacy, Sungkyunkwan University, Suwon16419, Republic of Korea
| | - Hongjun Jang
- Department of Chemistry, Duke University, Durham, NC27708
| | - Angela Rivera
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC27710
| | - Anna Floyd Averette
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC27710
| | - Eunchong Park
- Department of Integrative Immunobiology, Duke University Medical Center, Durham, NC27710
| | - Tzu-Chieh Liao
- Department of Integrative Immunobiology, Duke University Medical Center, Durham, NC27710
| | - Maria Ciofani
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC27710
- Department of Integrative Immunobiology, Duke University Medical Center, Durham, NC27710
| | - Jianli Wu
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC27710
| | - Jen-Tsan Ashley Chi
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC27710
| | - Ronald A. Venters
- Duke University Nuclear Magnetic Resonance Center, Duke University, Durham, NC27710
- Department of Radiology, Duke University Medical Center, Durham, NC27710
| | - Hyun-Ju Park
- School of Pharmacy, Sungkyunkwan University, Suwon16419, Republic of Korea
| | - William J. Steinbach
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR72202
| | - Praveen R. Juvvadi
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR72202
| | - Joseph Heitman
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC27710
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC27710
| | - Jiyong Hong
- Department of Chemistry, Duke University, Durham, NC27708
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC27710
| |
Collapse
|
34
|
Kang J, Zhu JQ, Wang Y, He Q. Effect of Immunosuppressive Regimens on Metabolic Dysfunction-associated Fatty Liver Disease Following Liver Transplantation. J Clin Exp Hepatol 2025; 15:102387. [PMID: 39268481 PMCID: PMC11388780 DOI: 10.1016/j.jceh.2024.102387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 07/31/2024] [Indexed: 09/15/2024] Open
Abstract
Background Metabolic dysfunction-associated fatty liver disease has been linked to negative outcomes in patients with end-stage liver disease following liver transplantation. However, the influence of immunosuppressive regimens on it has not been explored. Methods A retrospective analysis was conducted using the preoperative and postoperative data from patients with end-stage liver disease. The study compared three different groups: tacrolimus-based group, sirolimus-based group, and combined tacrolimus- and sirolimus-based regimens. Binary logistic regression analysis was employed to identify risk factors for metabolic dysfunction-associated fatty liver disease. Results A total of 171 patients participated in the study, consisting of 127 males and 44 females, with a mean age of 49.6 years. The prevalence of posttransplant metabolic dysfunction-associated fatty liver disease was 29.23%. Among the three groups, there were 111 liver transplant recipients in the tacrolimus-based group, 28 in the sirolimus-based group, and 32 in the combination group. A statistically significant difference was observed in the incidence of metabolic dysfunction-associated fatty liver disease (P < 0.05), whereas the other preoperative and postoperative parameters showed no significant differences. Multivariate analysis revealed that a low-calorie diet (95% confidence intervals: 0.15-0.90, P = 0.021) and a combination of tacrolimus- and sirolimus-based immunosuppressive regimen (95% confidence intervals: 1.01-2.77, P = 0.046) were associated with lower risk of posttransplant metabolic dysfunction-associated fatty liver disease. Conclusions Our study indicates that implementing a low-calorie diet and utilizing a combination of tacrolimus- and sirolimus-based immunosuppressive regimen can effectively lower the risk of posttransplant metabolic dysfunction-associated fatty liver disease following liver transplantation.
Collapse
Affiliation(s)
- Jing Kang
- Department of Internal Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, PR China
| | - Ji-Qiao Zhu
- Department of Hepatobiliary and Pancreaticosplenic Surgery, Beijing Organ Transplant Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, PR China
| | - Yan Wang
- Department of Hepatobiliary and Pancreaticosplenic Surgery, Beijing Organ Transplant Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, PR China
| | - Qiang He
- Department of Hepatobiliary and Pancreaticosplenic Surgery, Beijing Organ Transplant Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, PR China
| |
Collapse
|
35
|
Han JW, Park SH. Advancing immunosuppression in liver transplantation: the role of regulatory T cells in immune modulation and graft tolerance. CLINICAL TRANSPLANTATION AND RESEARCH 2024; 38:257-272. [PMID: 39696994 PMCID: PMC11732766 DOI: 10.4285/ctr.24.0059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 11/23/2024] [Accepted: 11/28/2024] [Indexed: 12/20/2024]
Abstract
Prolonged immunosuppressive therapy in liver transplantation (LT) is associated with significant adverse effects, such as nephrotoxicity, metabolic complications, and heightened risk of infection or malignancy. Regulatory T cells (Tregs) represent a promising target for inducing immune tolerance in LT, with the potential to reduce or eliminate the need for life-long immunosuppression. This review summarizes current knowledge on the roles of Tregs in LT, highlighting their mechanisms and the impact of various immunosuppressive agents on Treg stability and function. The liver's distinct immunological microenvironment, characterized by tolerogenic antigen-presenting cells and high levels of interleukin (IL)-10 and transforming growth factor-β, positions this organ as an ideal setting for Treg-mediated tolerance. We discuss Treg dynamics in LT, their association with rejection risk, and their utility as biomarkers of transplant outcomes. Emerging strategies, including the use of low-dose calcineurin inhibitors with mammalian target of rapamycin inhibitors, adoptive Treg therapy, and low-dose IL-2, aim to enhance Treg function while providing sufficient immunosuppression. Thus, the future of LT involves precision medicine approaches that integrate Treg monitoring with tailored immunosuppressive protocols to optimize long-term outcomes for LT recipients.
Collapse
Affiliation(s)
- Ji Won Han
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Su-Hyung Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| |
Collapse
|
36
|
Li C, Chen Y, Huang W, Qiu Y, Huang S, Zhou Y, Zhou F, Xu J, Ren X, Zhang J, Wang Z, Ding M, Ding K. Structure-Based Design of "Head-to-Tail" Macrocyclic PROTACs. JACS AU 2024; 4:4866-4882. [PMID: 39735913 PMCID: PMC11672125 DOI: 10.1021/jacsau.4c00831] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/29/2024] [Accepted: 11/06/2024] [Indexed: 12/31/2024]
Abstract
Macrocyclization is a compelling strategy for conventional drug design for improving biological activity, target specificity, and metabolic stability, but it was rarely applied to the design of PROTACs possibly due to the mechanism and structural complexity. Herein, we report the rational design of the first series of "Head-to-Tail" macrocyclic PROTACs. The resulting molecule SHD913 exhibited pronounced Brd4 protein degradation with low nM DC50 values while almost totally dismissing the "hook effect", which is a general character and common concern of a PROTAC, in multiple cancer cell lines. Further biological evaluation revealed that the compound exhibited positive cooperativity and induced de novo protein-protein interactions (PPIs) in both biophysical and cellular NanoBRET assays and outperformed macroPROTAC-1 that is the first reported macrocyclic Brd4 PROTAC, in cellular assays. In vitro liver microsomal stability evaluation suggested that SHD913 demonstrated improved metabolic stability in different species compared with the linear counterpart. The co-crystal structure of Brd4BD2: SHD913: VCB (VHL, Elongin C and Elongin B) complex determination and molecular dynamics (MD) simulation also elucidated details of the chemical-induced PPIs and highlighted the crucial contribution of restricted conformation of SHD913 to the ternary complex formation. These results collectively support that macrocyclization could be an attractive and feasible strategy for a new PROTAC design.
Collapse
Affiliation(s)
- Chungen Li
- State
Key Laboratory of Chemical Biology, Shanghai
Institute of Organic Chemistry, Chinese Academy of Sciences, No. 345 Lingling Road, Shanghai 200032, China
| | - Yihan Chen
- State
Key Laboratory of Chemical Biology, Shanghai
Institute of Organic Chemistry, Chinese Academy of Sciences, No. 345 Lingling Road, Shanghai 200032, China
- University
of Chinese Academy of Sciences, No. 1 Yanxihu Road, Huairou District, Beijing 101408, China
| | - Weixue Huang
- State
Key Laboratory of Chemical Biology, Shanghai
Institute of Organic Chemistry, Chinese Academy of Sciences, No. 345 Lingling Road, Shanghai 200032, China
| | - Yudi Qiu
- Key
Laboratory of Structure-Based Drugs Design and Discovery of Ministry
of Education, Shengyang Pharmaceutical University, Shenyang 110016, China
| | - Shengjie Huang
- International
Cooperative Laboratory of Traditional Chinese Medicine Modernization
and Innovative Drug Discovery of Chinese Ministry of Education (MOE),
Guangzhou City Key Laboratory of Precision Chemical Drug Development,
College of Pharmacy, Jinan University, No. 855 Xingye Avenue East, Guangzhou 511400, China
| | - Yang Zhou
- International
Cooperative Laboratory of Traditional Chinese Medicine Modernization
and Innovative Drug Discovery of Chinese Ministry of Education (MOE),
Guangzhou City Key Laboratory of Precision Chemical Drug Development,
College of Pharmacy, Jinan University, No. 855 Xingye Avenue East, Guangzhou 511400, China
| | - Fengtao Zhou
- International
Cooperative Laboratory of Traditional Chinese Medicine Modernization
and Innovative Drug Discovery of Chinese Ministry of Education (MOE),
Guangzhou City Key Laboratory of Precision Chemical Drug Development,
College of Pharmacy, Jinan University, No. 855 Xingye Avenue East, Guangzhou 511400, China
| | - Jian Xu
- Livzon Research
Institute, Livzon Pharmaceutical Group Inc., No. 38, Chuangye North Road, Jinwan
District, Zhuhai 519000, China
| | - Xiaomei Ren
- State
Key Laboratory of Chemical Biology, Shanghai
Institute of Organic Chemistry, Chinese Academy of Sciences, No. 345 Lingling Road, Shanghai 200032, China
| | - Jinwei Zhang
- State
Key Laboratory of Chemical Biology, Shanghai
Institute of Organic Chemistry, Chinese Academy of Sciences, No. 345 Lingling Road, Shanghai 200032, China
| | - Zhen Wang
- State
Key Laboratory of Chemical Biology, Shanghai
Institute of Organic Chemistry, Chinese Academy of Sciences, No. 345 Lingling Road, Shanghai 200032, China
| | - Ming Ding
- School
of Life Science and Technology, China Pharmaceutical
University, No. 639 Longmian
Avenue, Nanjing 211198, China
| | - Ke Ding
- State
Key Laboratory of Chemical Biology, Shanghai
Institute of Organic Chemistry, Chinese Academy of Sciences, No. 345 Lingling Road, Shanghai 200032, China
| |
Collapse
|
37
|
Zheng X, Qin S, Zhong M, Xu Q, Huai C, Qiu X. PPP3R1 Promoter Polymorphism (Allelic Variation) Affects Tacrolimus Treatment Efficacy by Modulating E2F6 Binding Affinity. Biomedicines 2024; 12:2896. [PMID: 39767802 PMCID: PMC11727355 DOI: 10.3390/biomedicines12122896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/02/2024] [Accepted: 12/17/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Tacrolimus is widely used as a first-line immunosuppressant in transplant immunology; however, its clinical application is constrained by the narrow therapeutic index and considerable interindividual variability. In this study, we identified the potential regulatory role of a novel PPP3R1 promoter polymorphism, rs4519508 C > T, in the tacrolimus pharmacodynamic pathway. METHODS Dual-luciferase reporter assays and bioinformatic analysis were applied to assess the impact of allelic variation. Electrophoretic mobility shift assays (EMSA) validated the altered binding of transcription factors. Quantitative real-time PCR (qRT-PCR), enzyme-linked immunosorbent assay (ELISA) and Western blots were used to determine the immunosuppressive effect of tacrolimus. RESULTS Assays revealed that rs4519508 C > T markedly enhanced PPP3R1 promoter activity. EMSA assays validated the binding of E2F6 to rs4519508 C (wild-type) and the binding was significantly weaker to the rs4519508 T (mutant-type). The overexpression of E2F6 significantly reduced the transcriptional activity and expression of PPP3R1 when the rs4519508 site presented as major C allele, an effect that was not observed with the rs4519508 T allele. Furthermore, the downregulation of E2F6 raises the level of downstream immune cytokines inhibited by TAC. CONCLUSIONS This study proposed that E2F6 suppresses the expression of PPP3R1, while rs4519508 C > T impairs the binding of E2F6, and thus elevates the level of PPP3R1, so that the inhibition of the downstream immune cytokines by TAC is attenuated. Our findings reported the potential regulatory role of a novel polymorphism, PPP3R1 rs4519508 C > T, which may serve as pharmacodynamic-associated pharmacogenetic biomarker indicating individual response variability of tacrolimus, and thus aid the clinical management of transplant immunology.
Collapse
Affiliation(s)
- Xinyi Zheng
- Department of Pharmacy, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai 200040, China; (X.Z.)
| | - Shengying Qin
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, No. 1954 Huashan Rd, Shanghai 200030, China
| | - Mingkang Zhong
- Department of Pharmacy, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai 200040, China; (X.Z.)
| | - Qinxia Xu
- Department of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Cong Huai
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, No. 1954 Huashan Rd, Shanghai 200030, China
| | - Xiaoyan Qiu
- Department of Pharmacy, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai 200040, China; (X.Z.)
| |
Collapse
|
38
|
Sousa IS, Tavares LFS, Silva BA, Moreno DSA, Alviano CS, Santos ALS, Kneipp LF. Calcineurin activity in Fonsecaea pedrosoi: tacrolimus and cyclosporine A inhibited conidia growth, filamentation and showed synergism with itraconazole. Braz J Microbiol 2024; 55:3643-3654. [PMID: 39044105 PMCID: PMC11711851 DOI: 10.1007/s42770-024-01463-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 07/11/2024] [Indexed: 07/25/2024] Open
Abstract
Fonsecaea pedrosoi is a melanized fungus that causes chromoblastomycosis (CBM), a tropical neglected disease responsible for chronic and disability-related subcutaneous mycosis. Given the challenging nature of CBM treatment, the study of new targets and novel bioactive drugs capable of improving patient life quality is urgent. In the present work, we detected a calcineurin activity in F. pedrosoi conidial form, employing primarily colorimetric, immunoblotting and flow cytometry assays. Our findings reveal that the calcineurin activity of F. pedrosoi was stimulated by Ca2+/calmodulin, inhibited by EGTA and specific inhibitors, such as tacrolimus (FK506) and cyclosporine A (CsA), and proved to be insensitive to okadaic acid. In addition, FK506 and CsA were able to affect the cellular viability and the fungal proliferation. This effect was corroborated by transmission electron microscopy that showed both calcineurin inhibitors promoted profound changes in the ultrastructure of conidia, causing mainly cytoplasm condensation and intense vacuolization that are clear indication of cell death. Our data indicated that FK506 exhibited the highest effectiveness, with a minimum inhibitory concentration (MIC) of 3.12 mg/L, whereas CsA required 15.6 mg/L to inhibit 100% of conidial growth. Interestingly, when both were combined with itraconazole, they demonstrated anti-F. pedrosoi activity, exhibiting a synergistic effect. Moreover, the fungal filamentation was affected after treatment with both calcineurin inhibitors. These data corroborate with other calcineurin studies in fungal cells and open up further discussions aiming to establish the role of this enzyme as a potential target for antifungal therapy against CBM infections.
Collapse
Affiliation(s)
- Ingrid S Sousa
- Laboratório de Taxonomia, Bioquímica e Bioprospecção de Fungos (LTBBF), Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, 21040-900, Brazil
| | - Lucilene F S Tavares
- Laboratório de Taxonomia, Bioquímica e Bioprospecção de Fungos (LTBBF), Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, 21040-900, Brazil
| | - Bianca A Silva
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes (LEAMER), Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, 21941-901, Brazil
| | - Daniela S A Moreno
- Laboratório de Estrutura de Microrganismos, IMPG, UFRJ, Rio de Janeiro, 21941-902, Brazil
| | - Celuta S Alviano
- Laboratório de Estrutura de Microrganismos, IMPG, UFRJ, Rio de Janeiro, 21941-902, Brazil
| | - André L S Santos
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes (LEAMER), Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, 21941-901, Brazil
- Rede Micologia RJ, Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ), Rio de Janeiro, 21941-901, Brazil
| | - Lucimar F Kneipp
- Laboratório de Taxonomia, Bioquímica e Bioprospecção de Fungos (LTBBF), Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, 21040-900, Brazil.
- Rede Micologia RJ, Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ), Rio de Janeiro, 21941-901, Brazil.
| |
Collapse
|
39
|
Berber M, Penton D. Calcineurin inhibitors and the renin-angiotensin-aldosterone system. Acta Physiol (Oxf) 2024; 240:e14248. [PMID: 39460458 DOI: 10.1111/apha.14248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 10/07/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024]
Abstract
Calcineurin inhibitors (CnIs) are effective immunosuppressants with decades of accumulated experience in treating immune disorders and, most notably, solid organ transplantation. While CnIs have significantly increased graft survival and transformed the patient standard of care, their use has been overshadowed by a number of undesired side effects. For instance, CnI-associated nephrotoxicity has been reported since early studies and remains a major therapeutic concern. The occurrence of several ion imbalances alongside hypertension was also noted early on, indicating the involvement of the renin-angiotensin-aldosterone system (RAAS) in CnI-mediated toxicity. However, the literature in this field is crowded with conflicting reports from clinical trials as well as studies using animal and invitro models. With this review, we aim to provide a structured and updated overview of the physiological and pathophysiological evidence supporting the involvement of the classical RAAS in CnI-associated toxicity.
Collapse
Affiliation(s)
- Mesut Berber
- Department of Pediatrics, Harvard Medical School and Division of Endocrinology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - David Penton
- Electrophysiology Facility, University of Zurich, Zurich, Switzerland
| |
Collapse
|
40
|
Ferreira PA. Personal essay of a rookie's journey with Bill Pak and his legacy: tales and perspectives on PI-PLC, NorpA and cyclophilin, NinaA - William L. Pak, PhD., 1932-2023: in memoriam. J Neurogenet 2024; 38:165-174. [PMID: 38913811 DOI: 10.1080/01677063.2024.2366455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 05/30/2024] [Indexed: 06/26/2024]
Abstract
The neurogenetics and vision community recently mourned William L. Pak, PhD, whose pioneering work spearheaded the genetic, electrophysiological, and molecular bases of biological processes underpinning vision. This essay provides a historical background to the daunting challenges and personal experiences that carved the path to seminal findings. It also reflects on the intellectual framework, mentoring philosophy, and inspirational legacy of Bill Pak's research. An emphasis and perspectives are placed on the discoveries and implications to date of the phosphatidylinositol-specific phospholipase C (PI-PLC), NorpA, and the cyclophilin, NinaA of the fruit fly, Drosophila melanogaster, and their respective mammalian homologues, PI-PLCβ4, and cyclophilin-related protein, Ran-binding protein 2 (Ranbp2) in critical biological processes and diseases of photoreceptors and other neurons.
Collapse
Affiliation(s)
- Paulo A Ferreira
- Departments of Ophthalmology and Pathology, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
41
|
Hu Q, Liu Y, Yue Q, Zhou S, Jin X, Lin F, Huang XJ, Zhuang J, Lu J, Gao X, Lee HY. Lenalidomide-induced pure red cell aplasia is associated with elevated expression of MHC-I molecules on erythrocytes. Nat Commun 2024; 15:10131. [PMID: 39578482 PMCID: PMC11584757 DOI: 10.1038/s41467-024-54571-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 11/15/2024] [Indexed: 11/24/2024] Open
Abstract
The RVd therapy, combining lenalidomide, bortezomib, and dexamethasone, is a mainstay treatment for multiple myeloma. A multiple myeloma patient developed pure red cell aplasia (PRCA) following RVd treatment, despite the absence of common PRCA triggers. In vitro analyses reveal lenalidomide as a pivotal disruptor of erythropoiesis. Single-cell transcriptome analysis unveils hyperactive CD8+ T cells and impaired erythropoiesis in the patient's bone marrow. Unexpectedly, the patient's erythroid cells display abnormally high expression of genes in the antigen presentation pathway, particularly those for major histocompatibility class I (MHC-I) molecules. Functional assays demonstrate that lenalidomide treatment further augmented MHC-I expression in the patient's erythroid cells. Blocking MHC-I or depleting T cells alleviates the defective erythropoiesis of PRCA, suggesting that the interaction between erythroid cells with elevated MHC-I and T cells in the bone marrow might contribute to PRCA. Taken together, our study implicates a mechanism underlying lenalidomide-induced PRCA in treating cancer patients.
Collapse
Affiliation(s)
- Qi Hu
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, 100871, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Yang Liu
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing, 100871, China
| | - Qiuyu Yue
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, 100871, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Shuo Zhou
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, 100871, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Xianghong Jin
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Fan Lin
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing, 100871, China
| | - Xiao-Jun Huang
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing, 100871, China
| | - Junling Zhuang
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China.
| | - Jin Lu
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing, 100871, China.
| | - Xiaofei Gao
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, 310030, Zhejiang, China.
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China.
| | - Hsiang-Ying Lee
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, 100871, China.
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China.
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing, 100871, China.
| |
Collapse
|
42
|
Chandler F, Reddy PAN, Bhutda S, Ross RL, Datta A, Walden M, Walker K, Di Donato S, Cassel JA, Prakesch MA, Aman A, Datti A, Campbell LJ, Foglizzo M, Bell L, Stein DN, Ault JR, Al-awar RS, Calabrese AN, Sicheri F, Del Galdo F, Salvino JM, Greenberg RA, Zeqiraj E. Molecular glues that inhibit deubiquitylase activity and inflammatory signalling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.07.611787. [PMID: 39282282 PMCID: PMC11398498 DOI: 10.1101/2024.09.07.611787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Deubiquitylases (DUBs) are crucial in cell signalling and are often regulated by interactions within protein complexes. The BRCC36 isopeptidase complex (BRISC) regulates inflammatory signalling by cleaving K63-linked polyubiquitin chains on Type I interferon receptors (IFNAR1). As a Zn2+-dependent JAMM/MPN DUB, BRCC36 is challenging to target with selective inhibitors. We discovered first-in-class inhibitors, termed BRISC molecular glues (BLUEs), which stabilise a 16-subunit BRISC dimer in an autoinhibited conformation, blocking active sites and interactions with the targeting subunit SHMT2. This unique mode of action results in selective inhibition of BRISC over related complexes with the same catalytic subunit, splice variants and other JAMM/MPN DUBs. BLUE treatment reduced interferon-stimulated gene expression in cells containing wild type BRISC, and this effect was absent when using structure-guided, inhibitor-resistant BRISC mutants. Additionally, BLUEs increase IFNAR1 ubiquitylation and decrease IFNAR1 surface levels, offering a potential new strategy to mitigate Type I interferon-mediated diseases. Our approach also provides a template for designing selective inhibitors of large protein complexes by promoting, rather than blocking, protein-protein interactions.
Collapse
Affiliation(s)
- Francesca Chandler
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Poli Adi Narayana Reddy
- The Wistar Cancer Center for Molecular Screening, The Wistar Institute, Philadelphia, PA, USA
| | - Smita Bhutda
- Department of Cancer Biology, Penn Center for Genome Integrity, Basser Center for BRCA, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rebecca L. Ross
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals, NHS Trust, Chapel Allerton Hospital, Leeds, UK
| | - Arindam Datta
- Department of Cancer Biology, Penn Center for Genome Integrity, Basser Center for BRCA, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Miriam Walden
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Kieran Walker
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Stefano Di Donato
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals, NHS Trust, Chapel Allerton Hospital, Leeds, UK
| | - Joel A. Cassel
- The Wistar Cancer Center for Molecular Screening, The Wistar Institute, Philadelphia, PA, USA
| | - Michael A. Prakesch
- Drug Discovery Program, Ontario Institute for Cancer Research, Toronto, ON, Canada
| | - Ahmed Aman
- Drug Discovery Program, Ontario Institute for Cancer Research, Toronto, ON, Canada
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - Alessandro Datti
- Department of Agriculture, Food, and Environmental Sciences, University of Perugia, Perugia, Italy
| | - Lisa J. Campbell
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Martina Foglizzo
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Lillie Bell
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Daniel N. Stein
- Department of Cancer Biology, Penn Center for Genome Integrity, Basser Center for BRCA, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - James R. Ault
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Rima S. Al-awar
- Drug Discovery Program, Ontario Institute for Cancer Research, Toronto, ON, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Antonio N. Calabrese
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Frank Sicheri
- Centre for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Francesco Del Galdo
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals, NHS Trust, Chapel Allerton Hospital, Leeds, UK
| | - Joseph M. Salvino
- The Wistar Cancer Center for Molecular Screening, The Wistar Institute, Philadelphia, PA, USA
| | - Roger A. Greenberg
- Department of Cancer Biology, Penn Center for Genome Integrity, Basser Center for BRCA, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elton Zeqiraj
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| |
Collapse
|
43
|
Dreizler JK, Meyners C, Hausch F. Toward Dual Targeting of Catalytic and Gatekeeper Pockets in Cyclophilins Using a Macrocyclic Scaffold. ACS Med Chem Lett 2024; 15:2012-2018. [PMID: 39563809 PMCID: PMC11571008 DOI: 10.1021/acsmedchemlett.4c00427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/30/2024] [Accepted: 10/16/2024] [Indexed: 11/21/2024] Open
Abstract
Cyclophilins, especially cyclophilin A, are involved in a variety of diseases, including the life cycle of many viruses. An advanced macrocyclic inhibitor of cyclophilin was reported to bind the catalytic pocket but not the neighboring gatekeeper pocket. Here we describe macrocyclic cyclophilin inhibitors bearing side chains designed to reach out to the gatekeeper pocket. After establishing a suitable synthesis allowing for late-stage modification of the relevant positions, we explored this exit vector. This culminated in a rigid ornithine-resembling analogue as a versatile building block, which was also incorporated into the macrocyclic scaffold. The use of amines as the gatekeeper-engaging modality was invalidated, but the exit vector was successfully established as a promising position for future modifications. Further work is needed to identify suitable motifs to simultaneously engage the catalytic and gatekeeper pockets in this highly developed macrocyclic scaffold.
Collapse
Affiliation(s)
- Johannes K Dreizler
- Department of Chemistry and Biochemistry Clemens-Schöpf-Institute, Technical University Darmstadt, Peter-Grünberg Straße 4, 64287 Darmstadt, Germany
| | - Christian Meyners
- Department of Chemistry and Biochemistry Clemens-Schöpf-Institute, Technical University Darmstadt, Peter-Grünberg Straße 4, 64287 Darmstadt, Germany
| | - Felix Hausch
- Department of Chemistry and Biochemistry Clemens-Schöpf-Institute, Technical University Darmstadt, Peter-Grünberg Straße 4, 64287 Darmstadt, Germany
- Centre for Synthetic Biology, Technical University Darmstadt, 64287 Darmstadt, Germany
| |
Collapse
|
44
|
Dome PA, Jeong P, Nam G, Jang H, Rivera A, Averette AF, Park E, Liao TC, Ciofani M, Wu J, Chi JTA, Venters RA, Park HJ, Steinbach WJ, Juvvadi PR, Heitman J, Hong J. Structure-guided design and synthesis of C22- and C32-modified FK520 analogs with enhanced activity against human pathogenic fungi. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.27.615491. [PMID: 39386510 PMCID: PMC11463406 DOI: 10.1101/2024.09.27.615491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Invasive fungal infections are a leading cause of death worldwide. Translating molecular insights into clinical benefits is challenging because fungal pathogens and their hosts share similar eukaryotic physiology. Consequently, current antifungal treatments have limited efficacy, may be poorly fungicidal in the host, can exhibit toxicity, and are increasingly compromised by emerging resistance. We have established that the phosphatase calcineurin (CaN) is required for invasive fungal disease and an attractive target for antifungal drug development. CaN is a druggable target, and there is vast clinical experience with the CaN inhibitors FK506 and cyclosporin A (CsA). However, while FK506 and its natural analog FK520 exhibit antifungal activity, they are also immunosuppressive in the host and thus not fungal-selective. We leverage our pathogenic fungal CaN-FK506-FKBP12 complex X-ray structures and biophysical data to support structure-based ligand design as well as structure-activity relationship analyses of broad-spectrum FK506/FK520 derivatives with potent antifungal activity and reduced immunosuppressive activity. Here we apply molecular docking studies to develop antifungal C22- or C32-modified FK520 derivatives with improved therapeutic index scores. Among them, the C32-modified FK520 derivative JH-FK-44 ( 7 ) demonstrates a significantly improved therapeutic index compared to JH-FK-08, our lead compound to date. NMR binding studies with C32-derivatives are consistent with our hypothesis that C32 modifications disrupt the hydrogen bonding network in the human complex while introducing favorable electrostatic and cation-π interactions with the fungal FKBP12 R86 residue. These findings further reinforce calcineurin inhibition as a promising strategy for antifungal therapy. Significance Invasive fungal infections cause significant mortality worldwide, and current antifungal treatments are often ineffective, toxic, or face growing resistance. This research identifies calcineurin (CaN), a critical protein for fungal survival, as a potential target for developing new antifungal drugs. Although existing CaN inhibitors such as FK506 (tacrolimus) and FK520 (ascomycin) possess antifungal properties, their immunosuppressive effects limit their clinical utility. By studying the structure of human and fungal FKBP12-FK506 or FK520 complexes with CaN, we have designed and synthesized modified FK520 derivatives with strong antifungal activity and reduced immunosuppressive effects. These new derivatives are expected to have significantly improved therapeutic profiles, offering hope for more effective and safer antifungal treatments.
Collapse
|
45
|
Chimura T, Manabe T. Ca2+-PP2B-PSD-95 axis: A novel regulatory mechanism of the phosphorylation state of Serine 295 of PSD-95. PLoS One 2024; 19:e0313441. [PMID: 39509447 PMCID: PMC11542788 DOI: 10.1371/journal.pone.0313441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 10/23/2024] [Indexed: 11/15/2024] Open
Abstract
The phosphorylation state of PSD-95 at Serine 295 (Ser295) is important for the regulation of synaptic plasticity. Although the activation of NMDA receptors (NMDARs), which initiates an intracellular calcium signaling cascade, decreases phosphorylated Ser295 (pS295) of PSD-95, the molecular mechanisms are not fully understood. We found that the calcium-activated protein phosphatase PP2B dephosphorylated pS295 not only in basal conditions but also in NMDAR-activated conditions in cultured neurons. The biochemical assay also revealed the dephosphorylation of pS295 by PP2B, consistently supporting the results obtained using neurons. The newly identified calcium signaling cascade "Ca2+-PP2B-PSD-95 axis" would play an important role in the molecular mechanism for NMDA receptor-dependent plasticity.
Collapse
Affiliation(s)
- Takahiko Chimura
- Department of Basic Medical Sciences, Institute of Medical Science, Division of Neuronal Network, University of Tokyo, Tokyo, Japan
| | - Toshiya Manabe
- Department of Basic Medical Sciences, Institute of Medical Science, Division of Neuronal Network, University of Tokyo, Tokyo, Japan
| |
Collapse
|
46
|
Zhong G, Chang X, Xie W, Zhou X. Targeted protein degradation: advances in drug discovery and clinical practice. Signal Transduct Target Ther 2024; 9:308. [PMID: 39500878 PMCID: PMC11539257 DOI: 10.1038/s41392-024-02004-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 08/19/2024] [Accepted: 09/28/2024] [Indexed: 11/08/2024] Open
Abstract
Targeted protein degradation (TPD) represents a revolutionary therapeutic strategy in disease management, providing a stark contrast to traditional therapeutic approaches like small molecule inhibitors that primarily focus on inhibiting protein function. This advanced technology capitalizes on the cell's intrinsic proteolytic systems, including the proteasome and lysosomal pathways, to selectively eliminate disease-causing proteins. TPD not only enhances the efficacy of treatments but also expands the scope of protein degradation applications. Despite its considerable potential, TPD faces challenges related to the properties of the drugs and their rational design. This review thoroughly explores the mechanisms and clinical advancements of TPD, from its initial conceptualization to practical implementation, with a particular focus on proteolysis-targeting chimeras and molecular glues. In addition, the review delves into emerging technologies and methodologies aimed at addressing these challenges and enhancing therapeutic efficacy. We also discuss the significant clinical trials and highlight the promising therapeutic outcomes associated with TPD drugs, illustrating their potential to transform the treatment landscape. Furthermore, the review considers the benefits of combining TPD with other therapies to enhance overall treatment effectiveness and overcome drug resistance. The future directions of TPD applications are also explored, presenting an optimistic perspective on further innovations. By offering a comprehensive overview of the current innovations and the challenges faced, this review assesses the transformative potential of TPD in revolutionizing drug development and disease management, setting the stage for a new era in medical therapy.
Collapse
Affiliation(s)
- Guangcai Zhong
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Xiaoyu Chang
- School of Pharmaceutical Sciences, Pingyuan Laboratory, Zhengzhou University, Zhengzhou, 450001, China
| | - Weilin Xie
- Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
| | - Xiangxiang Zhou
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China.
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China.
| |
Collapse
|
47
|
Yang Y, Cheng K, Xu G. Novel approaches to primary membranous nephropathy: Beyond the KDIGO guidelines. Eur J Pharmacol 2024; 982:176928. [PMID: 39182551 DOI: 10.1016/j.ejphar.2024.176928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/05/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
Primary membranous nephropathy (PMN) is an immune-mediated glomerular disease. Rituximab (RTX) is recommended as a first-line immunosuppressive therapy and shows high clinical efficacy, but the optimal doses remain controversial. Approximately 20%-40% of PMN patients experience RTX resistance and failure. Reduced bioavailability, RTX internalization and attack, anti-RTX antibody production, autoreactive B-cell reservoirs and chronic and irreversible renal damage may contribute to this problem. Therefore, new treatment modalities are needed to compensate for this deficit. New interventions and new dose combinations are being proposed. Multiple drug combination therapies show comparable clinical efficacy to conventional treatments by blocking the production of disease-causing antibodies in multiple directions, and can reduce single-agent doses without increasing adverse effects. New therapies that directly target B cells, plasma cells, and antibody production have shown encouraging results. In addition, new techniques for sweeping antibodies and chimeric antigen receptor T-cell therapy also may be promising strategies for PMN. Immunoadsorption could be used as an auxiliary choice for severe cases. This article explores new treatments for PMN and highlights possible mechanisms for potential new technologies that offer new ideas for treatment.
Collapse
Affiliation(s)
- Yang Yang
- Department of Nephrology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, PR China; Jiangxi Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang University, PR China
| | - Kaiqi Cheng
- The Third Hospital of Nanchang, Nanchang, PR China
| | - Gaosi Xu
- Department of Nephrology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, PR China.
| |
Collapse
|
48
|
Zhang R, Zheng Y, Xiang F, Zhou J. Inducing or enhancing protein-protein interaction to develop drugs: Molecular glues with various biological activity. Eur J Med Chem 2024; 277:116756. [PMID: 39191033 DOI: 10.1016/j.ejmech.2024.116756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 07/15/2024] [Accepted: 08/01/2024] [Indexed: 08/29/2024]
Abstract
Over the past two decades, molecular glues (MGs) have gradually attracted the attention of the pharmaceutical community with the advent of MG degraders such as IMiDs and indisulam. Such molecules degrade the target protein by promoting the interaction between the target protein and E3 ligase. In addition, as a chemical inducer, MGs promote the dimerization of homologous proteins and heterologous proteins to form ternary complexes, which have great prospects in regulating biological activities. This review focuses on the application of MGs in the field of drug development including protein-protein interaction (PPI) stability and protein degradation. We thoroughly analyze the structure of various MGs and the interactions between MGs and various biologically active molecules, thus providing new perspectives for the development of PPI stabilizers and new degraders.
Collapse
Affiliation(s)
- Rongyu Zhang
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, Department of Chemistry, Zhejiang Normal University, 688 Yingbin Road, Jinhua, 321004, PR China
| | - Yirong Zheng
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, Department of Chemistry, Zhejiang Normal University, 688 Yingbin Road, Jinhua, 321004, PR China
| | - Fengjiao Xiang
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, Department of Chemistry, Zhejiang Normal University, 688 Yingbin Road, Jinhua, 321004, PR China
| | - Jinming Zhou
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, Department of Chemistry, Zhejiang Normal University, 688 Yingbin Road, Jinhua, 321004, PR China.
| |
Collapse
|
49
|
Ren Y, Chen H, Zhao SY, Ma L, He QX, Gong WB, Wu JW, Yao HW, Wang ZX. Biochemical analyses reveal new insights into RCAN1/Rcn1 inhibition of calcineurin. FEBS J 2024; 291:4813-4829. [PMID: 39241105 DOI: 10.1111/febs.17266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 06/18/2024] [Accepted: 08/23/2024] [Indexed: 09/08/2024]
Abstract
Calcineurin is a serine/threonine protein phosphatase that is highly conserved from yeast to human and plays a critical role in many physiological processes. Regulators of calcineurin (RCANs) are a family of endogenous calcineurin regulators, which are capable of inhibiting the catalytic activity of calcineurin in vivo and in vitro. In this study, we first characterized the biochemical properties of yeast calcineurin and its endogenous regulator Rcn1, a yeast homolog of RCAN1. Our data show that Rcn1 inhibits yeast calcineurin toward pNPP substrate with a noncompetitive mode; and Rcn1 binds cooperatively to yeast calcineurin through multiple low-affinity interactions at several docking regions. Next, we reinvestigated the mechanism underlying the inhibition of mammalian calcineurin by RCAN1 using a combination of biochemical, biophysical, and computational methods. In contrast to previous observations, RCAN1 noncompetitively inhibits calcineurin phosphatase activity toward both pNPP and phospho-RII peptide substrates by targeting the enzyme active site in part. Re-analysis of previously reported kinetic data reveals that the RCAN1 concentrations used were too low to distinguish between the inhibition mechanisms [Chan B et al. (2005) Proc Natl Acad Sci USA 102, 13075]. The results presented in this study provide new insights into the interaction between calcineurin and RCAN1/Rcn1.
Collapse
Affiliation(s)
- Yan Ren
- Institute of Molecular Enzymology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
- Department of Biochemistry and Molecular Biology, Beijing Normal University, China
| | - Hui Chen
- Institute of Molecular Enzymology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| | - Shan-Yue Zhao
- Institute of Molecular Enzymology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| | - Lei Ma
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Qing-Xia He
- Institute of Molecular Enzymology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| | - Wei-Bin Gong
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Jia-Wei Wu
- Institute of Molecular Enzymology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| | - Hong-Wei Yao
- Institute of Molecular Enzymology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| | - Zhi-Xin Wang
- Institute of Molecular Enzymology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| |
Collapse
|
50
|
Martín-Sierra C, Martins R, Coucelo M, Abrantes AM, Caetano Oliveira R, Tralhão JG, Botelho MF, Furtado E, Domingues MR, Paiva A, Laranjeira P. Tumor Resection in Hepatic Carcinomas Restores Circulating T Regulatory Cells. J Clin Med 2024; 13:6011. [PMID: 39408071 PMCID: PMC11478317 DOI: 10.3390/jcm13196011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/07/2024] [Accepted: 10/07/2024] [Indexed: 10/20/2024] Open
Abstract
Background/Objectives: Cholangiocarcinoma (CCA) and hepatocellular carcinoma (HCC) represent major primary liver cancers, affecting one of the most vital organs in the human body. T regulatory (Treg) cells play an important role in liver cancers through the immunosuppression of antitumor immune responses. The current study focuses on the characterization of circulating natural killer (NK) cells and T cell subsets, including Treg cells, in CCA and HCC patients, before and after surgical tumor resection, in order to understand the effect of tumor resection on the homeostasis of peripheral blood NK cells and T cells. Methods: Whole blood assays were performed to monitor immune alterations and the functional competence of circulating lymphocytes in a group of ten healthy individuals, eight CCA patients, and twenty HCC patients, before and one month after the surgical procedure, using flow cytometry, cell sorting, and qRT-PCR. Results: Before tumor resection, both HCC and CCA patients display increased percentages of CD8+ Treg cells and decreased frequencies of circulating CD4+ Treg cells. Notwithstanding, no functional impairment was detected on circulating CD4+ Treg cells, neither in CCA nor in HCC patients. Interestingly, the frequency of peripheral CD4+ Treg cells increased from 0.55% ± 0.49 and 0.71% ± 0.54 (in CCA and HCC, respectively) at T0 to 0.99% ± 0.91 and 1.17% ± 0.33 (in CCA and HCC, respectively) at T1, following tumor resection. Conclusions: Our results suggest mechanisms of immune modulation induced by tumor resection.
Collapse
Affiliation(s)
- Carmen Martín-Sierra
- Flow Cytometry Unit, Department of Clinical Pathology, Hospitais da Universidade de Coimbra, Unidade Local de Saúde de Coimbra, 3000-076 Coimbra, Portugal;
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Center of Environmental Genetics of Oncobiology (CIMAGO), Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal; (R.M.); (M.C.); (A.M.A.); (R.C.O.); (J.G.T.); (M.F.B.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3000-061 Coimbra, Portugal
| | - Ricardo Martins
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Center of Environmental Genetics of Oncobiology (CIMAGO), Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal; (R.M.); (M.C.); (A.M.A.); (R.C.O.); (J.G.T.); (M.F.B.)
- Unidade Transplantação Hepática Pediátrica e de Adultos, Centro Hospitalar e Universitário de Coimbra, 3000-075 Coimbra, Portugal;
- Serviço de Cirurgia Geral, Unidade HBP, Centro Hospitalar e Universitário de Coimbra, 3000-075 Coimbra, Portugal
- University of Coimbra, Faculty of Medicine, Biophysics Institute, 3000-548 Coimbra, Portugal
| | - Margarida Coucelo
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Center of Environmental Genetics of Oncobiology (CIMAGO), Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal; (R.M.); (M.C.); (A.M.A.); (R.C.O.); (J.G.T.); (M.F.B.)
- Unidade Funcional de Hematologia Molecular, Serviço de Hematologia Clínica, Centro Hospitalar e Universitário de Coimbra, 3000-075 Coimbra, Portugal
| | - Ana Margarida Abrantes
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Center of Environmental Genetics of Oncobiology (CIMAGO), Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal; (R.M.); (M.C.); (A.M.A.); (R.C.O.); (J.G.T.); (M.F.B.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3000-061 Coimbra, Portugal
- University of Coimbra, Faculty of Medicine, Biophysics Institute, 3000-548 Coimbra, Portugal
| | - Rui Caetano Oliveira
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Center of Environmental Genetics of Oncobiology (CIMAGO), Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal; (R.M.); (M.C.); (A.M.A.); (R.C.O.); (J.G.T.); (M.F.B.)
- University of Coimbra, Faculty of Medicine, Biophysics Institute, 3000-548 Coimbra, Portugal
- Serviço de Anatomia Patológica, Centro Hospitalar e Universitário de Coimbra, 3000-075 Coimbra, Portugal
| | - José Guilherme Tralhão
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Center of Environmental Genetics of Oncobiology (CIMAGO), Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal; (R.M.); (M.C.); (A.M.A.); (R.C.O.); (J.G.T.); (M.F.B.)
- Clinical Academic Center of Coimbra (CACC), 3000-061 Coimbra, Portugal
- Unidade Transplantação Hepática Pediátrica e de Adultos, Centro Hospitalar e Universitário de Coimbra, 3000-075 Coimbra, Portugal;
- Serviço de Cirurgia Geral, Unidade HBP, Centro Hospitalar e Universitário de Coimbra, 3000-075 Coimbra, Portugal
- University of Coimbra, Faculty of Medicine, Biophysics Institute, 3000-548 Coimbra, Portugal
| | - Maria Filomena Botelho
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Center of Environmental Genetics of Oncobiology (CIMAGO), Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal; (R.M.); (M.C.); (A.M.A.); (R.C.O.); (J.G.T.); (M.F.B.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3000-061 Coimbra, Portugal
- University of Coimbra, Faculty of Medicine, Biophysics Institute, 3000-548 Coimbra, Portugal
| | - Emanuel Furtado
- Unidade Transplantação Hepática Pediátrica e de Adultos, Centro Hospitalar e Universitário de Coimbra, 3000-075 Coimbra, Portugal;
| | - Maria Rosário Domingues
- CESAM—Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal;
- Mass Spectrometry Center, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Artur Paiva
- Flow Cytometry Unit, Department of Clinical Pathology, Hospitais da Universidade de Coimbra, Unidade Local de Saúde de Coimbra, 3000-076 Coimbra, Portugal;
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Center of Environmental Genetics of Oncobiology (CIMAGO), Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal; (R.M.); (M.C.); (A.M.A.); (R.C.O.); (J.G.T.); (M.F.B.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3000-061 Coimbra, Portugal
- Instituto Politécnico de Coimbra, ESTESC-Coimbra Health School, Ciências Biomédicas Laboratoriais, 3046-854 Coimbra, Portugal
| | - Paula Laranjeira
- Flow Cytometry Unit, Department of Clinical Pathology, Hospitais da Universidade de Coimbra, Unidade Local de Saúde de Coimbra, 3000-076 Coimbra, Portugal;
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Center of Environmental Genetics of Oncobiology (CIMAGO), Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal; (R.M.); (M.C.); (A.M.A.); (R.C.O.); (J.G.T.); (M.F.B.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3000-061 Coimbra, Portugal
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal
| |
Collapse
|