1
|
Chaudhary B, Arya P, Sharma V, Kumar P, Singla D, Grewal AS. Targeting anti-apoptotic mechanisms in tumour cells: Strategies for enhancing Cancer therapy. Bioorg Chem 2025; 159:108388. [PMID: 40107036 DOI: 10.1016/j.bioorg.2025.108388] [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: 01/23/2025] [Revised: 03/05/2025] [Accepted: 03/13/2025] [Indexed: 03/22/2025]
Abstract
Anti-cancer drug's cytotoxicity is determined by their ability to induce predetermined cell demise, commonly called apoptosis. The cancer-causing cells are able to evade cell death, which has been affiliated with both malignancy as well as resistance to cancer treatments. In order to avoid cell death, cancerous tumour cells often produce an abundance of anti-apoptotic proteins, becoming "dependent" on them. Consequently, protein inhibitors of cell death may prove to be beneficial as pharmacological targets for the future creation of cancer therapies. This article examines the molecular routes of apoptosis, its clinical manifestations, anti-cancer therapy options that target the intrinsic mechanism of apoptosis, proteins that prevent cell death, and members of the B-lymphoma-2 subset. In addition, novel approaches to cell death are highlighted, including how curcumin mitigates chemotherapy-induced apoptosis in healthy tissues and the various ways melatonin modifies apoptosis to improve cancer treatment efficacy, particularly through the TNF superfamily. Cancer treatment-induced increases in anti-apoptotic proteins lead to drug resistance; yet, ligands that trigger cell death by inhibiting these proteins are expected to improve chemotherapy's efficacy. The potential of frequency-modulated dietary phytochemicals as a cancer therapeutic pathway, including autophagy and apoptosis, is also explored. This approach may be more efficient than inhibition alone in overcoming drug resistance. Consequently, this method has the potential to allow for lower medication concentrations, reducing cytotoxicity and unwanted side effects.
Collapse
Affiliation(s)
- Benu Chaudhary
- Shri Ram College of Pharmacy, Ramba, Karnal, Haryana, India
| | - Preeti Arya
- Shri Ram College of Pharmacy, Ramba, Karnal, Haryana, India
| | - Vikas Sharma
- Guru Gobind Singh College of Pharmacy, Yamuna Nagar, Haryana, India
| | - Parveen Kumar
- NIMS Institute of Pharmacy, NIMS University, Jaipur, Rajasthan, India
| | - Deepak Singla
- Guru Gobind Singh College of Pharmacy, Yamuna Nagar, Haryana, India
| | | |
Collapse
|
2
|
Yen CC, Chen PCH, Chen SC, Wu WC, Yen CH, Lin YC, Wu PK, Chen CM, Wang JY, Chao TC, Yang MH, Fletcher JA. Ferroptosis as a therapeutic vulnerability in MDM2 inhibition in dedifferentiated liposarcoma. Oncol Lett 2025; 29:269. [PMID: 40247991 PMCID: PMC12005077 DOI: 10.3892/ol.2025.15015] [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: 10/24/2024] [Accepted: 03/07/2025] [Indexed: 04/19/2025] Open
Abstract
Ferroptosis is a form of necrotic cell death characterized by phospholipid oxidation. The cystine-glutamate antiporter (xCT), composed of solute carrier family 7 member 11 (SLC7A11) and SLC3A2, imports cystine for glutathione synthesis. Glutathione peroxidase 4 (GPX4) requires glutathione to counteract lipid peroxidation and prevent ferroptosis. Erastin, an xCT inhibitor, and Ras-selective lethal small molecule 3 (RSL3), a GPX4 inhibitor, suppress GPX4 function and induce ferroptosis. Tumor protein p53 (TP53) has a paradoxical role in ferroptosis regulation. Mouse double minute 2 homolog (MDM2), a negative regulator of TP53, is a key oncogene in well-differentiated liposarcoma (WDLPS) and dedifferentiated liposarcoma (DDLPS). Therefore, the present study explored the role of ferroptosis in DDLPS treatment response and resistance. Publicly available expression profiles of WDLPS, DDLPS and adipose tissue were analyzed, and the differential expression of ferroptosis-related genes regulated by the MDM2-TP53 pathway was identified in WDLPS and DDLPS. In vitro experiments were performed to assess the effects of erastin and RSL3 on the viability, lipid peroxidation and apoptosis of DDLPS cell lines. The results revealed that erastin and RSL3 induced lipid peroxidation and apoptosis, thereby exerting cytotoxic effects. In addition, nutlin-3, an MDM2 inhibitor, was demonstrated to increase lipid peroxidation and cytotoxicity when applied prior to erastin treatment. Notably, nutlin-3 also upregulated SLC3A2 expression in DDLPS cell lines, thereby enhancing cystine uptake. This increase in cystine uptake was suppressed by erastin. In addition, nutlin-3-induced SLC3A2 upregulation was abolished by TP53 knockdown. Nutlin-3 combined with erastin or RSL3 reduced absolute p-4EBP-1 levels in NDDLS-1 cells and p-p70S6 levels in both cell lines, with no significant impact on the p-4EBP-1/4EBP-1 and p-p70S6/p70S6 ratios. These results indicate that ferroptosis is a therapeutic vulnerability in the response to MDM2 inhibition in DDLPS. Furthermore, combining MDM2 inhibitors with ferroptosis-inducing agents may provide a potential therapeutic strategy for DDLPS and the role of mTOR in the pro-apoptotic effect of these combinations deserve further investigation.
Collapse
Affiliation(s)
- Chueh-Chuan Yen
- Department of Medical Research, Division of Clinical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- Department of Oncology, Division of Medical Oncology, Center for Immuno-oncology, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- Department of Orthopedics and Traumatology, Therapeutical and Research Center of Musculoskeletal Tumor, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
| | - Paul Chih-Hsueh Chen
- Department of Orthopedics and Traumatology, Therapeutical and Research Center of Musculoskeletal Tumor, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
| | - San-Chi Chen
- Department of Oncology, Division of Medical Oncology, Center for Immuno-oncology, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- Department of Orthopedics and Traumatology, Therapeutical and Research Center of Musculoskeletal Tumor, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
- Institute of Clinical Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
| | - Wen-Chi Wu
- Department of Oncology, Division of Medical Oncology, Center for Immuno-oncology, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- Department of Orthopedics and Traumatology, Therapeutical and Research Center of Musculoskeletal Tumor, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
- Institute of Clinical Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
| | - Chiao-Han Yen
- Department of Medical Research, Division of Clinical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- Department of Oncology, Division of Medical Oncology, Center for Immuno-oncology, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
| | - Yung-Chan Lin
- Department of Medical Research, Division of Clinical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- Department of Oncology, Division of Medical Oncology, Center for Immuno-oncology, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
| | - Po-Kuei Wu
- Department of Orthopedics and Traumatology, Therapeutical and Research Center of Musculoskeletal Tumor, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
- Department of Orthopedics and Traumatology, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
| | - Chao-Ming Chen
- Department of Orthopedics and Traumatology, Therapeutical and Research Center of Musculoskeletal Tumor, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
- Department of Orthopedics and Traumatology, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
| | - Jir-You Wang
- Department of Orthopedics and Traumatology, Therapeutical and Research Center of Musculoskeletal Tumor, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- Department of Orthopedics and Traumatology, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- Institute of Traditional Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
| | - Ta-Chung Chao
- Department of Oncology, Division of Medical Oncology, Center for Immuno-oncology, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- Department of Orthopedics and Traumatology, Therapeutical and Research Center of Musculoskeletal Tumor, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
| | - Muh-Hwa Yang
- Department of Oncology, Division of Medical Oncology, Center for Immuno-oncology, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
- Institute of Clinical Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
| | | |
Collapse
|
3
|
Shemorry A, Besten WD, Mulvihill MM, Essenburg CJ, Blaquiere N, Kleinheinz T, Villemure E, Peale F, Deshmukh G, Maddalo D, Levy E, Yu K, Steensma MR, Tovar EA, Wolfrum E, Nagapudi K, Blake RA, Forrest WF, Staben ST, Graveel CR, Fairbrother WJ, Wertz IE. Engineering ERα degraders with pleiotropic ubiquitin ligase ligands maximizes therapeutic efficacy by co-opting distinct effector ligases. Cell Chem Biol 2025; 32:694-709.e35. [PMID: 40378821 DOI: 10.1016/j.chembiol.2025.04.008] [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: 06/07/2024] [Revised: 12/18/2024] [Accepted: 04/15/2025] [Indexed: 05/19/2025]
Abstract
Proximity-inducing compounds that modulate target protein homeostasis represent an emerging therapeutic strategy. While the inherent complexity of these bifunctional compounds presents certain challenges, their unique composition offers opportunities to co-opt specific cellular effectors to enhance therapeutic impact. In this study, we systematically evaluate a series of bifunctional degrader compounds engineered with the estrogen receptor-alpha (ERα) inhibitor endoxifen linked to various bioactive ubiquitin ligase ligands. Notably, ERα degraders containing pan-IAP antagonist ligands significantly reduced the proliferation of ERα-dependent cells compared to clinical-stage ERα degraders. These pan-IAP antagonist-based ERα degraders leverage distinct effector ligases to achieve dual therapeutic effects: They utilize XIAP within tumor cells to promote ERα degradation and activate cIAP1/2 in both tumor and immune cells to induce TNFα, which drives tumor cell death. Our findings illustrate a broader concept that co-opting the discrete functions of selected cellular effectors, while simultaneously modulating therapeutic target protein homeostasis, are dual strategies that can significantly enhance the efficacy of induced proximity therapeutics.
Collapse
Affiliation(s)
- Anna Shemorry
- Discovery Oncology, Genentech, South San Francisco, CA 94080, USA; Early Discovery Biochemistry, Genentech, South San Francisco, CA 94080, USA
| | - Willem den Besten
- Discovery Oncology, Genentech, South San Francisco, CA 94080, USA; Early Discovery Biochemistry, Genentech, South San Francisco, CA 94080, USA
| | - Melinda M Mulvihill
- Biochemical and Cellular Pharmacology, Genentech, South San Francisco, CA 94080, USA
| | - Curt J Essenburg
- Department of Cell Biology, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Nicole Blaquiere
- Department of Discovery Chemistry, Genentech, South San Francisco, CA 94080, USA
| | - Tracy Kleinheinz
- Biochemical and Cellular Pharmacology, Genentech, South San Francisco, CA 94080, USA
| | - Elisia Villemure
- Department of Discovery Chemistry, Genentech, South San Francisco, CA 94080, USA
| | - Frank Peale
- Department of Research Pathology, Genentech, South San Francisco, CA, USA
| | | | - Danilo Maddalo
- Translational Oncology, Genentech, South San Francisco, CA 94080, USA
| | - Elizabeth Levy
- Department of Small Molecule Pharmaceutical Sciences, Genentech, South San Francisco, CA 94080, USA
| | - Kebing Yu
- Microchemistry Proteomics and Lipidomics, Genentech, South San Francisco, CA 94080, USA
| | - Matthew R Steensma
- Department of Cell Biology, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Elizabeth A Tovar
- Department of Cell Biology, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Emily Wolfrum
- Bioinformatics and Biostatistics Core, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Karthik Nagapudi
- Department of Small Molecule Pharmaceutical Sciences, Genentech, South San Francisco, CA 94080, USA
| | - Robert A Blake
- Biochemical and Cellular Pharmacology, Genentech, South San Francisco, CA 94080, USA
| | | | - Steven T Staben
- Department of Discovery Chemistry, Genentech, South San Francisco, CA 94080, USA
| | - Carrie R Graveel
- Department of Cell Biology, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Wayne J Fairbrother
- Early Discovery Biochemistry, Genentech, South San Francisco, CA 94080, USA.
| | - Ingrid E Wertz
- Discovery Oncology, Genentech, South San Francisco, CA 94080, USA; Early Discovery Biochemistry, Genentech, South San Francisco, CA 94080, USA.
| |
Collapse
|
4
|
Jeong S, Cha J, Ahmed W, Kim J, Kim M, Hong KT, Choi W, Choi S, Yoo TH, An H, An SC, Lee J, Choi J, Kim S, Lee J, Lee S, Choi J, Kim JM. Development of MDM2-Targeting PROTAC for Advancing Bone Regeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2415626. [PMID: 40125646 PMCID: PMC12097015 DOI: 10.1002/advs.202415626] [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] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 02/20/2025] [Indexed: 03/25/2025]
Abstract
Proteolysis-targeting chimeras (PROTACs) degrade target proteins through the ubiquitin-proteasome system. To date, PROTACs are primarily used to treat various diseases; however, they have not been applied in regenerative therapy. Herein, this work introduces MDM2-targeting PROTACs customized for application in bone regeneration. An MDM2-PROTAC library is constructed by combining Nutlin-3 and CRBN ligands with various linker designs. Through a multistep validation process, this work develops MDM2-PROTACs (CL144 and CL174) that presented potent degradation efficiency and a robust inductive effect on the biomineralization. Next, this work performs whole-transcriptome analysis to dissect the biological effects of the CL144, and reveals the upregulation of osteogenic marker genes. Furthermore, CL144 effectively induced bone regeneration in bone graft and ovariectomy (OVX) models after local and systemic administration, respectively. In the OVX model, the combination treatment with CL144 and alendronate induced a synergistic effect. Overall, this study demonstrates the promising role of MDM2-PROTAC in promoting bone regeneration, marking the first step toward expanding the application of the PROTAC technology.
Collapse
Affiliation(s)
- Sol Jeong
- Department of Oral Microbiology and ImmunologySchool of Dentistry and Dental Research InstituteSeoul National UniversitySeoul08826Republic of Korea
| | - Jae‐Kook Cha
- Department of PeriodontologyResearch Institute of Periodontal RegenerationCollege of DentistryYonsei UniversitySeoul03722Republic of Korea
- Department of Oral MedicineInfectionand ImmunityHarvard School of Dental MedicineBoston02115USA
| | - Wasim Ahmed
- Department of Molecular Science and TechnologyAjou UniversityGyeonggi‐do16499Republic of Korea
| | - Jaewan Kim
- Department of Molecular Science and TechnologyAjou UniversityGyeonggi‐do16499Republic of Korea
| | - Minsup Kim
- TARS ScientificSeoul01717Republic of Korea
| | - Kyung Tae Hong
- Department of PharmacologyKorea University College of MedicineKorea UniversitySeoul02841Republic of Korea
| | - Wonji Choi
- Department of Molecular Science and TechnologyAjou UniversityGyeonggi‐do16499Republic of Korea
| | - Sunjoo Choi
- Department of Molecular Science and TechnologyAjou UniversityGyeonggi‐do16499Republic of Korea
| | - Tae Hyeon Yoo
- Department of Molecular Science and TechnologyAjou UniversityGyeonggi‐do16499Republic of Korea
| | - Hyun‑Ju An
- Department of Orthopaedic SurgeryCHA Bundang Medical CenterCHA University School of MedicineGyeonggi‐do13488Republic of Korea
| | - Seung Chan An
- Department of Orthopaedic SurgeryCHA Bundang Medical CenterCHA University School of MedicineGyeonggi‐do13488Republic of Korea
| | - Jaemin Lee
- Department of Orthopaedic SurgeryCHA Bundang Medical CenterCHA University School of MedicineGyeonggi‐do13488Republic of Korea
| | - Jimin Choi
- Department of PeriodontologyResearch Institute of Periodontal RegenerationCollege of DentistryYonsei UniversitySeoul03722Republic of Korea
| | - Sun‐Young Kim
- Department of Conservative Dentistry and Dental Research InstituteSchool of DentistrySeoul National UniversitySeoul08826Republic of Korea
| | - Jun‐Seok Lee
- Department of PharmacologyKorea University College of MedicineKorea UniversitySeoul02841Republic of Korea
| | - Soonchul Lee
- Department of Orthopaedic SurgeryCHA Bundang Medical CenterCHA University School of MedicineGyeonggi‐do13488Republic of Korea
| | - Junwon Choi
- Department of Molecular Science and TechnologyAjou UniversityGyeonggi‐do16499Republic of Korea
- Advanced College of Bio‐convergence EngineeringAjou UniversityGyeonggi‐do16499Republic of Korea
| | - Jin Man Kim
- Department of Oral Microbiology and ImmunologySchool of Dentistry and Dental Research InstituteSeoul National UniversitySeoul08826Republic of Korea
- Dental Multiomics CenterSchool of Dentistry and Dental Research InstituteSeoul National UniversityGwanak‐ro 1, Gwanak‐guSeoul08826Republic of Korea
- Innovative Pharmaceutical Sciences ProgramCollege of Transdisciplinary InnovationsSeoul National UniversityGwanak‐ro 1, Gwanak‐guSeoul08826Republic of Korea
| |
Collapse
|
5
|
Hamada A, Kita Y, Sakatani T, Nakamura K, Takada H, Ikeuchi R, Koike S, Masuda N, Murakami K, Sano T, Goto T, Saito R, Teramoto Y, Fujimoto M, Hatano N, Kamada M, Ogawa O, Kobayashi T. PTEN loss drives p53 LOH and immune evasion in a novel urothelial organoid model harboring p53 missense mutations. Oncogene 2025; 44:1336-1349. [PMID: 39987272 PMCID: PMC12052601 DOI: 10.1038/s41388-025-03311-5] [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: 08/01/2024] [Revised: 01/15/2025] [Accepted: 02/12/2025] [Indexed: 02/24/2025]
Abstract
Despite missense mutation accounts for over 60% of p53 alterations while homozygous deletion (HOM) for only 5% or less in advanced bladder cancer cases, most of the previously reported mouse models are deficient of p53. Accordingly, few studies have addressed the mechanisms of missense mutation occurrence and its functional advantage over HOM in bladder cancer development. Organoids derived from Krt5-expressing mouse urothelium (K5-mUrorganoid) demonstrated the crucial role of Pten loss in driving loss of wild-type allele of Trp53 (Trp53R172H/LOH), which conferred tumorigenic ability to K5-mUrorganoid in athymic mice. These tumors recapitulated the histological and genetic characteristics of the human basal-squamous subtype bladder cancer. Both Trp53R172H/Δ; PtenΔ/Δ and Trp53Δ/Δ; PtenΔ/Δ K5-mUrorganoids formed tumors in athymic mice, whereas only Trp53R172H/Δ; PtenΔ/Δ K5-mUrorganoid formed tumors even when directly inoculated in immunocompetent syngeneic mice. The absence of wild-type Trp53 was associated with upregulation of proliferative signaling, and the presence of a mutant Trp53 allele was associated with immune-excluded microenvironment. This study highlights the functional significance of p53 mutant LOH in bladder carcinogenesis conferring several hallmarks of cancer such as sustaining proliferative signaling and avoiding immune destruction, thus provides a novel immunocompetent mouse model of urothelial carcinoma harboring p53 mutations as a novel tool for cancer immunology research.
Collapse
Affiliation(s)
- Akihiro Hamada
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yuki Kita
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Toru Sakatani
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kenji Nakamura
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hideaki Takada
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ryosuke Ikeuchi
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Shuhei Koike
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Norihiko Masuda
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Department of Urology, Rakuwakai Otowa Hospital, Kyoto, Japan
| | - Kaoru Murakami
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takeshi Sano
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Department of Urology and Andrology, Kansai Medical University, Osaka, Japan
| | - Takayuki Goto
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ryoichi Saito
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yuki Teramoto
- Department of Diagnostic Pathology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masakazu Fujimoto
- Department of Diagnostic Pathology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Narumi Hatano
- Department of Biomedical Data Intelligence, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Mayumi Kamada
- Department of Biomedical Data Intelligence, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Osamu Ogawa
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Department of Urology, Otsu Red Cross Hospital, Shiga, Japan
| | - Takashi Kobayashi
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| |
Collapse
|
6
|
Lu M, Ren Y, Feng S, Wang S, Xia W, Gu B, Shen Y, Yue A, Li N, Zhang Y, Zhong J. MDM2 inhibitor induces apoptosis in colon cancer cells through activation of the CHOP-DR5 pathway, independent of p53 phenotype. Front Pharmacol 2025; 16:1508421. [PMID: 40264676 PMCID: PMC12011796 DOI: 10.3389/fphar.2025.1508421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 03/19/2025] [Indexed: 04/24/2025] Open
Abstract
Introduction Murine double minute 2 (MDM2), a key negative regulator of p53, forms a feedback loop with p53 to drive tumor progression, including colorectal cancer. Nutlin-3a, an MDM2 inhibitor, induces apoptosis in wild-type p53 tumors, but its effects on p53-mutated cancers and potential p53-independent apoptotic mechanisms remain unclear. Methods We investigated Nutlin-3a's effects on colon cancer cells with varying p53 phenotypes. Endoplasmic reticulum (ER) stress-associated CHOP was detected and knocked down to explore mechanisms. In vitro and in vivo experiments assessed Nutlin-3a's synergy with 5-fluorouracil and TRAIL. Results Nutlin-3a activated caspase-8-dependent extrinsic apoptosis in colon cancer cells via DR5 upregulation, independent of p53 status. ER stress and CHOP activation mediated DR5 induction, driven by calcium release. Combined Nutlin-3a treatment enhanced sensitivity to 5-fluorouracil and TRAIL in vitro and in vivo through caspase-8 pathway activation. Discussion These findings reveal a novel p53-independent apoptotic mechanism of Nutlin-3a involving ER stress and death receptor signaling. This pathway highlights Nutlin-3a's potential as an adjuvant therapy for colon cancer, even in p53-mutated tumors, by enhancing chemotherapeutic efficacy through extrinsic apoptosis.
Collapse
Affiliation(s)
- Manman Lu
- Department of Oncology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Yingli Ren
- College of Medicine, Henan Polytechnic University, Jiaozuo, China
| | - Sijia Feng
- Henan Province Engineering Technology Research Center of Tumor Diagnostic Biomarkers and RNA Interference Drugs, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Shenggen Wang
- Department of Oncology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Weiyue Xia
- Department of Oncology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Baoru Gu
- Department of Oncology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Yuhou Shen
- Department of Abdominal Surgical Oncology Ward 2, Xinxiang Central Hospital, Xinxiang, China
- Department of Abdominal Surgical Oncology Ward 2, The Fourth Clinical College of Xinxiang Medical University, Xinxiang, China
| | - Aimin Yue
- Department of Abdominal Surgical Oncology Ward 2, Xinxiang Central Hospital, Xinxiang, China
- Department of Abdominal Surgical Oncology Ward 2, The Fourth Clinical College of Xinxiang Medical University, Xinxiang, China
| | - Na Li
- Department of Oncology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Yongxi Zhang
- Department of Oncology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Province Engineering Technology Research Center of Tumor Diagnostic Biomarkers and RNA Interference Drugs, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Jiateng Zhong
- Department of Oncology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Province Engineering Technology Research Center of Tumor Diagnostic Biomarkers and RNA Interference Drugs, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
7
|
Bharti J, Gogu P, Pandey SK, Verma A, Yadav JP, Singh AK, Kumar P, Dwivedi AR, Pathak P. BRAF V600E in cancer: Exploring structural complexities, mutation profiles, and pathway dysregulation. Exp Cell Res 2025; 446:114440. [PMID: 39961465 DOI: 10.1016/j.yexcr.2025.114440] [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: 08/08/2024] [Revised: 11/14/2024] [Accepted: 02/10/2025] [Indexed: 02/23/2025]
Abstract
BRAF, a fundamental component of cellular signaling pathways regulating growth and survival, is frequently mutated in cancer development. Among entire BRAF mutations, the V600E substitution stands out as a dominant alteration in various malignancies, including melanoma, colorectal cancer, and thyroid cancer. Understanding the structural differences between wild-type BRAF and BRAFV600E is crucial for elucidating the molecular mechanisms underpinnings tumorigenesis and identifying dysregulation associated with the same. V600E mutation results in a constitutively active kinase domain, leading to dysregulated downstream signaling independent of extracellular stimuli. This sustained activation promotes cell proliferation, survival, angiogenesis, and hallmark features of the cancer cells. The study describes three distinct classes of BRAF mutations where Class 1 mutations predominantly involve point mutations within the BRAF gene, while Class 2 encompasses in-frame insertions and deletions, and Class 3 comprises gene fusions with large-scale chromosomal rearrangements. Further, we have discussed dysregulated pathways associated with mutation of BRAFV600E, which includes MAPK/ERK, PI3K/AKT/mTOR, TP53, DNA damage response, and WNT/β-Catenin from schematic representation. In the current review, we have shown how these dysregulated pathways play pivotal roles in tumorigenesis, tumor progression in BRAF-mutant cancers and highlighted the critical role of BRAF dysregulation in cancer development followed by its therapeutic implications of targeting dysregulated pathways in BRAF-driven malignancies.
Collapse
Affiliation(s)
- Jayhind Bharti
- Drug Discovery Laboratory, School of Pharmacy, GITAM (Deemed to be) University, Hyderabad Campus, 502329, India
| | - Priyadharshini Gogu
- Drug Discovery Laboratory, School of Pharmacy, GITAM (Deemed to be) University, Hyderabad Campus, 502329, India
| | | | - Amita Verma
- Bioorganic and Medicinal Chemistry Research Laboratory, Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj, 211007, India; School of Pharmacy, Graphic Era Hill University, Dehradun-248002, India
| | - Jagat Pal Yadav
- Bioorganic and Medicinal Chemistry Research Laboratory, Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj, 211007, India
| | - Ankit Kumar Singh
- Bioorganic and Medicinal Chemistry Research Laboratory, Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj, 211007, India; Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Ghudda, Bathinda, India
| | - Pradeep Kumar
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Ghudda, Bathinda, India
| | - Ashish Ranjan Dwivedi
- Drug Discovery Laboratory, School of Pharmacy, GITAM (Deemed to be) University, Hyderabad Campus, 502329, India.
| | - Prateek Pathak
- Drug Discovery Laboratory, School of Pharmacy, GITAM (Deemed to be) University, Hyderabad Campus, 502329, India.
| |
Collapse
|
8
|
Paronyan A, Muradyan N, Sargsyan A, Arakelov V, Arakelov G, Nazaryan K. In Silico Study of Pyrin and 14-3-3 Protein Modulation During FMF and PAAND. ACS OMEGA 2025; 10:3462-3473. [PMID: 39926484 PMCID: PMC11799992 DOI: 10.1021/acsomega.4c07386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 11/25/2024] [Accepted: 12/24/2024] [Indexed: 02/11/2025]
Abstract
Familial Mediterranean fever (FMF) is a genetically determined disease transmitted through autosomal recessive inheritance. Recently, a rare but similar disease to FMF, pyrin-associated autoinflammation with neutrophilic dermatosis (PAAND), has been discovered. PAAND is inherited dominantly and is characterized by neutrophilic dermatosis and recurrent fevers. The cause of the disease is point mutations in the MEFV gene. The pyrin protein is a product of this gene and is one of the main factors in the disease's progression. This paper examines the interaction between pyrin and the 14-3-3 protein and screens for modulators affecting their interaction. Regulating this interaction is crucial for understanding the mechanism of FMF development, specifically the disruption of this complexation, which leads to inflammatory responses. The pyrin-14-3-3 interaction is essential for designing potential drugs since weakening this interaction can result in inflammation. This research of in silico experiments identified low molecular weight chemical compounds that have a modulating effect on the tertiary structures of mutant variations of pyrin and 14-3-3 proteins. Studies have identified modulator molecules that interact with the FMF-associated mutant structure of pyrin (M694I) and 14-3-3τ. The chemical compounds that result from this process can be used as modulators and as a potential new basis for the development of therapeutic drugs.
Collapse
Affiliation(s)
- Adrine Paronyan
- Institute
of Molecular Biology of the National Academy of Sciences of the Republic
of Armenia (NAS RA), Yerevan 0014, Armenia
- Russian-Armenian
(Slavonic) University, Yerevan 0051, Armenia
| | - Nelli Muradyan
- Institute
of Molecular Biology of the National Academy of Sciences of the Republic
of Armenia (NAS RA), Yerevan 0014, Armenia
- International
Scientific-Educational Center of NAS RA, Yerevan 0019, Armenia
| | - Arsen Sargsyan
- Institute
of Molecular Biology of the National Academy of Sciences of the Republic
of Armenia (NAS RA), Yerevan 0014, Armenia
- Russian-Armenian
(Slavonic) University, Yerevan 0051, Armenia
| | - Vahram Arakelov
- Institute
of Molecular Biology of the National Academy of Sciences of the Republic
of Armenia (NAS RA), Yerevan 0014, Armenia
| | - Grigor Arakelov
- Institute
of Molecular Biology of the National Academy of Sciences of the Republic
of Armenia (NAS RA), Yerevan 0014, Armenia
- Russian-Armenian
(Slavonic) University, Yerevan 0051, Armenia
- International
Scientific-Educational Center of NAS RA, Yerevan 0019, Armenia
| | - Karen Nazaryan
- Institute
of Molecular Biology of the National Academy of Sciences of the Republic
of Armenia (NAS RA), Yerevan 0014, Armenia
- Russian-Armenian
(Slavonic) University, Yerevan 0051, Armenia
| |
Collapse
|
9
|
Andrysik Z, Espinosa JM. Harnessing p53 for targeted cancer therapy: new advances and future directions. Transcription 2025; 16:3-46. [PMID: 40031988 PMCID: PMC11970777 DOI: 10.1080/21541264.2025.2452711] [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: 11/28/2024] [Revised: 01/07/2025] [Accepted: 01/08/2025] [Indexed: 03/05/2025] Open
Abstract
The transcription factor p53 is the most frequently impaired tumor suppressor in human cancers. In response to various stress stimuli, p53 activates transcription of genes that mediate its tumor-suppressive functions. Distinctive characteristics of p53 outlined here enable a well-defined program of genes involved in cell cycle arrest, apoptosis, senescence, differentiation, metabolism, autophagy, DNA repair, anti-viral response, and anti-metastatic functions, as well as facilitating autoregulation within the p53 network. This versatile, anti-cancer network governed chiefly by a single protein represents an immense opportunity for targeted cancer treatment, since about half of human tumors retain unmutated p53. During the last two decades, numerous compounds have been developed to block the interaction of p53 with the main negative regulator MDM2. However, small molecule inhibitors of MDM2 only induce a therapeutically desirable apoptotic response in a limited number of cancer types. Moreover, clinical trials of the MDM2 inhibitors as monotherapies have not met expectations and have revealed hematological toxicity as a characteristic adverse effect across this drug class. Currently, combination treatments are the leading strategy for enhancing efficacy and reducing adverse effects of MDM2 inhibitors. This review summarizes efforts to identify and test therapeutics that work synergistically with MDM2 inhibitors. Two main types of drugs have emerged among compounds used in the following combination treatments: first, modulators of the p53-regulated transcriptome (including chromatin modifiers), translatome, and proteome, and second, drugs targeting the downstream pathways such as apoptosis, cell cycle arrest, DNA repair, metabolic stress response, immune response, ferroptosis, and growth factor signaling. Here, we review the current literature in this field, while also highlighting overarching principles that could guide target selection in future combination treatments.
Collapse
Affiliation(s)
- Zdenek Andrysik
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Joaquin M. Espinosa
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
10
|
Xie F, Niu Y, Chen X, Kong X, Yan G, Zhuang A, Li X, Lian L, Qin D, Zhang Q, Zhang R, Yang K, Xia X, Chen K, Xiao M, Yang C, Wu T, Shen Y, Yu C, Luo C, Lin SH, Li W. Ursodeoxycholic acid inhibits the uptake of cystine through SLC7A11 and impairs de novo synthesis of glutathione. J Pharm Anal 2025; 15:101068. [PMID: 39902457 PMCID: PMC11788867 DOI: 10.1016/j.jpha.2024.101068] [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: 03/19/2024] [Revised: 07/18/2024] [Accepted: 08/03/2024] [Indexed: 02/03/2025] Open
Abstract
Ursodeoxycholic acid (UDCA) is a naturally occurring, low-toxicity, and hydrophilic bile acid (BA) in the human body that is converted by intestinal flora using primary BA. Solute carrier family 7 member 11 (SLC7A11) functions to uptake extracellular cystine in exchange for glutamate, and is highly expressed in a variety of human cancers. Retroperitoneal liposarcoma (RLPS) refers to liposarcoma originating from the retroperitoneal area. Lipidomics analysis revealed that UDCA was one of the most significantly downregulated metabolites in sera of RLPS patients compared with healthy subjects. The augmentation of UDCA concentration (≥25 μg/mL) demonstrated a suppressive effect on the proliferation of liposarcoma cells. [15N2]-cystine and [13C5]-glutamine isotope tracing revealed that UDCA impairs cystine uptake and glutathione (GSH) synthesis. Mechanistically, UDCA binds to the cystine transporter SLC7A11 to inhibit cystine uptake and impair GSH de novo synthesis, leading to reactive oxygen species (ROS) accumulation and mitochondrial oxidative damage. Furthermore, UDCA can promote the anti-cancer effects of ferroptosis inducers (Erastin, RSL3), the murine double minute 2 (MDM2) inhibitors (Nutlin 3a, RG7112), cyclin dependent kinase 4 (CDK4) inhibitor (Abemaciclib), and glutaminase inhibitor (CB839). Together, UDCA functions as a cystine exchange factor that binds to SLC7A11 for antitumor activity, and SLC7A11 is not only a new transporter for BA but also a clinically applicable target for UDCA. More importantly, in combination with other antitumor chemotherapy or physiotherapy treatments, UDCA may provide effective and promising treatment strategies for RLPS or other types of tumors in a ROS-dependent manner.
Collapse
Affiliation(s)
- Fu'an Xie
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Yujia Niu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Xiaobing Chen
- Department of Retroperitoneal Tumor Surgery, Peking University International Hospital, Beijing, 102206, China
| | - Xu Kong
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Guangting Yan
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Aobo Zhuang
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Xi Li
- School of Public Health, Harvard University, Boston, MA, 02115, USA
| | - Lanlan Lian
- Department of Laboratory Medicine, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
| | - Dongmei Qin
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Quan Zhang
- National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Ruyi Zhang
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Kunrong Yang
- Laboratory of Biochemistry and Molecular Biology Research, Department of Clinical Laboratory, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Xiaogang Xia
- Department of Hepatobiliary Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Kun Chen
- Department of Hepatobiliary Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Mengmeng Xiao
- Department of General Surgery, Peking University People's Hospital, Beijing, 100032, China
| | - Chunkang Yang
- Department of Gastrointestinal Surgical Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Ting Wu
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
- Department of Retroperitoneal Tumor Surgery, Peking University International Hospital, Beijing, 102206, China
| | - Ye Shen
- Department of Management, Jiang Xia Blood Technology Co., Ltd., Shanghai, 200000, China
| | - Chundong Yu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Chenghua Luo
- Department of General Surgery, Peking University People's Hospital, Beijing, 100032, China
| | - Shu-Hai Lin
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Wengang Li
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
- Department of Hepatobiliary Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| |
Collapse
|
11
|
Han A, Liu J, Du P, Li W, Quan H, Lin Z, Chen L. Taraxasterol regulates p53 transcriptional activity to inhibit pancreatic cancer by inducing MDM2 ubiquitination degradation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156298. [PMID: 39671783 DOI: 10.1016/j.phymed.2024.156298] [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: 08/22/2024] [Revised: 11/11/2024] [Accepted: 11/25/2024] [Indexed: 12/15/2024]
Abstract
BACKGROUND Pancreatic cancer (PC) is a malignant tumor with complex development mechanisms and a poor prognosis. Taraxasterol (TAX), a pentacyclic triterpenoid plant sterol derived from Taraxacum mongolicum, has multiple biological activities including an anti-tumor effect. However, the mechanism by which TAX exerts its anticancer effects in PC remains unclear. PURPOSE This study aimed to elucidate the molecular mechanism by which TAX suppresses the proliferation of PC. METHODS The intersection of TAX and PC targets was obtained through network pharmacology. RNA-seq was used to identify TAX-induced differentially expressed genes in PC. Molecular docking, CETSA, western blot analysis, and qRT-PCR were performed to confirm the effectiveness of targets. The influence of TAX on PC was assessed by analyzing proliferation, apoptosis, and the cell cycle via MTT assay, colony formation assay, and flow cytometry, respectively. Co-IP assay and immunofluorescence assay were used to evaluate the effect of TAX on targeted genes. A nude mouse xenograft model was constructed to determine the inhibitory effects of TAX on PC in vivo. RESULTS TAX suppressed PC cell proliferation by promoting apoptosis and inducing cell cycle arrest in vitro and in vivo. Mechanistically, TAX interacted with MDM2, a critical regulator of proliferation, and decreased its stability by inducing ubiquitin-mediated degradation, which facilitates the nuclear translocation of p53 and downregulation of CXCL5 transcription, ultimately suppressing PC cell proliferation. CONCLUSION MDM2/p53/CXCL5 is the key pathway of TAX inhibiting the proliferation of PC cells.
Collapse
Affiliation(s)
- Anna Han
- Central Laboratory, Yanbian University Hospital, Yanji 133000, PR China; Key Laboratory Pathobiology, State Ethnic Affairs Commission, Yanbian University, Yanji 133000, PR China
| | - Jiajing Liu
- Key Laboratory Pathobiology, State Ethnic Affairs Commission, Yanbian University, Yanji 133000, PR China
| | - Pan Du
- Key Laboratory Pathobiology, State Ethnic Affairs Commission, Yanbian University, Yanji 133000, PR China
| | - Wenxuan Li
- Key Laboratory Pathobiology, State Ethnic Affairs Commission, Yanbian University, Yanji 133000, PR China
| | - Haiyan Quan
- Central Laboratory, Yanbian University Hospital, Yanji 133000, PR China
| | - Zhenhua Lin
- Central Laboratory, Yanbian University Hospital, Yanji 133000, PR China
| | - Liyan Chen
- Key Laboratory Pathobiology, State Ethnic Affairs Commission, Yanbian University, Yanji 133000, PR China.
| |
Collapse
|
12
|
Tomas F, Roux P, Gire V. Interaction of p53 with the Δ133p53α and Δ160p53α isoforms regulates p53 conformation and transcriptional activity. Cell Death Dis 2024; 15:845. [PMID: 39562560 PMCID: PMC11576908 DOI: 10.1038/s41419-024-07213-4] [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: 04/12/2024] [Revised: 10/29/2024] [Accepted: 10/31/2024] [Indexed: 11/21/2024]
Abstract
The TP53 gene encodes p53, a transcription factor involved in tumor suppression. However, TP53 also encodes other protein isoforms, some of which can disrupt the tumor suppressor functions of p53 even in the absence of TP53 mutations. In particular, elevated levels of the Δ133TP53 mRNA are detected in many cancer types and can be associated with poorer disease-free survival. We investigated the mechanisms of action of the two proteins translated from the Δ133TP53 mRNA: the Δ133p53α and Δ160p53α isoforms, both of which retain the oligomerization domain of p53. We discovered that the Δ133p53α and Δ160p53α isoforms adopt an altered conformation compared to full-length p53, exposing the PAb240 epitope (RHSVVV), which is inaccessible to the PAb240 antibody in the functional conformation of p53 (reactive to PAb1620). The Δ133p53α and/or Δ160p53α isoforms form hetero-oligomers with p53, regulating the stability, the conformation and the transcriptional activity of the p53 hetero-oligomers. Under basal conditions, Δ133p53α and Δ160p53α, in complex with p53, prevent proteasome-dependent degradation leading to the accumulation of PAb240 reactive Δ133p53α/Δ160p53α/p53 hetero-oligomers without increasing p53 transcriptional activity. Conversely, depletion of endogenous Δ133p53α isoforms in human fibroblasts is sufficient to restore p53 transcriptional activity, towards p53-target genes involved in cell cycle arrest. In the DNA damage response (DDR), PAb240 reactive Δ133p53α/Δ160p53α/p53 hetero-oligomers are highly phosphorylated at Ser15 compared to PAb1620-reactive p53 complexes devoid of Δ133p53α and Δ160p53α. This suggests that PAb240-reactive p53 hetero-oligomers integrate DNA damage signals. Δ133p53α accumulation is a late event in the DDR that depends on p53, but not on its transcriptional activation. The formation of Δ133p53α and p53 complexes increases at later DDR stages. We propose that Δ133p53α isoforms regulate p53 conformation as part of the normal p53 biology, modulating p53 activity and thereby adapting the cellular response to the cell signals.
Collapse
Affiliation(s)
- Fanny Tomas
- CRBM, University of Montpellier, CNRS, Montpellier, France
| | - Pierre Roux
- CRBM, University of Montpellier, CNRS, Montpellier, France
| | - Véronique Gire
- CRBM, University of Montpellier, CNRS, Montpellier, France.
| |
Collapse
|
13
|
Ahmadi SE, Rahimian E, Rahimi S, Zarandi B, Bahraini M, Soleymani M, Safdari SM, Shabannezhad A, Jaafari N, Safa M. From regulation to deregulation of p53 in hematologic malignancies: implications for diagnosis, prognosis and therapy. Biomark Res 2024; 12:137. [PMID: 39538363 PMCID: PMC11565275 DOI: 10.1186/s40364-024-00676-9] [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: 07/16/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
The p53 protein, encoded by the TP53 gene, serves as a critical tumor suppressor, playing a vital role in maintaining genomic stability and regulating cellular responses to stress. Dysregulation of p53 is frequently observed in hematological malignancies, significantly impacting disease progression and patient outcomes. This review aims to examine the regulatory mechanisms of p53, the implications of TP53 mutations in various hematological cancers, and emerging therapeutic strategies targeting p53. We conducted a comprehensive literature review to synthesize recent findings related to p53's multifaceted role in hematologic cancers, focusing on its regulatory pathways and therapeutic potential. TP53 mutations in hematological malignancies often lead to treatment resistance and poor prognosis. Current therapeutic strategies, including p53 reactivation and gene therapy, show promise in improving treatment outcomes. Understanding the intricacies of p53 regulation and the consequences of its mutations is essential for developing effective diagnostic and therapeutic strategies in hematological malignancies, ultimately enhancing patient care and survival.
Collapse
Affiliation(s)
- Seyed Esmaeil Ahmadi
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Elahe Rahimian
- Department of Medical Translational Oncology, National Center for Tumor Diseases (NCT) Dresden, Dresden, Germany
| | - Samira Rahimi
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Bahman Zarandi
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mehran Bahraini
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maral Soleymani
- Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Seyed Mehrab Safdari
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ashkan Shabannezhad
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Niloofar Jaafari
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Majid Safa
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
14
|
Nian Z, Dou Y, Shen Y, Liu J, Du X, Jiang Y, Zhou Y, Fu B, Sun R, Zheng X, Tian Z, Wei H. Interleukin-34-orchestrated tumor-associated macrophage reprogramming is required for tumor immune escape driven by p53 inactivation. Immunity 2024; 57:2344-2361.e7. [PMID: 39321806 DOI: 10.1016/j.immuni.2024.08.015] [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: 09/25/2023] [Revised: 03/29/2024] [Accepted: 08/22/2024] [Indexed: 09/27/2024]
Abstract
As the most frequent genetic alteration in cancer, more than half of human cancers have p53 mutations that cause transcriptional inactivation. However, how p53 modulates the immune landscape to create a niche for immune escape remains elusive. We found that cancer stem cells (CSCs) established an interleukin-34 (IL-34)-orchestrated niche to promote tumorigenesis in p53-inactivated liver cancer. Mechanistically, we discovered that Il34 is a gene transcriptionally repressed by p53, and p53 loss resulted in IL-34 secretion by CSCs. IL-34 induced CD36-mediated elevations in fatty acid oxidative metabolism to drive M2-like polarization of foam-like tumor-associated macrophages (TAMs). These IL-34-orchestrated TAMs suppressed CD8+ T cell-mediated antitumor immunity to promote immune escape. Blockade of the IL-34-CD36 axis elicited antitumor immunity and synergized with anti-PD-1 immunotherapy, leading to a complete response. Our findings reveal the underlying mechanism of p53 modulation of the tumor immune microenvironment and provide a potential target for immunotherapy of cancer with p53 inactivation.
Collapse
Affiliation(s)
- Zhigang Nian
- Department of Geriatrics, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230036, China; Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Institue of Immunology, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Yingchao Dou
- Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Institue of Immunology, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Yiqing Shen
- Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Institue of Immunology, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Jintang Liu
- Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Institue of Immunology, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Xianghui Du
- Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Institue of Immunology, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Yong Jiang
- Department of Anesthesiology, The first affiliated hospital of Anhui Medical University, Hefei, Anhui 230027, China
| | - Yonggang Zhou
- Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Institue of Immunology, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Binqing Fu
- Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Institue of Immunology, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Rui Sun
- Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Institue of Immunology, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Xiaohu Zheng
- Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Institue of Immunology, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Zhigang Tian
- Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Institue of Immunology, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Haiming Wei
- Department of Geriatrics, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230036, China; Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Institue of Immunology, University of Science and Technology of China, Hefei, Anhui 230027, China.
| |
Collapse
|
15
|
Gautam P, Ciuta I, Teif VB, Sinha SK. Predicting p53-dependent cell transitions from thermodynamic models. J Chem Phys 2024; 161:135101. [PMID: 39356070 DOI: 10.1063/5.0225166] [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: 06/23/2024] [Accepted: 09/18/2024] [Indexed: 10/03/2024] Open
Abstract
A cell's fate involves transitions among its various states, each defined by a distinct gene expression profile governed by the topology of gene regulatory networks, which are affected by 3D genome organization. Here, we develop thermodynamic models to determine the fate of a malignant cell as governed by the tumor suppressor p53 signaling network, taking into account long-range chromatin interactions in the mean-field approximation. The tumor suppressor p53 responds to stress by selectively triggering one of the potential transcription programs that influence many layers of cell signaling. These range from p53 phosphorylation to modulation of its DNA binding affinity, phase separation phenomena, and internal connectivity among cell fate genes. We use the minimum free energy of the system as a fundamental property of biological networks that influences the connection between the gene network topology and the state of the cell. We constructed models based on network topology and equilibrium thermodynamics. Our modeling shows that the binding of phosphorylated p53 to promoters of target genes can have properties of a first order phase transition. We apply our model to cancer cell lines ranging from breast cancer (MCF-7), colon cancer (HCT116), and leukemia (K562), with each one characterized by a specific network topology that determines the cell fate. Our results clarify the biological relevance of these mechanisms and suggest that they represent flexible network designs for switching between developmental decisions.
Collapse
Affiliation(s)
- Pankaj Gautam
- Theoretical and Computational Biophysical Chemistry Group, Department of Chemistry, Indian Institute of Technology Ropar, Rupnagar, Punjab 140001, India
| | - Isabella Ciuta
- School of Life Sciences, University of Essex, Wivenhoe Park, Colchester CO4 3SQ, United Kingdom
| | - Vladimir B Teif
- School of Life Sciences, University of Essex, Wivenhoe Park, Colchester CO4 3SQ, United Kingdom
| | - Sudipta Kumar Sinha
- Theoretical and Computational Biophysical Chemistry Group, Department of Chemistry, Indian Institute of Technology Ropar, Rupnagar, Punjab 140001, India
| |
Collapse
|
16
|
Tian X, El-Deiry WS. Integrated stress response (ISR) activation and apoptosis through HRI kinase by PG3 and other p53 pathway-restoring cancer therapeutics. Oncotarget 2024; 15:614-633. [PMID: 39288289 PMCID: PMC11407758 DOI: 10.18632/oncotarget.28637] [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: 01/14/2024] [Accepted: 08/01/2024] [Indexed: 09/19/2024] Open
Abstract
Restoration of the p53 pathway has been a long-term goal in the field of cancer research to treat tumors with mutated p53 and aggressive clinical behavior. p53 pathway restoration in p53-deficient cancers can be achieved by small molecules via p53-dependent or p53-independent processes. Hereafter p53-independent restoration of p53-pathway-signaling in p53-deficient/mutated tumors is referred to as 'restoration of the p53 pathway'. We compare activation of p53 target genes by novel compounds PG3 and PG3-Oc, that activate p53-target genes in a p53-independent manner, and four mutant p53-activating compounds while Nutlin-3a is used as negative control. PG3 and PG3-Oc upregulate p21, PUMA, and DR5 in five cancer cell lines with various p53 mutational statuses through ATF4 (Activating Transcriptional Factor 4) and integrated stress response (ISR) independent of p53. Mutant p53-targeting compounds induce expression of the 3 major downstream p53 target genes and ATF4 in a highly variable and cell-type-dependent manner. PG3 treatment activates ATF4 through ISR via kinase HRI (Heme-Regulated Inhibitor). ATF4 mediates upregulation of PUMA, p21, and NAG-1/GDF15 (Nonsteroidal anti-inflammatory drug-activated gene 1). We note that PUMA mediates apoptosis through activation of caspase-8 in HT29 cells and potentially caspase-10 in SW480 cells. We provide a novel mechanism engaged by PG3 to induce cell death via the HRI/ATF4/PUMA axis. Our results provide unique insights into the mechanism of action of PG3 as a novel cancer therapeutic targeting p53 pathway-like tumor suppression.
Collapse
Affiliation(s)
- Xiaobing Tian
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, RI 02912, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02912, USA
| | - Wafik S. El-Deiry
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, RI 02912, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02912, USA
- Department of Medicine, Hematology/Oncology Division, Lifespan Health System and Brown University, RI 02906, USA
| |
Collapse
|
17
|
Kim HS, Grimes SM, Chen T, Sathe A, Lau BT, Hwang GH, Bae S, Ji HP. Direct measurement of engineered cancer mutations and their transcriptional phenotypes in single cells. Nat Biotechnol 2024; 42:1254-1262. [PMID: 37697151 PMCID: PMC11324510 DOI: 10.1038/s41587-023-01949-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 08/15/2023] [Indexed: 09/13/2023]
Abstract
Genome sequencing studies have identified numerous cancer mutations across a wide spectrum of tumor types, but determining the phenotypic consequence of these mutations remains a challenge. Here, we developed a high-throughput, multiplexed single-cell technology called TISCC-seq to engineer predesignated mutations in cells using CRISPR base editors, directly delineate their genotype among individual cells and determine each mutation's transcriptional phenotype. Long-read sequencing of the target gene's transcript identifies the engineered mutations, and the transcriptome profile from the same set of cells is simultaneously analyzed by short-read sequencing. Through integration, we determine the mutations' genotype and expression phenotype at single-cell resolution. Using cell lines, we engineer and evaluate the impact of >100 TP53 mutations on gene expression. Based on the single-cell gene expression, we classify the mutations as having a functionally significant phenotype.
Collapse
Affiliation(s)
- Heon Seok Kim
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, Republic of Korea
- Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul, Republic of Korea
| | - Susan M Grimes
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Tianqi Chen
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Anuja Sathe
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Billy T Lau
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Gue-Ho Hwang
- Medical Research Center of Genomic Medicine Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Sangsu Bae
- Medical Research Center of Genomic Medicine Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hanlee P Ji
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Electrical Engineering, Stanford University, Stanford, CA, USA.
| |
Collapse
|
18
|
Wang W, Albadari N, Du Y, Fowler JF, Sang HT, Xian W, McKeon F, Li W, Zhou J, Zhang R. MDM2 Inhibitors for Cancer Therapy: The Past, Present, and Future. Pharmacol Rev 2024; 76:414-453. [PMID: 38697854 PMCID: PMC11068841 DOI: 10.1124/pharmrev.123.001026] [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: 08/22/2023] [Revised: 11/28/2023] [Accepted: 01/16/2024] [Indexed: 05/05/2024] Open
Abstract
Since its discovery over 35 years ago, MDM2 has emerged as an attractive target for the development of cancer therapy. MDM2's activities extend from carcinogenesis to immunity to the response to various cancer therapies. Since the report of the first MDM2 inhibitor more than 30 years ago, various approaches to inhibit MDM2 have been attempted, with hundreds of small-molecule inhibitors evaluated in preclinical studies and numerous molecules tested in clinical trials. Although many MDM2 inhibitors and degraders have been evaluated in clinical trials, there is currently no Food and Drug Administration (FDA)-approved MDM2 inhibitor on the market. Nevertheless, there are several current clinical trials of promising agents that may overcome the past failures, including agents granted FDA orphan drug or fast-track status. We herein summarize the research efforts to discover and develop MDM2 inhibitors, focusing on those that induce MDM2 degradation and exert anticancer activity, regardless of the p53 status of the cancer. We also describe how preclinical and clinical investigations have moved toward combining MDM2 inhibitors with other agents, including immune checkpoint inhibitors. Finally, we discuss the current challenges and future directions to accelerate the clinical application of MDM2 inhibitors. In conclusion, targeting MDM2 remains a promising treatment approach, and targeting MDM2 for protein degradation represents a novel strategy to downregulate MDM2 without the side effects of the existing agents blocking p53-MDM2 binding. Additional preclinical and clinical investigations are needed to finally realize the full potential of MDM2 inhibition in treating cancer and other chronic diseases where MDM2 has been implicated. SIGNIFICANCE STATEMENT: Overexpression/amplification of the MDM2 oncogene has been detected in various human cancers and is associated with disease progression, treatment resistance, and poor patient outcomes. This article reviews the previous, current, and emerging MDM2-targeted therapies and summarizes the preclinical and clinical studies combining MDM2 inhibitors with chemotherapy and immunotherapy regimens. The findings of these contemporary studies may lead to safer and more effective treatments for patients with cancers overexpressing MDM2.
Collapse
Affiliation(s)
- Wei Wang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy (W.W., Y.D., J.F.F., H.T.S., R.Z.), Drug Discovery Institute (W.W., R.Z.), Stem Cell Center, Department of Biology and Biochemistry (W.X., F.M.), University of Houston, Houston, Texas; College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee (N.A., W.L.); and Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas (J.Z.)
| | - Najah Albadari
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy (W.W., Y.D., J.F.F., H.T.S., R.Z.), Drug Discovery Institute (W.W., R.Z.), Stem Cell Center, Department of Biology and Biochemistry (W.X., F.M.), University of Houston, Houston, Texas; College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee (N.A., W.L.); and Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas (J.Z.)
| | - Yi Du
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy (W.W., Y.D., J.F.F., H.T.S., R.Z.), Drug Discovery Institute (W.W., R.Z.), Stem Cell Center, Department of Biology and Biochemistry (W.X., F.M.), University of Houston, Houston, Texas; College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee (N.A., W.L.); and Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas (J.Z.)
| | - Josef F Fowler
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy (W.W., Y.D., J.F.F., H.T.S., R.Z.), Drug Discovery Institute (W.W., R.Z.), Stem Cell Center, Department of Biology and Biochemistry (W.X., F.M.), University of Houston, Houston, Texas; College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee (N.A., W.L.); and Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas (J.Z.)
| | - Hannah T Sang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy (W.W., Y.D., J.F.F., H.T.S., R.Z.), Drug Discovery Institute (W.W., R.Z.), Stem Cell Center, Department of Biology and Biochemistry (W.X., F.M.), University of Houston, Houston, Texas; College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee (N.A., W.L.); and Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas (J.Z.)
| | - Wa Xian
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy (W.W., Y.D., J.F.F., H.T.S., R.Z.), Drug Discovery Institute (W.W., R.Z.), Stem Cell Center, Department of Biology and Biochemistry (W.X., F.M.), University of Houston, Houston, Texas; College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee (N.A., W.L.); and Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas (J.Z.)
| | - Frank McKeon
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy (W.W., Y.D., J.F.F., H.T.S., R.Z.), Drug Discovery Institute (W.W., R.Z.), Stem Cell Center, Department of Biology and Biochemistry (W.X., F.M.), University of Houston, Houston, Texas; College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee (N.A., W.L.); and Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas (J.Z.)
| | - Wei Li
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy (W.W., Y.D., J.F.F., H.T.S., R.Z.), Drug Discovery Institute (W.W., R.Z.), Stem Cell Center, Department of Biology and Biochemistry (W.X., F.M.), University of Houston, Houston, Texas; College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee (N.A., W.L.); and Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas (J.Z.)
| | - Jia Zhou
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy (W.W., Y.D., J.F.F., H.T.S., R.Z.), Drug Discovery Institute (W.W., R.Z.), Stem Cell Center, Department of Biology and Biochemistry (W.X., F.M.), University of Houston, Houston, Texas; College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee (N.A., W.L.); and Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas (J.Z.)
| | - Ruiwen Zhang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy (W.W., Y.D., J.F.F., H.T.S., R.Z.), Drug Discovery Institute (W.W., R.Z.), Stem Cell Center, Department of Biology and Biochemistry (W.X., F.M.), University of Houston, Houston, Texas; College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee (N.A., W.L.); and Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas (J.Z.)
| |
Collapse
|
19
|
Lin Z, Liu C, Yan Z, Cheng J, Wang X, Zhou F, Lyu X, Zhang S, Zhang D, Meng X, Zhao Y. Synthesis and biological evaluation of 4-imidazolidinone-containing compounds as potent inhibitors of the MDM2/p53 interaction. Eur J Med Chem 2024; 270:116366. [PMID: 38581730 DOI: 10.1016/j.ejmech.2024.116366] [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: 02/05/2024] [Revised: 03/19/2024] [Accepted: 03/27/2024] [Indexed: 04/08/2024]
Abstract
Inhibition of MDM2/p53 interaction with small-molecule inhibitors stabilizes p53 from MDM2 mediated degradation, which is a promising strategy for the treatment of cancer. In this report, a novel series of 4-imidazolidinone-containing compounds have been synthesized and tested in MDM2/p53 and MDM4/p53 FP binding assays. Upon SAR studies, compounds 2 (TB114) and 22 were identified as the most potent inhibitors of MDM2/p53 but not MDM4/p53 interactions. Both 2 and 22 exhibited strong antiproliferative activities in HCT-116 and MOLM-13 cell lines harboring wild type p53. Mechanistic studies show that 2 and 22 dose-dependently activated p53 and its target genes and induced apoptosis in cells based on the Western blot, qPCR, and flow cytometry assays. In addition, the antiproliferative activities of 2 and 22 were dependent on wild type p53, while they were not toxic to HEK-293 kidney cells. Furthermore, the on-target activities of 2 were general and applicable to other cancer cell lines with wild type p53. These attributes make 2 a good candidate for future optimization to discover a potential treatment of wild-type p53 cancer.
Collapse
Affiliation(s)
- Zhitong Lin
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China; State Key Laboratory of Drug Research and Small-Molecule Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road Shanghai, 201203, China
| | - Chen Liu
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China; State Key Laboratory of Drug Research and Small-Molecule Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road Shanghai, 201203, China
| | - Ziqin Yan
- State Key Laboratory of Drug Research and Small-Molecule Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road Shanghai, 201203, China
| | - Jing Cheng
- State Key Laboratory of Drug Research and Small-Molecule Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road Shanghai, 201203, China; University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China
| | - Xiancheng Wang
- State Key Laboratory of Drug Research and Small-Molecule Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road Shanghai, 201203, China; University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China
| | - Feilong Zhou
- State Key Laboratory of Drug Research and Small-Molecule Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road Shanghai, 201203, China
| | - Xilin Lyu
- State Key Laboratory of Drug Research and Small-Molecule Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road Shanghai, 201203, China
| | - Shiyan Zhang
- State Key Laboratory of Drug Research and Small-Molecule Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road Shanghai, 201203, China; University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China
| | - Daizhou Zhang
- Shandong Provincial Key Laboratory of Biopharmaceuticals, Shandong Academy of Pharmaceutical Sciences, Jinan, 250101, China
| | - Xiangjing Meng
- Shandong Provincial Key Laboratory of Biopharmaceuticals, Shandong Academy of Pharmaceutical Sciences, Jinan, 250101, China.
| | - Yujun Zhao
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China; State Key Laboratory of Drug Research and Small-Molecule Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road Shanghai, 201203, China; University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China; Shandong Provincial Key Laboratory of Biopharmaceuticals, Shandong Academy of Pharmaceutical Sciences, Jinan, 250101, China.
| |
Collapse
|
20
|
Esposito E, Pozza E, Contado C, Pula W, Bortolini O, Ragno D, Toldo S, Casciano F, Bondi A, Zauli E, Secchiero P, Zauli G, Melloni E. Microfluidic Fabricated Liposomes for Nutlin-3a Ocular Delivery as Potential Candidate for Proliferative Vitreoretinal Diseases Treatment. Int J Nanomedicine 2024; 19:3513-3536. [PMID: 38623081 PMCID: PMC11018138 DOI: 10.2147/ijn.s452134] [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: 12/12/2023] [Accepted: 03/09/2024] [Indexed: 04/17/2024] Open
Abstract
Purpose Proliferative vitreoretinal diseases (PVDs) represent a heterogeneous group of pathologies characterized by the presence of retinal proliferative membranes, in whose development retinal pigment epithelium (RPE) is deeply involved. As the only effective treatment for PVDs at present is surgery, we aimed to investigate the potential therapeutic activity of Nutlin-3a, a small non-genotoxic inhibitor of the MDM2/p53 interaction, on ARPE-19 cell line and on human RPE primary cells, as in vitro models of RPE and, more importantly, to formulate and evaluate Nutlin-3a loaded liposomes designed for ophthalmic administration. Methods Liposomes were produced using an innovative approach by a microfluidic device under selection of different conditions. Liposome size distribution was evaluated by photon correlation spectroscopy and centrifugal field flow fractionation, while the liposome structure was studied by transmission electron microscopy and Fourier-transform infrared spectroscopy. The Nutlin-3a entrapment capacity was evaluated by ultrafiltration and HPLC. Nutlin-3a biological effectiveness as a solution or loaded in liposomes was evaluated by viability, proliferation, apoptosis and migration assays and by morphological analysis. Results The microfluidic formulative study enabled the selection of liposomes composed of phosphatidylcholine (PC) 5.4 or 8.2 mg/mL and 10% ethanol, characterized by roundish vesicular structures with 150-250 nm mean diameters. Particularly, liposomes based on the lower PC concentration were characterized by higher stability. Nutlin-3a was effectively encapsulated in liposomes and was able to induce a significant reduction of viability and migration in RPE cell models. Conclusion Our results lay the basis for a possible use of liposomes for the ocular delivery of Nutlin-3a.
Collapse
Affiliation(s)
- Elisabetta Esposito
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Ferrara, I-44121, Italy
| | - Elena Pozza
- Department of Translational Medicine, University of Ferrara, Ferrara, I-44121, Italy
| | - Catia Contado
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Ferrara, I-44121, Italy
| | - Walter Pula
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Ferrara, I-44121, Italy
| | - Olga Bortolini
- Department of Environmental Sciences and Prevention, University of Ferrara, Ferrara, I-44121, Italy
| | - Daniele Ragno
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Ferrara, I-44121, Italy
| | - Sofia Toldo
- Department of Environmental Sciences and Prevention, University of Ferrara, Ferrara, I-44121, Italy
| | - Fabio Casciano
- Department of Translational Medicine and LTTA Centre, University of Ferrara, Ferrara, I-44121, Italy
| | - Agnese Bondi
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Ferrara, I-44121, Italy
| | - Enrico Zauli
- Department of Translational Medicine, University of Ferrara, Ferrara, I-44121, Italy
| | - Paola Secchiero
- Department of Translational Medicine and LTTA Centre, University of Ferrara, Ferrara, I-44121, Italy
| | - Giorgio Zauli
- Department of Environmental Sciences and Prevention, University of Ferrara, Ferrara, I-44121, Italy
| | - Elisabetta Melloni
- Department of Translational Medicine and LTTA Centre, University of Ferrara, Ferrara, I-44121, Italy
| |
Collapse
|
21
|
Nolt GL, Keeble AR, Wen Y, Strong AC, Thomas NT, Valentino TR, Brightwell CR, Murach KA, Patrizia S, Weinstabl H, Gollner A, McCarthy JJ, Fry CS, Franti M, Filareto A, Peterson CA, Dungan CM. Inhibition of p53-MDM2 binding reduces senescent cell abundance and improves the adaptive responses of skeletal muscle from aged mice. GeroScience 2024; 46:2153-2176. [PMID: 37872294 PMCID: PMC10828311 DOI: 10.1007/s11357-023-00976-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 10/09/2023] [Indexed: 10/25/2023] Open
Abstract
Skeletal muscle adaptation to external stimuli, such as regeneration following injury and hypertrophy in response to resistance exercise, are blunted with advanced age. The accumulation of senescent cells, along with defects in myogenic progenitor cell (MPC) proliferation, have been strongly linked as contributing factors to age-associated impairment in muscle adaptation. p53 plays an integral role in all these processes, as upregulation of p53 causes apoptosis in senescent cells and prevents mitotic catastrophe in MPCs from old mice. The goal of this study was to determine if a novel pharmaceutical agent (BI01), which functions by upregulating p53 through inhibition of binding to MDM2, the primary p53 regulatory protein, improves muscle regeneration and hypertrophy in old mice. BI01 effectively reduced the number of senescent cells in vitro but had no effect on MPC survival or proliferation at a comparable dose. Following repeated oral gavage with 2 mg/kg of BI01 (OS) or vehicle (OV), old mice (24 months) underwent unilateral BaCl2 injury in the tibialis anterior (TA) muscle, with PBS injections serving as controls. After 7 days, satellite cell number was higher in the TA of OS compared to OV mice, as was the expression of genes involved in ATP production. By 35 days, old mice treated with BI01 displayed reduced senescent cell burden, enhanced regeneration (higher muscle mass and fiber cross-sectional area) and restoration of muscle function relative to OV mice. To examine the impact of 2 mg/kg BI01 on muscle hypertrophy, the plantaris muscle was subjected to 28 days of mechanical overload (MOV) in OS and OV mice. In response to MOV, OS mice had larger plantaris muscles and muscle fibers than OV mice, particularly type 2b + x fibers, associated with reduced senescent cells. Together our data show that BI01 is an effective senolytic agent that may also augment muscle metabolism to enhance muscle regeneration and hypertrophy in old mice.
Collapse
Affiliation(s)
- Georgia L Nolt
- Department of Physiology, University of Kentucky, Lexington, KY, USA
- The Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| | - Alexander R Keeble
- Department of Physiology, University of Kentucky, Lexington, KY, USA
- The Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| | - Yuan Wen
- The Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
- Department of Physical Therapy, University of Kentucky, Lexington, KY, USA
| | - Aubrey C Strong
- The Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| | - Nicholas T Thomas
- The Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
- Department of Athletic Training and Clinical Nutrition, University of Kentucky, Lexington, KY, USA
| | - Taylor R Valentino
- Department of Physiology, University of Kentucky, Lexington, KY, USA
- The Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| | - Camille R Brightwell
- The Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
- Department of Athletic Training and Clinical Nutrition, University of Kentucky, Lexington, KY, USA
| | - Kevin A Murach
- Department of Health, Human Performance, and Recreation, University of Arkansas, Fayetteville, AR, USA
| | - Sini Patrizia
- Regenerative Medicine, Boehringer Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, CT, 06877, USA
| | - Harald Weinstabl
- Boehringer Ingelheim RCV, Boehringer Ingelheim Pharmaceuticals Inc., Vienna, Austria
| | - Andreas Gollner
- Boehringer Ingelheim RCV, Boehringer Ingelheim Pharmaceuticals Inc., Vienna, Austria
| | - John J McCarthy
- Department of Physiology, University of Kentucky, Lexington, KY, USA
- The Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| | - Christopher S Fry
- The Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
- Department of Athletic Training and Clinical Nutrition, University of Kentucky, Lexington, KY, USA
| | - Michael Franti
- Regenerative Medicine, Boehringer Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, CT, 06877, USA
| | - Antonio Filareto
- Regenerative Medicine, Boehringer Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, CT, 06877, USA.
| | - Charlotte A Peterson
- The Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
- Department of Physical Therapy, University of Kentucky, Lexington, KY, USA
| | - Cory M Dungan
- The Center for Muscle Biology, University of Kentucky, Lexington, KY, USA.
- Department of Physical Therapy, University of Kentucky, Lexington, KY, USA.
- Department of Health, Human Performance, and Recreation, Baylor University, One Bear Place #97313, Waco, TX, 76706, USA.
| |
Collapse
|
22
|
Abed A, Greene MK, Alsa’d AA, Lees A, Hindley A, Longley DB, McDade SS, Scott CJ. Nanoencapsulation of MDM2 Inhibitor RG7388 and Class-I HDAC Inhibitor Entinostat Enhances their Therapeutic Potential Through Synergistic Antitumor Effects and Reduction of Systemic Toxicity. Mol Pharm 2024; 21:1246-1255. [PMID: 38334409 PMCID: PMC10915795 DOI: 10.1021/acs.molpharmaceut.3c00926] [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: 10/04/2023] [Revised: 01/24/2024] [Accepted: 01/24/2024] [Indexed: 02/10/2024]
Abstract
Inhibitors of the p53-MDM2 interaction such as RG7388 have been developed to exploit latent tumor suppressive properties in p53 in 50% of tumors in which p53 is wild-type. However, these agents for the most part activate cell cycle arrest rather than death, and high doses in patients elicit on-target dose-limiting neutropenia. Recent work from our group indicates that combination of p53-MDM2 inhibitors with the class-I HDAC inhibitor Entinostat (which itself has dose-limiting toxicity issues) has the potential to significantly augment cell death in p53 wild-type colorectal cancer cells. We investigated whether coencapsulation of RG7388 and Entinostat within polymeric nanoparticles (NPs) could overcome efficacy and toxicity limitations of this drug combination. Combinations of RG7388 and Entinostat across a range of different molar ratios resulted in synergistic increases in cell death when delivered in both free drug and nanoencapsulated formats in all colorectal cell lines tested. Importantly, we also explored the in vivo impact of the drug combination on murine blood leukocytes, showing that the leukopenia induced by the free drugs could be significantly mitigated by nanoencapsulation. Taken together, this study demonstrates that formulating these agents within a single nanoparticle delivery platform may provide clinical utility beyond use as nonencapsulated agents.
Collapse
Affiliation(s)
- Anas Abed
- The
Patrick G Johnston Centre for Cancer Research, School of Medicine,
Dentistry and Biomedical Sciences, Queen’s
University Belfast, 97 Lisburn Road, Belfast BT9 7AE, United Kingdom
- Pharmacological
and Diagnostic Research Centre, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman 19111, Jordan
| | - Michelle K. Greene
- The
Patrick G Johnston Centre for Cancer Research, School of Medicine,
Dentistry and Biomedical Sciences, Queen’s
University Belfast, 97 Lisburn Road, Belfast BT9 7AE, United Kingdom
| | - Alhareth A. Alsa’d
- Pharmacological
and Diagnostic Research Centre, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman 19111, Jordan
- School
of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - Andrea Lees
- The
Patrick G Johnston Centre for Cancer Research, School of Medicine,
Dentistry and Biomedical Sciences, Queen’s
University Belfast, 97 Lisburn Road, Belfast BT9 7AE, United Kingdom
| | - Andrew Hindley
- Clinical
Haematology, Belfast City Hospital, 97 Lisburn Road, Belfast, BT9 7AB, United Kingdom
| | - Daniel B Longley
- The
Patrick G Johnston Centre for Cancer Research, School of Medicine,
Dentistry and Biomedical Sciences, Queen’s
University Belfast, 97 Lisburn Road, Belfast BT9 7AE, United Kingdom
| | - Simon S McDade
- The
Patrick G Johnston Centre for Cancer Research, School of Medicine,
Dentistry and Biomedical Sciences, Queen’s
University Belfast, 97 Lisburn Road, Belfast BT9 7AE, United Kingdom
| | - Christopher J. Scott
- The
Patrick G Johnston Centre for Cancer Research, School of Medicine,
Dentistry and Biomedical Sciences, Queen’s
University Belfast, 97 Lisburn Road, Belfast BT9 7AE, United Kingdom
| |
Collapse
|
23
|
Shin JH, Kim HR, Roe JS. A pipeline to characterize p53 effectors by integrative cistrome and transcriptome analysis in a genetically-defined organoid model. Genes Dis 2024; 11:512-515. [PMID: 37692486 PMCID: PMC10491866 DOI: 10.1016/j.gendis.2023.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 02/21/2023] [Accepted: 03/13/2023] [Indexed: 09/12/2023] Open
Affiliation(s)
- June-Ha Shin
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Republic of Korea
| | - Hwa-Ryeon Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Republic of Korea
| | - Jae-Seok Roe
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Republic of Korea
| |
Collapse
|
24
|
Obrador E, Moreno-Murciano P, Oriol-Caballo M, López-Blanch R, Pineda B, Gutiérrez-Arroyo JL, Loras A, Gonzalez-Bonet LG, Martinez-Cadenas C, Estrela JM, Marqués-Torrejón MÁ. Glioblastoma Therapy: Past, Present and Future. Int J Mol Sci 2024; 25:2529. [PMID: 38473776 PMCID: PMC10931797 DOI: 10.3390/ijms25052529] [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: 12/23/2023] [Revised: 02/10/2024] [Accepted: 02/16/2024] [Indexed: 03/14/2024] Open
Abstract
Glioblastoma (GB) stands out as the most prevalent and lethal form of brain cancer. Although great efforts have been made by clinicians and researchers, no significant improvement in survival has been achieved since the Stupp protocol became the standard of care (SOC) in 2005. Despite multimodality treatments, recurrence is almost universal with survival rates under 2 years after diagnosis. Here, we discuss the recent progress in our understanding of GB pathophysiology, in particular, the importance of glioma stem cells (GSCs), the tumor microenvironment conditions, and epigenetic mechanisms involved in GB growth, aggressiveness and recurrence. The discussion on therapeutic strategies first covers the SOC treatment and targeted therapies that have been shown to interfere with different signaling pathways (pRB/CDK4/RB1/P16ink4, TP53/MDM2/P14arf, PI3k/Akt-PTEN, RAS/RAF/MEK, PARP) involved in GB tumorigenesis, pathophysiology, and treatment resistance acquisition. Below, we analyze several immunotherapeutic approaches (i.e., checkpoint inhibitors, vaccines, CAR-modified NK or T cells, oncolytic virotherapy) that have been used in an attempt to enhance the immune response against GB, and thereby avoid recidivism or increase survival of GB patients. Finally, we present treatment attempts made using nanotherapies (nanometric structures having active anti-GB agents such as antibodies, chemotherapeutic/anti-angiogenic drugs or sensitizers, radionuclides, and molecules that target GB cellular receptors or open the blood-brain barrier) and non-ionizing energies (laser interstitial thermal therapy, high/low intensity focused ultrasounds, photodynamic/sonodynamic therapies and electroporation). The aim of this review is to discuss the advances and limitations of the current therapies and to present novel approaches that are under development or following clinical trials.
Collapse
Affiliation(s)
- Elena Obrador
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Paz Moreno-Murciano
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
| | - María Oriol-Caballo
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Rafael López-Blanch
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Begoña Pineda
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Julia Lara Gutiérrez-Arroyo
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon, Spain; (J.L.G.-A.); (A.L.); (C.M.-C.)
| | - Alba Loras
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon, Spain; (J.L.G.-A.); (A.L.); (C.M.-C.)
| | - Luis G. Gonzalez-Bonet
- Department of Neurosurgery, Castellon General University Hospital, 12004 Castellon, Spain;
| | - Conrado Martinez-Cadenas
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon, Spain; (J.L.G.-A.); (A.L.); (C.M.-C.)
| | - José M. Estrela
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
- Department of Physiology, Faculty of Pharmacy, University of Valencia, 46100 Burjassot, Spain
| | | |
Collapse
|
25
|
Stafylidis C, Vlachopoulou D, Kontandreopoulou CN, Diamantopoulos PΤ. Unmet Horizons: Assessing the Challenges in the Treatment of TP53-Mutated Acute Myeloid Leukemia. J Clin Med 2024; 13:1082. [PMID: 38398394 PMCID: PMC10889132 DOI: 10.3390/jcm13041082] [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: 12/27/2023] [Revised: 02/07/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
Acute myeloid leukemia (AML) remains a challenging hematologic malignancy. The presence of TP53 mutations in AML poses a therapeutic challenge, considering that standard treatments face significant setbacks in achieving meaningful responses. There is a pressing need for the development of innovative treatment modalities to overcome resistance to conventional treatments attributable to the unique biology of TP53-mutated (TP53mut) AML. This review underscores the role of TP53 mutations in AML, examines the current landscape of treatment options, and highlights novel therapeutic approaches, including targeted therapies, combination regimens, and emerging immunotherapies, as well as agents being explored in preclinical studies according to their potential to address the unique hurdles posed by TP53mut AML.
Collapse
Affiliation(s)
| | | | | | - Panagiotis Τ. Diamantopoulos
- Hematology Unit, First Department of Internal Medicine, Laikon General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (C.S.); (D.V.); (C.-N.K.)
| |
Collapse
|
26
|
Kim D, Min D, Kim J, Kim MJ, Seo Y, Jung BH, Kwon SH, Ro H, Lee S, Sa JK, Lee JY. Nutlin-3a induces KRAS mutant/p53 wild type lung cancer specific methuosis-like cell death that is dependent on GFPT2. J Exp Clin Cancer Res 2023; 42:338. [PMID: 38093368 PMCID: PMC10720203 DOI: 10.1186/s13046-023-02922-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 12/01/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND Oncogenic KRAS mutation, the most frequent mutation in non-small cell lung cancer (NSCLC), is an aggressiveness risk factor and leads to the metabolic reprogramming of cancer cells by promoting glucose, glutamine, and fatty acid absorption and glycolysis. Lately, sotorasib was approved by the FDA as a first-in-class KRAS-G12C inhibitor. However, sotorasib still has a derivative barrier, which is not effective for other KRAS mutation types, except for G12C. Additionally, resistance to sotorasib is likely to develop, demanding the need for alternative therapeutic strategies. METHODS KRAS mutant, and wildtype NSCLC cells were used in vitro cell analyses. Cell viability, proliferation, and death were measured by MTT, cell counting, colony analyses, and annexin V staining for FACS. Cell tracker dyes were used to investigate cell morphology, which was examined by holotomograpy, and confocal microscopes. RNA sequencing was performed to identify key target molecule or pathway, which was confirmed by qRT-PCR, western blotting, and metabolite analyses by UHPLC-MS/MS. Zebrafish and mouse xenograft model were used for in vivo analysis. RESULTS In this study, we found that nutlin-3a, an MDM2 antagonist, inhibited the KRAS-PI3K/Akt-mTOR pathway and disrupted the fusion of both autophagosomes and macropinosomes with lysosomes. This further elucidated non-apoptotic and catastrophic macropinocytosis associated methuosis-like cell death, which was found to be dependent on GFPT2 of the hexosamine biosynthetic pathway, specifically in KRAS mutant /p53 wild type NSCLC cells. CONCLUSION These results indicate the potential of nutlin-3a as an alternative agent for treating KRAS mutant/p53 wild type NSCLC cells.
Collapse
Affiliation(s)
- Dasom Kim
- Department of Pathology, Korea University College of Medicine, 73, Goryeodae-Ro, Seongbuk-Gu, Seoul, 02841, South Korea
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, South Korea
| | - Dongwha Min
- Department of Pathology, Korea University College of Medicine, 73, Goryeodae-Ro, Seongbuk-Gu, Seoul, 02841, South Korea
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, South Korea
| | - Joohee Kim
- Department of Biological Sciences, Sookmyung Women's University, Seoul, South Korea
| | - Min Jung Kim
- Department of Biological Sciences, Sookmyung Women's University, Seoul, South Korea
| | - Yerim Seo
- Center for Advanced Biomolecular Recognition, Korea Instiute of Science and Technology (KIST), Seoul, 02792, Korea
| | - Byung Hwa Jung
- Center for Advanced Biomolecular Recognition, Korea Instiute of Science and Technology (KIST), Seoul, 02792, Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul, 02792, South Korea
| | - Seung-Hae Kwon
- Korea Basic Science Institute, Seoul Center, Seoul, South Korea
| | - Hyunju Ro
- Department of Biological Sciences, College of Bioscience and Biotechnology, Chungnam National University, Daejeon, 34134, Korea
| | - Seoee Lee
- Department of Biological Sciences, College of Bioscience and Biotechnology, Chungnam National University, Daejeon, 34134, Korea
| | - Jason K Sa
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, South Korea
- Department of Biomedical Informatics, Korea University College of Medicine, Seoul, South Korea
| | - Ji-Yun Lee
- Department of Pathology, Korea University College of Medicine, 73, Goryeodae-Ro, Seongbuk-Gu, Seoul, 02841, South Korea.
| |
Collapse
|
27
|
Fang Y, Jiang Y, Wei L, Ma Q, Ren Z, Yuan Q, Wei DQ. DeepProSite: structure-aware protein binding site prediction using ESMFold and pretrained language model. Bioinformatics 2023; 39:btad718. [PMID: 38015872 PMCID: PMC10723037 DOI: 10.1093/bioinformatics/btad718] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 11/04/2023] [Accepted: 11/27/2023] [Indexed: 11/30/2023] Open
Abstract
MOTIVATION Identifying the functional sites of a protein, such as the binding sites of proteins, peptides, or other biological components, is crucial for understanding related biological processes and drug design. However, existing sequence-based methods have limited predictive accuracy, as they only consider sequence-adjacent contextual features and lack structural information. RESULTS In this study, DeepProSite is presented as a new framework for identifying protein binding site that utilizes protein structure and sequence information. DeepProSite first generates protein structures from ESMFold and sequence representations from pretrained language models. It then uses Graph Transformer and formulates binding site predictions as graph node classifications. In predicting protein-protein/peptide binding sites, DeepProSite outperforms state-of-the-art sequence- and structure-based methods on most metrics. Moreover, DeepProSite maintains its performance when predicting unbound structures, in contrast to competing structure-based prediction methods. DeepProSite is also extended to the prediction of binding sites for nucleic acids and other ligands, verifying its generalization capability. Finally, an online server for predicting multiple types of residue is established as the implementation of the proposed DeepProSite. AVAILABILITY AND IMPLEMENTATION The datasets and source codes can be accessed at https://github.com/WeiLab-Biology/DeepProSite. The proposed DeepProSite can be accessed at https://inner.wei-group.net/DeepProSite/.
Collapse
Affiliation(s)
- Yitian Fang
- State Key Laboratory of Microbial Metabolism, Shanghai-Islamabad-Belgrade Joint Innovation Center on Antibacterial Resistances, Joint International Research Laboratory of Metabolic & Developmental Sciences and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200040, China
- Peng Cheng Laboratory, Shenzhen 518055, China
| | - Yi Jiang
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Leyi Wei
- School of Software, Shandong University, Jinan, Shandong 250100, China
| | - Qin Ma
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | | | - Qianmu Yuan
- School of Computer Science and Engineering, Sun Yat-sen University, Guangzhou 510000, China
| | - Dong-Qing Wei
- State Key Laboratory of Microbial Metabolism, Shanghai-Islamabad-Belgrade Joint Innovation Center on Antibacterial Resistances, Joint International Research Laboratory of Metabolic & Developmental Sciences and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200040, China
- Peng Cheng Laboratory, Shenzhen 518055, China
| |
Collapse
|
28
|
Wu DC, Ku CC, Pan JB, Wuputra K, Yang YH, Liu CJ, Liu YC, Kato K, Saito S, Lin YC, Chong IW, Hsiao M, Hu HM, Kuo CH, Kuo KK, Lin CS, Yokoyama KK. Heterogeneity of Phase II Enzyme Ligands on Controlling the Progression of Human Gastric Cancer Organoids as Stem Cell Therapy Model. Int J Mol Sci 2023; 24:15911. [PMID: 37958895 PMCID: PMC10647227 DOI: 10.3390/ijms242115911] [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: 09/21/2023] [Revised: 10/26/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023] Open
Abstract
Gastric cancer (GC) organoids are frequently used to examine cell proliferation and death as well as cancer development. Invasion/migration assay, xenotransplantation, and reactive oxygen species (ROS) production were used to examine the effects of antioxidant drugs, including perillaldehyde (PEA), cinnamaldehyde (CA), and sulforaphane (SFN), on GC. PEA and CA repressed the proliferation of human GC organoids, whereas SFN enhanced it. Caspase 3 activities were also repressed on treatment with PEA and CA. Furthermore, the tumor formation and invasive activities were repressed on treatment with PEA and CA, whereas they were enhanced on treatment with SFN. These results in three-dimensional (3D)-GC organoids showed the different cancer development of phase II enzyme ligands in 2D-GC cells. ROS production and the expression of TP53, nuclear factor erythroid 2-related factor (NRF2), and Jun dimerization protein 2 were also downregulated on treatment with PEA and CA, but not SFN. NRF2 knockdown reversed the effects of these antioxidant drugs on the invasive activities of the 3D-GC organoids. Moreover, ROS production was also inhibited by treatment with PEA and CA, but not SFN. Thus, NRF2 plays a key role in the differential effects of these antioxidant drugs on cancer progression in 3D-GC organoids. PEA and CA can potentially be new antitumorigenic therapeutics for GC.
Collapse
Affiliation(s)
- Deng-Chyang Wu
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (D.-C.W.); (C.-C.K.); (J.-B.P.); (K.W.); (I.-W.C.); (C.-S.L.)
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (Y.-H.Y.); (C.-J.L.); (K.-K.K.)
- Cell Therapy and Research Center, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan;
- Division of General and Digestive Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan (C.-H.K.)
| | - Chia-Chen Ku
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (D.-C.W.); (C.-C.K.); (J.-B.P.); (K.W.); (I.-W.C.); (C.-S.L.)
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (Y.-H.Y.); (C.-J.L.); (K.-K.K.)
- Cell Therapy and Research Center, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan;
| | - Jia-Bin Pan
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (D.-C.W.); (C.-C.K.); (J.-B.P.); (K.W.); (I.-W.C.); (C.-S.L.)
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (Y.-H.Y.); (C.-J.L.); (K.-K.K.)
- Cell Therapy and Research Center, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan;
| | - Kenly Wuputra
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (D.-C.W.); (C.-C.K.); (J.-B.P.); (K.W.); (I.-W.C.); (C.-S.L.)
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (Y.-H.Y.); (C.-J.L.); (K.-K.K.)
- Cell Therapy and Research Center, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan;
| | - Ya-Han Yang
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (Y.-H.Y.); (C.-J.L.); (K.-K.K.)
- Division of General and Digestive Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan (C.-H.K.)
| | - Chung-Jung Liu
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (Y.-H.Y.); (C.-J.L.); (K.-K.K.)
- Cell Therapy and Research Center, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan;
- Division of General and Digestive Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan (C.-H.K.)
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Yi-Chang Liu
- Cell Therapy and Research Center, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan;
| | - Kohsuke Kato
- Department of Infection Biology, Graduate School of Comprehensive Human Sciences, The University of Tsukuba, Tsukuba 305-8577, Japan;
| | - Shigeo Saito
- Saito Laboratory of Cell Technology, Yaita 239-1571, Japan;
| | - Ying-Chu Lin
- School of Dentistry, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Inn-Wen Chong
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (D.-C.W.); (C.-C.K.); (J.-B.P.); (K.W.); (I.-W.C.); (C.-S.L.)
- Division of Pulmonary and Critical Care Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Michael Hsiao
- Genome Research Center, Academia Sinica, Nangan, Taipei 115, Taiwan;
| | - Huang-Ming Hu
- Division of General and Digestive Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan (C.-H.K.)
- Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 801, Taiwan
| | - Chao-Hung Kuo
- Division of General and Digestive Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan (C.-H.K.)
- Department of Internal Medicine, Kaohsiung Municipal Siaogang Hospital, Kaohsiung 812, Taiwan
| | - Kung-Kai Kuo
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (Y.-H.Y.); (C.-J.L.); (K.-K.K.)
- Division of General and Digestive Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan (C.-H.K.)
| | - Chang-Shen Lin
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (D.-C.W.); (C.-C.K.); (J.-B.P.); (K.W.); (I.-W.C.); (C.-S.L.)
| | - Kazunari K. Yokoyama
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (D.-C.W.); (C.-C.K.); (J.-B.P.); (K.W.); (I.-W.C.); (C.-S.L.)
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (Y.-H.Y.); (C.-J.L.); (K.-K.K.)
- Cell Therapy and Research Center, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan;
| |
Collapse
|
29
|
Wu D, Li Y, Li C, Zhong S, Liu B, Hang H, Wang H. MDM2 Antagonist Nutlin-3 Stimulates Global DNA Hydroxymethylation by Enhancing p53-TET1 Signaling Axis. ACS Chem Biol 2023; 18:2240-2248. [PMID: 37463352 DOI: 10.1021/acschembio.3c00247] [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: 07/20/2023]
Abstract
DNA hydroxymethylation is involved in many biological processes, including nuclear reprogramming, embryonic development, and tumor suppression. In this study, we report that an anticancer agent, nutlin-3, selectively stimulates global DNA hydroxymethylation in TP53 wild-type cancer cells as manifested by the elevation of 5-hydroxymethylcytosine (5hmC) in genomic DNA. In contrast, nutlin 3 fails to enhance DNA hydroxymethylation in TP53-mutated cancer cells. Consistently, nutlin-3 as a MDM2 antagonist only activates wild-type but not mutated TP53. Furthermore, nutlin-3 does not alter the expression of TET1 but slightly reduces the expression of TET2 and TET3 proteins. These TET family proteins are responsible for converting 5-methylcytosine (5mC) to 5hmC. Interestingly, TET1 knockdown could significantly block the nutlin-3-induced DNA hydroxymethylation as well as TP53 and P21 activation. Immunoprecipitation analysis supports that p53 strongly interacts with TET1 proteins. These results suggest that nutlin-3 activates TP53 and promotes p53-TET1 interaction. As positive feedback, the p53-TET1 interaction further enhances p53 activation and promotes apoptosis. Collectively, we demonstrate that nutlin-3 stimulates DNA hydroxymethylation and apoptosis via a positive feedback mechanism.
Collapse
Affiliation(s)
- Danni Wu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- China General Microbiological Culture Collection Center, State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yao Li
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Cuiping Li
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Shangwei Zhong
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Baodong Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Haiying Hang
- Key Laboratory for Protein and Peptide Pharmaceuticals, National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Hailin Wang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- China General Microbiological Culture Collection Center, State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- School of Environment, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310000, P. R. China
| |
Collapse
|
30
|
Muravev AA, Voloshina AD, Sapunova AS, Gabdrakhmanova FB, Lenina OA, Petrov KA, Shityakov S, Skorb EV, Solovieva SE, Antipin IS. Calix[4]arene-pyrazole conjugates as potential cancer therapeutics. Bioorg Chem 2023; 139:106742. [PMID: 37480816 DOI: 10.1016/j.bioorg.2023.106742] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/12/2023] [Accepted: 07/16/2023] [Indexed: 07/24/2023]
Abstract
Tumor selectivity is yet a challenge in chemotherapy-based cancer treatment. A series of calixarenes derivatized at the lower rim with 3-phenyl-1H-pyrazole units with variable upper-rim substituent and conformations of macrocyclic core, alkyl chain length between heterocycle and core, as well as phenolic monomer (5-(4-tert-butylphenyloxy)methoxy-3-phenyl-1H-pyrazole) have been synthesized and characterized in a range of therapeutically relevant cellular models (M-HeLa, MCF7, A-549, PC3, Chang liver, and Wi38) from different target organs/systems. Specific cytotoxicity for M-HeLa cells has been observed in tert-butylcalix[4]arene pyrazoles in 1,3-alternate (compound 7b) and partial cone (compound 7c) conformations with low mutagenicity and haemotoxicity and in vivo toxicity in mice. Compounds 7b,c have induced mitochondrial pathway of apoptosis of M-HeLa cells through caspase-9 activation preceded by the cell cycle arrest at G0/G1 phase. A concomitant overexpression of DNA damage markers in pyrazole-treated M-HeLa cells suggests that calixarene pyrazoles target DNA, which was supported by the presence of interactions between calixarenes and ctDNA at the air-water interface.
Collapse
Affiliation(s)
- Anton A Muravev
- Infochemistry Scientific Center, ITMO University, Lomonosov Str. 9, 191002 Saint Petersburg, Russia; Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, Arbuzov Str. 8, 420088 Kazan, Russia.
| | - Alexandra D Voloshina
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, Arbuzov Str. 8, 420088 Kazan, Russia
| | - Anastasia S Sapunova
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, Arbuzov Str. 8, 420088 Kazan, Russia
| | - Farida B Gabdrakhmanova
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, Arbuzov Str. 8, 420088 Kazan, Russia
| | - Oksana A Lenina
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, Arbuzov Str. 8, 420088 Kazan, Russia
| | - Konstantin A Petrov
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, Arbuzov Str. 8, 420088 Kazan, Russia
| | - Sergey Shityakov
- Infochemistry Scientific Center, ITMO University, Lomonosov Str. 9, 191002 Saint Petersburg, Russia
| | - Ekaterina V Skorb
- Infochemistry Scientific Center, ITMO University, Lomonosov Str. 9, 191002 Saint Petersburg, Russia
| | - Svetlana E Solovieva
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, Arbuzov Str. 8, 420088 Kazan, Russia
| | - Igor S Antipin
- Kazan Federal University, Kremlyovskaya Str. 18, 420008 Kazan, Russia
| |
Collapse
|
31
|
Psatha K, Kollipara L, Drakos E, Deligianni E, Brintakis K, Patsouris E, Sickmann A, Rassidakis GZ, Aivaliotis M. Interruption of p53-MDM2 Interaction by Nutlin-3a in Human Lymphoma Cell Models Initiates a Cell-Dependent Global Effect on Transcriptome and Proteome Level. Cancers (Basel) 2023; 15:3903. [PMID: 37568720 PMCID: PMC10417430 DOI: 10.3390/cancers15153903] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/22/2023] [Accepted: 06/28/2023] [Indexed: 08/13/2023] Open
Abstract
In most lymphomas, p53 signaling pathway is inactivated by various mechanisms independent to p53 gene mutations or deletions. In many cases, p53 function is largely regulated by alterations in the protein abundance levels by the action of E3 ubiquitin-protein ligase MDM2, targeting p53 to proteasome-mediated degradation. In the present study, an integrating transcriptomics and proteomics analysis was employed to investigate the effect of p53 activation by a small-molecule MDM2-antagonist, nutlin-3a, on three lymphoma cell models following p53 activation. Our analysis revealed a system-wide nutlin-3a-associated effect in all examined lymphoma types, identifying in total of 4037 differentially affected proteins involved in a plethora of pathways, with significant heterogeneity among lymphomas. Our findings include known p53-targets and novel p53 activation effects, involving transcription, translation, or degradation of protein components of pathways, such as a decrease in key members of PI3K/mTOR pathway, heat-shock response, and glycolysis, and an increase in key members of oxidative phoshosphorylation, autophagy and mitochondrial translation. Combined inhibition of HSP90 or PI3K/mTOR pathway with nutlin-3a-mediated p53-activation enhanced the apoptotic effects suggesting a promising strategy against human lymphomas. Integrated omic profiling after p53 activation offered novel insights on the regulatory role specific proteins and pathways may have in lymphomagenesis.
Collapse
Affiliation(s)
- Konstantina Psatha
- Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology, 70013 Heraklion, Greece; (K.P.); (E.D.)
- Department of Pathology, Medical School, University of Crete, 70013 Heraklion, Greece;
- First Department of Pathology, National and Kapodistrian University of Athens, 15772 Athens, Greece;
- Functional Proteomics and Systems Biology (FunPATh), Center for Interdisciplinary Research and Innovation (CIRI-AUTH), 54124 Thessaloniki, Greece
| | - Laxmikanth Kollipara
- Leibniz-Institut für Analytische Wissenschaften–ISAS–e.V., 44139 Dortmund, Germany; (L.K.); (A.S.)
| | - Elias Drakos
- Department of Pathology, Medical School, University of Crete, 70013 Heraklion, Greece;
| | - Elena Deligianni
- Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology, 70013 Heraklion, Greece; (K.P.); (E.D.)
| | - Konstantinos Brintakis
- Institute of Electronic Structure and Laser, Foundation for Research and Technology—Hellas, 71110 Heraklion, Greece;
| | - Eustratios Patsouris
- First Department of Pathology, National and Kapodistrian University of Athens, 15772 Athens, Greece;
| | - Albert Sickmann
- Leibniz-Institut für Analytische Wissenschaften–ISAS–e.V., 44139 Dortmund, Germany; (L.K.); (A.S.)
- Department of Chemistry, College of Physical Sciences, University of Aberdeen, Aberdeen AB24 3FX, UK
- Medizinische Fakultät, Medizinische Proteom-Center (MPC), Ruhr-Universität Bochum, 44801 Bochum, Germany
| | - George Z. Rassidakis
- Department of Oncology-Pathology, Karolinska Institute, 17164 Stockholm, Sweden;
- Department of Hematopathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
- Department of Clinical Pathology and Cancer Diagnostics, Karolinska University Hospital, Solna, 17176 Stockholm, Sweden
| | - Michalis Aivaliotis
- Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology, 70013 Heraklion, Greece; (K.P.); (E.D.)
- Functional Proteomics and Systems Biology (FunPATh), Center for Interdisciplinary Research and Innovation (CIRI-AUTH), 54124 Thessaloniki, Greece
- Basic and Translational Research Unit, Special Unit for Biomedical Research and Education, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
- Laboratory of Biological Chemistry, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| |
Collapse
|
32
|
Pellot Ortiz KI, Rechberger JS, Nonnenbroich LF, Daniels DJ, Sarkaria JN. MDM2 Inhibition in the Treatment of Glioblastoma: From Concept to Clinical Investigation. Biomedicines 2023; 11:1879. [PMID: 37509518 PMCID: PMC10377337 DOI: 10.3390/biomedicines11071879] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 06/28/2023] [Accepted: 06/30/2023] [Indexed: 07/30/2023] Open
Abstract
Inhibition of the interaction between MDM2 and p53 has emerged as a promising strategy for combating cancer, including the treatment of glioblastoma (GBM). Numerous MDM2 inhibitors have been developed and are currently undergoing rigorous testing for their potential in GBM therapy. Encouraging results from studies conducted in cell culture and animal models suggest that MDM2 inhibitors could effectively treat a specific subset of GBM patients with wild-type TP53 or functional p53. Combination therapy with clinically established treatment modalities such as radiation and chemotherapy offers the potential to achieve a more profound therapeutic response. Furthermore, an increasing array of other molecularly targeted therapies are being explored in combination with MDM2 inhibitors to increase the effects of individual treatments. While some MDM2 inhibitors have progressed to early phase clinical trials in GBM, their efficacy, alone and in combination, is yet to be confirmed. In this article, we present an overview of MDM2 inhibitors currently under preclinical and clinical investigation, with a specific focus on the drugs being assessed in ongoing clinical trials for GBM patients.
Collapse
Affiliation(s)
| | - Julian S Rechberger
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, USA
| | - Leo F Nonnenbroich
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA
- Hopp Children's Cancer Center Heidelberg (KiTZ), 69120 Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), 69120 Heidelberg, Germany
| | - David J Daniels
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, USA
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
33
|
Senapati J, Muftuoglu M, Ishizawa J, Abbas HA, Loghavi S, Borthakur G, Yilmaz M, Issa GC, Dara SI, Basyal M, Li L, Naqvi K, Pourebrahim R, Jabbour EJ, Kornblau SM, Short NJ, Pemmaraju N, Garcia-Manero G, Ravandi F, Khoury J, Daver N, Kantarjian HM, Andreeff M, DiNardo CD. A Phase I study of Milademetan (DS3032b) in combination with low dose cytarabine with or without venetoclax in acute myeloid leukemia: Clinical safety, efficacy, and correlative analysis. Blood Cancer J 2023; 13:101. [PMID: 37386016 PMCID: PMC10310786 DOI: 10.1038/s41408-023-00871-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/02/2023] [Accepted: 06/13/2023] [Indexed: 07/01/2023] Open
Abstract
In TP53 wild-type acute myeloid leukemia (AML), inhibition of MDM2 can enhance p53 protein expression and potentiate leukemic cell apoptosis. MDM2 inhibitor (MDM2i) monotherapy in AML has shown modest responses in clinical trials but combining options of MDM2i with other potent AML-directed agents like cytarabine and venetoclax could improve its efficacy. We conducted a phase I clinical trial (NCT03634228) to study the safety and efficacy of milademetan (an MDM2i) with low-dose cytarabine (LDAC)±venetoclax in adult patients with relapsed refractory (R/R) or newly diagnosed (ND; unfit) TP53 wild-type AML and performed comprehensive CyTOF analyses to interrogate multiple signaling pathways, the p53-MDM2 axis and the interplay between pro/anti-apoptotic molecules to identify factors that determine response and resistance to therapy. Sixteen patients (14 R/R, 2 N/D treated secondary AML) at a median age of 70 years (range, 23-80 years) were treated in this trial. Two patients (13%) achieved an overall response (complete remission with incomplete hematological recovery). Median cycles on trial were 1 (range 1-7) and at a median follow-up of 11 months, no patients remained on active therapy. Gastrointestinal toxicity was significant and dose-limiting (50% of patients ≥ grade 3). Single-cell proteomic analysis of the leukemia compartment revealed therapy-induced proteomic alterations and potential mechanisms of adaptive response to the MDM2i combination. The response was associated with immune cell abundance and induced the proteomic profiles of leukemia cells to disrupt survival pathways and significantly reduced MCL1 and YTHDF2 to potentiate leukemic cell death. The combination of milademetan, LDAC±venetoclax led to only modest responses with recognizable gastrointestinal toxicity. Treatment-induced reduction of MCL1 and YTHDF2 in an immune-rich milieu correlate with treatment response.
Collapse
Affiliation(s)
- Jayastu Senapati
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | | | - Jo Ishizawa
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | - Hussein A Abbas
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | - Sanam Loghavi
- Department of Hematopathology, MD Anderson Cancer Center, Houston, TX, USA
| | - Gautam Borthakur
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | - Musa Yilmaz
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | - Ghayas C Issa
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | - Samuel I Dara
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | - Mahesh Basyal
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | - Li Li
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | - Kiran Naqvi
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | | | - Elias J Jabbour
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | | | - Nicholas J Short
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | - Naveen Pemmaraju
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | | | - Farhad Ravandi
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | - Joseph Khoury
- Department of Hematopathology, MD Anderson Cancer Center, Houston, TX, USA
| | - Naval Daver
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | | | - Michael Andreeff
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA.
| | | |
Collapse
|
34
|
Wermelinger GF, Rubini L, da Fonseca ACC, Ouverney G, de Oliveira RPRF, de Souza AS, Forezi LSM, Limaverde-Sousa G, Pinheiro S, Robbs BK. A Novel MDM2-Binding Chalcone Induces Apoptosis of Oral Squamous Cell Carcinoma. Biomedicines 2023; 11:1711. [PMID: 37371806 DOI: 10.3390/biomedicines11061711] [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: 05/25/2023] [Revised: 06/07/2023] [Accepted: 06/10/2023] [Indexed: 06/29/2023] Open
Abstract
Oral squamous cell carcinoma (OSCC) represents ~90% of all oral cancers, being the eighth most common cancer in men. The overall 5-year survival rate is only 39% for metastatic cancers, and currently used chemotherapeutics can cause important side effects. Thus, there is an urgency in developing new and effective anti-cancer agents. As both chalcones and 1,2,3-triazoles are valuable pharmacophores/privileged structures in the search for anticancer compounds, in this work, new 1,2,3-triazole-chalcone hybrids were synthesized and evaluated against oral squamous cell carcinoma. By using different in silico, in vitro, and in vivo approaches, we demonstrated that compound 1f has great cytotoxicity and selectivity against OSCC (higher than carboplatin and doxorubicin) and other cancer cells in addition to showing minimal toxicity in mice. Furthermore, we demonstrate that induced cell death occurs by apoptosis and cell cycle arrest at the G2/M phase. Moreover, we found that 1f has a potential affinity for MDM2 protein, similar to the known ligand nutlin-3, and presents a better selectivity, pharmacological profile, and potential to be orally absorbed and is not a substrate of Pg-P when compared to nutlin-3. Therefore, we conclude that 1f is a good lead for a new chemotherapeutic drug against OSCC and possibly other types of cancers.
Collapse
Affiliation(s)
- Guilherme Freimann Wermelinger
- Basic Science Department, Health Institute of Nova Friburgo, Fluminense Federal University, Nova Friburgo 28625-650, RJ, Brazil
| | - Lucas Rubini
- Basic Science Department, Health Institute of Nova Friburgo, Fluminense Federal University, Nova Friburgo 28625-650, RJ, Brazil
| | - Anna Carolina Carvalho da Fonseca
- Postgraduate Program in Dentistry, Health Institute of Nova Friburgo, Fluminense Federal University, Nova Friburgo 28625-650, RJ, Brazil
| | - Gabriel Ouverney
- Postgraduate Program in Applied Science for Health Products, Faculty of Pharmacy, Fluminense Federal University, Niteroi 24020-141, RJ, Brazil
| | - Rafael P R F de Oliveira
- Department of Organic Chemistry, Chemistry Institute, Fluminense Federal University, Niteroi 24020-141, RJ, Brazil
| | - Acácio S de Souza
- Department of Organic Chemistry, Chemistry Institute, Fluminense Federal University, Niteroi 24020-141, RJ, Brazil
| | - Luana S M Forezi
- Department of Organic Chemistry, Chemistry Institute, Fluminense Federal University, Niteroi 24020-141, RJ, Brazil
| | - Gabriel Limaverde-Sousa
- Oswaldo Cruz Institute, Oswaldo Cruz Foundation, FIOCRUZ, Rio de Janeiro 21040-900, RJ, Brazil
| | - Sergio Pinheiro
- Department of Organic Chemistry, Chemistry Institute, Fluminense Federal University, Niteroi 24020-141, RJ, Brazil
| | - Bruno Kaufmann Robbs
- Basic Science Department, Health Institute of Nova Friburgo, Fluminense Federal University, Nova Friburgo 28625-650, RJ, Brazil
| |
Collapse
|
35
|
Zhou MY, Bui NQ, Charville GW, Ganjoo KN, Pan M. Treatment of De-Differentiated Liposarcoma in the Era of Immunotherapy. Int J Mol Sci 2023; 24:ijms24119571. [PMID: 37298520 DOI: 10.3390/ijms24119571] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 05/30/2023] [Accepted: 05/30/2023] [Indexed: 06/12/2023] Open
Abstract
Well-differentiated/de-differentiated liposarcoma (WDLPS/DDLPS) is one of the most common histologic subtypes of soft tissue sarcoma (STS); however, treatment options remain limited. WDLPS and DDLPS both exhibit the characteristic amplification of chromosome region 12q13-15, which contains the genes CDK4 and MDM2. DDLPS exhibits higher amplification ratios of these two and carries additional genomic lesions, including the amplification of chromosome region 1p32 and chromosome region 6q23, which may explain the more aggressive biology of DDLPS. WDLPS does not respond to systemic chemotherapy and is primarily managed with local therapy, including multiple resections and debulking procedures whenever clinically feasible. In contrast, DDLPS can respond to chemotherapy drugs and drug combinations, including doxorubicin (or doxorubicin in combination with ifosfamide), gemcitabine (or gemcitabine in combination with docetaxel), trabectedin, eribulin, and pazopanib. However, the response rate is generally low, and the response duration is usually short. This review highlights the clinical trials with developmental therapeutics that have been completed or are ongoing, including CDK4/6 inhibitors, MDM2 inhibitors, and immune checkpoint inhibitors. This review will also discuss the current landscape in assessing biomarkers for identifying tumors sensitive to immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Maggie Y Zhou
- Sarcoma Program, Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Nam Q Bui
- Sarcoma Program, Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Gregory W Charville
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Kristen N Ganjoo
- Sarcoma Program, Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Minggui Pan
- Sarcoma Program, Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94304, USA
| |
Collapse
|
36
|
Adams CM, Mitra R, Xiao Y, Michener P, Palazzo J, Chao A, Gour J, Cassel J, Salvino JM, Eischen CM. Targeted MDM2 Degradation Reveals a New Vulnerability for p53-Inactivated Triple-Negative Breast Cancer. Cancer Discov 2023; 13:1210-1229. [PMID: 36734633 PMCID: PMC10164114 DOI: 10.1158/2159-8290.cd-22-1131] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 12/29/2022] [Accepted: 01/31/2023] [Indexed: 02/04/2023]
Abstract
Triple-negative breast cancers (TNBC) frequently inactivate p53, increasing their aggressiveness and therapy resistance. We identified an unexpected protein vulnerability in p53-inactivated TNBC and designed a new PROteolysis TArgeting Chimera (PROTAC) to target it. Our PROTAC selectively targets MDM2 for proteasome-mediated degradation with high-affinity binding and VHL recruitment. MDM2 loss in p53 mutant/deleted TNBC cells in two-dimensional/three-dimensional culture and TNBC patient explants, including relapsed tumors, causes apoptosis while sparing normal cells. Our MDM2-PROTAC is stable in vivo, and treatment of TNBC xenograft-bearing mice demonstrates tumor on-target efficacy with no toxicity to normal cells, significantly extending survival. Transcriptomic analyses revealed upregulation of p53 family target genes. Investigations showed activation and a required role for TAp73 to mediate MDM2-PROTAC-induced apoptosis. Our data, challenging the current MDM2/p53 paradigm, show MDM2 is required for p53-inactivated TNBC cell survival, and PROTAC-targeted MDM2 degradation is an innovative potential therapeutic strategy for TNBC and superior to existing MDM2 inhibitors. SIGNIFICANCE p53-inactivated TNBC is an aggressive, therapy-resistant, and lethal breast cancer subtype. We designed a new compound targeting an unexpected vulnerability we identified in TNBC. Our MDM2-targeted degrader kills p53-inactivated TNBC cells, highlighting the requirement for MDM2 in TNBC cell survival and as a new therapeutic target for this disease. See related commentary by Peuget and Selivanova, p. 1043. This article is highlighted in the In This Issue feature, p. 1027.
Collapse
Affiliation(s)
- Clare M. Adams
- Department of Pharmacology, Physiology and Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Ramkrishna Mitra
- Department of Pharmacology, Physiology and Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | | | - Peter Michener
- Department of Pharmacology, Physiology and Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Juan Palazzo
- Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Allen Chao
- The Wistar Institute, Philadelphia, PA, USA
| | | | | | | | - Christine M. Eischen
- Department of Pharmacology, Physiology and Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
37
|
Zhu Y, Gu L, Lin X, Zhou X, Lu B, Liu C, Li Y, Prochownik EV, Karin M, Wang F, Li Y. P53 deficiency affects cholesterol esterification to exacerbate hepatocarcinogenesis. Hepatology 2023; 77:1499-1511. [PMID: 35398929 PMCID: PMC11186660 DOI: 10.1002/hep.32518] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 04/01/2022] [Accepted: 04/04/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS Cholesterol ester (CE) biosynthesis and homeostasis play critical roles in many cancers, including HCC, but their exact mechanistic contributions to HCC disease development require further study. APPROACH AND RESULTS Here, we report on a proposed role of tumor suppressor P53 in its repressing ubiquitin-specific peptidase 19 (USP19) and sterol O-acyltransferase (SOAT) 1, which maintains CE homeostasis. USP19 enhances cholesterol esterification and contributes to hepatocarcinogenesis (HCG) by deubiquitinating and stabilizing SOAT1. Loss of either SOAT1 or USP19 dramatically attenuates cholesterol esterification and HCG in P53-deficient mice fed with either a normal chow diet or a high-cholesterol, high-fat diet (HCHFD). SOAT1 inhibitor avasimibe has more inhibitory effect on HCC progression in HCHFD-maintained P53-deficient mice when compared to the inhibitors of de novo cholesterol synthesis. Consistent with our findings in the mouse model, the P53-USP19-SOAT1 signaling axis is also dysregulated in human HCCs. CONCLUSIONS Collectively, our findings demonstrate that SOAT1 participates in HCG by increasing cholesterol esterification, thus indicating that SOAT1 is a potential biomarker and therapeutic target in P53-deficient HCC.
Collapse
Affiliation(s)
- Yahui Zhu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
- School of Medicine, Chongqing University, Chongqing, China
| | - Li Gu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| | - Xi Lin
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| | - Xinyi Zhou
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| | - Bingjun Lu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| | - Cheng Liu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| | - Yajun Li
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| | - Edward V. Prochownik
- Division of Hematology/Oncology, Children’s Hospital of Pittsburgh of UPMC, The Department of Microbiology and Molecular Genetics, The Pittsburgh Liver Research Center and The Hillman Cancer Center of UPMC, The University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Michael Karin
- Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Fubing Wang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Youjun Li
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| |
Collapse
|
38
|
Tatavosian R, Donovan MG, Galbraith MD, Duc HN, Szwarc MM, Joshi MU, Frieman A, Bilousova G, Cao Y, Smith KP, Song K, Rachubinski AL, Andrysik Z, Espinosa JM. Cell differentiation modifies the p53 transcriptional program through a combination of gene silencing and constitutive transactivation. Cell Death Differ 2023; 30:952-965. [PMID: 36681780 PMCID: PMC10070495 DOI: 10.1038/s41418-023-01113-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 01/03/2023] [Accepted: 01/06/2023] [Indexed: 01/22/2023] Open
Abstract
The p53 transcription factor is a master regulator of cellular responses to stress that is commonly inactivated in diverse cancer types. Despite decades of research, the mechanisms by which p53 impedes tumorigenesis across vastly different cellular contexts requires further investigation. The bulk of research has been completed using in vitro studies of cancer cell lines or in vivo studies in mouse models, but much less is known about p53 action in diverse non-transformed human tissues. Here, we investigated how different cellular states modify the p53 transcriptional program in human cells through a combination of computational analyses of publicly available large-scale datasets and in vitro studies using an isogenic system consisting of induced pluripotent stem cells (iPSCs) and two derived lineages. Analysis of publicly available mRNA expression and genetic dependency data demonstrated wide variation in terms of expression and function of a core p53 transcriptional program across various tissues and lineages. To monitor the impact of cell differentiation on the p53 transcriptome within an isogenic cell culture system, we activated p53 by pharmacological inhibition of its negative regulator MDM2. Using cell phenotyping assays and genome wide transcriptome analyses, we demonstrated that cell differentiation confines and modifies the p53 transcriptional network in a lineage-specific fashion. Although hundreds of p53 target genes are transactivated in iPSCs, only a small fraction is transactivated in each of the differentiated lineages. Mechanistic studies using small molecule inhibitors and genetic knockdowns revealed the presence of two major regulatory mechanisms contributing to this massive heterogeneity across cellular states: gene silencing by epigenetic regulatory complexes and constitutive transactivation by lineage-specific transcription factors. Altogether, these results illuminate the impact of cell differentiation on the p53 program, thus advancing our understanding of how this tumor suppressor functions in different contexts.
Collapse
Affiliation(s)
- Roubina Tatavosian
- Department of Pharmacology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Linda Crnic Institute for Down Syndrome, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Micah G Donovan
- Department of Pharmacology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Matthew D Galbraith
- Department of Pharmacology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Linda Crnic Institute for Down Syndrome, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Huy N Duc
- Functional Genomics Facility, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Maria M Szwarc
- Department of Pharmacology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Molishree U Joshi
- Functional Genomics Facility, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Amy Frieman
- Charles C. Gates Center for Regenerative Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Ganna Bilousova
- Charles C. Gates Center for Regenerative Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Yingqiong Cao
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Keith P Smith
- Linda Crnic Institute for Down Syndrome, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Kunhua Song
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Angela L Rachubinski
- Linda Crnic Institute for Down Syndrome, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Section of Developmental Pediatrics, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Zdenek Andrysik
- Department of Pharmacology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
| | - Joaquin M Espinosa
- Department of Pharmacology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
- Linda Crnic Institute for Down Syndrome, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
- Functional Genomics Facility, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
| |
Collapse
|
39
|
Szwarc MM, Guarnieri AL, Joshi M, Duc HN, Laird MC, Pandey A, Khanal S, Dohm E, Bui AK, Sullivan KD, Galbraith MD, Andrysik Z, Espinosa JM. FAM193A is a positive regulator of p53 activity. Cell Rep 2023; 42:112230. [PMID: 36897777 PMCID: PMC10164416 DOI: 10.1016/j.celrep.2023.112230] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/21/2022] [Accepted: 02/21/2023] [Indexed: 03/11/2023] Open
Abstract
Inactivation of the p53 tumor suppressor, either by mutations or through hyperactivation of repressors such as MDM2 and MDM4, is a hallmark of cancer. Although many inhibitors of the p53-MDM2/4 interaction have been developed, such as Nutlin, their therapeutic value is limited by highly heterogeneous cellular responses. We report here a multi-omics investigation of the cellular response to MDM2/4 inhibitors, leading to identification of FAM193A as a widespread regulator of p53 function. CRISPR screening identified FAM193A as necessary for the response to Nutlin. FAM193A expression correlates with Nutlin sensitivity across hundreds of cell lines. Furthermore, genetic codependency data highlight FAM193A as a component of the p53 pathway across diverse tumor types. Mechanistically, FAM193A interacts with MDM4, and FAM193A depletion stabilizes MDM4 and inhibits the p53 transcriptional program. Last, FAM193A expression is associated with better prognosis in multiple malignancies. Altogether, these results identify FAM193A as a positive regulator of p53.
Collapse
Affiliation(s)
- Maria M Szwarc
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Anna L Guarnieri
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Molishree Joshi
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Functional Genomics Facility, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Huy N Duc
- Functional Genomics Facility, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Madison C Laird
- Functional Genomics Facility, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Ahwan Pandey
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
| | - Santosh Khanal
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Emily Dohm
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Aimee K Bui
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kelly D Sullivan
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Pediatrics, Section of Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Matthew D Galbraith
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Zdenek Andrysik
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| | - Joaquin M Espinosa
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Functional Genomics Facility, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
40
|
Chen Y, Yang P, Wang J, Gao S, Xiao S, Zhang W, Zhu M, Wang Y, Ke X, Jing H. p53 directly downregulates the expression of CDC20 to exert anti-tumor activity in mantle cell lymphoma. Exp Hematol Oncol 2023; 12:28. [PMID: 36882855 PMCID: PMC9990225 DOI: 10.1186/s40164-023-00381-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 02/07/2023] [Indexed: 03/09/2023] Open
Abstract
BACKGROUND Cell cycle dysregulation characterized by cyclin D1 overexpression is common in mantle cell lymphoma (MCL), while mitotic disorder was less studied. Cell division cycle 20 homologue (CDC20), an essential mitotic regulator, was highly expressed in various tumors. Another common abnormality in MCL is p53 inactivation. Little was known about the role of CDC20 in MCL tumorigenesis and the regulatory relationship between p53 and CDC20 in MCL. METHODS CDC20 expression was detected in MCL patients and MCL cell lines harboring mutant p53 (Jeko and Mino cells) and wild-type p53 (Z138 and JVM2 cells). Z138 and JVM2 cells were treated with CDC20 inhibitor apcin, p53 agonist nutlin-3a, or in combination, and then cell proliferation, cell apoptosis, cell cycle, cell migration and invasion were determined by CCK-8, flow cytometry and Transwell assays. The regulatory mechanism between p53 and CDC20 was revealed by dual-luciferase reporter gene assay and CUT&Tag technology. The anti-tumor effect, safety and tolerability of nutlin-3a and apcin were investigated in vivo in the Z138-driven xenograft tumor model. RESULTS CDC20 was overexpressed in MCL patients and cell lines compared with their respective controls. The typical immunohistochemical marker of MCL patients, cyclin D1, was positively correlated with CDC20 expression. CDC20 high expression indicated unfavorable clinicopathological features and poor prognosis in MCL patients. In Z138 and JVM2 cells, either apcin or nutlin-3a treatment could inhibit cell proliferation, migration and invasion, and induce cell apoptosis and cell cycle arrest. GEO analysis, RT-qPCR and WB results showed that p53 expression was negatively correlated with CDC20 expression in MCL patients, Z138 and JVM2 cells, while this relationship was not observed in p53-mutant cells. Dual-luciferase reporter gene assay and CUT&Tag assay revealed mechanistically that CDC20 was transcriptionally repressed by p53 through directly binding p53 to CDC20 promoter from - 492 to + 101 bp. Moreover, combined treatment of nutlin-3a and apcin showed better anti-tumor effect than single treatment in Z138 and JVM2 cells. Administration of nutlin-3a/apcin alone or in combination confirmed their efficacy and safety in tumor-bearing mice. CONCLUSIONS Our study validates the essential role of p53 and CDC20 in MCL tumorigenesis, and provides a new insight for MCL therapeutics through dual-targeting p53 and CDC20.
Collapse
Affiliation(s)
- Yingtong Chen
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, 49 Huayuan North Road, Haidian District, Beijing, 100191, China.,Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, 49 Huayuan North Road, Haidian District, Beijing, 100191, China
| | - Ping Yang
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, 49 Huayuan North Road, Haidian District, Beijing, 100191, China
| | - Jing Wang
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, 49 Huayuan North Road, Haidian District, Beijing, 100191, China
| | - Shuang Gao
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, 49 Huayuan North Road, Haidian District, Beijing, 100191, China
| | - Shiyu Xiao
- Department of Gastroenterology, Peking University Third Hospital, 49 Huayuan North Road, Haidian District, Beijing, 100191, China.,Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, 49 Huayuan North Road, Haidian District, Beijing, 100191, China
| | - Weilong Zhang
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, 49 Huayuan North Road, Haidian District, Beijing, 100191, China
| | - Mingxia Zhu
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, 49 Huayuan North Road, Haidian District, Beijing, 100191, China
| | - Yanfang Wang
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, 49 Huayuan North Road, Haidian District, Beijing, 100191, China
| | - Xiaoyan Ke
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, 49 Huayuan North Road, Haidian District, Beijing, 100191, China.
| | - Hongmei Jing
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, 49 Huayuan North Road, Haidian District, Beijing, 100191, China.
| |
Collapse
|
41
|
Sampson C, Wang Q, Otkur W, Zhao H, Lu Y, Liu X, Piao H. The roles of E3 ubiquitin ligases in cancer progression and targeted therapy. Clin Transl Med 2023; 13:e1204. [PMID: 36881608 PMCID: PMC9991012 DOI: 10.1002/ctm2.1204] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 02/07/2023] [Accepted: 02/13/2023] [Indexed: 03/08/2023] Open
Abstract
Ubiquitination is one of the most important post-translational modifications which plays a significant role in conserving the homeostasis of cellular proteins. In the ubiquitination process, ubiquitin is conjugated to target protein substrates for degradation, translocation or activation, dysregulation of which is linked to several diseases including various types of cancers. E3 ubiquitin ligases are regarded as the most influential ubiquitin enzyme owing to their ability to select, bind and recruit target substrates for ubiquitination. In particular, E3 ligases are pivotal in the cancer hallmarks pathways where they serve as tumour promoters or suppressors. The specificity of E3 ligases coupled with their implication in cancer hallmarks engendered the development of compounds that specifically target E3 ligases for cancer therapy. In this review, we highlight the role of E3 ligases in cancer hallmarks such as sustained proliferation via cell cycle progression, immune evasion and tumour promoting inflammation, and in the evasion of apoptosis. In addition, we summarise the application and the role of small compounds that target E3 ligases for cancer treatment along with the significance of targeting E3 ligases as potential cancer therapy.
Collapse
Affiliation(s)
- Chibuzo Sampson
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of SciencesDalianChina
- University of Chinese Academy of SciencesBeijingChina
| | - Qiuping Wang
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of SciencesDalianChina
| | - Wuxiyar Otkur
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of SciencesDalianChina
| | - Haifeng Zhao
- Department of OrthopedicsDalian Second People's HospitalDalianChina
| | - Yun Lu
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of SciencesDalianChina
- Department of StomatologyDalian Medical UniversityDalianChina
| | - Xiaolong Liu
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of SciencesDalianChina
| | - Hai‐long Piao
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of SciencesDalianChina
- University of Chinese Academy of SciencesBeijingChina
| |
Collapse
|
42
|
Wang Z, Mačáková M, Bugai A, Kuznetsov SG, Hassinen A, Lenasi T, Potdar S, Friedel CC, Barborič M. P-TEFb promotes cell survival upon p53 activation by suppressing intrinsic apoptosis pathway. Nucleic Acids Res 2023; 51:1687-1706. [PMID: 36727434 PMCID: PMC9976905 DOI: 10.1093/nar/gkad001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/27/2022] [Accepted: 01/03/2023] [Indexed: 02/03/2023] Open
Abstract
Positive transcription elongation factor b (P-TEFb) is the crucial player in RNA polymerase II (Pol II) pause release that has emerged as a promising target in cancer. Because single-agent therapy may fail to deliver durable clinical response, targeting of P-TEFb shall benefit when deployed as a combination therapy. We screened a comprehensive oncology library and identified clinically relevant antimetabolites and Mouse double minute 2 homolog (MDM2) inhibitors as top compounds eliciting p53-dependent death of colorectal cancer cells in synergy with selective inhibitors of P-TEFb. While the targeting of P-TEFb augments apoptosis by anti-metabolite 5-fluorouracil, it switches the fate of cancer cells by the non-genotoxic MDM2 inhibitor Nutlin-3a from cell-cycle arrest to apoptosis. Mechanistically, the fate switching is enabled by the induction of p53-dependent pro-apoptotic genes and repression of P-TEFb-dependent pro-survival genes of the PI3K-AKT signaling cascade, which stimulates caspase 9 and intrinsic apoptosis pathway in BAX/BAK-dependent manner. Finally, combination treatments trigger apoptosis of cancer cell spheroids. Together, co-targeting of P-TEFb and suppressors of intrinsic apoptosis could become a viable strategy to eliminate cancer cells.
Collapse
Affiliation(s)
- Zhijia Wang
- Department of Biochemistry and Developmental Biology, University of Helsinki, Helsinki FIN-00014, Finland
| | - Monika Mačáková
- Department of Biochemistry and Developmental Biology, University of Helsinki, Helsinki FIN-00014, Finland
| | - Andrii Bugai
- Department of Biochemistry and Developmental Biology, University of Helsinki, Helsinki FIN-00014, Finland.,Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus C, Denmark
| | - Sergey G Kuznetsov
- High-Throughput Biomedicine Unit (HTB), Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki FIN-00014, Finland
| | - Antti Hassinen
- High Content Imaging and Analysis Unit (HCA), Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki FIN-00014, Finland
| | - Tina Lenasi
- Department of Biochemistry and Developmental Biology, University of Helsinki, Helsinki FIN-00014, Finland
| | - Swapnil Potdar
- High-Throughput Biomedicine Unit (HTB), Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki FIN-00014, Finland
| | - Caroline C Friedel
- Institute for Informatics, Ludwig-Maximilians-Universität München, 80333 Munich, Germany
| | - Matjaž Barborič
- Department of Biochemistry and Developmental Biology, University of Helsinki, Helsinki FIN-00014, Finland
| |
Collapse
|
43
|
Nakamura S, Kibe M, Takehara T, Suzuki T. Direct Catalytic Enantioselective Reaction of α-Isocyanoacetonitriles with Ketimines Using Cinchona Alkaloid Amide-Cu(II) Catalysts. Org Lett 2023; 25:1040-1044. [PMID: 36749377 DOI: 10.1021/acs.orglett.3c00259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The first enantioselective reaction of α-isocyanoacetonitriles was developed. The reaction of various α-isocyanoacetonitriles with ketimines using cinchona alkaloid amide-Cu(II) catalysts afforded imidazolines with consecutive tetrasubstituted stereogenic carbon centers in good yields and high diastereo- and enantioselectivities. The stereoselectivity of the reaction is explained on the basis of the control experiment and density functional theory (DFT) calculations. The products were subsequently converted into chiral compounds. This process highlights the possible use of α-isocyanoacetonitriles for asymmetric and organic syntheses.
Collapse
Affiliation(s)
- Shuichi Nakamura
- Department of Life Science and Applied Chemistry, Graduate School of Engineering, Nagoya Institute of Technology, Gokiso, Showa-ku, Nagoya 466-8555, Japan
| | - Masato Kibe
- Department of Life Science and Applied Chemistry, Graduate School of Engineering, Nagoya Institute of Technology, Gokiso, Showa-ku, Nagoya 466-8555, Japan
| | - Tsunayoshi Takehara
- The Institute of Scientific and Industrial Research, Osaka University, 8-1 Mihogaoka, Ibaraki-shi, Osaka 567-0047, Japan
| | - Takeyuki Suzuki
- The Institute of Scientific and Industrial Research, Osaka University, 8-1 Mihogaoka, Ibaraki-shi, Osaka 567-0047, Japan
| |
Collapse
|
44
|
Andrysik Z, Sullivan KD, Kieft JS, Espinosa JM. PPM1D suppresses p53-dependent transactivation and cell death by inhibiting the Integrated Stress Response. Nat Commun 2022; 13:7400. [PMID: 36456590 PMCID: PMC9715646 DOI: 10.1038/s41467-022-35089-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 11/17/2022] [Indexed: 12/03/2022] Open
Abstract
The p53 transcription factor is a master regulator of cellular stress responses inhibited by repressors such as MDM2 and the phosphatase PPM1D. Activation of p53 with pharmacological inhibitors of its repressors is being tested in clinical trials for cancer therapy, but efficacy has been limited by poor induction of tumor cell death. We demonstrate that dual inhibition of MDM2 and PPM1D induces apoptosis in multiple cancer cell types via amplification of the p53 transcriptional program through the eIF2α-ATF4 pathway. PPM1D inhibition induces phosphorylation of eIF2α, ATF4 accumulation, and ATF4-dependent enhancement of p53-dependent transactivation upon MDM2 inhibition. Dual inhibition of p53 repressors depletes heme and induces HRI-dependent eIF2α phosphorylation. Pharmacological induction of eIF2α phosphorylation synergizes with MDM2 inhibition to induce cell death and halt tumor growth in mice. These results demonstrate that PPM1D inhibits both the p53 network and the integrated stress response controlled by eIF2α-ATF4, with clear therapeutic implications.
Collapse
Affiliation(s)
- Zdenek Andrysik
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
| | - Kelly D Sullivan
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Department of Pediatrics, Section of Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Jeffrey S Kieft
- Department of Biochemistry and Molecular Genetics and RNA Bioscience Initiative, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Joaquin M Espinosa
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
| |
Collapse
|
45
|
Zhao M, Nie P, Guo Y, Chen H. Pulmonary artery intimal sarcoma: A rare cause of filling defects in pulmonary arteries. Am J Med Sci 2022; 364:655-660. [PMID: 35588894 DOI: 10.1016/j.amjms.2022.05.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 10/07/2021] [Accepted: 05/11/2022] [Indexed: 01/25/2023]
Abstract
Pulmonary artery intimal sarcomas are very rare and arise from primitive pluripotent mesenchymal cells. They are often misdiagnosed as pulmonary thromboembolism, leading to futile anticoagulation treatment and delayed diagnosis. We present a case of a patient who showed nonspecific pulmonary symptoms and characteristic imaging manifestation. Progressive symptoms and additional imaging led to the suspicion of a pulmonary artery intimal sarcoma, which was finally confirmed by pathological biopsy. This case serves as a reminder to consider pulmonary artery intimal sarcomas in the differential diagnosis of patients with dyspnea and filling defects on computed tomography pulmonary angiography or contrast-enhanced computed tomography.
Collapse
Affiliation(s)
- Mimi Zhao
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China, 266003
| | - Pei Nie
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China, 266003
| | - Yonghua Guo
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China, 266003
| | - Haisong Chen
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China, 266003.
| |
Collapse
|
46
|
An HG, Shin S, Lee B, Kwon Y, Kwon TU, Kwon YJ, Chun YJ. Induction of synergistic apoptosis by tetramethoxystilbene and nutlin-3a in human cervical cancer cells. Toxicol Res 2022; 38:591-600. [PMID: 36277372 PMCID: PMC9532473 DOI: 10.1007/s43188-022-00150-4] [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: 07/12/2022] [Revised: 08/03/2022] [Accepted: 08/03/2022] [Indexed: 11/26/2022] Open
Abstract
2,4,3',5'-Tetramethoxystilbene (TMS) is a selective inhibitor of cytochrome P450 1B1 to block the conversion from estradiol to 4-OH-estradiol. Several studies suggested that TMS may act as a potent anti-cancer agent for hormone-related cancer including cervical cancer. Nutlin-3a is a cis-imidazoline analog that interferes with the interaction between mouse double minute 2 homolog (MDM2) and the tumor suppressor p53. The purpose of the study was to compare the cytotoxic effect of TMS and nutlin-3a treatment individually and in combination in HeLa cells. To assess the potential synergistic effects between TMS and nutlin-3a, low concentrations of TMS and nutlin-3a were simultaneously treated in HeLa cells. Based on cell viability, apoptosis assays, and the increase in cleaved caspase-3 and poly (ADP-ribose) polymerase cleavage, it was demonstrated that the combination with TMS and nutlin-3a exerts a synergistic effect on cancer cell death. Isobologram analysis of HeLa cells noted synergism between TMS and nutlin-3a. The combined treatment increased the expression of mitochondrial pro-apoptotic factors such as Bax and Bak, and decreased the expression of the XIAP. In addition, combination treatment significantly enhanced the translocation of AIF to the nucleus in HeLa cells. In conclusion, the results demonstrate that the combination of TMS and nutlin-3a induces synergistic apoptosis in HeLa cells, suggesting the possibility that this combination can be applied as a novel therapeutic strategy for cervical cancer.
Collapse
Affiliation(s)
- Hong-Gyu An
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, 06974 Seoul, Republic of Korea
| | - Sangyun Shin
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, 06974 Seoul, Republic of Korea
| | - Boyoung Lee
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, 06974 Seoul, Republic of Korea
| | - Yeonju Kwon
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, 06974 Seoul, Republic of Korea
| | - Tae-Uk Kwon
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, 06974 Seoul, Republic of Korea
| | - Yeo-Jung Kwon
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, 06974 Seoul, Republic of Korea
| | - Young-Jin Chun
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, 06974 Seoul, Republic of Korea
| |
Collapse
|
47
|
Wang B, Gao J, Zhao Z, Zhong X, Cui H, Hou H, Zhang Y, Zheng J, Di J, Liu Y. Identification of a small-molecule RPL11 mimetic that inhibits tumor growth by targeting MDM2-p53 pathway. Mol Med 2022; 28:109. [PMID: 36071402 PMCID: PMC9450376 DOI: 10.1186/s10020-022-00537-x] [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: 05/12/2022] [Accepted: 08/29/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Targeting ribosome biogenesis to activate p53 has recently emerged as a therapeutic strategy in human cancer. Among various ribosomal proteins, RPL11 centralizes the nucleolar stress-sensing pathway by binding MDM2, leading to MDM2 inactivation and p53 activation. Therefore, the identification of MDM2-binding RPL11-mimetics would be valuable for anti-cancer therapeutics. METHODS Based on the crystal structure of the interface between RPL11 and MDM2, we have identified 15 potential allosteric modulators of MDM2 through the virtual screening. RESULTS One of these compounds, named S9, directly binds MDM2 and competitively inhibits the interaction between RPL11 and MDM2, leading to p53 stabilization and activation. Moreover, S9 inhibits cancer cell proliferation in vitro and in vivo. Mechanistic study reveals that MDM2 is required for S9-induced G2 cell cycle arrest and apoptosis, whereas p53 contributes to S9-induced apoptosis. CONCLUSIONS Putting together, S9 may serve as a lead compound for the development of an anticancer drug that specifically targets RPL11-MDM2-p53 pathway.
Collapse
Affiliation(s)
- Bingwu Wang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of Oncology, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China
| | - Jian Gao
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China.,Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221000, China
| | - Zhongjun Zhao
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Xuefei Zhong
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Hao Cui
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Hui Hou
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Yanping Zhang
- Department of Radiation and Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, 450 West Drive, Chapel Hill, USA
| | - Junnian Zheng
- Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China. .,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China.
| | - Jiehui Di
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China. .,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China. .,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China.
| | - Yong Liu
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China. .,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China. .,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China. .,The State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China.
| |
Collapse
|
48
|
The RING finger protein family in health and disease. Signal Transduct Target Ther 2022; 7:300. [PMID: 36042206 PMCID: PMC9424811 DOI: 10.1038/s41392-022-01152-2] [Citation(s) in RCA: 90] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 07/31/2022] [Accepted: 08/09/2022] [Indexed: 02/05/2023] Open
Abstract
Ubiquitination is a highly conserved and fundamental posttranslational modification (PTM) in all eukaryotes regulating thousands of proteins. The RING (really interesting new gene) finger (RNF) protein, containing the RING domain, exerts E3 ubiquitin ligase that mediates the covalent attachment of ubiquitin (Ub) to target proteins. Multiple reviews have summarized the critical roles of the tripartite-motif (TRIM) protein family, a subgroup of RNF proteins, in various diseases, including cancer, inflammatory, infectious, and neuropsychiatric disorders. Except for TRIMs, since numerous studies over the past decades have delineated that other RNF proteins also exert widespread involvement in several diseases, their importance should not be underestimated. This review summarizes the potential contribution of dysregulated RNF proteins, except for TRIMs, to the pathogenesis of some diseases, including cancer, autoimmune diseases, and neurodegenerative disorder. Since viral infection is broadly involved in the induction and development of those diseases, this manuscript also highlights the regulatory roles of RNF proteins, excluding TRIMs, in the antiviral immune responses. In addition, we further discuss the potential intervention strategies targeting other RNF proteins for the prevention and therapeutics of those human diseases.
Collapse
|
49
|
Lin H, Wang L, Liu Z, Long K, Kong M, Ye D, Chen X, Wang K, Wu KKL, Fan M, Song E, Wang C, Hoo RLC, Hui X, Hallenborg P, Piao H, Xu A, Cheng KKY. Hepatic MDM2 Causes Metabolic Associated Fatty Liver Disease by Blocking Triglyceride-VLDL Secretion via ApoB Degradation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2200742. [PMID: 35524581 PMCID: PMC9284139 DOI: 10.1002/advs.202200742] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/15/2022] [Indexed: 05/06/2023]
Abstract
Dysfunctional triglyceride-very low-density lipoprotein (TG-VLDL) metabolism is linked to metabolic-associated fatty liver disease (MAFLD); however, the underlying cause remains unclear. The study shows that hepatic E3 ubiquitin ligase murine double minute 2 (MDM2) controls MAFLD by blocking TG-VLDL secretion. A remarkable upregulation of MDM2 is observed in the livers of human and mouse models with different levels of severity of MAFLD. Hepatocyte-specific deletion of MDM2 protects against high-fat high-cholesterol diet-induced hepatic steatosis and inflammation, accompanied by a significant elevation in TG-VLDL secretion. As an E3 ubiquitin ligase, MDM2 targets apolipoprotein B (ApoB) for proteasomal degradation through direct protein-protein interaction, which leads to reduced TG-VLDL secretion in hepatocytes. Pharmacological blockage of the MDM2-ApoB interaction alleviates dietary-induced hepatic steatohepatitis and fibrosis by inducing hepatic ApoB expression and subsequent TG-VLDL secretion. The effect of MDM2 on VLDL metabolism is p53-independent. Collectively, these findings suggest that MDM2 acts as a negative regulator of hepatic ApoB levels and TG-VLDL secretion in MAFLD. Inhibition of the MDM2-ApoB interaction may represent a potential therapeutic approach for MAFLD treatment.
Collapse
Affiliation(s)
- Huige Lin
- Department of Health Technology and InformaticsThe Hong Kong Polytechnic UniversityHung HomKowloonHong Kong
| | - Lin Wang
- Department of Health Technology and InformaticsThe Hong Kong Polytechnic UniversityHung HomKowloonHong Kong
- The State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongPokfulamHong Kong
- Department of MedicineThe University of Hong KongPokfulamHong Kong
| | - Zhuohao Liu
- The State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongPokfulamHong Kong
- Department of MedicineThe University of Hong KongPokfulamHong Kong
- Department of NeurosurgeryShenzhen HospitalSouthern Medical UniversityShenzhen518000P. R. China
| | - Kekao Long
- Department of Health Technology and InformaticsThe Hong Kong Polytechnic UniversityHung HomKowloonHong Kong
| | - Mengjie Kong
- Department of Health Technology and InformaticsThe Hong Kong Polytechnic UniversityHung HomKowloonHong Kong
| | - Dewei Ye
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of EducationGuangdong Pharmaceutical UniversityGuangzhou510000P. R. China
| | - Xi Chen
- Department of Health Technology and InformaticsThe Hong Kong Polytechnic UniversityHung HomKowloonHong Kong
| | - Kai Wang
- Department of Health Technology and InformaticsThe Hong Kong Polytechnic UniversityHung HomKowloonHong Kong
| | - Kelvin KL Wu
- Department of Health Technology and InformaticsThe Hong Kong Polytechnic UniversityHung HomKowloonHong Kong
| | - Mengqi Fan
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of EducationGuangdong Pharmaceutical UniversityGuangzhou510000P. R. China
| | - Erfei Song
- Department of Metabolic and Bariatric SurgeryThe First Affiliated Hospital of Jinan UniversityGuangzhou510000P. R. China
| | - Cunchuan Wang
- Department of Metabolic and Bariatric SurgeryThe First Affiliated Hospital of Jinan UniversityGuangzhou510000P. R. China
| | - Ruby LC Hoo
- The State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongPokfulamHong Kong
- Department of Pharmacology and PharmacyThe University of Hong KongPokfulamHong Kong
| | - Xiaoyan Hui
- The State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongPokfulamHong Kong
- Department of MedicineThe University of Hong KongPokfulamHong Kong
| | - Philip Hallenborg
- Department of Biochemistry and Molecular BiologyUniversity of Southern DenmarkSouthern Denmark5230Denmark
| | - Hailong Piao
- Dalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116000P. R. China
| | - Aimin Xu
- The State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongPokfulamHong Kong
- Department of MedicineThe University of Hong KongPokfulamHong Kong
- Department of Pharmacology and PharmacyThe University of Hong KongPokfulamHong Kong
| | - Kenneth KY Cheng
- Department of Health Technology and InformaticsThe Hong Kong Polytechnic UniversityHung HomKowloonHong Kong
| |
Collapse
|
50
|
Li H, Wang Z, Fang X, Zeng W, Yang Y, Jin L, Wei X, Qin Y, Wang C, Liang W. Poroptosis: A form of cell death depending on plasma membrane nanopores formation. iScience 2022; 25:104481. [PMID: 35712073 PMCID: PMC9194171 DOI: 10.1016/j.isci.2022.104481] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 04/19/2022] [Accepted: 05/24/2022] [Indexed: 12/13/2022] Open
Abstract
Immunogenic cell death (ICD) in malignant cells can decrease tumor burden and activate antitumor immune response to obtain lasting antitumor immunity, leading to the elimination of distant metastases and prevention of recurrence. Here, we reveal that ppM1 peptide is capable of forming irreparable transmembrane pores on tumor cell membrane, leading to ICD which we name poroptosis. Poroptosis is directly dependent on cell membrane nanopores regardless of the upstream signaling of cell death. ppM1-induced poroptosis was characterized by the sustained release of intracellular LDH. This unique feature is distinct from other well-characterized types of acute necrosis induced by freezing-thawing (F/T) and detergents, which leads to the burst release of intracellular LDH. Our results suggested that steady transmembrane-nanopore-mediated subacute cell death played a vital role in subsequent activated immunity that transforms to an antitumor immune microenvironment. Selectively generating poroptosis in cancer cell could be a promise strategy for cancer therapy.
Collapse
Affiliation(s)
- Hao Li
- Protein & Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, P. R. China
- University of the Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Zihao Wang
- Protein & Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, P. R. China
- University of the Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Xiaocui Fang
- Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, and Key Laboratory of Standardization and Measurement for Nanotechnology, National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing 100190, P. R. China
- University of the Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Wenfeng Zeng
- Protein & Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, P. R. China
- University of the Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Yanlian Yang
- Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, and Key Laboratory of Standardization and Measurement for Nanotechnology, National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing 100190, P. R. China
- University of the Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Lingtao Jin
- Department of Molecular Medicine, UT Health San Antonio, San Antonio, TX 78229, USA
| | - Xiuli Wei
- Protein & Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, P. R. China
| | - Yan Qin
- Protein & Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, P. R. China
| | - Chen Wang
- Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, and Key Laboratory of Standardization and Measurement for Nanotechnology, National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing 100190, P. R. China
- University of the Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Wei Liang
- Protein & Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, P. R. China
- University of the Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|